[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (23)

Search Parameters:
Keywords = dedifferentiated thyroid cancer

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
8 pages, 1576 KiB  
Article
No Correlation between PD-L1 and NIS Expression in Lymph Node Metastatic Papillary Thyroid Carcinoma
by Lévay Bernadett, Kiss Alexandra, Fröhlich Georgina, Tóth Erika, Slezák András, Péter Ilona, Oberna Ferenc and Dohán Orsolya
Diagnostics 2024, 14(17), 1858; https://doi.org/10.3390/diagnostics14171858 - 26 Aug 2024
Viewed by 1073
Abstract
Approximately 90% of thyroid cancers are differentiated thyroid cancers (DTCs), originating from follicular epithelial cells. Out of these, 90% are papillary thyroid cancer (PTC), and 10% are follicular thyroid cancer (FTC). The standard care procedure for PTC includes surgery, followed by radioiodine (RAI) [...] Read more.
Approximately 90% of thyroid cancers are differentiated thyroid cancers (DTCs), originating from follicular epithelial cells. Out of these, 90% are papillary thyroid cancer (PTC), and 10% are follicular thyroid cancer (FTC). The standard care procedure for PTC includes surgery, followed by radioiodine (RAI) ablation and thyroid-stimulating hormone (TSH) suppressive therapy. Globally, treating radioiodine-refractory DTC poses a challenge. During malignant transformation, thyroid epithelial cells often lose their ability to absorb radioiodine due to impaired membrane targeting or lack of NIS (sodium/iodide symporter) expression. Recent reports show an increase in PD-L1 (programmed death ligand 1) expression in thyroid cancer cells during dedifferentiation. However, no research exists wherein NIS and PD-L1 expression are analyzed together in thyroid cancer. Therefore, we aimed to investigate and correlate PD-L1 and NIS expression within primary tumor samples of lymph node metastatic PTC. We analyzed the expression of hNIS (human sodium/iodide symporter) and PD-L1 in primary tumor samples from metastatic PTC patients using immunohistochemistry. Immunohistochemistry analysis of PD-L1 and NIS was conducted in 89 and 86 PTC cases, respectively. Any subcellular NIS localization was counted as a positive result. PD-L1 expression was absent in 25 tumors, while 58 tumors displayed PD-L1 expression in 1–50% of their cells; in 6 tumors, over 50% of the cells tested positive for PD-L1. NIS immunohistochemistry was performed for 86 primary papillary carcinomas, with 51 out of 86 tumors showcasing NIS expression. Only in seven cases was NIS localized in the plasma membrane; in most tumors, NIS was primarily found in the intracytoplasmic membrane compartments. In the case of PD-L1 staining, cells showing linear membrane positivity of any intensity were counted as positive. The evaluation of NIS immunostaining was simpler: cells showing staining of any intensity of cytoplasmic or membranous fashion were counted as positive. The number of NIS positive cells can be further divided into cytoplasmic and membrane positive compartments. There was no observed correlation between PD-L1 and NIS expression. We can speculate that the manipulation of the PD-1/PD-L1 axis using anti-PD-L1 or anti-PD-1 antibodies could reinstate the functional expression of NIS. However, based on our study, the only conclusion that can be drawn is that there is no correlation between the percentage of NIS- or PD-L1-expressing tumor cells in the primary tumor of lymph node metastatic PTC. Full article
(This article belongs to the Special Issue Advances in the Diagnosis and Management of Thyroid Cancer)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) Papillary thyroid carcinoma showing typical architectural and cytomorphological features (black arrow) (HE: hematoxylin–eosin staining). (<b>B</b>) Papillary thyroid carcinoma cells featuring membranous PD-L1 staining (black arrow) and normal follicular cells showing no staining (blue arrow) (PD-L1 IHC: immunohistochemistry). (<b>C</b>) Normal follicular epithelial cells exhibiting heterogenous NIS staining on their membrane (black arrow) compared with tumor cells exhibiting intracellular NIS. (<b>A</b>) HE staining; (<b>B</b>) PD-L1 IHC; (<b>C</b>) NIS IHC. (40× magnification; all samples were taken from the same tumor sample).</p>
Full article ">Figure 2
<p>PD-L1 expression (TPS) depending on capsular (<b>a</b>), vascular (<b>b</b>), and lymphatic invasion (<b>c</b>).</p>
Full article ">
14 pages, 3276 KiB  
Article
Comparative Uptake Patterns of Radioactive Iodine and [18F]-Fluorodeoxyglucose (FDG) in Metastatic Differentiated Thyroid Cancers
by Devan Diwanji, Emmanuel Carrodeguas, Youngho Seo, Hyunseok Kang, Myat Han Soe, Janet M. Chiang, Li Zhang, Chienying Liu, Spencer C. Behr and Robert R. Flavell
J. Clin. Med. 2024, 13(13), 3963; https://doi.org/10.3390/jcm13133963 - 6 Jul 2024
Viewed by 1281
Abstract
Background: Metastatic differentiated thyroid cancer (DTC) represents a molecularly heterogeneous group of cancers with varying radioactive iodine (RAI) and [18F]-fluorodeoxyglucose (FDG) uptake patterns potentially correlated with the degree of de-differentiation through the so-called “flip-flop” phenomenon. However, it is unknown if RAI [...] Read more.
Background: Metastatic differentiated thyroid cancer (DTC) represents a molecularly heterogeneous group of cancers with varying radioactive iodine (RAI) and [18F]-fluorodeoxyglucose (FDG) uptake patterns potentially correlated with the degree of de-differentiation through the so-called “flip-flop” phenomenon. However, it is unknown if RAI and FDG uptake patterns correlate with molecular status or metastatic site. Materials and Methods: A retrospective analysis of metastatic DTC patients (n = 46) with radioactive 131-iodine whole body scan (WBS) and FDG-PET imaging between 2008 and 2022 was performed. The inclusion criteria included accessible FDG-PET and WBS studies within 1 year of each other. Studies were interpreted by two blinded radiologists for iodine or FDG uptake in extrathyroidal sites including lungs, lymph nodes, and bone. Cases were stratified by BRAF V600E mutation status, histology, and a combination of tumor genotype and histology. The data were analyzed by McNemar’s Chi-square test. Results: Lung metastasis FDG uptake was significantly more common than iodine uptake (WBS: 52%, FDG: 84%, p = 0.04), but no significant differences were found for lymph or bone metastases. Lung metastasis FDG uptake was significantly more prevalent in the papillary pattern sub-cohort (WBS: 37%, FDG: 89%, p = 0.02) than the follicular pattern sub-cohort (WBS: 75%, FDG: 75%, p = 1.00). Similarly, BRAF V600E+ tumors with lung metastases also demonstrated a preponderance of FDG uptake (WBS: 29%, FDG: 93%, p = 0.02) than BRAF V600E− tumors (WBS: 83%, FDG: 83%, p = 1.00) with lung metastases. Papillary histology featured higher FDG uptake in lung metastasis (WBS: 39%, FDG: 89%, p = 0.03) compared with follicular histology (WBS: 69%, FDG: 77%, p = 1.00). Patients with papillary pattern disease, BRAF V600E+ mutation, or papillary histology had reduced agreement between both modalities in uptake at all metastatic sites compared with those with follicular pattern disease, BRAF V600E− mutation, or follicular histology. Low agreement in lymph node uptake was observed in all patients irrespective of molecular status or histology. Conclusions: The pattern of FDG-PET and radioiodine uptake is dependent on molecular status and metastatic site, with those with papillary histology or BRAF V600E+ mutation featuring increased FDG uptake in distant metastasis. Further study with an expanded cohort may identify which patients may benefit from specific imaging modalities to recognize and surveil metastases. Full article
(This article belongs to the Section Oncology)
Show Figures

Figure 1

Figure 1
<p>WBS and FDG uptake stratified by molecular subtype. WBS: radioactive iodine whole body scan; FDG: <sup>18</sup>Fluorodeoxyglucose-PET; PP: papillary pattern; FP: follicular pattern. (<b>A</b>) No significant differences were observed in lymphatic tissue metastatic radiotracer uptake in any cohort. (<b>B</b>) Significantly increased FDG over iodine uptake was observed in the entire cohort in lung tissue and papillary pattern (PP) sub-cohort, but not in the follicular pattern (FP) sub-cohort. (<b>C</b>) No significant differences in radiotracer uptake were observed in bone in any cohort. (<b>D</b>) Table summary with proportions of patients which have metastases at any given site and proportions of patients that had FDG or WBS positivity. Asterisk (*) indicates <span class="html-italic">p</span> &lt; 0.05; ns = not significant.</p>
Full article ">Figure 2
<p>WBS and FDG uptake stratified by <span class="html-italic">BRAF</span> V600E status. WBS: radioactive iodine whole body scan; FDG: <sup>18</sup>Fluorodeoxyglucose-PET. Only patients with <span class="html-italic">BRAF</span> testing are included in the cohort analysis (purple and pink for “WBS Tested” and “FDG Tested”, respectively). (<b>A</b>) No significant differences were observed in lymphatic tissue metastatic radiotracer uptake in any cohort. (<b>B</b>) Significantly increased FDG over iodine uptake was observed in the entire <span class="html-italic">BRAF</span> tested cohort in lung tissue and <span class="html-italic">BRAF+</span> sub-cohort, but not in the <span class="html-italic">BRAF</span>− sub-cohort. (<b>C</b>) No significant differences in radiotracer uptake were observed in bone in any cohort. (<b>D</b>) Table summary with proportions of patients which have metastases at any given site and proportions of patients that had FDG or WBS positivity. Asterisk (*) indicates <span class="html-italic">p</span> &lt; 0.05; ns = not significant.</p>
Full article ">Figure 3
<p>Example WBS and FDG PET scans of DTC lung metastasis. RAI WBS: radioactive iodine whole body scan; FDG: <sup>18</sup>Fluorodeoxyglucose-PET. (<b>A</b>,<b>B</b>) Top row images are exemplary of DTC lung metastasis with FDG &gt; WBS uptake in a patient with <span class="html-italic">BRAF+</span> disease, illustrative of the “flip-flop” phenomenon in a less differentiated tumor. Despite clear focal FDG avidity, iodine radiotracer is at background levels in WBS. (<b>C</b>,<b>D</b>) Middle row images demonstrate a concordant <span class="html-italic">BRAF+</span> case with iodine and FDG radiotracer avidity in the lung. (<b>E</b>,<b>F</b>) Bottom row images demonstrate <span class="html-italic">a BRAF</span>− case showing both low level FDG and iodine avidity in lung metastasis, both clearly above background levels, although more conspicuous by WBS.</p>
Full article ">Figure 4
<p>WBS and FDG uptake stratified by histology. (<b>A</b>) No significant differences were observed in lymphatic tissue metastatic radiotracer uptake in any cohort. (<b>B</b>) Significantly increased FDG over iodine uptake was observed in the entire cohort in lung tissue and papillary sub-cohort, but not in the follicular sub-cohort. (<b>C</b>) No significant differences in radiotracer uptake were observed in bone in any cohort. (<b>D</b>) Table summary with proportions of patients which have metastases at any given site and proportions of patients that had FDG or WBS positivity. Asterisk (*) indicates <span class="html-italic">p</span> &lt; 0.05; ns = not significant.</p>
Full article ">Figure 5
<p>Agreement between imaging modalities by papillary or follicular pattern. Agreement is measured by percent discordance between WBS and FDG in patients who had positive uptake by at least one imaging modality. Discordance is inversely proportional to agreement. Below each bar chart, pie charts express the classification of each case into WBS+/FDG+, WBS+ only, and FDG+ only at each metastatic site. Discordance is the calculated sum of WBS+ only and FDG+ only. (<b>A</b>) In the entire cohort, percent discordant rates for lymph nodes (79%), lung (64%), and bone (31%). (<b>B</b>) In patients with papillary patterned thyroid cancer, lymph node (88%), lung (74%), and bone (75%). (<b>C</b>) In patients with follicular patterned thyroid cancer, lymph node (63%), lung (42%), and bone (17%).</p>
Full article ">Figure 6
<p>Schematic of recommended initial imaging modalities by molecular classification and histology.</p>
Full article ">
16 pages, 2185 KiB  
Review
Molecular Perspectives in Radioactive Iodine Theranostics: Current Redifferentiation Protocols for Mis-Differentiated Thyroid Cancer
by Seza A. Gulec, Cristina Benites and Maria E. Cabanillas
J. Clin. Med. 2024, 13(13), 3645; https://doi.org/10.3390/jcm13133645 - 21 Jun 2024
Viewed by 1743
Abstract
Thyroid cancer molecular oncogenesis involves functional dedifferentiation. The initiating genomic alterations primarily affect the MAPK pathway signal transduction and generate an enhanced ERK output, which in turn results in suppression of the expression of transcription of the molecules of iodine metabolomics. The clinical [...] Read more.
Thyroid cancer molecular oncogenesis involves functional dedifferentiation. The initiating genomic alterations primarily affect the MAPK pathway signal transduction and generate an enhanced ERK output, which in turn results in suppression of the expression of transcription of the molecules of iodine metabolomics. The clinical end result of these molecular alterations is an attenuation in theranostic power of radioactive iodine (RAI). The utilization of RAI in systemic therapy of metastatic disease requires restoration of the functional differentiation. This concept has been accomplished by modulation of MAPK signaling. Objective responses have been demonstrated in metastatic disease settings. RAI-refractoriness in “differentiated thyroid cancers” remains a clinical problem despite optimized RAI administration protocols. Functional mis-differentiation and associated RAI-indifference are the underlying primary obstacles. MAPK pathway modulation offers a potential for reversal of RAI-indifference and combat refractoriness. This review presents the latest clinical experience and protocols for the redifferentiation of radioiodine-refractory mis-differentiated thyroid cancer, providing a comprehensive overview of the current protocols and intervention strategies used by leading institutions. Timing and techniques of imaging, thyrotropin (TSH) stimulation methods, and redifferentiation agents are presented. The efficacy and limitations of various approaches are discussed, providing an overview of the advantages and disadvantages associated with each of the protocols. Full article
(This article belongs to the Special Issue New Strategies in the Treatment of Thyroid Carcinoma)
Show Figures

