Next Article in Journal
Modulation of Photosystem II Function in Celery via Foliar-Applied Salicylic Acid during Gradual Water Deficit Stress
Previous Article in Journal
Determination of Matrix Metalloproteinase 2 in Biological Samples Using a 3D Stochastic Microsensor Based on Graphene Oxide/AuNanoparticles/(Z)-N-(pyridin-4-yl-methyl) Octadec-9-enamide
Previous Article in Special Issue
Mitochondrial One-Carbon Metabolism and Alzheimer’s Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Biomarkers in Thyroid Cancer: Emerging Opportunities from Non-Coding RNAs and Mitochondrial Space

1
Department of Head and Neck Surgery, Clinica INDISA, Santiago 7520440, Chile
2
Faculty of Medicine, Universidad Andres Bello, Santiago 8370071, Chile
3
Department of Basic and Clinical Oncology, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
4
Institute of Biomedical Sciences, Faculty of Medicine and Faculty of Life Sciences, Universidad Andres Bello, Santiago 8370071, Chile
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(12), 6719; https://doi.org/10.3390/ijms25126719
Submission received: 20 April 2024 / Revised: 1 June 2024 / Accepted: 14 June 2024 / Published: 18 June 2024
(This article belongs to the Special Issue Mitochondrial Metabolism Alterations in Health and Disease)

Abstract

:
Thyroid cancer diagnosis primarily relies on imaging techniques and cytological analyses. In cases where the diagnosis is uncertain, the quantification of molecular markers has been incorporated after cytological examination. This approach helps physicians to make surgical decisions, estimate cancer aggressiveness, and monitor the response to treatments. Despite the availability of commercial molecular tests, their widespread use has been hindered in our experience due to cost constraints and variability between them. Thus, numerous groups are currently evaluating new molecular markers that ultimately will lead to improved diagnostic certainty, as well as better classification of prognosis and recurrence. In this review, we start reviewing the current preoperative testing methodologies, followed by a comprehensive review of emerging molecular markers. We focus on micro RNAs, long non-coding RNAs, and mitochondrial (mt) signatures, including mtDNA genes and circulating cell-free mtDNA. We envision that a robust set of molecular markers will complement the national and international clinical guides for proper assessment of the disease.

1. Introduction

Thyroid cancer accounts for 1 to 4% of all malignancies worldwide. Its incidence has increased over the years, from 1.5% before 1996 to 6.8% after 1996, with an increase of three times per decade [1] (for a definition of epidemiological terms, see Table 1). In Chile, the estimated number of new cases per year is 1300 [2]. This increase in incidence has been attributed to a higher diagnosis rate, mainly due to the implementation of imaging techniques (diagnosis achieved in 50% of cases), as well as genetic and environmental factors, such as obesity and the increase in iodine consumption [1]. Histologically, thyroid cancer develops in two cell types, with more than 90% derived from follicular cells (epithelial, responsible for the uptake of iodine and production of thyroid hormones), and 3–5% derived from parafollicular cells (associated with synthesis and production of calcitonin).
According to the WHO 2022 guidelines, thyroid cancers have been categorized into three primary groups based on their level of differentiation. (1) Low grade (differentiated) cancers, which include papillary thyroid carcinoma (PTC), follicular thyroid carcinoma (FTC) and oncocytic carcinoma. The prognosis for these cancers is generally favorable. (2) High-grade cancers, which include poorly differentiated thyroid carcinoma (PDTC) and high-grade differentiated thyroid carcinoma (DHGTC). These cancers are more aggressive, presenting poorer outcomes, than low-grade cancers. (3) Anaplastic thyroid cancer (ATC), which is the most undifferentiated and aggressive form of thyroid cancer with a very poor prognosis [3]. Papillary thyroid cancer is the most common and has several histological subtypes (diffuse sclerosing, tall cell, columnar cell, solid, and hobnail among others) and clinical behavior [4]. Although we acknowledge different tumor types of thyroid carcinomas, we mostly focus on the papillary thyroid carcinoma in this review.

1.1. Diagnosis and Uncertainty in Thyroid Cancer

The classic methods for diagnosing thyroid cancer are through images (ultrasound—TIRADS classification) and fine-needle aspiration (cytology) [5,6]. The latter allows classifying the probability of thyroid cancer according to the “Bethesda Criteria” from I to VI: I, non-diagnosed or inadequate; II, benign; III, atypia of undetermined significance; IV, follicular neoplasia; V, suspicious for malignancy; and VI, malignant. In the latest update (2023), each category receives a combined risk of malignancy (ROM), with a mean of 13%, 4%, 22%, 30%, 74%, and 97%, respectively [6]. Handling cases based on an indeterminate cytological study is not solely determined by the Bethesda classification. In fact, the application of this method alone has not been able to provide clinically applicable pre-surgical recommendations, either in terms of follow-up or diagnostic surgical resection. As a result, international guidelines incorporate clinical, ultrasound, and cytological characteristics, as well as molecular markers [7]. The latter tests are based on genetic mutation analysis, gene expression evaluation, and microRNA-based classifiers (specific panels are not disclosed). Genetic mutations related to the mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) pathway are at the forefront of research in this area [8]. Clinical and molecular tools for guiding the diagnosis and proposing therapeutic options for thyroid cancer patients based on molecular characterization are limited and have not been updated so far. Some new tumorigenic and prognostic factors have been added to the study of other tumors, and more recently, to thyroid cancer [5,9,10]. The application of these markers in clinical decision-making should be studied in detail. This review provides a comprehensive overview of potential new markers with higher clinical relevance in thyroid cancer.

1.2. Current Preoperative Molecular Markers and Commercial Tests

There are molecular tests and tumor markers to study thyroid cancer, particularly the papillary thyroid carcinoma, but there is also an availability of molecular tests to study the other histological types of thyroid neoplasia [7,11,12]. Molecular genetic markers, such as mutations in MAPK pathways, including ALK, NTRK, RET, RAS, and BRAFV600E, have been studied for the early detection of papillary cancer [13]. Mutations in RAS, BRAFK601E, PAX8/PPAR and less common PTEN have been observed in follicular thyroid cancer [14,15,16]. For less differentiated tumors with a worse prognosis, such as anaplastic thyroid cancer, where TERT and P53 mutations are common, the integration of these markers will provide better diagnostic and prognostic value.
The indication to perform a molecular study arises in order to safely assign a benign status for an indeterminate thyroid nodule, i.e., reported as Bethesda III or IV [17,18], leading to the avoidance of unnecessary surgical procedure. Molecular studies also allow prognostic stratification and thus modify management decisions, although in our experience routine practice is difficult to massify due to costs and variability between tests [5]. Testing for single mutations, such as BRAFV600E or RET/PTC translocations, has yielded good specificity (100%) but poor sensitivity (50–60%) [19,20], hence panels detecting gene mutation profiles or changes in expression levels have arisen. It has been proposed that, while mutation tests are good rule-in tests for thyroid cancer, gene expression classifier testing is a good rule-out test for thyroid cancer [13]. The use of gene mutation profiling panels, such as a panel detecting mutations found in 70% of thyroid cancers (BRAFV600E, HRAS codon 61, KRAS codons 12/13, NRAS codon 61, RET/PTC1, RET/PTC3, and PAX8/PPARG, [9]), has yielded a sensitivity of >90% [21]. A gene classifier test, capable of classifying changes in the expression levels of up to 167 genes, has a significant negative predictive value but little positive predictive value (52%) [9].
Regarding commercially available molecular tests used in clinical practice, Table 2 summarizes the main features of Afirma®, Thyroseq®, Thyroid Print, and ThyGeNEXT®, all belonging to a group that identifies genetic anomalies through sequencing and gene expression.
While Afirma GSC is a rule-out test focused on Bethesda III/IV samples, Afirma XA (Xpression Atlas) is used for GSC suspicious and Bethesda V/VI samples [23]. Studies have shown that complementation of the GEC (Gene Expression Classifier) with the Afirma GSC approach leads to higher specificity without compromising sensitivity [31]. GSC can also be used on its own for benign calls [32,33], and XA improves stratification [34]. When used in combination, GSC and XC presented a high positive and negative predictive value and decreased the number of surgeries in 75% of instances [22]. Thyroseq V3 (2018) is a test based on 112 thyroid-related genes, looking for point mutations, indels, gene fusions, copy number alterations, and abnormal gene expression [25]. In return, Thyroseq reports a “genomic classifier” to separate malignant from benign lesions, preventing surgery in most patients [24]. Regarding Thyroid Print, its recent validation from two multicenter and independent clinical trials has given it a specificity of 88%, a negative predictive value of 95%, and a positive predictive value of 78%. Meanwhile, a study reported a positive predictive value of 71% despite a smaller sample [30]. In Chile, Olmos et al. [29] carried out a prospective cohort study using 1272 fine-needle aspirations in a single-health center. They studied indeterminate nodules (Bethesda III and IV) using the Thyroid Print classifier and found benign nodules (comparable to Bethesda II) in 67% of cases after a 15-month follow-up. For patients with suspicious results after the test, the risk of malignancy was estimated at 71% compared to the histological result, making the risk of malignancy comparable to Bethesda [29]. Finally, the ThyGeNEXT test looks for mutations in selected genes (ALK, BRAF, GNAS, HRAS, KRAS, NRAS, PIK3CA, PTEN, RET, and TERT promoter genes) plus fusion genes (ALK, BRAF, NTRK, PPARG, RET, PAX8, TBP, USP33, and THADA) [27], although this panel is often used in combination with a panel of microRNAs (miRNAs) named ThyraMIR to improve stratification and predictive value (more below) [26].
The current molecular classifiers have theoretically higher performance in thyroid cancer diagnosis than cytology, with sensitivities ranging from 91–94% and specificities between 68% and 90%. These molecular classifiers have been shown to provide favorable outcomes in cytologically indeterminate nodules, primarily by averting diagnostic surgeries. They achieve this through high negative predictive values, indicating a low likelihood of cancer when test results are negative. However, their widespread application is hindered by the limitation of larger studies outside the United States, either due to the high acquisition costs or logistical terms of commercialization due to patents from the country of origin (USA) and other factors that could be the real causes of exemption from diagnostic surgery in their reports. Despite the use of biological markers in indeterminate nodules following Bethesda classification, there is still no genetic characterization that works independently to select a specific treatment. However, both European and American guidelines suggest interpretation at the clinician’s discretion to guide management if they are available for use [5].
The large genetic panels available on the market, as well as the study of specific mutations (which may or may not be included in the panels), have not only complementary applications with cytology but also in determining the degree of aggressiveness and response to treatment. The mutations BRAFV600E, TERT, and RAS are the most widely studied in relation to these clinical variables [35,36]. Genetic mutations have been observed in various types of thyroid cancer, which have affected the different clinical pillars related to this condition. The presence of BRAFV600E is associated with the classic papillary type and its high cell subtype [35]. It is often linked to locoregional invasion, lymph node metastasis (especially cervical and lung), and refractoriness to radiotherapy. Mutations in RAS and RET are both related to invasive behavior (RAS mostly through the hematogenous route and consequently to the bone) but with differences [37]. Mutations in RAS can be present in different types of thyroid cancers (papillary and follicular), with the papillary type having the highest frequency at 38%. These mutations retain susceptibility to radioactive iodine therapy. Additionally, in follicular cell cancers, RET/PTC fusions (particularly RET/PTC1 and RET/PTC3) have been identified, especially in the papillary type in pediatric and young adult populations. Their presence indicates a certain behavior but still responds well to radioiodine treatment [35,38]. On the other hand, point mutations in RET (specific base pair changes) have lower susceptibility to remission with I131 and are found in medullary thyroid cancer, mainly in hereditary cases, and to a lesser extent in sporadic cases [35].
Based on various histopathological findings in large patient cohorts, the use of molecular markers has been incorporated specifically after cytological diagnosis to determine aggressiveness and response to treatment. However, this is limited to anaplastic and medullary cancer. In the context of anaplastic thyroid cancer, the identification of the BRAF mutation V600E is incorporated into the initial management process to recommend the use of selective BRAF/MEK inhibitors (dabrafenib/trametinib, respectively), particularly if the cancer is not entirely unresectable [35]. When it comes to medullary cancer, detecting the germline mutation in the RET gene is used to determine whether it is hereditary [39].

1.3. Stratifying the Risk of Disease Recurrence

The standard treatment for differentiated thyroid carcinoma (DTC) usually involves surgery, followed by radioiodine (RI) ablation if necessary. The American Thyroid Association (ATA) suggests classifying patients into three risk groups (high, intermediate, and low) based on the risk of disease recurrence or persistence [17]. In low-risk patients, radioiodine therapy is generally not recommended as a complementary treatment. These patients have a low mortality rate, with a recurrence or persistence risk of about 3% [40]. The ATA guidelines consider the BRAF gene mutation as one of the factors to predict intermediate risk of recurrence if it is associated with multifocal micropapillary thyroid cancer. Other risk factors are determined exclusively from the histopathological results following surgery. However, the BRAF gene mutation alone cannot determine the use of I131 as it depends on the histological characterization [35]. However, studies have shown that the presence of BRAF downregulates iodine–sodium transporters, regardless of whether it is a classic subtype of papillary thyroid cancer or a cell type, making them resistant to iodine treatment. Therefore, the molecular characterization of thyroid cancer is improved by attempting radiotherapy and, if refractory, scaling up to selective NTRK inhibitors. This reflects the usefulness of incorporating biomarkers into stratification plans, contrasting with the selective use offered by ATA [41,42]. It is worth noting that the markers we described earlier are not the only ones available. New tumor factors have been incorporated into various studies, with and without applications in thyroid cancer.