Figure 1

Figure 1
<p>MAPK-ERK pathway and its ERK-mediated feedback regulation. A constitutive activation of the MAPK pathway results in enhanced ERK output. The intranuclear interactions of the ERK-mediated transcriptional program is rather complicated. There are two major directions for ERK-mediated transcriptional modifications. First is the generic oncogenic program producing the hallmarks of cancer phenotype: Panel (A). The second, the subject of this review, is the disruption of follicular functional differentiation via interruption of expression of genes associated with thyroid functional differentiation: Panel (B), a heat map depicting physiologic expression levels of iodine handling genes. From left to right: normal thyroid tissue, RAS-mutated cancers, non-RAS/non-BRAF cancers, and BRAF-mutated cancers. Darker colors indicate suppressed expression of genes. The lighter (brighter) colors of the iodine metabolic transcriptome under the “normal” column indicate regular expression of respective genes. ERK has feedback control over the MAPK pathway via RAS and RAF. For RAS-mutated cancers, ERK feedback inhibition works via both RAS and RAF. MEK-only inhibition is considered adequate for clinical redifferentiation. ERK feedback inhibition does not work with mutated BRAF. To achieve a clinically effective redifferentiation, MEK and BRAF combined inhibition is often required. Created with Biorender.com (<a href="https://app.biorender.com" target="_blank">https://app.biorender.com</a>, Accessed 1 May 2024).</p>
Full article ">Figure 2
<p>Signal transduction pathways leading to generic phenotypic expressions of oncogenesis (A) and specific iodine transcriptomic expression (B). Panel (B) is a heat map depicting physiologic expression levels of iodine handling genes. From left to right: normal thyroid tissue, RAS-mutated cancers, non-RAS/non-BRAF cancers, and BRAF-mutated cancers. Darker colors indicate suppressed expression of genes. The lighter (brighter) colors of the iodine metabolic transcriptome under the “normal” column indicate regular expression of respective genes. The MAPK-ERK pathway is well established and studied for effective modulation towards redifferentiation. The PI3K-mTOR pathway’s role in functional dedifferentiation and redifferentiation interventions are under investigation. <b>I</b>–<b>III</b>: MAPK-ERK pathway interventions. BRAF inhibitors: dabrafenib, vemurafenib, and encorafenib; MEK inhibitors: trametinib, cobimetinib, binimetinib, and selumetinib. IV–V: PI3K-mTOR pathway interventions. mTOR inhibitor: everolimus; PI3K inhibitor: copanlisib. <b>VI</b>–<b>VII</b>: RTK fusion oncoprotein interventions. RET fusion inhibitors: selpercatinib and pralsetinib; NTRK fusion inhibitors: larotrectinib and entrectinib. <b>VIII</b>–<b>XI</b>: RTK interventions. Multikinase inhibitors: lenvatinib: FGFR, PDGFR, VEGFR, and C-KIT; sorafenib: PDGFR, VEGFR, and C-KIT. Cabozantinib: MET and VEGFR2. The RAS molecule is activated by multiple TKRs and serves as a common inducting molecule for downstream signaling. Further specifics, including FDA approval status, of the therapeutic agents listed in this figure are listed in <a href="#jcm-13-03645-t001" class="html-table">Table 1</a>. Created with Biorender.com (<a href="https://app.biorender.com" target="_blank">https://app.biorender.com</a>, Accessed 1 May 2024).</p>
Full article ">Figure 3
<p>A successful redifferentiation of a BRAF (+) RAI-indifferent metastatic thyroid cancer with combined BRAF and MEK inhibition treatment. The first column of images on the left is the representative views of I-124 PET/CT obtained prior to redifferentiation treatment. The column in the middle demonstrates the views of I-124 PET/CT obtained after the redifferentiation treatment. The last column on the right is the post-treatment whole-body scan with 150 mCi activity (MCRC series).</p>
Full article ">Figure 4
<p>A flow chart for a comprehensive redifferentiation protocol. The pre-redifferentiation work-up includes (1) evaluation of the metastatic disease pattern and volume with functional/anatomic imaging, (2) Tg panel, (3) genomic and molecular profiling, and (4) determination of the clinically appropriate redifferentiation agent(s). Pre-redifferentiation work-up is completed 3–6 weeks prior to committing the redifferentiation drug therapy. The <span class="html-italic">evaluation for RAI-refractory disease</span> is performed by using I-124 PET/CT imaging. Preparation for imaging involves TSH stimulation. This can be performed with the rhTSH or withdrawal protocol. In the latter, the patients receive rhTSH injections on the first two days of the week block. I-124, 1–2 mCi, is administered on the third day. A complete image data set includes 3-day imaging for determination of cumulated activity (lesional and whole-body/bone marrow) and established MIRD voxel dosimetry. Each imaging time point also provides additional technical information (2–4 h imaging for calibration, 24 h imaging for conventional uptake determination, and 48 h is for SUV-based single-time-point predictive dosimetry). The patients then start the <span class="html-italic">redifferentiation drug therapy</span>. An <span class="html-italic">evaluation of redifferentiation</span> is performed in the fifth week block. This, essentially, is similar to I-124 imaging and the dosimetry protocol used prior to initiating the redifferentiation treatment. A decision to proceed with an anticipated RAI therapy is made at the end of this week. The TSH induction protocol is kept consistent for imaging and therapy interventions. With the rhTSH choice, the patients receive rhTSH injections on the first two days of the sixth week block. The <span class="html-italic">RAI therapy</span> is administered on the third day of the protocol week. The administered activity is determined by institutional preferences, typically in the range of 150–300 mCi I-131. The conventional post-treatment RAI scan, whole-body, and SPECT scan are performed on day 7 of the RAI administration. A 48 hour imaging is helpful for validation of pretreatment dosimetric evaluation. The restaging work-up is performed 3–6 m post-RAI-treatment and includes functional and anatomic imaging as well as the Tg panel.</p>
Full article ">
12 pages, 248 KiB  
Review
Advances in the Development of Positron Emission Tomography Tracers for Improved Detection of Differentiated Thyroid Cancer
by Hannelore Iris Coerts, Bart de Keizer and Frederik Anton Verburg
Cancers 2024, 16(7), 1401; https://doi.org/10.3390/cancers16071401 - 2 Apr 2024
Cited by 2 | Viewed by 1885
Abstract
Thyroid cancer poses a significant challenge in clinical management, necessitating precise diagnostic tools and treatment strategies for optimal patient outcomes. This review explores the evolving field of radiotracers in the diagnosis and management of thyroid cancer, focusing on prostate-specific membrane antigen (PSMA)-based radiotracers, [...] Read more.
Thyroid cancer poses a significant challenge in clinical management, necessitating precise diagnostic tools and treatment strategies for optimal patient outcomes. This review explores the evolving field of radiotracers in the diagnosis and management of thyroid cancer, focusing on prostate-specific membrane antigen (PSMA)-based radiotracers, fibroblast activation protein inhibitor (FAPI)-based radiotracers, Arg-Gly-Asp (RGD)-based radiotracers, and 18F-tetrafluoroborate (18F-TFB). PSMA-based radiotracers, initially developed for prostate cancer imaging, have shown promise in detecting thyroid cancer lesions; however, their detection rate is lower than 18F-FDG PET/CT. FAPI-based radiotracers, targeting fibroblast activation protein highly expressed in tumors, offer potential in the detection of lymph nodes and radioiodine-resistant metastases. RGD-based radiotracers, binding to integrin αvβ3 found on tumor cells and angiogenic blood vessels, demonstrate diagnostic accuracy in detecting radioiodine-resistant thyroid cancer metastases. 18F-TFB emerges as a promising PET tracer for imaging of lymph node metastases and recurrent DTC, offering advantages over traditional methods. Overall, these radiotracers show promise in enhancing diagnostic accuracy, patient stratification, and treatment selection in differentiated thyroid cancer, warranting further research and clinical validation. Given the promising staging capabilities of 18F-TFB and the efficacy of FAP-targeting tracers in advanced, potentially dedifferentiated cases, continued investigation in these domains is justified. Full article
(This article belongs to the Special Issue Thyroid Cancer: Diagnosis, Prognosis and Treatment)
20 pages, 4467 KiB  
Article
Modeling RET-Rearranged Non-Small Cell Lung Cancer (NSCLC): Generation of Lung Progenitor Cells (LPCs) from Patient-Derived Induced Pluripotent Stem Cells (iPSCs)
by Paul Marcoux, Jin Wook Hwang, Christophe Desterke, Jusuf Imeri, Annelise Bennaceur-Griscelli and Ali G. Turhan
Cells 2023, 12(24), 2847; https://doi.org/10.3390/cells12242847 - 15 Dec 2023
Cited by 2 | Viewed by 1776
Abstract
REarranged during Transfection (RET) oncogenic rearrangements can occur in 1–2% of lung adenocarcinomas. While RET-driven NSCLC models have been developed using various approaches, no model based on patient-derived induced pluripotent stem cells (iPSCs) has yet been described. Patient-derived iPSCs hold great promise for [...] Read more.
REarranged during Transfection (RET) oncogenic rearrangements can occur in 1–2% of lung adenocarcinomas. While RET-driven NSCLC models have been developed using various approaches, no model based on patient-derived induced pluripotent stem cells (iPSCs) has yet been described. Patient-derived iPSCs hold great promise for disease modeling and drug screening. However, generating iPSCs with specific oncogenic drivers, like RET rearrangements, presents challenges due to reprogramming efficiency and genotypic variability within tumors. To address this issue, we aimed to generate lung progenitor cells (LPCs) from patient-derived iPSCs carrying the mutation RETC634Y, commonly associated with medullary thyroid carcinoma. Additionally, we established a RETC634Y knock-in iPSC model to validate the effect of this oncogenic mutation during LPC differentiation. We successfully generated LPCs from RETC634Y iPSCs using a 16-day protocol and detected an overexpression of cancer-associated markers as compared to control iPSCs. Transcriptomic analysis revealed a distinct signature of NSCLC tumor repression, suggesting a lung multilineage lung dedifferentiation, along with an upregulated signature associated with RETC634Y mutation, potentially linked to poor NSCLC prognosis. These findings were validated using the RETC634Y knock-in iPSC model, highlighting key cancerous targets such as PROM2 and C1QTNF6, known to be associated with poor prognostic outcomes. Furthermore, the LPCs derived from RETC634Y iPSCs exhibited a positive response to the RET inhibitor pralsetinib, evidenced by the downregulation of the cancer markers. This study provides a novel patient-derived off-the-shelf iPSC model of RET-driven NSCLC, paving the way for exploring the molecular mechanisms involved in RET-driven NSCLC to study disease progression and to uncover potential therapeutic targets. Full article
(This article belongs to the Section Stem Cells)
Show Figures

Figure 1

Figure 1
<p>Generation of lung progenitor cells (LPCs) from iRET<sup>C634Y</sup> is associated with the overexpression of FOXA2 and NKX2-1. (<b>A</b>) Schematic representation of the differentiation protocol from iPSC to NKX2-1<sup>+</sup> lung progenitor cells (LPCs). (<b>B</b>) Morphology of <span class="html-italic">RET<sup>C634Y</sup></span> mutated iPSC (iRET<sup>C634Y</sup>) and its isogenic CRISPR control (iRET<sup>CTRL</sup>) during LPC differentiation at definitive endoderm (DE), anterior foregut endoderm (AFE), and LPC stages. Magnification 10×; scale bar 100 μm. (<b>C</b>) Immunostaining of LPCs derived from iRET<sup>C634Y</sup> and iRET<sup>CTRL</sup> iPSCs showing the expression of TP63 (green), DAPI (blue) or merged. (<b>D</b>–<b>F</b>) Expression of the differentiation markers specific to each stage; (<b>D</b>) DE, (<b>E</b>) AFE, and (<b>F</b>) LPC; quantified by qRT-PCR. Fold change (2<sup>−ΔΔCt</sup>) was normalized to iPSC stage. Differentiation experiments were performed three times for each condition. <span class="html-italic">p</span>-values were calculated at each stage using a two-tailed Student’s <span class="html-italic">t</span>-test. ns, not significant; * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001; **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 2
<p><span class="html-italic">RET<sup>C634Y</sup></span>-dependent gene signature during iPSC-derived LPC differentiation predicts a major transcriptional repression in NSCLC associated with a lung multilineage dedifferentiation. (<b>A</b>) Method for analyzing the <span class="html-italic">RET<sup>C634Y</sup></span>-dependent signature during iPSC-derived LPC differentiation. (<b>B</b>) Unsupervised principal component analysis based on <span class="html-italic">RET<sup>C634Y</sup></span>-dependent gene signature can stratify tumoral and normal lung adjacent samples from GSE44077 transcriptome dataset. (<b>C</b>) Volcano plot of differential expressed gene analysis between tumor and lung adjacent tissue of GSE44077 restricted to <span class="html-italic">RET<sup>C634Y</sup></span>-dependent gene signature (filter fixed over 0.5 log2 of fold change). (<b>D</b>) Barplot of functional enrichment performed with <span class="html-italic">RET<sup>C634Y</sup></span>-dependent repressed signature on Gene Ontology Cellular Component (GO-CC) database. (<b>E</b>) Functional enrichment network highlighting the implication of connected components like focal adhesion, anchoring junction, and synapse in <span class="html-italic">RET<sup>C634Y</sup></span>-dependent repressed signature in NSCLC tumors. (<b>F</b>) Barplot of functional enrichment performed on Co-expression Lung Atlas database with <span class="html-italic">RET<sup>C634Y</sup></span>-dependent repressed NSCLC signature. (<b>G</b>) Functional enrichment network identifying a lung multilineage implication of <span class="html-italic">RET<sup>C634Y</sup></span>-dependent repressed signature in NSCLC.</p>
Full article ">Figure 3
<p>Adverse lung cancer prognosis for patients overexpressing <span class="html-italic">RET<sup>C634Y</sup></span>-dependent activated signature. (<b>A</b>) Barplot of functional enrichment performed with <span class="html-italic">RET<sup>C634Y</sup></span>-dependent activated signature on DisGeNET disease database. (<b>B</b>) Lung cancer related networks of genes found upregulated in NSCLC with <span class="html-italic">RET<sup>C634Y</sup></span>-dependent activated model integration. (<b>C</b>) Oncoprint of the RET 10 genes signature in the transcriptome of the TCGA lung adenocarcinoma cohort (510 patients/510 samples). (<b>D</b>) Kaplan–Meier curve and log-rank test analysis assessing the overall survival (OS) of lung adenocarcinoma patients, comparing those with (red) and without (blue) the overexpression of RET 10 genes signature. (<b>E</b>) Barplot of univariate overall survival analysis for the individual genes of RET 10 genes signature. The proportion of patient deaths among those exhibiting gene overexpression (purple) and the corresponding median overall survival (blue) are displayed. (<b>F</b>) Expression of two cancer markers associated with adverse prognosis quantified by qRT-PCR. Fold changes (2<sup>−ΔΔCt</sup>) have been normalized to iPSC stage. Experiments were performed three times. <span class="html-italic">p</span>-values were calculated using a two-tailed Student’s <span class="html-italic">t</span>-test. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 4
<p>LPCs generated from <span class="html-italic">RET<sup>C634Y</sup></span> knock-in iPSCs are also associated with an overexpression of FOXA2 and NKX2-1. (<b>A</b>) qRT-PCR quantification of RET mRNA in LPCs derived from PB68-WT and PB68-RET<sup>C634Y</sup> iPSCs. (<b>B</b>) Immunostaining of LPCs derived from PB68-WT and PB68-RET<sup>C634Y</sup> iPSCs showing the expression of pRET (green) and DAPI (blue). (<b>C</b>) Expression of the differentiation markers specific to each stage quantified by qRT-PCR. Fold changes (2<sup>−ΔΔCt</sup>) have been normalized to iPSC stage. Differentiation experiments were performed three times for each condition. <span class="html-italic">p</span>-values were calculated at each stage using a two-tailed Student’s <span class="html-italic">t</span>-test. ns, not significant; ns: non-significant, * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001; **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 5
<p><span class="html-italic">RET<sup>C634Y</sup></span> knock-in (RET-KI) induced a metastatic and fibroblastic lung adenocarcinoma expression signature in iPSC-derived LPCs. (<b>A</b>) Method for analyzing the RET-KI dependent signature during iPSC-derived LPC differentiation. (<b>B</b>) Functional enrichment on a single-cell atlas of metastatic lung adenocarcinoma with the genes upregulated specifically during RET-KI LPC differentiation. (<b>C</b>) Fibroblastic functional enriched network drawn during RET-KI LPC differentiation. (<b>D</b>) Oncoprint of RET-KI markers found overexpressed in the transcriptome of the TCGA lung adenocarcinoma cohort (510 patients/510 samples). (<b>E</b>) Kaplan–Meier curve and log-rank test analysis assessing the overall survival (OS) of lung adenocarcinoma patients, comparing those with (red) and without (blue) the overexpression of RET-KI markers. (<b>F</b>) Expression of C1QTNF6 and PROM2, two cancers markers associated with adverse prognosis, quantified by qRT-PCR at LPC stage. Fold changes (2<sup>−ΔΔCt</sup>) were normalized to iPSC stage. Experiments were performed three times. <span class="html-italic">p</span>-values were calculated using a two-tailed Student’s <span class="html-italic">t</span>-test. **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 6
<p>RET inhibitor pralsetinib treatment has a specific inhibitory effect on the genes upregulated by <span class="html-italic">RET<sup>C634Y</sup></span>mutation. (<b>A</b>,<b>B</b>) Expression of <span class="html-italic">FOXA2</span>, <span class="html-italic">NKX2-1</span>, <span class="html-italic">C1QTNF6</span>, <span class="html-italic">PROM2</span> quantified by qRT-PCR in iRET model (<b>A</b>) and PB68 model (<b>B</b>) with and without daily 10 nM pralsetinib treatment. Fold changes (2<sup>−ΔΔCt</sup>) have been normalized to iPSC stage. Experiments were performed three times. Two-ways ANOVA was performed to test the effect of cell lines and pralsetinib treatment. For each combination of cell lines and genes, a Sidak’s multiple comparisons test was performed to test the effect of pralsetinib treatment as compared to WT. ns: non-significant, * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001; **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">
22 pages, 8652 KiB  
Article
Unraveling the Roles of miR-204-5p and HMGA2 in Papillary Thyroid Cancer Tumorigenesis
by Cindy Van Branteghem, Alice Augenlicht, Pieter Demetter, Ligia Craciun and Carine Maenhaut
Int. J. Mol. Sci. 2023, 24(13), 10764; https://doi.org/10.3390/ijms241310764 - 28 Jun 2023
Cited by 4 | Viewed by 2659
Abstract
Thyroid cancer is the most common endocrine malignant tumor with an increasing incidence rate. Although differentiated types of thyroid cancer generally present good clinical outcomes, some dedifferentiate into aggressive and lethal forms. However, the molecular mechanisms governing aggressiveness and dedifferentiation are still poorly [...] Read more.
Thyroid cancer is the most common endocrine malignant tumor with an increasing incidence rate. Although differentiated types of thyroid cancer generally present good clinical outcomes, some dedifferentiate into aggressive and lethal forms. However, the molecular mechanisms governing aggressiveness and dedifferentiation are still poorly understood. Aberrant expression of miRNAs is often correlated to tumor development, and miR-204-5p has previously been identified in papillary thyroid carcinoma as downregulated and associated with aggressiveness. This study aimed to explore its role in thyroid tumorigenesis. To address this, gain-of-function experiments were performed by transiently transfecting miR-204-5p in thyroid cancer cell lines. Then, the clinical relevance of our data was evaluated in vivo. We prove that this miRNA inhibits cell invasion by regulating several targets associated with an epithelial-mesenchymal transition, such as SNAI2, TGFBR2, SOX4 and HMGA2. HMGA2 expression is regulated by the MAPK pathway but not by the PI3K, IGF1R or TGFβ pathways, and the inhibition of cell invasion by miR-204-5p involves direct binding and repression of HMGA2. Finally, we confirmed in vivo the relationship between miR-204-5p and HMGA2 in human PTC and a corresponding mouse model. Our data suggest that HMGA2 inhibition offers promising perspectives for thyroid cancer treatment. Full article
(This article belongs to the Special Issue Recent Advances in Thyroid Cancer, 2nd Edition)
Show Figures