2. Emerging Molecular Markers for Thyroid Cancer: Opportunities from the Non-Coding Genome

The sequencing of the human genome revealed that most RNAs in eukaryotes are “non-coding” (ncRNA), as they do not have the potential to produce proteins [43]. Thus, 75% of the information contained in DNA is transcribed to RNAs, but only 2% is translated to proteins [44]. Originally, non-coding RNAs were interpreted as transcriptional noise [45]; however, the significant role played by these RNAs has been confirmed, both in biological mechanisms [46,47] and in the development of diseases, including cancer [48,49]. With the advancements and reduced costs in omics technologies, alongside the development of lab-on-chip devices, detecting changes in gene expression or mutations is becoming increasingly accessible and affordable [50,51].

2.1. Micro RNAs

MicroRNAs (miRNAs) are non-coding RNAs that are synthesized from longer transcripts and processed into 19- to 25-nucleotide oligonucleotides through a step-wise mechanism (reviewed in [52]). By binding to target mRNAs, miRNAs repress translation and induce mRNA decay, significantly influencing the overall gene expression profile in cells. In thyroid cancer, miRNA expression profiles analyzed through quantitative PCR have been employed to stratify tumors, with diagnostic tests such as ThyraMIR and mir-THYpe being notable examples.
As mentioned before, ThyraMIR is often used as a complement to the ThyGeNEXT test [26]. ThyraMIR analyzes changes in the expression of 11 miRNAs and their association with malignancy (Table 3) [53]. In a single-health center study, ThyGeNEXT + ThyraMIR showed a 52,94% of positive predictive value [22], and 95% of diagnostic yield [54]. Although testing positive is associated with malignancy in surgical pathology, the ThyraMIR classifier failed to differentiate between benign and malignant RAS-mutated tumors [55]. As for mir-THYpe, it analyzes 11 miRNAs, where 6 are used as normalizers (let-7a, miR-103, miR-125a-5p, let-7b, miR-145, and RNU48) and 5 as discriminators of malignancy (Table 3) [56]. mir-THYpe reached average values of sensitivity 89.3%, specificity 81.6%, NPV 95%, and PPV 66%, as shown by a prospective, non-interventional multicenter study carried out on 440 patients. These results allowed clinicians to reduce unnecessary surgery in 52.5% of cases [57].
It is envisioned that an increased array of molecular biomarkers will enhance the reliability of thyroid cancer tests and offer clinicians more options for stratifying indeterminate thyroid nodules. With the growing number of studies investigating the molecular mechanisms underlying thyroid cancer, additional miRNAs are emerging as potential alternatives for future diagnostic tests [59]. Interestingly, DICER, the endoribonuclease responsible for generating mature miRNAs from stem-loop miRNAs [52], may harbor pathogenic somatic mutations that alter miRNA-5p to miRNA-3p production in thyroid cancer [62].
Potential miRNA candidates for diagnosis of thyroid cancer are described in Table 4. Based on the number of studies performed with each miRNA in cancer cells, the miR-1, miR-2, miR-7, and miR144 arise as the most robust and promising biomarkers to be included in future diagnostic tests. With the advancement and refinement of small RNA purification techniques, coupled with the availability of commercial adapters, quantification of miRNAs via real-time PCR or next-generation sequencing platforms has become a routine procedure [50].

2.2. Long Non-Coding RNAs (lncRNAs)

Long non-coding RNAs (lncRNAs) are transcripts longer than 200 nucleotides that do not encode proteins. These RNAs are often spliced, capped, and polyadenylated [47]. lncRNAs can modulate gene expression through various mechanisms, including direct binding to the DNA, interacting with epigenetic modifiers, and recruiting transcription factors to regulatory regions. Additionally, lncRNAs regulate miRNA function by capturing them and preventing them from targeting mRNAs in cells. This mechanism prevents miRNAs from blocking translation. Consequently, when lncRNA are dysregulated in disease, associated changes in gene expression may occur at both the transcriptional and post-transcriptional level [84].
Most lncRNAs exhibit cell-specific expression patterns and are typically expressed at low levels [85]. Alterations in their expression have been linked to the development and progression of various diseases, including thyroid cancer [86]. While lncRNAs have been studied in the context of thyroid cancer, their clinical application remains in the research phase. Using long non-coding RNAs (lncRNAs) as biomarkers offers several advantages over proteins, particularly in terms of quantification. Designing, acquiring, and standardizing primers for real-time PCR is often simpler and more straightforward than developing antibodies. Additionally, while messenger RNA levels do not always correlate with protein amounts due to post-transcriptional mechanisms, non-coding RNAs are generally considered more consistent as final products. Although lncRNAs can undergo modifications, such as adenosine methylation, the implications of these modifications are still being explored.
Table 5 presents potential lncRNA candidates for use as biomarkers in thyroid cancer. While all of them may be deemed promising molecules, HOTAIR and MALAT1 stand out as robust candidates due to the substantial support from numerous studies across various cancers [87], along with PTCSC3 as an RNA originally discovered in thyroid cancer [88]. HOTAIR is a lncRNA with an essential role in the repression of early development-related genes in adult individuals, collaborating with epigenetic complexes [89]. In papillary thyroid cancer, HOTAIR has been found to be upregulated, leading to increased growth, migration, and invasive properties [76,79,90]. Patients with lower levels of HOTAIR have better survival rates [91]. Regarding MALAT1, this lncRNA is widely recognized among cancer researchers. Initially discovered as one of the most overexpressed RNAs in lung cancer patients with metastasis, MALAT1 has been found overexpressed in numerous other tumor types, including thyroid [87,90]. MALAT11 regulates expression of target genes by directly binding to the genome and collaborating with epigenetic complexes. By functioning also as a competing endogenous RNA, it captures miRNAs in the nucleus [92,93]. Thus, MALAT1 increases the expression of proteins associated with proliferation, migration, and angiogenesis, such as FGF2. This association links MALAT1 to a prognosis of metastasis and early locoregional invasion [94]. In anaplastic thyroid carcinoma, MALAT1 knockdown leads to reduced cell proliferation, invasion, and migration [95], like the effects observed with BRAF and RET gene alterations. This phenomenon has been particularly noted in medullary thyroid cancer [94]. However, its application in clinical practice is currently limited, as it has only been tested in vitro in thyroid tissue [95]. Finally, regarding PTCSC3, this lncRNA was initially identified as thyroid-specific [88], although subsequent research has revealed its presence in other cancers [96]. In thyroid cancer cells, it is frequently downregulated [88], and its overexpression has been associated with growth inhibition, cell cycle arrest, and increased apoptosis [97]

2.3. Emerging RNA Molecules: Circular and Double-Stranded RNAs

In recent years, RNA variants that were originally disregarded as mis-splicing artifacts or by-products have been found to play significant roles in biological models. Advances in RNA-sequencing techniques and bioinformatics have revealed that circular and double-stranded RNAs participate in various biological mechanisms [107,108]. Circular RNAs are generated by back-splicing of pre-mRNAs and can function by sponging miRNAs and interacting with proteins (see [108] for review). By designing primers flanking the back-splice site, circular RNAs can be quantified specifically. Examples of circular RNAs found to be dysregulated in thyroid cancer cells include circITGA7, circRAPGEF5, circ_0058129, and circ_0005273 [74,81,83]. However, further research is required to gather robust data and propose circular RNAs as clinical biomarkers.
Double-stranded RNAs (dsRNAs), initially associated with viral infections, accumulate in vertebrate cells during the dysregulation of various cellular processes, including cancer [108] These dsRNAs originate from activated long and short interspersed elements, endogenous retroviruses, and mitochondrial transcripts. Unlike dsRNAs derived from viruses, endogenous dsRNAs often evade degradation and remain localized within the nucleus and mitochondria. Their quantification requires antibodies or dsRNA-binding proteins [109], making their incorporation into routine clinical assays more challenging compared to miRNAs or lncRNAs. Currently, no promising candidates among double-stranded RNAs have been identified as potential biomarkers for thyroid cancer diagnosis. Further research will uncover robust biomarkers that could aid in diagnosing cancer patients in the future.

3. Opportunities beyond the Nucleus: Mitochondrial Signatures

The proliferative and invasive capacity of cancer cells involves a reprogramming of the intermediary and energy metabolism of tumor cells, where mitochondria are the key players. Beyond energy generation, mitochondria are considered a powerful signaling platform for tumor anabolism and have an impact on tumor redox environment, calcium homeostasis, transcriptional and epigenetic regulation, and cell death pathways. In fact, several anti-cancer treatments disrupt mitochondrial function to trigger cancer cell death [110]. The proper work of mitochondria is sustained by two fundamental processes: first, the oxidative phosphorylation system (OXPHOS), which is the engine of energy generation in the form of ATP from the oxidation of glucose and fatty acid molecules. When mitochondria generate ATP, reactive oxygen species (ROS) are also formed as a by-product which are responsible for many of the aging-associated diseases, including cancer [111]. Second is the mitochondrial life cycle. This cycle refers to mitochondrial dynamics (MtDy), i.e., fusion and fission events of mitochondria; mitophagy, which corresponds to the selective degradation of damaged mitochondria or those that are no longer needed; and Mitochondrial Biogenesis, which involves mtDNA replication, protein synthesis and membrane biosynthesis [112].

3.1. mtDNA Signatures

Mitochondria contain multiple copies of mtDNA, which can range from tens to hundreds depending on the physiological and pathophysiological conditions of cells. The mtDNA is highly polymorphic and maternally inherited, which configures everyone with its own molecular maternal surname, called haplogroups, which correlate with the geographic origins of populations. The oldest haplogroups are from Africa and, with geographic migration and climate adaptations, the haplogroups from Europe, Asia, and America have evolved [113]. The type of haplogroup grants adaptive advantages and/or susceptibility to disease [114]. The mtDNA is a 16 kb circular DNA and encodes for 37 genes (13 mRNA, 2 rRNA, and 22 tRNA). The mitochondrial mRNAs encode for 13 core proteins of OXPHOS, which play a critical role in the proper assembly and functioning of the electron transport chain (ETC) for the generation of the mitochondrial membrane potential and, therefore, for the ATP synthesis. As a by-product of ETC functioning, ROS are generated. Since mtDNA molecules are in the same place in which ROS are produced, that is in the mitochondrial matrix, they are prone to undergo point mutations, deletions, and a reduction in their copy number. The mutation rate of mtDNA is higher compared to the nuclear genome [115] and these mutations accumulate as organisms age. The co-existence of wild type mtDNA with mutant mtDNA is known as heteroplasmy [116,117]. mtDNA alterations are associated with severe mitochondrial dysfunction and diseases. Thus, the integrity of mtDNA is pivotal for cell survival and health of the organism [118,119].
Recycling of oxidized and damaged mitochondria, (known as mitophagy) is critical for maintaining the copy number and integrity of mtDNA while keeping reduced levels of heteroplasmy [120,121,122]. Elevated mtDNA heteroplasmy leads to increased ROS which damages the bioenergetic capacity of mitochondria and alters mitophagy, favoring the development of diseases such as cancer [123,124]. Thus, both mtDNA and mitophagy are interconnected and dependent on each other. When mitochondria are stressed and/or mitophagy is dysfunctional, mtDNA may leak out from mitochondria and cells to reach plasma circulation, where it can be detected. The amount and features of this circulating cell-free mtDNA (ccf-mtDNA) constitute a parameter of mitochondrial dysfunction and disease.
In the context of a disease, the detection and analysis of mtDNA in terms of copy number, heteroplasmy, and haplogroups, as well as the levels of ccf-mtDNA, which are all measurable parameters, may configure a genomic signature and a powerful diagnostic tool in cancer. In fact, mtDNA copy number alterations contribute to the development of different types of carcinomas, such as lung cancer [124], breast cancer [125], hepatic cell carcinomas [126], gastric cancers [127], bladder, esophageal, kidney, and head/neck squamous carcinomas [125]. An increased mtDNA copy number may promote cancer progression by enhancing mitochondrial oxidative phosphorylation, as has also been observed in colorectal cancer [128]. Thyroid tumor cells also present an increase in the mtDNA copy number [129]. However, the relationship of mtDNA copy number with the risk of cancer is still inconclusive.
In some cancer cells, the striking accumulation of mitochondria leads to swollen cells with distinctive granular cytoplasmic eosinophilia forming an “oncocytic tumor” [130]. These tumors present two characteristic mutations: a deletion of 4977 bp called “common deletion” [131] and Complex 1 mtDNA mutations (loss of function and missense) that affect nearly 60% of patients [132,133,134]. Regarding papillary thyroid cancer, it was recently reported that this type of tumor also presents mitochondrial accumulation, DNA mutations, and loss of complex I integrity [135]. As for the mitochondrial accumulation, the Tall Cell subtype of Papillary Cell Carcinoma is considered “mitochondria-rich”, indicating that there is a spectrum in mitochondria content that can be indicative of disease progression [135]. In thyroid cancer, an increase has also been reported in the mtDNA heteroplasmy at the level of both point mutations and deletions, mainly affecting the respiratory complex I and IV [136]. Sasarman et al. [137] also reported mutations in MT-CYB encoding for Cyt b (subunit of complex III) and MT-TL1 encoding the mitochondrial tRNALeu in cancer. In addition, mtDNA mutations affecting the complex I subunit, ND1 (G3842A), ND4 (A11708G), and ND5(12418insA), have been correlated with liver cancer progression, and the ND2 (T4216C) mutation with colorectal cancer [138,139,140].
On the other hand, the haplogroup M has been associated with breast cancer [25] while, in thyroid cancer, a preponderance has been observed a preponderance of the haplogroup D4a in Chinese patients [141]. and notably, the haplogroup K in Europeans has been shown to be protective against thyroid cancer [142]. Su et al. [143] reported three sub-haplogroups of N (A4, B4a and B4g) and eight single-nucleotide polymorphisms (mtSNPs) (A16164G, C16266T, G5460A, T6680C, G9123A, A14587G, T16362C, and G709A) associate with the occurrence of papillary thyroid cancer.
Regarding the ccf-mtDNA, accumulating evidence suggests that plasma or serum ccf-mtDNA levels are significantly different between cancer patients and healthy individuals. Thus, the level of ccf-mtDNA has been shown to be significantly higher in patients with lung cancer than in healthy patients [144]. In patients infected with hepatitis B virus (HBV), a lower content of ccf-mtDNA had a significantly increased risk of hepatocellular carcinoma as compared with HBV patients with higher ccf-mtDNA [145]. In metastatic colorectal cancer, a decrease in ccf-mtDNA has also been shown, as compared with healthy individuals [146]. Similar results were found in patients with papillary thyroid carcinoma, who presented decreased ccf-mtDNA levels in plasma [147]. Interestingly, the amount of ccf-DNA in blood has been used as a potential non-invasive biomarker for diagnostic and prognostic evaluation in a wide variety of cancers, because it has been shown to be increased or decreased in the serum of patients with various malignant tumors as compared with healthy patients [148,149].
ccf-mtDNA is widely recognized as a favorable option over nuclear-circulating DNA because it is more abundant and then easy to detect and analyze, and because changes in ccf-mtDNA are closely related to the occurrence and development of tumors in terms of copy number and integrity [150]. Thus, the detection and analysis of ccf-mtDNA may provide important information for diagnosis and prognosis in thyroid cancer patients, which may reduce the number of unnecessary thyroid surgical procedures.