Figure 1

Figure 1
<p>miR-204-5p is downregulated in human thyroid cancer and thyroid cancer-derived cell lines. (<b>a</b>–<b>c</b>) Analysis of miR-204-5p expression levels using thyroid miR-seq data from The Cancer Genome Atlas (TCGA), (<b>a</b>) in normal samples (n = 59), thyroid cancer tissues (n = 502) and metastatic samples (n = 8). *** <span class="html-italic">p</span> &lt; 0.001 vs. normal; (<b>b</b>) in PTC tumors harboring the BRAF<sup>V600E</sup> mutation (BRAF Positive, n = 212) and those carrying other types of mutations (other, n = 248), *** <span class="html-italic">p</span> &lt; 0.001; (<b>c</b>) in PTC tumors according to tumor stage, * <span class="html-italic">p</span> &lt; 0.05. (<b>d</b>) Analysis of miR-204-5p expression levels by RT-qPCR in PTC and adjacent normal thyroid tissues (NT) (n = 7). * <span class="html-italic">p</span> &lt; 0.05. (<b>e</b>) and in PTC-derived thyroid cancer cell lines including TPC-1, K1 and BCPAP (n = 3 for each cell line) compared to a pool of 8 normal human thyroid tissues (NP). * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001 vs. NP.</p>
Full article ">Figure 2
<p>miR-204-5p overexpression inhibits invasion in PTC cell lines: (<b>a</b>) Cell migration and invasion were analyzed in TPC-1 (n = 6) and BCPAP (n = 3) cells three days after transfection, in non-transfected cells (NT), in miR-204-5p overexpressing (miR-204-5p) and mimicked negative control (miR-NC) transfected cells by counting five random fields. (<b>a</b>) Representative image after the invasion assay is shown in the right panel). * <span class="html-italic">p</span> &lt; 0.05 vs. miR-NC. (<b>b</b>) EdU incorporation was analyzed in TPC-1 and BCPAP cells by flow cytometry three days after transfection in non-transfected cells (NT), and cells transfected with miR-204-5p mimic (miR-204-5p) or mimic negative control (miR-NC) cells (n = 3). (<b>c</b>) Cell apoptosis was analyzed by western blotting with cleaved CASPASE-3 and cleaved PARP antibodies and quantified by ImageJ in TPC-1 and BCPAP cells three days after transfection, in cells transfected with miR-204-5p mimic (miR-204-5p) or mimic negative control (miR-NC). TPC-1 and BCPAP cells treated with staurosporine for 20 h were used as a positive control (C+). Cleaved CASPASE-3 expression was normalized with β-ACTIN expression; for PARP, the proportion of cleaved PARP relative to total PARP (cleaved and non-cleaved) was determined.</p>
Full article ">Figure 3
<p>Gene expression analysis reveals a lot of EMT genes whose expression is dysregulated following miR-204-5p expression. (<b>a</b>) Heatmap of the microarray results showing the genes differentially expressed between TPC-1 cells overexpressing miR-204-5p or mimic negative control (miR-NC) and which were validated by luciferase reporter assay as being miR-204-5p targets. Fold change for each gene was defined using gene expression ratios between miR-204-5p and miR-NC transfected cells. The colors of each probe represent the amplitude of the fold changes, as shown in the legend. (<b>b</b>) Venn diagram of predicted and validated targets of miR-204-5p from three online databases. Genes common between the intersection of the three databases and the microarray results and known to be involved in cell adhesion or invasion are shown in red.</p>
Full article ">Figure 4
<p>miR-204-5p inhibits SOX4, TGFBR2, SNAI2 and HMGA2 expression in TPC-1 and BCPAP cells. (<b>a</b>,<b>b</b>) TPC-1 and BCPAP cells were transfected with miR-204-5p or mimic negative control (miR-NC), and SOX4, TGFBR2, SNAI2 and HMGA2 mRNA and protein expressions were analyzed by RT-qPCR and western blotting, respectively (n ≥ 3). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001 (<b>c</b>) HMGA2 protein expression was also assessed by immunostaining with DAPI (blue) and HMGA2 antibody (green) 3 days after transfection. Nuclei were stained with DAPI (blue) (n = 3). The right panels represent the quantification of protein expression. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 5
<p>HMGA2 expression is regulated by the MAPK signalling pathway (<b>a</b>) Western blot analysis of total AKT and ERK and of the phosphorylated forms 24 h after treatment with trametinib (MEKi), GDC0941 (PI3Ki) and dabrafenib (BRAFi) in TPC-1 and BCPAP cells. Right panel: Quantification of the phosphorylated forms according to the different treatments. * <span class="html-italic">p</span> &lt; 0.05 ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001 vs. DMSO. (<b>b</b>) HMGA2 mRNA expression was analyzed by RT-qPCR in TPC-1 and BCPAP cells treated with different inhibitors during 24 h: trametinib (MEKi), GDC0941 (PI3Ki), dabrafenib (BRAFi), trametinib and dabrafenib (MEKi + BRAFi), BMS-754807 (IGF1Ri) (NT: non treated cells). DMSO treatment was used as a negative control. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001 vs. DMSO.</p>
Full article ">Figure 6
<p>HMGA2 silencing and MAPK signalling inhibition decrease the invasion capabilities of TPC-1 and BCPAP cells. (<b>a</b>) The predicted binding site of miR-204-5p to HMGA2 3′UTR (HMGA2-WT) and corresponding mutated sequence (HMGA2-MUT). Grey content corresponds to mutated nucleotides. (<b>b</b>) Relative percentage of luciferase activity following co-transfection of TPC-1 cells with HMGA2-WT or HMGA2-MUT vector and miR-204 mimic (HMGA2-WT-204) or mimic negative control (HMGA2-WT-NC). (<b>c</b>) HMGA2 mRNA expression levels were evaluated by RT-qPCR in a pool of 8 normal human thyroid tissues (NP) and three thyroid cancer cell lines: TPC-1, BCPAP and K1 (n = 3). * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 vs. NP. (<b>d</b>) Migration and invasion analysis in TPC-1 and BCPAP cells three days after transfection with a siRNA against HMGA2 (siRNA HMGA2) and a control siRNA (siRNA NC), and in non-transfected TPC-1 and BCPAP cells treated with trametinib (MEKi) for 24 h. The percentage of invasion was defined by the mean of invasion cell count divided by the mean of migration cell count x 100 (n = 3). * <span class="html-italic">p</span> &lt; 0.05. (<b>e</b>,<b>f</b>) HMGA2 mRNA and protein expression analysis by RT-qPCR and immunocytofluorescence, respectively, in TPC-1 (n = 4) and BCPAP (n = 3) cells, three days after transfection with HMGA2 antibody (green) (NT = non-transfected cells). * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 vs. siRNA NC.</p>
Full article ">Figure 7
<p>Inhibiting DNA methylation increases miR-204-5p and TRPM3 mRNA expression. (<b>a</b>) Correlation analysis between TRPM3 mRNA and miR-204-5p expression levels in the thyroid cancer samples from The Cancer Genome Atlas (TCGA). R = Pearson correlation. (<b>b</b>,<b>c</b>) Expression levels of miR-204-5p (<b>b</b>) and TRPM3 (<b>c</b>) were analyzed in TPC-1 and BCPAP cells by RT-qPCR after treatment with DMSO, 5′-aza-2-deoxycytidine (5′AZA), trametinib (MEKi), GDC0941 (PI3Ki) or dabrafenib (BRAFi) for 24 h and compared to non-treated cells (NT) (n = 5). * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 vs. DMSO. (<b>d</b>) RT-qPCR analysis of mRNA expression of miR-204-5p targets (HMGA2, SOX4, SNAI2) in non-treated (NT) TPC-1 (n = 5) and BCPAP cells (n = 3) and after treatment with 5′-aza-2-deoxycytidine (5′AZA) or DMSO for 24 h. ZEB1 mRNA expression was not a miR-204-5p target and was used as a negative control. * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 vs. DMSO.</p>
Full article ">Figure 8
<p>HMGA2 is overexpressed in human papillary thyroid cancer. (<b>a</b>–<b>d</b>): Analysis of HMGA2 mRNA expression using thyroid RNAseq data from The Cancer Genome Atlas (TCGA) (<b>a</b>) in normal tissues (n = 57), primary tumors (n = 502) and metastatic samples (n = 8), *** <span class="html-italic">p</span> &lt; 0.001 vs. normal tissue; (<b>b</b>) in primary tumors harboring the BRAF<sup>V600E</sup> mutation (BRAF Positive, n = 216) and those with other mutations (other, n = 211), *** <span class="html-italic">p</span> &lt; 0.001 and (<b>c</b>) according to tumor stage. * <span class="html-italic">p</span> &lt; 0.05. (<b>d</b>) Correlation analysis between HMGA2 mRNA and miR-204-5p expression in the thyroid cancer samples from The Cancer Genome Atlas (TCGA). R = Pearson correlation. (<b>e</b>) HMGA2 mRNA expression analyzed by RT-qPCR in 7 independent PTCs and normal adjacent tissues (NT). * <span class="html-italic">p</span> &lt; 0.05 vs. NT. (<b>f</b>) Immunostaining of HMGA2 (green) in PTC and normal thyroid (n = 3). Epithelial cells were stained with cytokeratin 8 antibody (KRT8, red) and nuclei with DAPI (blue). The % of HMGA2 expressing epithelial cells was evaluated by the ratio: several cells positive for both HMGA2 and KRT8/number of KRT8 positive cells ×100.</p>
Full article ">Figure 9
<p>miR-204-5p is downregulated while HMGA2 is overexpressed in the thyroid of RET/PTC3 mice. (<b>a</b>) RT-qPCR analysis of miR-204-5p and HMGA2 mRNA expression in 2- and 6-month-old RET/PTC3 (RET) mice thyroids (n = 11 for two months and n = 9 for six months) and a pool of 3 normal thyroids (WT) (n = 6). ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001 vs. WT. (<b>b</b>) Correlation analysis between HMGA2 mRNA and miR-204-5p expression in 2- and 6-months old RET/PTC3 thyroids. R = Pearson correlation. (<b>c</b>) Western blot analysis and quantification of HMGA2 protein expression in 2-month-old wild type (WT) and RET/PTC3 (RET) thyroids (n = 5 for WT and n = 9 for RET). *** <span class="html-italic">p</span> &lt; 0.001 vs. wild-type thyroids. (<b>d</b>) Analysis of HMGA2 expression by immunostaining with HMGA2 antibody (green) and cytokeratin eight antibody (KRT8, red) and DAPI (blue, nuclei staining) in 2- and 6-month-old thyroids (n = 7). The % of HMGA2 expressing cells among the epithelial cells was defined by the ratio: several cells positive for both HMGA2 and KRT8/number of KRT8 positive cells ×100.</p>
Full article ">Figure 9 Cont.
<p>miR-204-5p is downregulated while HMGA2 is overexpressed in the thyroid of RET/PTC3 mice. (<b>a</b>) RT-qPCR analysis of miR-204-5p and HMGA2 mRNA expression in 2- and 6-month-old RET/PTC3 (RET) mice thyroids (n = 11 for two months and n = 9 for six months) and a pool of 3 normal thyroids (WT) (n = 6). ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001 vs. WT. (<b>b</b>) Correlation analysis between HMGA2 mRNA and miR-204-5p expression in 2- and 6-months old RET/PTC3 thyroids. R = Pearson correlation. (<b>c</b>) Western blot analysis and quantification of HMGA2 protein expression in 2-month-old wild type (WT) and RET/PTC3 (RET) thyroids (n = 5 for WT and n = 9 for RET). *** <span class="html-italic">p</span> &lt; 0.001 vs. wild-type thyroids. (<b>d</b>) Analysis of HMGA2 expression by immunostaining with HMGA2 antibody (green) and cytokeratin eight antibody (KRT8, red) and DAPI (blue, nuclei staining) in 2- and 6-month-old thyroids (n = 7). The % of HMGA2 expressing cells among the epithelial cells was defined by the ratio: several cells positive for both HMGA2 and KRT8/number of KRT8 positive cells ×100.</p>
Full article ">
14 pages, 3549 KiB  
Review
Role of Truncated O-GalNAc Glycans in Cancer Progression and Metastasis in Endocrine Cancers
by Diluka Pinto and Rajeev Parameswaran
Cancers 2023, 15(13), 3266; https://doi.org/10.3390/cancers15133266 - 21 Jun 2023
Cited by 8 | Viewed by 2805
Abstract
Glycans are an essential part of cells, playing a fundamental role in many pathophysiological processes such as cell differentiation, adhesion, motility, signal transduction, host–pathogen interactions, tumour cell invasion, and metastasis development. These glycans are also able to exert control over the changes in [...] Read more.
Glycans are an essential part of cells, playing a fundamental role in many pathophysiological processes such as cell differentiation, adhesion, motility, signal transduction, host–pathogen interactions, tumour cell invasion, and metastasis development. These glycans are also able to exert control over the changes in tumour immunogenicity, interfering with tumour-editing events and leading to immune-resistant cancer cells. The incomplete synthesis of O-glycans or the formation of truncated glycans such as the Tn-antigen (Thomsen nouveau; GalNAcα- Ser/Thr), its sialylated version the STn-antigen (sialyl-Tn; Neu5Acα2–6GalNAcα-Ser/Thr) and the elongated T-antigen (Thomsen–Friedenreich; Galβ1-3GalNAcα-Ser/Thr) has been shown to be associated with tumour progression and metastatic state in many human cancers. Prognosis in various human cancers is significantly poor when they dedifferentiate or metastasise. Recent studies in glycobiology have shown truncated O-glycans to be a hallmark of cancer cells, and when expressed, increase the oncogenicity by promoting dedifferentiation, risk of metastasis by impaired adhesion (mediated by selectins and integrins), and resistance to immunological killing by NK cells. Insight into these truncated glycans provides a complimentary and attractive route for cancer antigen discovery. The recent emergence of immunotherapies against cancers is predicted to harness the potential of using such agents against cancer-associated truncated glycans. In this review, we explore the role of truncated O-glycans in cancer progression and metastasis along with some recent studies on the role of O-glycans in endocrine cancers affecting the thyroid and adrenal gland. Full article
(This article belongs to the Special Issue Glycosylation in Cancer—Biomarkers and Targeted Therapies)
Show Figures

Figure 1

Figure 1
<p>Synthesis of truncated <span class="html-italic">O</span>-GalNAc glycans (Tn, T, sialyl T and sialyl Tn antigens). The synthesis begins with attachment of GalNAc to serine or threonine to form Tn, which can be sialylated to form STn or extended to form core 1 glycan. The core 1 glycans then undergo sialylation or are converted to core glycan or form Lewis antigens. Abbreviations: ppGalNAcTs—polypeptide GalNAc- transferases; core 2 GlcNAc T—β1,6 N-Acetylglucosaminyltransferase; Ser/Thr—serine/threonine; UDP—uridine diphosphate; sLe<sup>x</sup>—sialyl Lewis antigen x; sLe<sup>a</sup>—sialyl Lewis antigen a. The truncated core <span class="html-italic">O</span>-glycans commonly seen in cancers are boxed in red.</p>
Full article ">Figure 2
<p>Role of various truncated <span class="html-italic">O</span>-glycans in cancer progression and metastasis. Cancer cells lose their adhesive mechanisms between cells, between cells and matrix to break free, and migrate through the extracellular membrane. The cancer cells enter the vascular channels and evade the immune system attack to survive and migrate through the systemic circulation. Using lectin glycan interactions mediated by selectins and sialyl Lewis antigens, the cancer cells extravasate through the endothelium and basement membrane and proliferate at a distant site.</p>
Full article ">Figure 3
<p>Kaplan–Meier survival curves in patients with adrenocortical carcinoma with positive and negative Helix pomatia agglutinin (HPA) immunolabelling. <span class="html-italic">p</span> = 0.002 (log rank test). Used with permission from Parameswaran et al. [<a href="#B36-cancers-15-03266" class="html-bibr">36</a>].</p>
Full article ">Figure 4
<p>HPA labelling showing intense staining in cytoplasm and cell surface of the cancer cells in adrenocortical carcinoma. Panel (<b>a</b>) high-power 20× magnification. Panel (<b>b</b>) 40× magnification. Used with permission from Parameswaran et al. [<a href="#B36-cancers-15-03266" class="html-bibr">36</a>].</p>
Full article ">
28 pages, 2012 KiB  
Review
Multi-Omics and Management of Follicular Carcinoma of the Thyroid
by Thifhelimbilu Emmanuel Luvhengo, Ifongo Bombil, Arian Mokhtari, Maeyane Stephens Moeng, Demetra Demetriou, Claire Sanders and Zodwa Dlamini
Biomedicines 2023, 11(4), 1217; https://doi.org/10.3390/biomedicines11041217 - 19 Apr 2023
Cited by 6 | Viewed by 5925
Abstract
Follicular thyroid carcinoma (FTC) is the second most common cancer of the thyroid gland, accounting for up to 20% of all primary malignant tumors in iodine-replete areas. The diagnostic work-up, staging, risk stratification, management, and follow-up strategies in patients who have FTC are [...] Read more.
Follicular thyroid carcinoma (FTC) is the second most common cancer of the thyroid gland, accounting for up to 20% of all primary malignant tumors in iodine-replete areas. The diagnostic work-up, staging, risk stratification, management, and follow-up strategies in patients who have FTC are modeled after those of papillary thyroid carcinoma (PTC), even though FTC is more aggressive. FTC has a greater propensity for haematogenous metastasis than PTC. Furthermore, FTC is a phenotypically and genotypically heterogeneous disease. The diagnosis and identification of markers of an aggressive FTC depend on the expertise and thoroughness of pathologists during histopathological analysis. An untreated or metastatic FTC is likely to de-differentiate and become poorly differentiated or undifferentiated and resistant to standard treatment. While thyroid lobectomy is adequate for the treatment of selected patients who have low-risk FTC, it is not advisable for patients whose tumor is larger than 4 cm in diameter or has extensive extra-thyroidal extension. Lobectomy is also not adequate for tumors that have aggressive mutations. Although the prognosis for over 80% of PTC and FTC is good, nearly 20% of the tumors behave aggressively. The introduction of radiomics, pathomics, genomics, transcriptomics, metabolomics, and liquid biopsy have led to improvements in the understanding of tumorigenesis, progression, treatment response, and prognostication of thyroid cancer. The article reviews the challenges that are encountered during the diagnostic work-up, staging, risk stratification, management, and follow-up of patients who have FTC. How the application of multi-omics can strengthen decision-making during the management of follicular carcinoma is also discussed. Full article
(This article belongs to the Special Issue Thyroid Cancer: From Biology to Therapeutic Opportunities)
Show Figures

Figure 1

Figure 1
<p>Classification and proportional rate of occurrence of follicular carcinoma and other thyroid malignancies.</p>
Full article ">Figure 2
<p>Pathways in the development, progression, metastasis, and de-differentiation of follicular neoplasms of the thyroid. The initiating event may be a gain mutation or re-arrangement in the genes that regulate the transmembrane tyrosine kinase receptors or involve intracellular kinases. The <span class="html-italic">MARK</span>, <span class="html-italic">PI3K/AKT/mTOR</span> and <span class="html-italic">RET</span> pathways are involved in the early phase of tumorigenesis. The <span class="html-italic">RAS</span> mutation is an upstream event for FTC, FA, FVPTC, and rarely PTC. <span class="html-italic">RAF</span> mutations are seen more frequently in PTC. The <span class="html-italic">p53, Wnt</span>, <span class="html-italic">PTEN</span>, <span class="html-italic">TERT,</span> and other mutations or rearrangements occur later as WDTC progresses towards PDTC or ATC. (Created using <a href="http://BioRender.com" target="_blank">BioRender.com</a> accessed on 13 February 2023).</p>
Full article ">Figure 3
<p>Progressive increase in the levels of complexity from machine learning to CNN in AI programs.</p>
Full article ">Figure 4
<p>Components of the multi-omics that can be integrated and analyzed using the AI platform for computer-aided decision making in the management of FTC.</p>
Full article ">
21 pages, 4170 KiB  
Article
Epigenetic Silencing of LRP2 Is Associated with Dedifferentiation and Poor Survival in Multiple Solid Tumor Types
by Martin Q. Rasmussen, Gitte Tindbæk, Morten Muhlig Nielsen, Camilla Merrild, Torben Steiniche, Jakob Skou Pedersen, Søren K. Moestrup, Søren E. Degn and Mette Madsen
Cancers 2023, 15(6), 1830; https://doi.org/10.3390/cancers15061830 - 17 Mar 2023
Cited by 3 | Viewed by 3252
Abstract
More than 80% of human cancers originate in epithelial tissues. Loss of epithelial cell characteristics are hallmarks of tumor development. Receptor-mediated endocytosis is a key function of absorptive epithelial cells with importance for cellular and organismal homeostasis. LRP2 (megalin) is the largest known [...] Read more.
More than 80% of human cancers originate in epithelial tissues. Loss of epithelial cell characteristics are hallmarks of tumor development. Receptor-mediated endocytosis is a key function of absorptive epithelial cells with importance for cellular and organismal homeostasis. LRP2 (megalin) is the largest known endocytic membrane receptor and is essential for endocytosis of various ligands in specialized epithelia, including the proximal tubules of the kidney, the thyroid gland, and breast glandular epithelium. However, the role and regulation of LRP2 in cancers that arise from these tissues has not been delineated. Here, we examined the expression of LRP2 across 33 cancer types in The Cancer Genome Atlas. As expected, the highest levels of LRP2 were found in cancer types that arise from LRP2-expressing absorptive epithelial cells. However, in a subset of tumors from these cancer types, we observed epigenetic silencing of LRP2. LRP2 expression showed a strong inverse correlation to methylation of a specific CpG site (cg02361027) in the first intron of the LRP2 gene. Interestingly, low expression of LRP2 was associated with poor patient outcome in clear cell renal cell carcinoma, papillary renal cell carcinoma, mesothelioma, papillary thyroid carcinoma, and invasive breast carcinoma. Furthermore, loss of LRP2 expression was associated with dedifferentiated histological and molecular subtypes of these cancers. These observations now motivate further studies on the functional role of LRP2 in tumors of epithelial origin and the potential use of LRP2 as a cancer biomarker. Full article
(This article belongs to the Special Issue The Role of Genetic and Epigenetic Aberrations in Cancer)
Show Figures