3.2. Mitophagic Gene Signature

In cancer metabolism, mitophagy not only enables the removal of dysfunctional, depolarized, or damaged mitochondria, but also allows the reduction of mitochondrial biomass as an adaptive response to metabolic stress, such as hypoxia, starvation, and DNA damage that are present in the tumor. Besides providing all the essential nutrients to support tumor biology, through carbohydrate, protein, lipid, and nucleotide degradation [151], mitophagy is also involved in the control of ROS and calcium homeostasis for cell signaling and differentiation, and in the regulation of the balance between fermentative glycolysis and OXPHOS energy metabolic pathways [152,153].
In cancer patients, it has been observed that mitophagy processes are deregulated, but whether they behave as promoters of tumor formation or as suppressors seems to be highly dependent on the type of cancer and its metabolic context [151] (Table 6). In this regard, cancer stem cells, which are associated with the promotion of metastasis, therapy resistance, and tumor dormancy, make the most of mitophagy to sustain their self-renewal properties [154]. In lung or pancreatic cancer cells, the loss of the mitophagic proteins ATG7 or ATG5 significantly decreases the aggressiveness of the tumor due to the accumulation of defective mitochondria and an excessive accumulation of lipids, accounting for a greater alteration in the ß-oxidation of fatty acids [155].
Cancer cells are also characterized by the stabilization of the hypoxia-induced transcription factor HIF-1α, one of the main factors responsible for the change in energy metabolism from the oxidative metabolism to the aerobic glycolytic metabolism, also known as the Warburg effect [151]. Notably, HIF-1α also activates the transcriptional regulation of the mitophagy receptors NIX and BNIP3, which would decrease mitochondrial biomass, reducing the cells’ oxygen demand and thus promoting survival under hypoxic conditions. The HIF-1α signaling pathway is more active during the proliferation and metastasis of cancer cells, which is why high HIF-1α expression is associated with a poor clinical prognosis. Results found in U87 and Glio6 glioma cells cultured under hypoxia showed that 32 genes were upregulated, including the mitochondrial nuclear-encoded genes BNIP3, NIX, and FAM162A [174]. BNIP3 and NIX have been involved both in apoptosis and mitophagy [175,176,177,178,179,180,181], and FAM162A, only in apoptosis [182]. However, it has been mentioned as belonging to the same BH3-only family as NIX and BNIP3. Toustrup et al. [183] and Sorensen et al. [184] defined 15-gene hypoxia classifiers based on gene expression to predict prognosis and therapeutic response in various cancer types, because hypoxia is a common feature in solid tumors. They assessed four head and neck squamous cell carcinomas, three esophagus cancer cell lines, three prostate cancer cell lines, and six colon cancer cell lines, and FAM162A, BNIP3, and NIX genes were overexpressed [183,184]. To date, there is no information about those genes in thyroid cancer. Interestingly, FAM162A has been shown to be highly expressed in the SiHa uterine cervix cancer (UCC) cell line when HIF-1a is overexpressed. However, far from inducing apoptosis and cell death, the UCC SiHa cells increased their proliferation and migration capacities [185]. The expression of FAM162A was investigated in 85 human tissue samples of UCC and over 70% were positive for FAM162A by immunohistochemistry as well as for HIF-1a, suggesting that FAM162A participates in a molecular mechanism of hypoxia-regulated tumor development. In fact, FAM162A-positive tumors are associated with an advanced stage of cancer, parametrial invasion, and tumor recurrence, according to the International Federation of Gynecology and Obstetrics. In addition, the overall survival rate tended to be poorer in patients with FAM162A expression than in those without FAM162A expression [186].

4. Conclusions

Promising molecular markers for thyroid cancer associated with non-coding nuclear genes and mitochondria have been reviewed and proposed. From a diagnostic perspective, identifying novel molecular markers can enhance the accuracy of preoperative fine-needle aspiration samples. Among the increasing number of alternatives, and based on the number of studies performed with each RNA, miR-1, miR-2, miR-7, and miR-144, along with the long non-coding RNAs HOTAIR, MALAT1, and PTCSC3 emerge as the most robust and promising candidates. Regarding mitochondria, measurements and genetic analysis of mtDNA, as well as the expression of mitophagic proteins FAM162A, NIX, and BNIP3, will contribute to a better understanding of the biology and development of thyroid cancer and support current cancer diagnosis and prognosis, benefiting patients. It is envisioned that these novel biomarkers will complement the commonly used cancer risk/probability classification (Bethesda). Their results can then be compared with the definitive biopsy findings of patients operated on for thyroid cancer (Bethesda V and VI) and those operated on for suspected thyroid cancer (Bethesda III and IV). Thus, based on classifications of prognosis and recurrence, the novel molecular markers reviewed here can serve as indicators of aggressiveness in patients with papillary thyroid cancer. This can complement the guidelines from the National (Chilean Department of Health) and International (ATA and TNM) clinical guides for a more accurate assessment of the disease [187,188].

Author Contributions

Conceptualization, P.C., R.A. and A.A.E.; writing, C.C., I.B., P.C., R.A. and A.A.E. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by UNIVERSIDAD ANDRES BELLO, grant number NUCLEO UNAB DI-03-22/NUC (to P.C., R.A., A.A.E.); and ANID FONDECYT REGULAR, grant number 1240853 (to R.A.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Kitahara, C.M.; Sosa, J.A. Understanding the ever-changing incidence of thyroid cancer. Nat. Rev. Endocrinol. 2020, 16, 617–618. [Google Scholar] [CrossRef] [PubMed]
  2. Sapunar, Z.J.; Ferrer, R.P. Epidemiología del cáncer de tiroides en un Instituto Oncológico. Efecto de las nuevas recomendaciones clínicas. Rev. Médica Chile 2020, 148, 573–581. [Google Scholar] [CrossRef] [PubMed]
  3. Jung, C.K.; Bychkov, A.; Kakudo, K. Update from the 2022 World Health Organization Classification of Thyroid Tumors: A Standardized Diagnostic Approach. Endocrinol. Metab. 2022, 37, 703–718. [Google Scholar] [CrossRef] [PubMed]
  4. Coca-Pelaz, A.; Shah, J.P.; Hernandez-Prera, J.C.; Ghossein, R.A.; Rodrigo, J.P.; Hartl, D.M.; Olsen, K.D.; Shaha, A.R.; Zafereo, M.; Suarez, C.; et al. Papillary Thyroid Cancer-Aggressive Variants and Impact on Management: A Narrative Review. Adv. Ther. 2020, 37, 3112–3128. [Google Scholar] [CrossRef] [PubMed]
  5. Durante, C.; Hegedus, L.; Czarniecka, A.; Paschke, R.; Russ, G.; Schmitt, F.; Soares, P.; Solymosi, T.; Papini, E. 2023 European Thyroid Association Clinical Practice Guidelines for thyroid nodule management. Eur. Thyroid J. 2023, 12, e230067. [Google Scholar] [CrossRef] [PubMed]
  6. Ali, S.Z.; Baloch, Z.W.; Cochand-Priollet, B.; Schmitt, F.C.; Vielh, P.; VanderLaan, P.A. The 2023 Bethesda System for Reporting Thyroid Cytopathology. Thyroid 2023, 33, 1039–1044. [Google Scholar] [CrossRef] [PubMed]
  7. Abdullah, M.I.; Junit, S.M.; Ng, K.L.; Jayapalan, J.J.; Karikalan, B.; Hashim, O.H. Papillary Thyroid Cancer: Genetic Alterations and Molecular Biomarker Investigations. Int. J. Med. Sci. 2019, 16, 450–460. [Google Scholar] [CrossRef] [PubMed]
  8. Schubert, L.; Mariko, M.L.; Clerc, J.; Huillard, O.; Groussin, L. MAPK Pathway Inhibitors in Thyroid Cancer: Preclinical and Clinical Data. Cancers 2023, 15, 710. [Google Scholar] [CrossRef]
  9. Alexander, E.K.; Kennedy, G.C.; Baloch, Z.W.; Cibas, E.S.; Chudova, D.; Diggans, J.; Friedman, L.; Kloos, R.T.; LiVolsi, V.A.; Mandel, S.J.; et al. Preoperative diagnosis of benign thyroid nodules with indeterminate cytology. N. Engl. J. Med. 2012, 367, 705–715. [Google Scholar] [CrossRef]
  10. Liu, Y.; Khan, S.; Li, L.; Ten Hagen, T.L.M.; Falahati, M. Molecular mechanisms of thyroid cancer: A competing endogenous RNA (ceRNA) point of view. Biomed. Pharmacother. 2022, 146, 112251. [Google Scholar] [CrossRef]
  11. Macerola, E.; Poma, A.M.; Vignali, P.; Proietti, A.; Ugolini, C.; Torregrossa, L.; Basolo, A.; Elisei, R.; Santini, F.; Basolo, F. Predictive Biomarkers in Thyroid Cancer. Front. Oncol. 2022, 12, 901004. [Google Scholar] [CrossRef] [PubMed]
  12. Silaghi, C.A.; Lozovanu, V.; Georgescu, C.E.; Georgescu, R.D.; Susman, S.; Nasui, B.A.; Dobrean, A.; Silaghi, H. Thyroseq v3, Afirma GSC, and microRNA Panels Versus Previous Molecular Tests in the Preoperative Diagnosis of Indeterminate Thyroid Nodules: A Systematic Review and Meta-Analysis. Front. Endocrinol. 2021, 12, 649522. [Google Scholar] [CrossRef] [PubMed]
  13. Lee, K.; Anastasopoulou, C.; Chandran, C.; Cassaro, S. Thyroid Cancer. 1 May 2023. Available online: https://www.ncbi.nlm.nih.gov/books/NBK459299/ (accessed on 21 January 2024).
  14. Matrone, A.; Citro, F.; Gambale, C.; Prete, A.; Minaldi, E.; Ciampi, R.; Ramone, T.; Materazzi, G.; Torregrossa, L.; Elisei, R. BRAF K601E Mutation in Oncocytic Carcinoma of the Thyroid: A Case Report and Literature Review. J. Clin. Med. 2023, 12, 6970. [Google Scholar] [CrossRef] [PubMed]
  15. Bandargal, S.; Rajab, M.; Forest, V.I.; Pusztaszeri, M.P.; Hier, M.P.; da Silva, S.D.; Payne, R.J. Characteristics of PTEN Mutation in Thyroid Tumours: A Retrospective Chart Review. Cancers 2023, 15, 1575. [Google Scholar] [CrossRef] [PubMed]
  16. Kondo, T.; Ezzat, S.; Asa, S.L. Pathogenetic mechanisms in thyroid follicular-cell neoplasia. Nat. Rev. Cancer 2006, 6, 292–306. [Google Scholar] [CrossRef] [PubMed]
  17. Haugen, B.R.; Alexander, E.K.; Bible, K.C.; Doherty, G.M.; Mandel, S.J.; Nikiforov, Y.E.; Pacini, F.; Randolph, G.W.; Sawka, A.M.; Schlumberger, M.; et al. 2015 American Thyroid Association Management Guidelines for Adult Patients with Thyroid Nodules and Differentiated Thyroid Cancer: The American Thyroid Association Guidelines Task Force on Thyroid Nodules and Differentiated Thyroid Cancer. Thyroid 2016, 26, 1–133. [Google Scholar] [CrossRef] [PubMed]
  18. Sahli, Z.T.; Smith, P.W.; Umbricht, C.B.; Zeiger, M.A. Preoperative Molecular Markers in Thyroid Nodules. Front. Endocrinol. 2018, 9, 179. [Google Scholar] [CrossRef] [PubMed]
  19. Adeniran, A.J.; Hui, P.; Chhieng, D.C.; Prasad, M.L.; Schofield, K.; Theoharis, C. BRAF mutation testing of thyroid fine-needle aspiration specimens enhances the predictability of malignancy in thyroid follicular lesions of undetermined significance. Acta Cytol. 2011, 55, 570–575. [Google Scholar] [CrossRef] [PubMed]
  20. Kim, S.K.; Hwang, T.S.; Yoo, Y.B.; Han, H.S.; Kim, D.L.; Song, K.H.; Lim, S.D.; Kim, W.S.; Paik, N.S. Surgical results of thyroid nodules according to a management guideline based on the BRAF(V600E) mutation status. J. Clin. Endocrinol. Metab. 2011, 96, 658–664. [Google Scholar] [CrossRef]
  21. Nikiforov, Y.E.; Ohori, N.P.; Hodak, S.P.; Carty, S.E.; LeBeau, S.O.; Ferris, R.L.; Yip, L.; Seethala, R.R.; Tublin, M.E.; Stang, M.T.; et al. Impact of mutational testing on the diagnosis and management of patients with cytologically indeterminate thyroid nodules: A prospective analysis of 1056 FNA samples. J. Clin. Endocrinol. Metab. 2011, 96, 3390–3397. [Google Scholar] [CrossRef]
  22. Kandil, E.; Metz, T.A.; Issa, P.P.; Aboueisha, M.; Omar, M.; Attia, A.S.; Chabot, B.; Hussein, M.; Moroz, K.; Shama, M.; et al. Diagnostic Performance of Afirma and Interpace Diagnostics Genetic Testing in Indeterminate Thyroid Nodules: A Single Center Study. Cancers 2023, 15, 2098. [Google Scholar] [CrossRef] [PubMed]
  23. Krane, J.F.; Cibas, E.S.; Endo, M.; Marqusee, E.; Hu, M.I.; Nasr, C.E.; Waguespack, S.G.; Wirth, L.J.; Kloos, R.T. The Afirma Xpression Atlas for thyroid nodules and thyroid cancer metastases: Insights to inform clinical decision-making from a fine-needle aspiration sample. Cancer Cytopathol. 2020, 128, 452–459. [Google Scholar] [CrossRef]
  24. Desai, D.; Lepe, M.; Baloch, Z.W.; Mandel, S.J. ThyroSeq v3 for Bethesda III and IV: An institutional experience. Cancer Cytopathol. 2021, 129, 164–170. [Google Scholar] [CrossRef] [PubMed]
  25. Nikiforova, M.N.; Mercurio, S.; Wald, A.I.; Barbi de Moura, M.; Callenberg, K.; Santana-Santos, L.; Gooding, W.E.; Yip, L.; Ferris, R.L.; Nikiforov, Y.E. Analytical performance of the ThyroSeq v3 genomic classifier for cancer diagnosis in thyroid nodules. Cancer 2018, 124, 1682–1690. [Google Scholar] [CrossRef]
  26. Alzahrani, A.S. Clinical use of Molecular Data in Thyroid Nodules and Cancer. J. Clin. Endocrinol. Metab. 2023, 108, 2759–2771. [Google Scholar] [CrossRef]
  27. Alzumaili, B.; Sadow, P.M. Update on Molecular Diagnostics in Thyroid Pathology: A Review. Genes 2023, 14, 1314. [Google Scholar] [CrossRef] [PubMed]
  28. Gonzalez, H.E.; Martinez, J.R.; Vargas-Salas, S.; Solar, A.; Veliz, L.; Cruz, F.; Arias, T.; Loyola, S.; Horvath, E.; Tala, H.; et al. A 10-Gene Classifier for Indeterminate Thyroid Nodules: Development and Multicenter Accuracy Study. Thyroid 2017, 27, 1058–1067. [Google Scholar] [CrossRef]
  29. Olmos, R.; Dominguez, J.M.; Vargas-Salas, S.; Mosso, L.; Fardella, C.E.; Gonzalez, G.; Baudrand, R.; Guarda, F.; Valenzuela, F.; Arteaga, E.; et al. ThyroidPrint(R): Clinical utility for indeterminate thyroid cytology. Endocr. Relat. Cancer 2023, 30, e220409. [Google Scholar] [CrossRef] [PubMed]
  30. Zafereo, M.; McIver, B.; Vargas-Salas, S.; Dominguez, J.M.; Steward, D.L.; Holsinger, F.C.; Kandil, E.; Williams, M.; Cruz, F.; Loyola, S.; et al. A Thyroid Genetic Classifier Correctly Predicts Benign Nodules with Indeterminate Cytology: Two Independent, Multicenter, Prospective Validation Trials. Thyroid 2020, 30, 704–712. [Google Scholar] [CrossRef]
  31. Harrell, R.M.; Eyerly-Webb, S.A.; Golding, A.C.; Edwards, C.M.; Bimston, D.N. Statistical Comparison of Afirma Gsc and Afirma Gec Outcomes in a Community Endocrine Surgical Practice: Early Findings. Endocr. Pract. 2019, 25, 161–164. [Google Scholar] [CrossRef]
  32. Angell, T.E.; Heller, H.T.; Cibas, E.S.; Barletta, J.A.; Kim, M.I.; Krane, J.F.; Marqusee, E. Independent Comparison of the Afirma Genomic Sequencing Classifier and Gene Expression Classifier for Cytologically Indeterminate Thyroid Nodules. Thyroid 2019, 29, 650–656. [Google Scholar] [CrossRef] [PubMed]
  33. Wei, S.; Veloski, C.; Sharda, P.; Ehya, H. Performance of the Afirma genomic sequencing classifier versus gene expression classifier: An institutional experience. Cancer Cytopathol. 2019, 127, 720–724. [Google Scholar] [CrossRef] [PubMed]
  34. Munoz-Zuluaga, C.A.; Heymann, J.J.; Solomon, J.P.; Patel, A.; Siddiqui, M.T.; Scognamiglio, T.; Gokozan, H.N. Use of the Afirma Xpression Atlas for cytologically indeterminate, Afirma Genomic Sequencing Classifier suspicious thyroid nodules: Clinicopathologic analysis with postoperative molecular testing. Am. J. Clin. Pathol. 2024, 161, 463–468. [Google Scholar] [CrossRef] [PubMed]
  35. Boucai, L.; Zafereo, M.; Cabanillas, M.E. Thyroid Cancer: A Review. JAMA 2024, 331, 425–435. [Google Scholar] [CrossRef] [PubMed]
  36. Marrero Rodríguez, M.T.; Sinconegui Gómez, B.; Cruz Cruz, A. Marcadores moleculares en el cáncer de tiroides. Rev. Cuba. Endocrinol. 2015, 26, 93–104. [Google Scholar]
  37. Nguyen, B.; Fong, C.; Luthra, A.; Smith, S.A.; DiNatale, R.G.; Nandakumar, S.; Walch, H.; Chatila, W.K.; Madupuri, R.; Kundra, R.; et al. Genomic characterization of metastatic patterns from prospective clinical sequencing of 25,000 patients. Cell 2022, 185, 563–575.e511. [Google Scholar] [CrossRef] [PubMed]
  38. Zeindl-Eberhart, E.; Liebmann, S.; Jungblut, P.R.; Mattow, J.; Schmid, M.; Kerler, R.; Rabes, H.M. Influence of RET/PTC1 and RET/PTC3 oncoproteins in radiation-induced papillary thyroid carcinomas on amounts of cytoskeletal protein species. Amino Acids 2011, 41, 415–425. [Google Scholar] [CrossRef] [PubMed]
  39. Larouche, V.; Akirov, A.; Thomas, C.M.; Krzyzanowska, M.K.; Ezzat, S. A primer on the genetics of medullary thyroid cancer. Curr. Oncol. 2019, 26, 389–394. [Google Scholar] [CrossRef]