Figure 1

Figure 1
<p><span class="html-italic">LRP2</span> expression across human cancers. Boxplot of <span class="html-italic">LRP2</span> expression (RSEM norm_count log2 transformed with offset of 1) across each cancer type in the TCGA Pan-Cancer dataset. Boxplot lines represent lower quartile, median, and upper quartile. Whiskers represent 1.5 times above or below interquartile range. Points reflect outliers. Full list of cancer type abbreviations is provided in <a href="#app1-cancers-15-01830" class="html-app">Table S1</a>.</p>
Full article ">Figure 2
<p><span class="html-italic">LRP2</span> expression is largely restricted to malignant cells in the tumor microenvironment. (<b>A</b>) UMAP visualization of single-cell RNA sequencing data integrated across 8 advanced renal cell carcinoma tumors (subsampling 10.000 cells) and colored by lineage. (<b>B</b>) Log-normalized expression of <span class="html-italic">LRP2</span> overlaid on UMAP from panel A. (<b>C</b>) UMAP visualization of single-cell RNA sequencing data integrated across 26 primary breast tumors (subsampling 10.000 cells) and colored by major cell type. (<b>D</b>) Log-normalized expression of <span class="html-italic">LRP2</span> overlaid on UMAP from panel C. CAF: cancer associated fibroblasts.</p>
Full article ">Figure 3
<p>Immunohistochemical analysis of LRP2 protein expression in 12 luminal A invasive ductal carcinomas. Representative images of sections from 12 different invasive ductal carcinomas labeled with a monoclonal mouse anti-human LRP2 antibody (1:100) are shown. Magnification: 20×. Scale bar: 100 µm. Similar images from labeling of sections from the same 12 luminal A invasive ductal carcinomas with a polyclonal rabbit anti-human LRP2 antibody are shown in <a href="#app1-cancers-15-01830" class="html-app">Figure S6</a>.</p>
Full article ">Figure 4
<p>Low <span class="html-italic">LRP2</span> expression is associated with poor survival in multiple cancers. <span class="html-italic">LRP2</span> expression hazard ratio with 95% confidence intervals is shown for each cancer type. Hazard ratio &lt; 1 indicates decreased risk with higher <span class="html-italic">LRP2</span>. Hazard ratio &gt; 1 indicates increased risk with higher <span class="html-italic">LRP2</span>. Cancer types are ranked (top to bottom) according to their median expression of <span class="html-italic">LRP2</span>. <span class="html-italic">p</span> value from the likelihood-ratio test is shown for significant cancer types. No data are shown for seven cancer types where analysis was not performed due to insufficient event number. Full list of cancer-type abbreviations is provided in <a href="#app1-cancers-15-01830" class="html-app">Table S1</a>. Source data provided in <a href="#app1-cancers-15-01830" class="html-app">Table S2</a>.</p>
Full article ">Figure 5
<p>Low <span class="html-italic">LRP2</span> expression is associated with poor outcome in multiple cancers. (<b>A</b>–<b>G</b>) Kaplan–Meier curve of <span class="html-italic">LRP2</span><sup>high</sup> and <span class="html-italic">LRP2</span><sup>low</sup> groups stratified based on the lower quartile of <span class="html-italic">LRP2</span> expression in each dataset (upper panel) and risk table (lower panel). <span class="html-italic">p</span> value from the log-rank test is shown.</p>
Full article ">Figure 6
<p>CpG methylation in the <span class="html-italic">LRP2</span> gene locus. Heatmap of CpG site values from the 19 CpG sites in the <span class="html-italic">LRP2</span> gene included in the Illumina450K TCGA Pan-Cancer dataset. <span class="html-italic">LRP2</span> β values for each CpG site in KIRC, KIRP, MESO, THCA, BRCA, LUAD, and SKCM are shown. CpG ID, CpG position on chromosome 2 (GRCh38) and location relative to the full-length <span class="html-italic">LRP2</span> transcript (ENST00000649046) are shown below the heatmap. cg02361027 at position 169360890, which shows a strong inverse correlation to <span class="html-italic">LRP2</span> expression, is highlighted in red.</p>
Full article ">Figure 7
<p><span class="html-italic">LRP2</span> silencing is associated with methylation of a specific CpG site in the first intron. (<b>A</b>–<b>G</b>) Scatter plots of <span class="html-italic">LRP2</span> expression (RSEM norm_count log2 transformed with offset of 1) versus methylation β values for the CpG site cg02361027. Pearson’s R and corresponding <span class="html-italic">p</span> values are shown together with the linear regression line and equation.</p>
Full article ">Figure 8
<p><span class="html-italic">LRP2</span> silencing is associated with tumor dedifferentiation. (<b>A</b>–<b>C</b>) Volcano plot of differential gene-expression analysis of <span class="html-italic">LRP2</span><sup>low</sup> versus <span class="html-italic">LRP2</span><sup>high</sup> tumors (lower quartile cutoff) in KIRC (panel A), KIRP (panel B), and BRCA (panel C). Genes downregulated in <span class="html-italic">LRP2</span><sup>low</sup> tumors (FDR &lt; 0.01 and log2 fold change &lt; −1) are shown in blue. Genes upregulated in <span class="html-italic">LRP2</span><sup>low</sup> tumors (FDR &lt; 0.01 and log2 fold change &gt; 1) are shown in orange. Non-significant genes (FDR &gt; 0.01) are shown in black. (<b>D</b>–<b>F</b>) Enrichment plots from gene set enrichment analysis showing selected top pathways downregulated in <span class="html-italic">LRP2</span><sup>low</sup> tumors in KIRC (panel D), KIRP (panel E), and BRCA (panel F). (<b>G</b>) Boxplot of <span class="html-italic">LRP2</span> expression across PAM50 molecular subtypes in breast cancer cohort METABRIC. (<b>H</b>) Boxplot of <span class="html-italic">LRP2</span> expression (RSEM norm_count log2 transformed with offset of 1) across histological subtypes in TCGA MESO. Boxplot lines represent lower quartile, median, and upper quartile. Whiskers represent 1.5 times above or below interquartile range. Points reflect outliers. (<b>I</b>) Scatter plot of correlation between <span class="html-italic">LRP2</span> expression (RSEM norm_count log2 transformed with offset of 1) and 16-gene tumor differentiation score in TCGA THCA. Pearson’s R and corresponding <span class="html-italic">p</span> values are shown together with the linear regression line and equation. Wilcox rank-sum test: * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01, **** <span class="html-italic">p</span> ≤ 0.0001. TDS: tumor differentiation score.</p>
Full article ">
11 pages, 1979 KiB  
Article
Extra-Cellular Vesicles Derived from Thyroid Cancer Cells Promote the Epithelial to Mesenchymal Transition (EMT) and the Transfer of Malignant Phenotypes through Immune Mediated Mechanisms
by Stefania Mardente, Michele Aventaggiato, Elena Splendiani, Emanuela Mari, Alessandra Zicari, Giuseppina Catanzaro, Agnese Po, Lucia Coppola and Marco Tafani
Int. J. Mol. Sci. 2023, 24(3), 2754; https://doi.org/10.3390/ijms24032754 - 1 Feb 2023
Cited by 6 | Viewed by 2222
Abstract
Thyroid cancer is the most common endocrine cancer, and its incidence is increasing in many countries around the world. Among thyroid cancers, the papillary thyroid cancer (PTC) histotype is particularly prevalent. A small percentage of papillary tumors is associated with metastases and aggressive [...] Read more.
Thyroid cancer is the most common endocrine cancer, and its incidence is increasing in many countries around the world. Among thyroid cancers, the papillary thyroid cancer (PTC) histotype is particularly prevalent. A small percentage of papillary tumors is associated with metastases and aggressive behavior due to de-differentiation obtained through the epithelial–mesenchymal transition (EMT) by which epithelial thyroid cells acquire a fibroblast-like morphology, reduce cellular adhesion, increase motility and expression of mesenchymal proteins. The tumor microenvironment plays an important role in promoting an aggressive phenotype through hypoxia and the secretion of HMGB1 and other factors. Hypoxia has been shown to drastically change the tumor cell phenotype and has been associated with increasing metastatic and migratory behavior. Cells transfer information to neighboring cells or distant locations by releasing extracellular membrane vesicles (EVs) that contain key molecules, such as mRNAs, microRNAs (miRNAs), and proteins, that are able to modify protein expression in recipient cells. In this study, we investigated the potential role of EVs released by the anaplastic cancer cell line CAL-62 in inducing a malignant phenotype in a papillary cancer cell line (BCPAP). Full article
(This article belongs to the Special Issue Cell Signaling and Immune Targets in Cancer)
Show Figures

Figure 1

Figure 1
<p>Extra-cellular microvesicles (EVs) in untreated CAL-62 cells observed by TEM. Transmission electron micrographs of CAL-62 cells with EVs, after 24 h culture with 5% FBS under normoxic conditions. Measurements of extra-cellular vesicles in the range of 40–200 nm are indicated (Bar: 100 µm).</p>
Full article ">Figure 2
<p>HMGB1 OD values in the different samples from CAL-62 cells and BCPAP cells cultured under normoxic and hypoxic conditions. ELISA analysis of (<b>A</b>) cell lysates, (<b>B</b>) supernatants and EVs lysates (100 µL/sample). <span class="html-italic">p</span> &lt; 0.05 in populations indicated with asterisks (*). <span class="html-italic">p</span> &lt; 0.005 in populations indicated with asterisks (***).</p>
Full article ">Figure 3
<p>CAL-62 cell-derived EVs content and impact on BCPAP cells. Left Panel (<b>A</b>) miRNAs expression in EVs extracted from CAL-62 cell supernatants, normalized with U6. Right panel (<b>B</b>) confocal microscopy images of CAL-62 EVs internalized by BCPAP cells. Upper lane: control BCPAP cells. Lower lane: BCPAP cells treated with CAL-62 EVs. EVs labeled with PKH26 (red), nuclei stained with DAPI (blue), and cytoplasmic actin stained with phalloidin (green). Internalization reached its maximum after 2 h of exposure. Bar, 48.5 μm. (<b>C</b>) Relative expression of miRNAs normalized with U6. Asterisks indicate <span class="html-italic">p</span> values * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.005, and **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 4
<p>Effects of CAL-62 EVs on expression of EMT proteins in BCPAP cells (<b>A</b>) representative Western blot showing increase in YAP and vimentin in BCPAP cells treated with EVs from CAL-62 cells. Data are presented as mean ± s.d. from triplicate experiments. (<b>B</b>) Invasion test (indicative figure). (<b>C</b>) wound closure results of three independent experiments. (<b>D</b>) Proliferation rate increases in BCPAP cells treated with CAL-62 EVs. Asterisks indicate <span class="html-italic">p</span> values * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.005, and **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">
11 pages, 519 KiB  
Review
Genetic Changes in Thyroid Cancers and the Importance of Their Preoperative Detection in Relation to the General Treatment and Determination of the Extent of Surgical Intervention—A Review
by Jiri Hlozek, Barbora Pekova, Jan Rotnágl, Richard Holý and Jaromir Astl
Biomedicines 2022, 10(7), 1515; https://doi.org/10.3390/biomedicines10071515 - 27 Jun 2022
Cited by 19 | Viewed by 3257
Abstract
Carcinomas of the thyroid gland are some of the most common malignancies of the endocrine system. The causes of tumor transformation are genetic changes in genes encoding cell signaling pathways that lead to an imbalance between cell proliferation and apoptosis. Some mutations have [...] Read more.
Carcinomas of the thyroid gland are some of the most common malignancies of the endocrine system. The causes of tumor transformation are genetic changes in genes encoding cell signaling pathways that lead to an imbalance between cell proliferation and apoptosis. Some mutations have been associated with increased tumor aggressiveness, metastatic lymph node spread, tendency to dedifferentiate, and/or reduced efficiency of radioiodine therapy. The main known genetic causes of thyroid cancer include point mutations in the BRAF, RAS, TERT, RET, and TP53 genes and the fusion genes RET/PTC, PAX8/PPAR-γ, and NTRK. Molecular genetic testing of the fine needle aspiration cytology of the thyroid tissue in the preoperative period or of the removed thyroid tissue in the postoperative period is becoming more and more common in selected institutions. Positive detection of genetic changes, thus, becomes a diagnostic and prognostic factor and a factor that determines the extent of the surgical and nonsurgical treatment. The findings of genetic research on thyroid cancer are now beginning to be applied to clinical practice. In preoperative molecular diagnostics, the aggressiveness of cancers with the most frequently occurring mutations is correlated with the extent of the planned surgical treatment (radicality of surgery, neck dissection, etc.). However, clear algorithms are not established for the majority of genetic alterations. This review aims to provide a basic overview of the findings of the most commonly occurring gene mutations in thyroid cancer and to discuss the current recommendations on the extent of surgical and biological treatment concerning preoperatively detected genetic changes. Full article
(This article belongs to the Special Issue Recent Advances in Thyroid Cancer: From Diagnosis to Treatment)
Show Figures

Figure 1

Figure 1
<p>Simlified scheme of cell signaling pathways important in thyroid cancerogenesis (RTK—growth factor receptortyrosine kinase; RAS, BRAF, MEK, ERK, PI3K—signal molecules; Akt—protein kinase B; mTOR—mechanistic target of rapamycin; PTEN—phosphatase and tensin homolog; <span class="html-italic">PAX8/PPAR-γ</span>—fusion gene).</p>
Full article ">
15 pages, 1279 KiB  
Review
Redox Homeostasis in Thyroid Cancer: Implications in Na+/I Symporter (NIS) Regulation
by Juliana Cazarin, Corinne Dupuy and Denise Pires de Carvalho
Int. J. Mol. Sci. 2022, 23(11), 6129; https://doi.org/10.3390/ijms23116129 - 30 May 2022
Cited by 15 | Viewed by 3266
Abstract
Radioiodine therapy (RAI) is a standard and effective therapeutic approach for differentiated thyroid cancers (DTCs) based on the unique capacity for iodide uptake and accumulation of the thyroid gland through the Na+/I symporter (NIS). However, around 5–15% of DTC patients [...] Read more.
Radioiodine therapy (RAI) is a standard and effective therapeutic approach for differentiated thyroid cancers (DTCs) based on the unique capacity for iodide uptake and accumulation of the thyroid gland through the Na+/I symporter (NIS). However, around 5–15% of DTC patients may become refractory to radioiodine, which is associated with a worse prognosis. The loss of RAI avidity due to thyroid cancers is attributed to cell dedifferentiation, resulting in NIS repression by transcriptional and post-transcriptional mechanisms. Targeting the signaling pathways potentially involved in this process to induce de novo iodide uptake in refractory tumors is the rationale of “redifferentiation strategies”. Oxidative stress (OS) results from the imbalance between ROS production and depuration that favors a pro-oxidative environment, resulting from increased ROS production, decreased antioxidant defenses, or both. NIS expression and function are regulated by the cellular redox state in cancer and non-cancer contexts. In addition, OS has been implicated in thyroid tumorigenesis and thyroid cancer cell dedifferentiation. Here, we review the main aspects of redox homeostasis in thyrocytes and discuss potential ROS-dependent mechanisms involved in NIS repression in thyroid cancer. Full article
(This article belongs to the Special Issue Molecular Translational Research on Thyroid Cancer)
Show Figures