  40. Parvathareddy, S.K.; Siraj, A.K.; Qadri, Z.; Ahmed, S.O.; DeVera, F.; Al-Sobhi, S.; Al-Dayel, F.; Al-Kuraya, K.S. Lymph node ratio is superior to AJCC N stage for predicting recurrence in papillary thyroid carcinoma. Endocr. Connect. 2022, 11, e210518. [Google Scholar] [CrossRef]
  41. Biondi, B.; Cooper, D.S. Benefits of thyrotropin suppression versus the risks of adverse effects in differentiated thyroid cancer. Thyroid 2010, 20, 135–146. [Google Scholar] [CrossRef]
  42. Chakravarty, D.; Santos, E.; Ryder, M.; Knauf, J.A.; Liao, X.H.; West, B.L.; Bollag, G.; Kolesnick, R.; Thin, T.H.; Rosen, N.; et al. Small-molecule MAPK inhibitors restore radioiodine incorporation in mouse thyroid cancers with conditional BRAF activation. J. Clin. Investig. 2011, 121, 4700–4711. [Google Scholar] [CrossRef]
  43. Kung, J.T.; Colognori, D.; Lee, J.T. Long noncoding RNAs: Past, present, and future. Genetics 2013, 193, 651–669. [Google Scholar] [CrossRef]
  44. Consortium, E.P.; Birney, E.; Stamatoyannopoulos, J.A.; Dutta, A.; Guigo, R.; Gingeras, T.R.; Margulies, E.H.; Weng, Z.; Snyder, M.; Dermitzakis, E.T.; et al. Identification and analysis of functional elements in 1% of the human genome by the ENCODE pilot project. Nature 2007, 447, 799–816. [Google Scholar] [CrossRef] [PubMed]
  45. Ohno, S. Simplicity of mammalian regulatory systems. Dev. Biol. 1972, 27, 131–136. [Google Scholar] [CrossRef]
  46. Andergassen, D.; Rinn, J.L. From genotype to phenotype: Genetics of mammalian long non-coding RNAs in vivo. Nat. Rev. Genet. 2022, 23, 229–243. [Google Scholar] [CrossRef]
  47. Mattick, J.S.; Amaral, P.P.; Carninci, P.; Carpenter, S.; Chang, H.Y.; Chen, L.L.; Chen, R.; Dean, C.; Dinger, M.E.; Fitzgerald, K.A.; et al. Long non-coding RNAs: Definitions, functions, challenges and recommendations. Nat. Rev. Mol. Cell Biol. 2023, 24, 430–447. [Google Scholar] [CrossRef]
  48. Disney, M.D. Targeting RNA with Small Molecules To Capture Opportunities at the Intersection of Chemistry, Biology, and Medicine. J. Am. Chem. Soc. 2019, 141, 6776–6790. [Google Scholar] [CrossRef] [PubMed]
  49. Zhang, F.; Lupski, J.R. Non-coding genetic variants in human disease. Hum. Mol. Genet. 2015, 24, R102–R110. [Google Scholar] [CrossRef] [PubMed]
  50. Dhuli, K.; Medori, M.C.; Donato, K.; Donato, K.; Maltese, P.E.; Tanzi, B.; Tezzele, S.; Mareso, C.; Miertus, J.; Generali, D.; et al. Omics sciences and precision medicine in thyroid cancer. Clin. Ther. 2023, 174, 11–20. [Google Scholar] [CrossRef]
  51. Venkatesalu, S.; Dilliyappan, S.; Satish Kumar, A.; Palaniyandi, T.; Baskar, G.; Ravi, M.; Sivaji, A. Prospectives and retrospectives of microfluidics devices and lab-on-A-chip emphasis on cancer. Clin. Chim. Acta 2024, 552, 117646. [Google Scholar] [CrossRef]
  52. Bofill-De Ros, X.; Vang Orom, U.A. Recent progress in miRNA biogenesis and decay. RNA Biol. 2024, 21, 1–8. [Google Scholar] [CrossRef] [PubMed]
  53. Finkelstein, S.D.; Sistrunk, J.W.; Malchoff, C.; Thompson, D.V.; Kumar, G.; Timmaraju, V.A.; Repko, B.; Mireskandari, A.; Evoy-Goodman, L.A.; Massoll, N.A.; et al. A Retrospective Evaluation of the Diagnostic Performance of an Interdependent Pairwise MicroRNA Expression Analysis with a Mutation Panel in Indeterminate Thyroid Nodules. Thyroid 2022, 32, 1362–1371. [Google Scholar] [CrossRef] [PubMed]
  54. Labourier, E.; Fahey, T.J., 3rd. Preoperative molecular testing in thyroid nodules with Bethesda VI cytology: Clinical experience and review of the literature. Diagn. Cytopathol. 2021, 49, E175–E180. [Google Scholar] [CrossRef] [PubMed]
  55. Tumati, A.; Egan, C.E.; Lee-Saxton, Y.J.; Marshall, T.E.; Lee, J.; Jain, K.; Heymann, J.J.; Gokozan, H.; Azar, S.A.; Schwarz, J.; et al. Clinical utility of a microRNA classifier in cytologically indeterminate thyroid nodules with RAS mutations: A multi-institutional study. Surgery 2024, 175, 234–240. [Google Scholar] [CrossRef] [PubMed]
  56. Santos, M.T.D.; Buzolin, A.L.; Gama, R.R.; Silva, E.; Dufloth, R.M.; Figueiredo, D.L.A.; Carvalho, A.L. Molecular Classification of Thyroid Nodules with Indeterminate Cytology: Development and Validation of a Highly Sensitive and Specific New miRNA-Based Classifier Test Using Fine-Needle Aspiration Smear Slides. Thyroid 2018, 28, 1618–1626. [Google Scholar] [CrossRef] [PubMed]
  57. Santos, M.T.; Rodrigues, B.M.; Shizukuda, S.; Oliveira, A.F.; Oliveira, M.; Figueiredo, D.L.A.; Melo, G.M.; Silva, R.A.; Fainstein, C.; Dos Reis, G.F.; et al. Clinical decision support analysis of a microRNA-based thyroid molecular classifier: A real-world, prospective and multicentre validation study. EBioMedicine 2022, 82, 104137. [Google Scholar] [CrossRef] [PubMed]
  58. Mian, C.; Pennelli, G.; Fassan, M.; Balistreri, M.; Barollo, S.; Cavedon, E.; Galuppini, F.; Pizzi, M.; Vianello, F.; Pelizzo, M.R.; et al. MicroRNA profiles in familial and sporadic medullary thyroid carcinoma: Preliminary relationships with RET status and outcome. Thyroid 2012, 22, 890–896. [Google Scholar] [CrossRef]
  59. Rogucki, M.; Sidorkiewicz, I.; Niemira, M.; Dzieciol, J.B.; Buczynska, A.; Adamska, A.; Siewko, K.; Kosciuszko, M.; Maliszewska, K.; Wojcicka, A.; et al. Expression Profile and Diagnostic Significance of MicroRNAs in Papillary Thyroid Cancer. Cancers 2022, 14, 2679. [Google Scholar] [CrossRef]
  60. Ignjatovic Jocic, V.; Jankovic Miljus, J.; Isic Dencic, T.; Zivaljevic, V.; Tatic, S.; Doric, I.; Selemetjev, S. Expression of pY397-FAK and Its miR Regulators Drive Dedifferentiation in the Thyroid Neoplasia Spectrum. Cells 2023, 12, 1721. [Google Scholar] [CrossRef]
  61. Fuziwara, C.S.; Kimura, E.T. MicroRNA Deregulation in Anaplastic Thyroid Cancer Biology. Int. J. Endocrinol. 2014, 2014, 743450. [Google Scholar] [CrossRef]
  62. Ricarte-Filho, J.C.; Casado-Medrano, V.; Reichenberger, E.; Spangler, Z.; Scheerer, M.; Isaza, A.; Baran, J.; Patel, T.; MacFarland, S.P.; Brodeur, G.M.; et al. DICER1 RNase IIIb domain mutations trigger widespread miRNA dysregulation and MAPK activation in pediatric thyroid cancer. Front. Endocrinol. 2023, 14, 1083382. [Google Scholar] [CrossRef] [PubMed]
  63. Di, W.; Li, Q.; Shen, W.; Guo, H.; Zhao, S. The long non-coding RNA HOTAIR promotes thyroid cancer cell growth, invasion and migration through the miR-1-CCND2 axis. Am. J. Cancer Res. 2017, 7, 1298–1309. [Google Scholar] [PubMed]
  64. Leone, V.; D’Angelo, D.; Rubio, I.; de Freitas, P.M.; Federico, A.; Colamaio, M.; Pallante, P.; Medeiros-Neto, G.; Fusco, A. MiR-1 is a tumor suppressor in thyroid carcinogenesis targeting CCND2, CXCR4, and SDF-1alpha. J. Clin. Endocrinol. Metab. 2011, 96, E1388–E1398. [Google Scholar] [CrossRef] [PubMed]
  65. Yip, L.; Kelly, L.; Shuai, Y.; Armstrong, M.J.; Nikiforov, Y.E.; Carty, S.E.; Nikiforova, M.N. MicroRNA signature distinguishes the degree of aggressiveness of papillary thyroid carcinoma. Ann. Surg. Oncol. 2011, 18, 2035–2041. [Google Scholar] [CrossRef] [PubMed]
  66. Han, J.Y.; Guo, S.; Wei, N.; Xue, R.; Li, W.; Dong, G.; Li, J.; Tian, X.; Chen, C.; Qiu, S.; et al. ciRS-7 Promotes the Proliferation and Migration of Papillary Thyroid Cancer by Negatively Regulating the miR-7/Epidermal Growth Factor Receptor Axis. Biomed Res. Int. 2020, 2020, 9875636. [Google Scholar] [CrossRef]
  67. Macerola, E.; Poma, A.M.; Proietti, A.; Rago, T.; Romani, R.; Vignali, P.; Ugolini, C.; Torregrossa, L.; Basolo, A.; Santini, F.; et al. Down-regulation of miR-7-5p and miR-548ar-5p predicts malignancy in indeterminate thyroid nodules negative for BRAF and RAS mutations. Endocrine 2022, 76, 677–686. [Google Scholar] [CrossRef] [PubMed]
  68. Guo, Y.; Zheng, H.; Yin, J.; Wang, H. Rs4911154 of circ-ITCH aggravated tumor malignancy of thyroid nodules via the circ-ITCH/miR-22-3p/CBL axis. Sci. Rep. 2021, 11, 18491. [Google Scholar] [CrossRef]
  69. Braun, J.; Hoang-Vu, C.; Dralle, H.; Huttelmaier, S. Downregulation of microRNAs directs the EMT and invasive potential of anaplastic thyroid carcinomas. Oncogene 2010, 29, 4237–4244. [Google Scholar] [CrossRef]
  70. Chen, C.; Zhou, L.; Wang, H.; Chen, J.; Li, W.; Liu, W.; Shen, M.; Liu, H.; Fu, X. Long noncoding RNA CNALPTC1 promotes cell proliferation and migration of papillary thyroid cancer via sponging miR-30 family. Am. J. Cancer Res. 2018, 8, 192–206. [Google Scholar]
  71. Guan, H.; Liang, W.; Xie, Z.; Li, H.; Liu, J.; Liu, L.; Xiu, L.; Li, Y. Down-regulation of miR-144 promotes thyroid cancer cell invasion by targeting ZEB1 and ZEB2. Endocrine 2015, 48, 566–574. [Google Scholar] [CrossRef]
  72. Liu, J.; Feng, L.; Zhang, H.; Zhang, J.; Zhang, Y.; Li, S.; Qin, L.; Yang, Z.; Xiong, J. Effects of miR-144 on the sensitivity of human anaplastic thyroid carcinoma cells to cisplatin by autophagy regulation. Cancer Biol. Ther. 2018, 19, 484–496. [Google Scholar] [CrossRef] [PubMed]
  73. Sun, J.; Shi, R.; Zhao, S.; Li, X.; Lu, S.; Bu, H.; Ma, X.; Su, C. E2F8, a direct target of miR-144, promotes papillary thyroid cancer progression via regulating cell cycle. J. Exp. Clin. Cancer Res. 2017, 36, 40. [Google Scholar] [CrossRef]
  74. Liu, W.; Zhao, J.; Jin, M.; Zhou, M. circRAPGEF5 Contributes to Papillary Thyroid Proliferation and Metastatis by Regulation miR-198/FGFR1. Mol. Ther. Nucleic Acids 2019, 14, 609–616. [Google Scholar] [CrossRef] [PubMed]
  75. Li, G.; Kong, Q. LncRNA LINC00460 promotes the papillary thyroid cancer progression by regulating the LINC00460/miR-485-5p/Raf1 axis. Biol. Res. 2019, 52, 61. [Google Scholar] [CrossRef]
  76. Xia, F.; Xia, W.; Yu, X. LncRNA HOTAIR Influences the Growth, Migration, and Invasion of Papillary Thyroid Carcinoma via Affection on the miR-488-5p/NUP205 Axis. Technol. Cancer Res. Treat. 2020, 19, 1533033820962125. [Google Scholar] [CrossRef] [PubMed]
  77. Feng, L.; Yang, B.; Tang, X.D. Long noncoding RNA LINC00460 promotes carcinogenesis via sponging miR-613 in papillary thyroid carcinoma. J. Cell Physiol. 2019, 234, 11431–11439. [Google Scholar] [CrossRef]
  78. Zou, X.; Guo, Z.H.; Li, Q.; Wang, P.S. Long Noncoding RNA LINC00460 Modulates MMP-9 to Promote Cell Proliferation, Invasion and Apoptosis by Targeting miR-539 in Papillary Thyroid Cancer. Cancer Manag. Res. 2020, 12, 199–207. [Google Scholar] [CrossRef]
  79. Guo, R.; Ning, Y.; Ma, Y.; Lin, Q.; Shen, N.; Shi, P. Long non-coding RNA HOTAIR/microRNA-761 sponge regulates PPME1 and further influences cell biological functions in thyroid carcinoma. Laryngoscope Investig. Otolaryngol. 2021, 6, 438–445. [Google Scholar] [CrossRef]
  80. Jiao, D.; Guo, F.; Fu, Q. MicroRNA-873 inhibits the progression of thyroid cancer by directly targeting ZEB1. Mol. Med. Rep. 2019, 20, 1986–1993. [Google Scholar] [CrossRef]
  81. Tan, X.; Zhao, J.; Lou, J.; Zheng, W.; Wang, P. Hsa_circ_0058129 regulates papillary thyroid cancer development via miR-873-5p/follistatin-like 1 axis. J. Clin. Lab. Anal. 2022, 36, e24401. [Google Scholar] [CrossRef]
  82. Wang, Z.; Liu, W.; Wang, C.; Ai, Z. miR-873-5p Inhibits Cell Migration and Invasion of Papillary Thyroid Cancer via Regulation of CXCL16. Onco Targets Ther. 2020, 13, 1037–1046. [Google Scholar] [CrossRef] [PubMed]
  83. Zhang, W.; Zhang, H.; Zhao, X. circ_0005273 promotes thyroid carcinoma progression by SOX2 expression. Endocr. Relat. Cancer 2020, 27, 11–21. [Google Scholar] [CrossRef] [PubMed]
  84. Ferrer, J.; Dimitrova, N. Transcription regulation by long non-coding RNAs: Mechanisms and disease relevance. Nat. Rev. Mol. Cell Biol. 2024, 25, 396–415. [Google Scholar] [CrossRef] [PubMed]
  85. Gloss, B.S.; Dinger, M.E. The specificity of long noncoding RNA expression. Biochim. Biophys. Acta 2016, 1859, 16–22. [Google Scholar] [CrossRef] [PubMed]
  86. Lu, W.; Xu, Y.; Xu, J.; Wang, Z.; Ye, G. Identification of differential expressed lncRNAs in human thyroid cancer by a genome-wide analyses. Cancer Med. 2018, 7, 3935–3944. [Google Scholar] [CrossRef] [PubMed]
  87. Malakoti, F.; Targhazeh, N.; Karimzadeh, H.; Mohammadi, E.; Asadi, M.; Asemi, Z.; Alemi, F. Multiple function of lncRNA MALAT1 in cancer occurrence and progression. Chem. Biol. Drug Des. 2023, 101, 1113–1137. [Google Scholar] [CrossRef] [PubMed]
  88. Jendrzejewski, J.; He, H.; Radomska, H.S.; Li, W.; Tomsic, J.; Liyanarachchi, S.; Davuluri, R.V.; Nagy, R.; de la Chapelle, A. The polymorphism rs944289 predisposes to papillary thyroid carcinoma through a large intergenic noncoding RNA gene of tumor suppressor type. Proc. Natl. Acad. Sci. USA 2012, 109, 8646–8651. [Google Scholar] [CrossRef] [PubMed]
  89. Rinn, J.L.; Kertesz, M.; Wang, J.K.; Squazzo, S.L.; Xu, X.; Brugmann, S.A.; Goodnough, L.H.; Helms, J.A.; Farnham, P.J.; Segal, E.; et al. Functional demarcation of active and silent chromatin domains in human HOX loci by noncoding RNAs. Cell 2007, 129, 1311–1323. [Google Scholar] [CrossRef]
  90. Islam, F.; Zhou, Y.; Lam, A.K. Long Non-Coding RNAs Profiling Using Microarray in Papillary Thyroid Carcinoma. Methods Mol. Biol. 2022, 2534, 135–148. [Google Scholar] [CrossRef]
  91. Chen, X.; Jin, J.; Zheng, L.; Sheng, Y.; Sun, J. Correlations of HOTAIR expression with pathological stage, CT characteristics and prognosis of patients with papillary thyroid carcinoma. J. BUON 2021, 26, 259–265. [Google Scholar]
  92. Arratia, F.; Fierro, C.; Blanco, A.; Fuentes, S.; Nahuelquen, D.; Montecino, M.; Rojas, A.; Aguilar, R. Selective Concurrence of the Long Non-Coding RNA MALAT1 and the Polycomb Repressive Complex 2 to Promoter Regions of Active Genes in MCF7 Breast Cancer Cells. Curr. Issues Mol. Biol. 2023, 45, 4735–4748. [Google Scholar] [CrossRef] [PubMed]
  93. West, J.A.; Davis, C.P.; Sunwoo, H.; Simon, M.D.; Sadreyev, R.I.; Wang, P.I.; Tolstorukov, M.Y.; Kingston, R.E. The long noncoding RNAs NEAT1 and MALAT1 bind active chromatin sites. Mol. Cell 2014, 55, 791–802. [Google Scholar] [CrossRef] [PubMed]
  94. Fu, S.; Wang, Y.; Li, H.; Chen, L.; Liu, Q. Regulatory Networks of LncRNA MALAT-1 in Cancer. Cancer Manag. Res. 2020, 12, 10181–10198. [Google Scholar] [CrossRef]
  95. Gou, L.; Zou, H.; Li, B. Long noncoding RNA MALAT1 knockdown inhibits progression of anaplastic thyroid carcinoma by regulating miR-200a-3p/FOXA1. Cancer Biol. Ther. 2019, 20, 1355–1365. [Google Scholar] [CrossRef] [PubMed]
  96. Ghasemian, M.; Poodineh, J. A review on the biological roles of LncRNA PTCSC3 in cancerous and non-cancerous disorders. Cancer Cell Int. 2023, 23, 184. [Google Scholar] [CrossRef]
  97. Fan, M.; Li, X.; Jiang, W.; Huang, Y.; Li, J.; Wang, Z. A long non-coding RNA, PTCSC3, as a tumor suppressor and a target of miRNAs in thyroid cancer cells. Exp. Ther. Med. 2013, 5, 1143–1146. [Google Scholar] [CrossRef]
  98. Liu, Y.; Zhang, H.; Wang, H.; Du, J.; Dong, P.; Liu, M.; Lin, Y. Long non-coding RNA DUXAP8 promotes the cell proliferation, migration, and invasion of papillary thyroid carcinoma via miR-223-3p mediated regulation of CXCR4. Bioengineered 2021, 12, 496–506. [Google Scholar] [CrossRef]
  99. Pang, R.; Yang, S. lncRNA DUXAP8 inhibits papillary thyroid carcinoma cell apoptosis via sponging the miR-20b-5p/SOS1 axis. Oncol. Rep. 2021, 45, 64. [Google Scholar] [CrossRef]
  100. Zhou, Q.; Feng, J.; Yin, S.; Ma, S.; Wang, J.; Yi, H. LncRNA FAM230B promotes the metastasis of papillary thyroid cancer by sponging the miR-378a-3p/WNT5A axis. Biochem. Biophys. Res. Commun. 2021, 546, 83–89. [Google Scholar] [CrossRef]
  101. Liao, G.; Huang, Z.; Gan, T.; Wu, C.; Wang, X.; Li, D. Long non-coding RNA nuclear enriched abundant transcript 1 (NEAT1) modulates inhibitor of DNA binding 1 (ID1) to facilitate papillary thyroid carcinoma development by sponging microRNA-524-5p. Bioengineered 2022, 13, 13201–13212. [Google Scholar] [CrossRef]
  102. Sun, W.; Wang, Z.; Qin, Y.; Ji, X.; Huang, J.; Zhang, F.; Wang, Z.; Dong, W.; He, L.; Zhang, H. NEAT1_2/RRAD/EHF Positive Feedback Loop Facilitates Aerobic Glycolysis in Papillary Thyroid Cancer Cells. Endocrinology 2023, 164, bqad085. [Google Scholar] [CrossRef] [PubMed]
  103. Zhou, Y.; Liu, S.; Luo, Y.; Zhang, M.; Jiang, X.; Xiong, Y. IncRNA MAPKAPK5-AS1 promotes proliferation and migration of thyroid cancer cell lines by targeting miR-519e-5p/YWHAH. Eur. J. Histochem. 2020, 64, 3177. [Google Scholar] [CrossRef] [PubMed]
  104. Zhang, X.; Li, D.; Jia, C.; Cai, H.; Lv, Z.; Wu, B. METTL14 promotes tumorigenesis by regulating lncRNA OIP5-AS1/miR-98/ADAMTS8 signaling in papillary thyroid cancer. Cell Death Dis. 2021, 12, 617. [Google Scholar] [CrossRef] [PubMed]
  105. Cai, T.; He, Y.; Peng, B. IncRNA XIST Stimulates Papillary Thyroid Cancer Development through the miR-330-3p/PDE5A Axis. Crit. Rev. Eukaryot. Gene Expr. 2023, 33, 13–26. [Google Scholar] [CrossRef] [PubMed]
  106. Liu, H.; Deng, H.; Zhao, Y.; Li, C.; Liang, Y. LncRNA XIST/miR-34a axis modulates the cell proliferation and tumor growth of thyroid cancer through MET-PI3K-AKT signaling. J. Exp. Clin. Cancer Res. 2018, 37, 279. [Google Scholar] [CrossRef] [PubMed]
  107. Pisignano, G.; Michael, D.C.; Visal, T.H.; Pirlog, R.; Ladomery, M.; Calin, G.A. Going circular: History, present, and future of circRNAs in cancer. Oncogene 2023, 42, 2783–2800. [Google Scholar] [CrossRef] [PubMed]
  108. Chen, Y.G.; Hur, S. Cellular origins of dsRNA, their recognition and consequences. Nat. Rev. Mol. Cell Biol. 2022, 23, 286–301. [Google Scholar] [CrossRef]
  109. Sadeq, S.; Al-Hashimi, S.; Cusack, C.M.; Werner, A. Endogenous Double-Stranded RNA. Noncoding RNA 2021, 7, 15. [Google Scholar] [CrossRef] [PubMed]
  110. Chen, D.S.; Mellman, I. Elements of cancer immunity and the cancer-immune set point. Nature 2017, 541, 321–330. [Google Scholar] [CrossRef]
  111. Brillo, V.; Chieregato, L.; Leanza, L.; Muccioli, S.; Costa, R. Mitochondrial Dynamics, ROS, and Cell Signaling: A Blended Overview. Life 2021, 11, 332. [Google Scholar] [CrossRef]
  112. Elorza, A.A.; Soffia, J.P. mtDNA Heteroplasmy at the Core of Aging-Associated Heart Failure. An Integrative View of OXPHOS and Mitochondrial Life Cycle in Cardiac Mitochondrial Physiology. Front. Cell Dev. Biol. 2021, 9, 625020. [Google Scholar] [CrossRef] [PubMed]
  113. Chocron, E.S.; Munkacsy, E.; Pickering, A.M. Cause or casualty: The role of mitochondrial DNA in aging and age-associated disease. Biochim. Biophys. Acta Mol. Basis Dis. 2019, 1865, 285–297. [Google Scholar] [CrossRef] [PubMed]
  114. Picard, M.; Wallace, D.C.; Burelle, Y. The rise of mitochondria in medicine. Mitochondrion 2016, 30, 105–116. [Google Scholar] [CrossRef] [PubMed]
  115. Soltys, D.T.; Pereira, C.P.M.; Rowies, F.T.; Farfel, J.M.; Grinberg, L.T.; Suemoto, C.K.; Leite, R.E.P.; Rodriguez, R.D.; Ericson, N.G.; Bielas, J.H.; et al. Lower mitochondrial DNA content but not increased mutagenesis associates with decreased base excision repair activity in brains of AD subjects. Neurobiol. Aging 2019, 73, 161–170. [Google Scholar] [CrossRef]
  116. Lakshmanan, L.N.; Yee, Z.; Ng, L.F.; Gunawan, R.; Halliwell, B.; Gruber, J. Clonal expansion of mitochondrial DNA deletions is a private mechanism of aging in long-lived animals. Aging Cell 2018, 17, e12814. [Google Scholar] [CrossRef] [PubMed]
  117. Nissanka, N.; Minczuk, M.; Moraes, C.T. Mechanisms of Mitochondrial DNA Deletion Formation. Trends Genet. 2019, 35, 235–244. [Google Scholar] [CrossRef] [PubMed]
  118. Busch, K.B.; Kowald, A.; Spelbrink, J.N. Quality matters: How does mitochondrial network dynamics and quality control impact on mtDNA integrity? Philos. Trans. R. Soc. Lond. B Biol. Sci. 2014, 369, 20130442. [Google Scholar] [CrossRef] [PubMed]
  119. Trifunovic, A.; Wredenberg, A.; Falkenberg, M.; Spelbrink, J.N.; Rovio, A.T.; Bruder, C.E.; Bohlooly, Y.M.; Gidlof, S.; Oldfors, A.; Wibom, R.; et al. Premature ageing in mice expressing defective mitochondrial DNA polymerase. Nature 2004, 429, 417–423. [Google Scholar] [CrossRef]
  120. Medeiros, T.C.; Graef, M. Autophagy determines mtDNA copy number dynamics during starvation. Autophagy 2019, 15, 178–179. [Google Scholar] [CrossRef]
  121. Medeiros, T.C.; Thomas, R.L.; Ghillebert, R.; Graef, M. Autophagy balances mtDNA synthesis and degradation by DNA polymerase POLG during starvation. J. Cell Biol. 2018, 217, 1601–1611. [Google Scholar] [CrossRef]
  122. Siegmund, S.E.; Yang, H.; Sharma, R.; Javors, M.; Skinner, O.; Mootha, V.; Hirano, M.; Schon, E.A. Low-dose rapamycin extends lifespan in a mouse model of mtDNA depletion syndrome. Hum. Mol. Genet. 2017, 26, 4588–4605. [Google Scholar] [CrossRef]
  123. Ahlqvist, K.J.; Leoncini, S.; Pecorelli, A.; Wortmann, S.B.; Ahola, S.; Forsstrom, S.; Guerranti, R.; De Felice, C.; Smeitink, J.; Ciccoli, L.; et al. MtDNA mutagenesis impairs elimination of mitochondria during erythroid maturation leading to enhanced erythrocyte destruction. Nat. Commun. 2015, 6, 6494. [Google Scholar] [CrossRef]
  124. Hosgood, H.D., 3rd; Liu, C.S.; Rothman, N.; Weinstein, S.J.; Bonner, M.R.; Shen, M.; Lim, U.; Virtamo, J.; Cheng, W.L.; Albanes, D.; et al. Mitochondrial DNA copy number and lung cancer risk in a prospective cohort study. Carcinogenesis 2010, 31, 847–849. [Google Scholar] [CrossRef] [PubMed]
  125. Thyagarajan, B.; Wang, R.; Nelson, H.; Barcelo, H.; Koh, W.P.; Yuan, J.M. Mitochondrial DNA copy number is associated with breast cancer risk. PLoS ONE 2013, 8, e65968. [Google Scholar] [CrossRef] [PubMed]
  126. Yamada, S.; Nomoto, S.; Fujii, T.; Kaneko, T.; Takeda, S.; Inoue, S.; Kanazumi, N.; Nakao, A. Correlation between copy number of mitochondrial DNA and clinico-pathologic parameters of hepatocellular carcinoma. Eur. J. Surg. Oncol. 2006, 32, 303–307. [Google Scholar] [CrossRef]