Figure 1

Figure 1
<p>The role of oxidative stress in thyroid carcinogenesis. A pro-oxidant environment is associated with thyroid tumorigenesis, and NADPH oxidases have been described as important ROS sources. Ionizing radiation, a risk factor for thyroid cancer, induces DUOX1-dependent H<sub>2</sub>O<sub>2</sub> production, resulting in DNA damage and potentially genomic instability. NOX4 is upregulated in PTCs and is positively regulated by the oncogenes BRAF<sup>V600E</sup> and HRAS<sup>V12</sup>. Increased NOX4 has been implicated in thyroid cancer dedifferentiation and genomic instability. DUOX2 is the source of H<sub>2</sub>O<sub>2</sub> for thyroid hormone biosynthesis in the apical membrane of thyrocytes, but its role in thyroid carcinogenesis is unclear.</p>
Full article ">Figure 2
<p>Mechanisms hypothetically involved in NIS redox regulation: (1) PAX8 oxidation results in reduced PAX8 DNA binding activity and the repression of <span class="html-italic">NIS</span> transcription; (2) ROS might mediate alterations of epigenetic events also promoting <span class="html-italic">NIS</span> transcriptional repression; (3) ROS might directly oxidize NIS protein or indirectly change the phosphorylation pattern of NIS protein, resulting in decreased NIS activity; (4) ROS might activate pathways involved in NIS endocytosis and autophagy, promoting NIS internalization and degradation.</p>
Full article ">
16 pages, 1683 KiB  
Review
To Detach, Migrate, Adhere, and Metastasize: CD97/ADGRE5 in Cancer
by Gabriela Aust, Leyu Zheng and Marianne Quaas
Cells 2022, 11(9), 1538; https://doi.org/10.3390/cells11091538 - 4 May 2022
Cited by 17 | Viewed by 4722
Abstract
Tumorigenesis is a multistep process, during which cells acquire a series of mutations that lead to unrestrained cell growth and proliferation, inhibition of cell differentiation, and evasion of cell death. Growing tumors stimulate angiogenesis, providing them with nutrients and oxygen. Ultimately, tumor cells [...] Read more.
Tumorigenesis is a multistep process, during which cells acquire a series of mutations that lead to unrestrained cell growth and proliferation, inhibition of cell differentiation, and evasion of cell death. Growing tumors stimulate angiogenesis, providing them with nutrients and oxygen. Ultimately, tumor cells invade the surrounding tissue and metastasize; a process responsible for about 90% of cancer-related deaths. Adhesion G protein-coupled receptors (aGPCRs) modulate the cellular processes closely related to tumor cell biology, such as adhesion and detachment, migration, polarity, and guidance. Soon after first being described, individual human aGPCRs were found to be involved in tumorigenesis. Twenty-five years ago, CD97/ADGRE5 was discovered to be induced in one of the most severe tumors, dedifferentiated anaplastic thyroid carcinoma. After decades of research, the time has come to review our knowledge of the presence and function of CD97 in cancer. In summary, CD97 is obviously induced or altered in many tumor entities; this has been shown consistently in nearly one hundred published studies. However, its high expression at circulating and tumor-infiltrating immune cells renders the systemic targeting of CD97 in tumors difficult. Full article
Show Figures

Figure 1

Figure 1
<p><b>Common features of human CD97</b>. (<b>a</b>) <b>Schematic structural organization of CD97.</b> The figure depicts the 835 amino acid full-length CD97(EGF1-5) isoform without the signal peptide. CD97 has a tripartite structure with the ECD, containing tandemly-arranged EGF-like repeats and the GAIN domain, the 7TM helices, and the ICD. The potential N-glycosylation sites in the EGF-like repeats are indicated. EGF2-5 can bind Ca<sup>2+</sup>, which is important for CD97 interactions. CD97 is self-cleaved at the GAIN domain-covered GPS, resulting in a bipartite structure with the NTF and CTF. Circulating sCD97 likely is the released NTF. The N-terminus of the CTF represents the integrated TA sequence. The ICD contains many phosphorylation sites. Four are confirmed in more than five records in which this modification was assigned using proteomic discovery mass spectrometry (<a href="http://phosphosite.org" target="_blank">phosphosite.org</a>, accessed on 4 February 2022). In CD97(EGF1-5), these sites are S818, T825, S831, and S833. The ICD ends up in a PBM. For abbreviations and further explanations, see the running text. (<b>b</b>) <b>Exon/intron and protein structure of the three CD97/<span class="html-italic">ADGRE5</span> isoforms.</b> Alternative splicing results in isoforms with three to five EGF-like repeats with distinct binding properties. The isoforms are named CD97(EGF125), EGF(1235), and EGF(1-5), according to the EGF-like repeats present (<a href="http://wormweb.org" target="_blank">wormweb.org</a>, accessed on 15 March 2022). Intron: line; exon: solid bar; 5′/3′-UTR: empty bar; signal peptide: red; EGF-like repeats: blue; GAIN: grey; 7TM: black; ICD: orange; scale bar: 1000 base pairs (<b>c</b>) <b>Interaction partners with the CD97 ECD.</b> CD55 is a glycophosphatidylinositol (GPI)-anchored transmembrane receptor with four short consensus repeats (SCRs). The first three SCRs interact with at least three EGF-like repeats of CD97. Thy-1/CD90 is a small, heavily N-glycosylated GPI-anchored transmembrane receptor with a single extracellular immunoglobulin (Ig) domain. Thy-1 binding to CD97 on polymorphonuclear cells (PMNC) is calcium-independent and occurs through the GAIN domain. Binding to the glycosaminoglycan side chain chondroitin sulfate B (CS B), a component of the extracellular matrix and of cell surfaces proteoglycans, is mediated by the fourth EGF-like repeat of CD97. Thus, CS B interacts only with CD97(EGF1-5). Binding is Ca<sup>2+</sup>-dependent. Soluble recombinant CD97 interacts with integrins on endothelial cells (ECs) via its RGD motif and at least three EGF-like repeats. CS B can act synergistically. Interacting domains are indicated in dark blue. (<b>d</b>) <b><span class="html-italic">ADGRE5</span> scRNAseq analysis in healthy normal human tissues.</b> The analysis comprises all protein-coding genes in 144 individual cell type clusters (The Human Protein Atlas, <a href="http://proteinatlas.org" target="_blank">proteinatlas.org</a>; accessed on 7 February 2022). In the heat map, only tissues containing the clusters “T-cells” and “macrophages” are considered. Additionally, the tissue-specific cell type cluster with the highest <span class="html-italic">ADGRE5</span> level is included (e.g., fat/adipocytes, lung/alveolar cells type 2, endometrium/smooth muscle cells, spleen/plasma cells, stomach/gastric mucus-secreting cells). Log2 transcripts per million (TPM) + 1 values are given. The figure was created with <a href="http://BioRender.com" target="_blank">BioRender.com</a> (accessed on 26 April 2022).</p>
Full article ">Figure 2
<p><b>CD97/<span class="html-italic">ADGRE5</span> in human cancer.</b> (<b>a</b>) <b><span class="html-italic">ADGRE5</span> levels in tumor-derived cell lines</b>, grouped by lineage-derivation, in part with subtypes such as lung cell lines (Cancer Cell Line Encyclopedia, <a href="http://depmap.org" target="_blank">depmap.org</a>; accessed on 15 February 2022). Except for small cell lung carcinomas (SCLCs, neuroendocrine tumors) and neuroblastoma, a malignant pediatric tumor of the peripheral (sympathetic) nerve system (PNS), most cell lines have moderate to high <span class="html-italic">ADGRE5</span>. Log2(TPM + 1) values are given. Ca (carcinoma); central nervous system (CNS); none small cell lung carcinoma (NSCLC) (<b>b</b>) <b>Frequency of <span class="html-italic">ADGRE5</span> gene alterations,</b> including somatic mutations, gene amplification, and deletion among various cancer (sub)types. The mutation frequency of <span class="html-italic">ADGRE5</span> in cancer is low (cBioPortal for Cancer Genomics, <a href="http://cbioportal.org" target="_blank">cbioportal.org</a>; accessed on 28 March 2022; PanCancer Studies, <span class="html-italic">n</span> = 76,639 samples). (<b>c</b>) <b>CD97 localization and functions in tumors.</b> In normal epithelium, as in enterocytes, CD97 localizes to E-cadherin-based adherens junctions (left, insert), likely maintaining <span class="html-italic">intercellular adhesion</span>, whereas in (colorectal) tumor cells, it frequently disappears from these cell contacts and accumulates inside the cells (middle, insert), where its function is not clarified. In several tumor entities, such as colorectal, gastric, and gall bladder carcinoma, CD97 is enhanced in tumor buds, appearing as scattered tumor cells in histological sections, and/or in cells at the tumor invasion front compared with cells in the tumor center, indicating a key role of CD97 in <span class="html-italic">tumor invasion</span>. Mechano-dependent phosphorylation at the CD97 PBM modulates <span class="html-italic">cellular detachment</span>. pCD97 appears in situ in scattered colorectal tumor cells and leukocytes, i.e., cells that dissociate from other cells or from the ECM during migration and invasion. Detachment likely occurs intracellularly at the PBM, not at the ECD, as indicated by lost membrane patches of detaching cells seen in vitro (right insert). (<b>d</b>) <b>CD97-regulated signaling cascades and <span class="html-italic">functions</span> in tumors.</b> CD97 is involved in <span class="html-italic">epithelial-mesenchymal transition (EMT).</span> In colorectal cancers, junctional proteins such as E-cadherin, β-catenin (β-cat), and CD97 frequently disappear from adherens junctions. β-catenin emerges in the cytoplasm and translocates into the nuclei, now acting as a transcriptional co-activator driving carcinogenesis. <span class="html-italic">Tumor migration and invasion.</span> CD97 heterodimerizes and functionally synergizes with LPAR1 to promote tumor cell (transendothelial) migration and invasion. The association activates the heterotrimeric G-protein Gα12/13. Upon GDP-GTP exchange, this complex dissociates into Gα12/13 and Gβ/γ subunits. The Gα12/13 subunit activates RHO; thus, stimulating various downstream signaling molecules (e.g., ERK/AKT and ROCK), to finally result in tumor cell migration and invasion. In one scenario, activated platelets release dense granules, causing disruption of the endothelial barrier-enabling tumor cell extravasation and metastasis. <span class="html-italic">Cell detachment.</span> Mechanical forces induce phosphorylation of CD97 by protein kinase C (PKC) and/or D (PKD) at its intracellular PBM, disrupting CD97 binding to the scaffold proteins such as DLG1. In parallel, the actin cytoskeleton is modulated and cells detach, which is necessary for enhanced tumor cell migration and invasion. <span class="html-italic">Tumor angiogenesis.</span> In experimental studies, CD97 interacts with integrin on ECs via its RGD motif and the EGF-like repeats to promote the angiogenesis associated with tumor progression and inflammation, and modulates angiogenesis through upregulation of MMP-9 by inducing N-cadherin expression. The figure was created with <a href="http://BioRender.com" target="_blank">BioRender.com</a> (accessed on 26 April 2022).</p>
Full article ">
17 pages, 2050 KiB  
Article
Targeting GLI1 Transcription Factor for Restoring Iodine Avidity with Redifferentiation in Radioactive-Iodine Refractory Thyroid Cancers
by Ji Min Oh, Ramya Lakshmi Rajendran, Prakash Gangadaran, Chae Moon Hong, Ju Hye Jeong, Jaetae Lee and Byeong-Cheol Ahn
Cancers 2022, 14(7), 1782; https://doi.org/10.3390/cancers14071782 - 31 Mar 2022
Cited by 2 | Viewed by 2728
Abstract
Radioactive-iodine (RAI) therapy is the mainstay for patients with recurrent and metastatic thyroid cancer. However, many patients exhibit dedifferentiation characteristics along with lack of sodium iodide symporter (NIS) functionality, low expression of thyroid-specific proteins, and poor RAI uptake, leading to poor prognosis. Previous [...] Read more.
Radioactive-iodine (RAI) therapy is the mainstay for patients with recurrent and metastatic thyroid cancer. However, many patients exhibit dedifferentiation characteristics along with lack of sodium iodide symporter (NIS) functionality, low expression of thyroid-specific proteins, and poor RAI uptake, leading to poor prognosis. Previous studies have demonstrated the effect of GLI family zinc finger 1 (GLI1) inhibition on tumor growth and apoptosis. In this study, we investigated the role of GLI1 in the context of redifferentiation and improvement in the efficacy of RAI therapy for thyroid cancer. We evaluated GLI1 expression in several thyroid cancer cell lines and selected TPC-1 and SW1736 cell lines showing the high expression of GLI. We performed GLI1 knockdown and evaluated the changes of thyroid-specific proteins expression, RAI uptake and I-131-mediated cytotoxicity. The effect of GANT61 (GLI1 inhibitor) on endogenous NIS expression was also assessed. Endogenous NIS expression upregulated by inhibiting GLI1, in addition, increased expression level in plasma membrane. Also, GLI1 knockdown increased expression of thyroid-specific proteins. Restoration of thyroid-specific proteins increased RAI uptake and I-131-mediated cytotoxic effect. Treatment with GANT61 also increased expression of endogenous NIS. Targeting GLI1 can be a potential strategy with redifferentiation for restoring RAI avidity in dedifferentiated thyroid cancers. Full article
(This article belongs to the Special Issue Thyroid Carcinoma)
Show Figures