  127. Liao, L.M.; Baccarelli, A.; Shu, X.O.; Gao, Y.T.; Ji, B.T.; Yang, G.; Li, H.L.; Hoxha, M.; Dioni, L.; Rothman, N.; et al. Mitochondrial DNA copy number and risk of gastric cancer: A report from the Shanghai Women’s Health Study. Cancer Epidemiol. Biomarkers Prev. 2011, 20, 1944–1949. [Google Scholar] [CrossRef] [PubMed]
  128. Sun, X.; Zhan, L.; Chen, Y.; Wang, G.; He, L.; Wang, Q.; Zhou, F.; Yang, F.; Wu, J.; Wu, Y.; et al. Increased mtDNA copy number promotes cancer progression by enhancing mitochondrial oxidative phosphorylation in microsatellite-stable colorectal cancer. Signal Transduct. Target. Ther. 2018, 3, 8. [Google Scholar] [CrossRef]
  129. Mambo, E.; Chatterjee, A.; Xing, M.; Tallini, G.; Haugen, B.R.; Yeung, S.C.; Sukumar, S.; Sidransky, D. Tumor-specific changes in mtDNA content in human cancer. Int. J. Cancer 2005, 116, 920–924. [Google Scholar] [CrossRef]
  130. Tallini, G. Oncocytic tumours. Virchows Arch. 1998, 433, 5–12. [Google Scholar] [CrossRef]
  131. Maximo, V.; Soares, P.; Lima, J.; Cameselle-Teijeiro, J.; Sobrinho-Simoes, M. Mitochondrial DNA somatic mutations (point mutations and large deletions) and mitochondrial DNA variants in human thyroid pathology: A study with emphasis on Hurthle cell tumors. Am. J. Pathol. 2002, 160, 1857–1865. [Google Scholar] [CrossRef]
  132. Gasparre, G.; Porcelli, A.M.; Bonora, E.; Pennisi, L.F.; Toller, M.; Iommarini, L.; Ghelli, A.; Moretti, M.; Betts, C.M.; Martinelli, G.N.; et al. Disruptive mitochondrial DNA mutations in complex I subunits are markers of oncocytic phenotype in thyroid tumors. Proc. Natl. Acad. Sci. USA 2007, 104, 9001–9006. [Google Scholar] [CrossRef]
  133. Gopal, R.K.; Kubler, K.; Calvo, S.E.; Polak, P.; Livitz, D.; Rosebrock, D.; Sadow, P.M.; Campbell, B.; Donovan, S.E.; Amin, S.; et al. Widespread Chromosomal Losses and Mitochondrial DNA Alterations as Genetic Drivers in Hurthle Cell Carcinoma. Cancer Cell 2018, 34, 242–255.e5. [Google Scholar] [CrossRef]
  134. Lyu, L.; Wang, Q.; Song, S.; Li, L.; Zhou, H.; Li, M.; Jiang, Z.; Zhou, C.; Chen, G.; Lyu, J.; et al. Oncocytic tumors are marked by enhanced mitochondrial content and mtDNA mutations of complex I in Chinese patients. Mitochondrion 2019, 45, 1–6. [Google Scholar] [CrossRef]
  135. Tsybrovskyy, O.; De Luise, M.; de Biase, D.; Caporali, L.; Fiorini, C.; Gasparre, G.; Carelli, V.; Hackl, D.; Imamovic, L.; Haim, S.; et al. Papillary thyroid carcinoma tall cell variant shares accumulation of mitochondria, mitochondrial DNA mutations, and loss of oxidative phosphorylation complex I integrity with oncocytic tumors. J. Pathol. Clin. Res. 2022, 8, 155–168. [Google Scholar] [CrossRef] [PubMed]
  136. Rogounovitch, T.; Saenko, V.; Yamashita, S. Mitochondrial DNA and human thyroid diseases. Endocr. J. 2004, 51, 265–277. [Google Scholar] [CrossRef] [PubMed]
  137. Sasarman, F.; Antonicka, H.; Shoubridge, E.A. The A3243G tRNALeu(UUR) MELAS mutation causes amino acid misincorporation and a combined respiratory chain assembly defect partially suppressed by overexpression of EFTu and EFG2. Hum. Mol. Genet. 2008, 17, 3697–3707. [Google Scholar] [CrossRef] [PubMed]
  138. Akouchekian, M.; Houshmand, M.; Akbari, M.H.; Kamalidehghan, B.; Dehghan, M. Analysis of mitochondrial ND1 gene in human colorectal cancer. J. Res. Med. Sci. 2011, 16, 50–55. [Google Scholar] [PubMed]
  139. Lin, Y.H.; Lim, S.N.; Chen, C.Y.; Chi, H.C.; Yeh, C.T.; Lin, W.R. Functional Role of Mitochondrial DNA in Cancer Progression. Int. J. Mol. Sci. 2022, 23, 1659. [Google Scholar] [CrossRef]
  140. Yin, P.H.; Wu, C.C.; Lin, J.C.; Chi, C.W.; Wei, Y.H.; Lee, H.C. Somatic mutations of mitochondrial genome in hepatocellular carcinoma. Mitochondrion 2010, 10, 174–182. [Google Scholar] [CrossRef] [PubMed]
  141. Fang, H.; Shen, L.; Chen, T.; He, J.; Ding, Z.; Wei, J.; Qu, J.; Chen, G.; Lu, J.; Bai, Y. Cancer type-specific modulation of mitochondrial haplogroups in breast, colorectal and thyroid cancer. BMC Cancer 2010, 10, 421. [Google Scholar] [CrossRef] [PubMed]
  142. Dabravolski, S.A.; Nikiforov, N.G.; Zhuravlev, A.D.; Orekhov, N.A.; Mikhaleva, L.M.; Orekhov, A.N. The Role of Altered Mitochondrial Metabolism in Thyroid Cancer Development and Mitochondria-Targeted Thyroid Cancer Treatment. Int. J. Mol. Sci. 2021, 23, 460. [Google Scholar] [CrossRef] [PubMed]
  143. Su, X.; Wang, W.; Ruan, G.; Liang, M.; Zheng, J.; Chen, Y.; Wu, H.; Fahey, T.J.; Guan, M.; Teng, L. A Comprehensive Characterization of Mitochondrial Genome in Papillary Thyroid Cancer. Int. J. Mol. Sci. 2016, 17, 1594. [Google Scholar] [CrossRef]
  144. Bulgakova, O.; Kussainova, A.; Kakabayev, A.; Aripova, A.; Baikenova, G.; Izzotti, A.; Bersimbaev, R. The level of free-circulating mtDNA in patients with radon-induced lung cancer. Environ. Res. 2022, 207, 112215. [Google Scholar] [CrossRef] [PubMed]
  145. Li, L.; Hann, H.W.; Wan, S.; Hann, R.S.; Wang, C.; Lai, Y.; Ye, X.; Evans, A.; Myers, R.E.; Ye, Z.; et al. Cell-free circulating mitochondrial DNA content and risk of hepatocellular carcinoma in patients with chronic HBV infection. Sci. Rep. 2016, 6, 23992. [Google Scholar] [CrossRef] [PubMed]
  146. Meddeb, R.; Dache, Z.A.A.; Thezenas, S.; Otandault, A.; Tanos, R.; Pastor, B.; Sanchez, C.; Azzi, J.; Tousch, G.; Azan, S.; et al. Quantifying circulating cell-free DNA in humans. Sci. Rep. 2019, 9, 5220. [Google Scholar] [CrossRef] [PubMed]
  147. Perdas, E.; Stawski, R.; Kaczka, K.; Nowak, D.; Zubrzycka, M. Altered levels of circulating nuclear and mitochondrial DNA in patients with Papillary Thyroid Cancer. Sci. Rep. 2019, 9, 14438. [Google Scholar] [CrossRef] [PubMed]
  148. Ellinger, J.; Albers, P.; Muller, S.C.; von Ruecker, A.; Bastian, P.J. Circulating mitochondrial DNA in the serum of patients with testicular germ cell cancer as a novel noninvasive diagnostic biomarker. BJU Int. 2009, 104, 48–52. [Google Scholar] [CrossRef] [PubMed]
  149. Seefeld, M.; El Tarhouny, S.; Fan, A.X.; Hahn, S.; Holzgreve, W.; Zhong, X.Y. Parallel assessment of circulatory cell-free DNA by PCR and nucleosomes by ELISA in breast tumors. Int. J. Biol. Markers 2008, 23, 69–73. [Google Scholar] [CrossRef] [PubMed]
  150. Reznik, E.; Miller, M.L.; Senbabaoglu, Y.; Riaz, N.; Sarungbam, J.; Tickoo, S.K.; Al-Ahmadie, H.A.; Lee, W.; Seshan, V.E.; Hakimi, A.A.; et al. Mitochondrial DNA copy number variation across human cancers. eLife 2016, 5, e10769. [Google Scholar] [CrossRef]
  151. Vara-Perez, M.; Felipe-Abrio, B.; Agostinis, P. Mitophagy in Cancer: A Tale of Adaptation. Cells 2019, 8, 493. [Google Scholar] [CrossRef]
  152. Bassnett, S. Lens organelle degradation. Exp. Eye Res. 2002, 74, 1–6. [Google Scholar] [CrossRef] [PubMed]
  153. Pua, H.H.; Guo, J.; Komatsu, M.; He, Y.W. Autophagy is essential for mitochondrial clearance in mature T lymphocytes. J. Immunol. 2009, 182, 4046–4055. [Google Scholar] [CrossRef] [PubMed]
  154. Pei, S.; Minhajuddin, M.; Adane, B.; Khan, N.; Stevens, B.M.; Mack, S.C.; Lai, S.; Rich, J.N.; Inguva, A.; Shannon, K.M.; et al. AMPK/FIS1-Mediated Mitophagy Is Required for Self-Renewal of Human AML Stem Cells. Cell Stem Cell 2018, 23, 86–100.e106. [Google Scholar] [CrossRef] [PubMed]
  155. Kimmelman, A.C.; White, E. Autophagy and Tumor Metabolism. Cell Metab. 2017, 25, 1037–1043. [Google Scholar] [CrossRef] [PubMed]
  156. Wang, M.; Luan, S.; Fan, X.; Wang, J.; Huang, J.; Gao, X.; Han, D. The emerging multifaceted role of PINK1 in cancer biology. Cancer Sci. 2022, 113, 4037–4047. [Google Scholar] [CrossRef] [PubMed]
  157. Hou, H.; Er, P.; Cheng, J.; Chen, X.; Ding, X.; Wang, Y.; Chen, X.; Yuan, Z.; Pang, Q.; Wang, P.; et al. High expression of FUNDC1 predicts poor prognostic outcomes and is a promising target to improve chemoradiotherapy effects in patients with cervical cancer. Cancer Med. 2017, 6, 1871–1881. [Google Scholar] [CrossRef]
  158. Li, J.; Agarwal, E.; Bertolini, I.; Seo, J.H.; Caino, M.C.; Ghosh, J.C.; Kossenkov, A.V.; Liu, Q.; Tang, H.Y.; Goldman, A.R.; et al. The mitophagy effector FUNDC1 controls mitochondrial reprogramming and cellular plasticity in cancer cells. Sci. Signal 2020, 13, eaaz8240. [Google Scholar] [CrossRef]
  159. Yuan, Q.; Sun, N.; Zheng, J.; Wang, Y.; Yan, X.; Mai, W.; Liao, Y.; Chen, X. Prognostic and Immunological Role of FUN14 Domain Containing 1 in Pan-Cancer: Friend or Foe? Front. Oncol. 2019, 9, 1502. [Google Scholar] [CrossRef] [PubMed]
  160. Humpton, T.J.; Alagesan, B.; DeNicola, G.M.; Lu, D.; Yordanov, G.N.; Leonhardt, C.S.; Yao, M.A.; Alagesan, P.; Zaatari, M.N.; Park, Y.; et al. Oncogenic KRAS Induces NIX-Mediated Mitophagy to Promote Pancreatic Cancer. Cancer Discov. 2019, 9, 1268–1287. [Google Scholar] [CrossRef]
  161. Fei, P.; Wang, W.; Kim, S.H.; Wang, S.; Burns, T.F.; Sax, J.K.; Buzzai, M.; Dicker, D.T.; McKenna, W.G.; Bernhard, E.J.; et al. Bnip3L is induced by p53 under hypoxia, and its knockdown promotes tumor growth. Cancer Cell 2004, 6, 597–609. [Google Scholar] [CrossRef]
  162. Burton, T.R.; Gibson, S.B. The role of Bcl-2 family member BNIP3 in cell death and disease: NIPping at the heels of cell death. Cell Death Differ. 2009, 16, 515–523. [Google Scholar] [CrossRef] [PubMed]
  163. Giatromanolaki, A.; Koukourakis, M.I.; Sowter, H.M.; Sivridis, E.; Gibson, S.; Gatter, K.C.; Harris, A.L. BNIP3 expression is linked with hypoxia-regulated protein expression and with poor prognosis in non-small cell lung cancer. Clin. Cancer Res. 2004, 10, 5566–5571. [Google Scholar] [CrossRef]
  164. Sowter, H.M.; Ferguson, M.; Pym, C.; Watson, P.; Fox, S.B.; Han, C.; Harris, A.L. Expression of the cell death genes BNip3 and NIX in ductal carcinoma in situ of the breast; correlation of BNip3 levels with necrosis and grade. J. Pathol. 2003, 201, 573–580. [Google Scholar] [CrossRef]
  165. Chourasia, A.H.; Tracy, K.; Frankenberger, C.; Boland, M.L.; Sharifi, M.N.; Drake, L.E.; Sachleben, J.R.; Asara, J.M.; Locasale, J.W.; Karczmar, G.S.; et al. Mitophagy defects arising from BNip3 loss promote mammary tumor progression to metastasis. EMBO Rep. 2015, 16, 1145–1163. [Google Scholar] [CrossRef]
  166. Erkan, M.; Kleeff, J.; Esposito, I.; Giese, T.; Ketterer, K.; Buchler, M.W.; Giese, N.A.; Friess, H. Loss of BNIP3 expression is a late event in pancreatic cancer contributing to chemoresistance and worsened prognosis. Oncogene 2005, 24, 4421–4432. [Google Scholar] [CrossRef] [PubMed]
  167. Li, X.; Lyu, Y.; Li, J.; Wang, X. AMBRA1 and its role as a target for anticancer therapy. Front. Oncol. 2022, 12, 946086. [Google Scholar] [CrossRef]
  168. Lv, S.; Zhao, X.; Zhang, E.; Yan, Y.; Ma, X.; Li, N.; Zou, Q.; Sun, L.; Song, T. Lysine demethylase KDM1A promotes cell growth via FKBP8-BCL2 axis in hepatocellular carcinoma. J. Biol. Chem. 2022, 298, 102374. [Google Scholar] [CrossRef]