Figure 1

Figure 1
<p>Investigation of hedgehog signaling pathway in thyroid cancer-derived cell lines. Three papillary thyroid cancer cell lines (BCPAP, K1, TPC-1) and three anaplastic thyroid cancer cell lines (BHT101, CAL62, SW1736) were subjected to western blot analysis. (<b>A</b>) Evaluation of the hedgehog signaling pathway. (<b>B</b>) Quantitative analysis of GLI1 expression in several thyroid cancer cell lines. ß-actin was used as an internal control. Mean ± standard deviation (SD) values from three independent experiments are presented. (<b>C</b>) Confirmation of NIS protein expression level in thyroid cancer-derived cell lines. ß-actin was used as an internal control. Raw data is presented in <a href="#app1-cancers-14-01782" class="html-app">Figures S2–S4</a>.</p>
Full article ">Figure 2
<p>Changes in endogenous NIS expression and its localization in GLI1-inhibited thyroid cancer cells. Both TPC-1 and SW1736 cells were treated with scrambled siRNA, GLI1 siRNA or NIS siRNA for 48 h. (<b>A</b>) Changes in NIS and GLI1 expression induced by GLI1 siRNA treatment in TPC-1 cells. GAPDH was used as an internal control. Mean ± SD values from five independent experiments are presented. *** <span class="html-italic">p</span> &lt; 0.001 (Student’s <span class="html-italic">t</span>-test). (<b>B</b>) Evaluation of NIS expression via GLI1 knockdown in SW1736 cells. Mean ± SD values from five independent experiments are presented. * <span class="html-italic">p</span> &lt; 0.05 (Student’s <span class="html-italic">t</span>-test). Western blot analysis for NIS protein with scrambled siRNA, GLI1 siRNA or NIS siRNA treatment to TPC-1 cells (<b>C</b>) and SW1736 cells (<b>D</b>). Immunofluorescence images showing localization of endogenous NIS expression in TPC-1 cells (<b>E</b>) and SW1736 cells (<b>F</b>). Scale bar: 20 µm. Expression of endogenous NIS protein in plasma membrane fraction by GLI1 knockdown in TPC-1 cells (<b>G</b>) and SW1736 cells (<b>H</b>). Caveolin-1 were used as loading controls for plasma membrane proteins. Raw data is presented in <a href="#app1-cancers-14-01782" class="html-app">Figures S5–S10</a>.</p>
Full article ">Figure 3
<p>Evaluation of the expression of thyroid-specific proteins and transcription factors in thyroid cancer-derived cells with GLI1 knockdown. Both TPC-1 and SW1736 cells were treated with scrambled siRNA or GLI1 siRNA for 48 h and changes in the expression of thyroid-specific proteins were evaluated. (<b>A</b>) Western blot analysis showing expression of thyroid-specific proteins (thyroperoxidase (TPO) and TSH receptor (TSHR)) and transcription factors (PAX-8 and TTF-1) in TPC-1 cells. (<b>B</b>) Quantitative analysis of western blots. GAPDH was used as an internal control. Mean ± SD values from at least three independent experiments are reported. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 (Student’s <span class="html-italic">t</span>-test). (<b>C</b>) Changes of thyroid-specific proteins and transcription factors expression in SW1736 cells with GLI1 knockdown. (<b>D</b>) Quantitative analysis of thyroid-specific proteins and transcription factors expression in SW1736 cells. Mean ± SD values from at least three independent experiments are presented. * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001 (Student’s <span class="html-italic">t</span>-test). Raw data is presented in <a href="#app1-cancers-14-01782" class="html-app">Figures S11–S14</a>.</p>
Full article ">Figure 4
<p>Verification of the extent of I-125 accumulation and I-131-mediated cytotoxic effect in GLI1-inhibited thyroid cancer cells. Both TPC-1 and SW1736 cells were treated with scrambled siRNA or GLI1 siRNA for 48 h. (<b>A</b>) For I-125 uptake assay, cells were treated with 37 kBq carrier-free I-125 and 100 μM sodium iodide at 37 °C for 30 min. Potassium perchlorate (KCIO<sub>4</sub>) was used as a competitive inhibitor of iodide transport. Upper–TPC-1 cells; Lower–SW1736 cells. The results are expressed as mean ± SD values of the experiment performed in triplicates. * <span class="html-italic">p</span> &lt; 0.05, NS: Not Significant (by Student’s <span class="html-italic">t</span>-test). (<b>B</b>) After incubation of siRNA in TPC-1 cells, the cells were incubated with or without 50 µCi/ml I-131 supplemented with 30 μM NaI for 7 h at 37 °C. Images about I-131 clonogenic assay. (<b>C</b>) Quantitative analysis based on I-131 clonogenic assay. The results are expressed as mean ± SD values of the experiment performed in triplicates. *** <span class="html-italic">p</span> &lt; 0.001, ** <span class="html-italic">p</span> &lt; 0.01, NS: Not Significant (by Student’s <span class="html-italic">t</span>-test).</p>
Full article ">Figure 5
<p>Efficacy of GANT61 in restoring endogenous NIS expression in thyroid cancer cells. Both TPC-1 and SW1736 cells were exposed to GANT61. (<b>A</b>) Results of cell viability assay showing time- and dose-dependent effects of GANT61 in TPC-1 cells. Mean ± SD values from three optical density (OD) is reported. (<b>B</b>) Endogenous NIS expression in whole cell lysate after treatment with GANT61. GAPDH was used as a loading control. The results are expressed as mean ± SD values of the experiment performed in quintuplicates. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, NS: Not Significant (by Student’s <span class="html-italic">t</span>-test). (<b>C</b>) Immunofluorescence images for monitoring changes of expression and localization with endogenous NIS in thyroid cancer-derived cells treated with GANT61. (<b>D</b>) Change of endogenous NIS expression in plasma membrane proteins. Caveolin-1 were used as loading controls for plasma membrane proteins. Raw data is presented in <a href="#app1-cancers-14-01782" class="html-app">Figures S15 and S16</a>.</p>
Full article ">
18 pages, 4611 KiB  
Article
Comprehensive Analysis of the Prognosis and Drug Sensitivity of Differentiation-Related lncRNAs in Papillary Thyroid Cancer
by Wenlong Wang, Ning Bai and Xinying Li
Cancers 2022, 14(5), 1353; https://doi.org/10.3390/cancers14051353 - 7 Mar 2022
Cited by 10 | Viewed by 3246
Abstract
Dedifferentiation is the main concern associated with radioactive iodine (RAI) refractoriness in patients with papillary thyroid cancer (PTC), and the underlying mechanisms of PTC dedifferentiation remain unclear. The present work aimed to identify a useful signature to indicate dedifferentiation and further explore its [...] Read more.
Dedifferentiation is the main concern associated with radioactive iodine (RAI) refractoriness in patients with papillary thyroid cancer (PTC), and the underlying mechanisms of PTC dedifferentiation remain unclear. The present work aimed to identify a useful signature to indicate dedifferentiation and further explore its role in prognosis and susceptibility to chemotherapy drugs. A total of five prognostic-related DR-lncRNAs were selected to establish a prognostic-predicting model, and corresponding risk scores were closely associated with the infiltration of immune cells and immune checkpoint blockade. Moreover, we built an integrated nomogram based on DR-lncRNAs and age that showed a strong ability to predict the 3- and 5-year overall survival. Interestingly, drug sensitivity analysis revealed that the low-risk group was more sensitive to Bendamustine and TAS-6417 than the high-risk group. In addition, knockdown of DR-lncRNAs (DPH6-DT) strongly promoted cell proliferation, invasion, and migration via PI3K-AKT signal pathway in vitro. Furthermore, DPH6-DT downregulation also increased the expression of vimentin and N-cadherin during epithelial-mesenchymal transition. This study firstly confirms that DR-lncRNAs play a vital role in the prognosis and immune cells infiltration in patients with PTC, as well as a predictor of the drugs’ chemosensitivity. Based on our results, DR-lncRNAs can serve as a promising prognostic biomarkers and treatment targets. Full article
(This article belongs to the Special Issue Biomarkers of Thyroid Cancer)
Show Figures

Figure 1

Figure 1
<p>Study flow chart.</p>
Full article ">Figure 2
<p>The landscape of DR-lncRNAs regulators. (<b>A</b>) Expression of 16 differentiation related regulators in normal and tumor samples; (<b>B</b>) The relationship between differentiation-related regulators and DR-lncRNAs. (<b>C</b>) Differential expression of 5 DR-lncRNAs regulators. *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 3
<p>Prognostic value of risk score. (<b>A</b>) Survival analysis of patients in the low-risk and high-risk groups. (<b>B</b>) Distributions of survival status and risk scores. (<b>C</b>) Heatmap distribution of risk scores and clinicopathological characteristics of the two groups. (<b>D</b>) The AUC of the risk score. AUC: the area under the receiver operating characteristic curve.</p>
Full article ">Figure 4
<p>The correlation between risk signature and immune cell infiltration. (<b>A</b>) CIBERSORT. (<b>B</b>) ssGSEA. (<b>C</b>) MCP counter. **** <span class="html-italic">p</span> &lt; 0.0001, *** <span class="html-italic">p</span> &lt; 0.001, ** <span class="html-italic">p</span> &lt; 0.01, and * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 5
<p>Drug sensitivity analysis. (<b>A</b>) Correlations between IC50 for different drugs and differentiation-related regulators. (<b>B</b>) Compared the efficiency of the chosen drugs in relation to the risk score. * <span class="html-italic">p</span> &lt; 0.05 and ns: no significance.</p>
Full article ">Figure 6
<p>Construction and evaluation of an integrated nomogram. (<b>A</b>) Nomogram was developed based on DR-lncRNA-based risk scores and age. (<b>B</b>) Calibration plots were performed to evaluate the predictive performance of 3- and 5-year OS. (<b>C</b>) The AUC value of the nomogram. (<b>D</b>) DCA of the nomogram. OS: overall survival, AUC: the area under the receiver operating characteristic curve. DCA: decisions curve analysis.</p>
Full article ">Figure 7
<p>Differential expression of prognostic related DR-lncRNAs. (<b>A</b>) Twenty paired PTC samples and adjacent normal tissues. (<b>B</b>) Nine thyroid cells and normal thyroid follicular epithelial cells. (<b>C</b>) PTC, ATC, and normal thyroid follicular epithelial cells. ATC: anaplastic thyroid cancer. PTC: papillary thyroid cancer. *** <span class="html-italic">p</span> &lt; 0.001, ** <span class="html-italic">p</span> &lt; 0.01, * <span class="html-italic">p</span> &lt; 0.05 and no significance.</p>
Full article ">Figure 8
<p>Depletion of <span class="html-italic">DHP6-DT</span> promoted proliferation and metastasis by activating the PI3K-AKT signaling pathway. (<b>A</b>) IHH-4 and KTC-1 cells were transfected with different si<span class="html-italic">DPH6-DT</span> and scramble vector (control). (<b>B</b>) Knockdown of <span class="html-italic">DPH6-DT</span> enhanced cell viability by CCK-8 assay. (<b>C</b>) Deficiency of <span class="html-italic">DPH6-DT</span> dramatically increased cell proliferation by EdU assay. (<b>D</b>) Knockdown of <span class="html-italic">DPH6-DT</span> accelerated cell migration and invasion. (<b>E</b>) Western blot for the EMT and PI3K-AKT signal pathway related protein expression upon the knockdown of DPH6-DT. EMT: epithelial-mesenchymal transition. *** <span class="html-italic">p</span> &lt; 0.001, ** <span class="html-italic">p</span> &lt; 0.01, and * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 9
<p>Brief summary for the molecular mechanism of DR-lncRNAs. EMT: epithelial-mesenchymal transition. ATC: anaplastic thyroid cancer. PTC: papillary thyroid cancer. Biorender, available online: <a href="https://app.biorender.com" target="_blank">https://app.biorender.com</a> (accessed on 2 November 2021).</p>
Full article ">
Back to TopTop