  169. Fong, S.; Mounkes, L.; Liu, Y.; Maibaum, M.; Alonzo, E.; Desprez, P.Y.; Thor, A.D.; Kashani-Sabet, M.; Debs, R.J. Functional identification of distinct sets of antitumor activities mediated by the FKBP gene family. Proc. Natl. Acad. Sci. USA 2003, 100, 14253–14258. [Google Scholar] [CrossRef] [PubMed]
  170. Singh, K.; Roy, M.; Prajapati, P.; Lipatova, A.; Sripada, L.; Gohel, D.; Singh, A.; Mane, M.; Godbole, M.M.; Chumakov, P.M.; et al. NLRX1 regulates TNF-alpha-induced mitochondria-lysosomal crosstalk to maintain the invasive and metastatic potential of breast cancer cells. Biochim. Biophys. Acta Mol. Basis Dis. 2019, 1865, 1460–1476. [Google Scholar] [CrossRef]
  171. Nagai-Singer, M.A.; Morrison, H.A.; Woolls, M.K.; Leedy, K.; Imran, K.M.; Tupik, J.D.; Allen, I.C. NLRX1 functions as a tumor suppressor in Pan02 pancreatic cancer cells. Front. Oncol. 2023, 13, 1155831. [Google Scholar] [CrossRef]
  172. Zhang, H.; Yin, C.; Liu, X.; Bai, X.; Wang, L.; Xu, H.; Ju, J.; Zhang, L. Prohibitin 2/PHB2 in Parkin-Mediated Mitophagy: A Potential Therapeutic Target for Non-Small Cell Lung Carcinoma. Med. Sci. Monit. 2020, 26, e923227. [Google Scholar] [CrossRef] [PubMed]
  173. Nunez-Vazquez, S.; Saura-Esteller, J.; Sanchez-Vera, I.; Guilbaud, E.; Cosialls, A.M.; Pons, G.; Ricci, J.E.; Iglesias-Serret, D.; Marchetti, S.; Gil, J. The prohibitin-binding compound fluorizoline inhibits mitophagy in cancer cells. Oncogenesis 2021, 10, 64. [Google Scholar] [CrossRef] [PubMed]
  174. Nissou, M.F.; El Atifi, M.; Guttin, A.; Godfraind, C.; Salon, C.; Garcion, E.; van der Sanden, B.; Issartel, J.P.; Berger, F.; Wion, D. Hypoxia-induced expression of VE-cadherin and filamin B in glioma cell cultures and pseudopalisade structures. J. Neurooncol. 2013, 113, 239–249. [Google Scholar] [CrossRef] [PubMed]
  175. Zhang, J.; Ney, P.A. Role of BNIP3 and NIX in cell death, autophagy, and mitophagy. Cell Death Differ. 2009, 16, 939–946. [Google Scholar] [CrossRef] [PubMed]
  176. Sowter, H.M.; Ratcliffe, P.J.; Watson, P.; Greenberg, A.H.; Harris, A.L. HIF-1-dependent regulation of hypoxic induction of the cell death factors BNIP3 and NIX in human tumors. Cancer Res. 2001, 61, 6669–6673. [Google Scholar] [PubMed]
  177. Guo, K.; Searfoss, G.; Krolikowski, D.; Pagnoni, M.; Franks, C.; Clark, K.; Yu, K.T.; Jaye, M.; Ivashchenko, Y. Hypoxia induces the expression of the pro-apoptotic gene BNIP3. Cell Death Differ. 2001, 8, 367–376. [Google Scholar] [CrossRef] [PubMed]
  178. Sandoval, H.; Thiagarajan, P.; Dasgupta, S.K.; Schumacher, A.; Prchal, J.T.; Chen, M.; Wang, J. Essential role for Nix in autophagic maturation of erythroid cells. Nature 2008, 454, 232–235. [Google Scholar] [CrossRef] [PubMed]
  179. Aerbajinai, W.; Giattina, M.; Lee, Y.T.; Raffeld, M.; Miller, J.L. The proapoptotic factor Nix is coexpressed with Bcl-xL during terminal erythroid differentiation. Blood 2003, 102, 712–717. [Google Scholar] [CrossRef] [PubMed]
  180. Bellot, G.; Garcia-Medina, R.; Gounon, P.; Chiche, J.; Roux, D.; Pouyssegur, J.; Mazure, N.M. Hypoxia-induced autophagy is mediated through hypoxia-inducible factor induction of BNIP3 and BNIP3L via their BH3 domains. Mol. Cell Biol. 2009, 29, 2570–2581. [Google Scholar] [CrossRef]
  181. Daido, S.; Kanzawa, T.; Yamamoto, A.; Takeuchi, H.; Kondo, Y.; Kondo, S. Pivotal role of the cell death factor BNIP3 in ceramide-induced autophagic cell death in malignant glioma cells. Cancer Res. 2004, 64, 4286–4293. [Google Scholar] [CrossRef]
  182. Lee, M.J.; Kim, J.Y.; Suk, K.; Park, J.H. Identification of the hypoxia-inducible factor 1 alpha-responsive HGTD-P gene as a mediator in the mitochondrial apoptotic pathway. Mol. Cell Biol. 2004, 24, 3918–3927. [Google Scholar] [CrossRef] [PubMed]
  183. Toustrup, K.; Sorensen, B.S.; Nordsmark, M.; Busk, M.; Wiuf, C.; Alsner, J.; Overgaard, J. Development of a hypoxia gene expression classifier with predictive impact for hypoxic modification of radiotherapy in head and neck cancer. Cancer Res. 2011, 71, 5923–5931. [Google Scholar] [CrossRef] [PubMed]
  184. Sorensen, B.S.; Knudsen, A.; Wittrup, C.F.; Nielsen, S.; Aggerholm-Pedersen, N.; Busk, M.; Horsman, M.; Hoyer, M.; Bouchelouche, P.N.; Overgaard, J.; et al. The usability of a 15-gene hypoxia classifier as a universal hypoxia profile in various cancer cell types. Radiother. Oncol. 2015, 116, 346–351. [Google Scholar] [CrossRef] [PubMed]
  185. Tang, B.; Qu, Y.; Zhao, F.; Mao, M.; Tang, J.; Li, X.; Ferriero, D.; Mu, D. In vitro effects of hypoxia-inducible factor 1alpha on the biological characteristics of the SiHa uterine cervix cancer cell line. Int. J. Gynecol. Cancer 2009, 19, 898–904. [Google Scholar] [CrossRef] [PubMed]
  186. Cho, Y.E.; Kim, J.Y.; Kim, Y.J.; Kim, Y.W.; Lee, S.; Park, J.H. Expression and clinicopathological significance of human growth and transformation-dependent protein (HGTD-P) in uterine cervical cancer. Histopathology 2010, 57, 479–482. [Google Scholar] [CrossRef] [PubMed]
  187. Basolo, F.; Torregrossa, L.; Giannini, R.; Miccoli, M.; Lupi, C.; Sensi, E.; Berti, P.; Elisei, R.; Vitti, P.; Baggiani, A.; et al. Correlation between the BRAF V600E mutation and tumor invasiveness in papillary thyroid carcinomas smaller than 20 millimeters: Analysis of 1060 cases. J. Clin. Endocrinol. Metab. 2010, 95, 4197–4205. [Google Scholar] [CrossRef]
  188. Xing, M. Genetic-guided Risk Assessment and Management of Thyroid Cancer. Endocrinol. Metab. Clin. N. Am. 2019, 48, 109–124. [Google Scholar] [CrossRef]
Table 1. Definition of epidemiological and clinical terms used in this text.
Table 1. Definition of epidemiological and clinical terms used in this text.
TermDefinition
IncidenceNumber of new cases of disease in a population during a period
SensitivityThe probability that the test result will be positive when disease is present
SpecificityThe probability that the test result will be negative when disease is absent
Positive Predictive Value (PPV)The probability of having the disease with a positive test result
Negative Predictive Value (NPV)Probability of not having the disease, with a negative test
Diagnostic YieldProportion of true positives cases within the study cohort, with a specific test
Table 2. Summary of the main characteristics of genetic classifiers.
Table 2. Summary of the main characteristics of genetic classifiers.
TestType of TestNo. of Genes 1Other BiomarkersClinical ApplicationRef.
Afirma GSC/Afirma XARNA NGS (mRNA Expression) 1115 GenesMutation Hotspots + Fusions + Loss of heterozygosity Afirma GSC is used to evaluate thyroid nodules with indeterminate cytology (Bethesda III/IV) to rule out benignity and thus help avoid unnecessary surgeries in benign cases. Therefore, it has diagnostic applications and can guide definitive treatment. Afirma XA is used for suspicious GSC samples and Bethesda V/VI.[5,22,23]
Thyroseq V3 Targeted DNA and RNA NGS 112 Genes>120 Fusions + 10 Copy Number Alterations + 19 Genes (Expression) Distinguishes between malignant and benign lesions, helping to avoid unnecessary surgery during the diagnostic phase.[24,25]
ThyGeNEXT/ThyraMIR Targeted NGS + miRNA Expression 10 Genes 28 Fusions + 10 miRNAs (Expression) To improve the risk stratification and diagnostic accuracy of indeterminate thyroid nodules. It detects mutations and micro RNAs associated with thyroid cancer and helps in selecting the right treatment. However, what sets it apart from other classifiers is its utilization for long-term monitoring. It is often combined with ThyraMIR, to enhance stratification and predictive value.[5,26,27]
Thyroid Print Quantitative Real-Time PCR (mRNA Expression) 10 Genes N/ADetects changes in gene expression to classify thyroid nodules’ malignancy. It provides additional information for diagnostic evaluation, which may influence treatment planning.[28,29,30]
1 Due to confidentiality policies, only the number of genes available in commercial classifiers is disclosed.
Table 3. miRNAs tested in commercial tests.
Table 3. miRNAs tested in commercial tests.
miRNAThyraMIR® mir-THYpe®Status in Thyroid Cancer
miR-21+ Promoter of cancer. Higher in tumor [53,58]
miR-29+ Suppressor of cancer. Reduced in tumor [53]
miR-31+ Promoter of cancer. Higher in tumor [53,59]
miR-138+ Suppressor of cancer. Reduced in tumor [53,60]
miR-139+ Suppressor of cancer. Reduced in tumor [53]
miR-146++Promoter of cancer. Higher in tumor [53,56,59,61]
miR-152 +Promoter of cancer. Higher in tumor [56,59]
miR-155++Suppressor of cancer. Reduced in tumor [53,56]
miR-181 +Discriminator [56]
miR-200 +Discriminator [56]
miR-204+ Suppressor of cancer. Reduced in tumor [53]
miR-222+ Promoter of cancer. Higher in tumor [53,61]
miR-375+ Promoter of cancer. Higher in tumor [53,58]
miR-551+ Promoter of cancer. Higher in tumor [53,59]
Table 4. Potential miRNAs biomarkers for thyroid cancer.
Table 4. Potential miRNAs biomarkers for thyroid cancer.
miRNAExpression in CancermRNA TargetReference
miR-1 DownregulatedCCND2, CXCR4 and MET[63,64,65]
miR-7 DownregulatedMETTL7B, EGFR[60,66,67]
miR-2UpregulatedCBL and beta-catenin[68]
miR-30DownregulatedBCL9, SNAI1, SMAD2, TGFBR1 and VIM[69,70]
miR-144DownregulatedTGFA, ZEB1, ZEB2, E2F8 and WWTR1[71,72,73]
miR-198DownregulatedFGFR1, E2F8[74]
miR-485DownregulatedRAF1[75]
miR-488DownregulatedNUP205[76]
miR-613DownregulatedSPHK2[77]
miR-539DownregulatedMMP9[78]
miR-761DownregulatedPPME[79]
miR-873DownregulatedZEB1, CXCL16, FSTL1[80,81,82]
miR-1183DownregulatedSOX2[83]
Table 5. Potential long non-coding RNA biomarkers for thyroid cancer.
Table 5. Potential long non-coding RNA biomarkers for thyroid cancer.
lncRNAExpression in CancerFunctionReference
DUXAP8UpregulatedIncreases proliferation, migration, and invasion.[98,99]
FAM230BUpregulatedPromotes migration and invasion in cancer cells through a mechanism involving the Wnt pathway.[100]
HOTAIRUpregulatedControls early development- related genes. Increases growth, migration, and invasive.[76,79,90,91]
LINC00460UpregulatedPromotes cell proliferation, invasive properties, and epithelial-to-mesenchymal transition.[75,77,78]
MALAT1UpregulatedIncreases cell proliferation, invasion, and migration.[87,90,94,95]
NEAT1UpregulatedFacilitates glycolytic metabolism, cell proliferation, and metastasis.[90,101,102]
MAPKAPK5-AS1UpregulatedIncreases proliferation and invasion.[103]
OIP5-AS1UpregulatedPromotes cell proliferation, migration, and invasion.[104]
PTCSC3DownregulatedTumor suppressor.[88,97]
XISTUpregulatedInactivates X chromosome in female cells. Promotes proliferation and invasion.[105,106]
Table 6. Mitophagic genes involved in cancer 1.
Table 6. Mitophagic genes involved in cancer 1.
Mitophagic
Protein
FunctionType of CancerRef.
Pink/ParkinPro-tumorigenicBreast, cervical, ovarian, lung, glioma, esophagus, melanoma[156]
Anti-tumorigenicLiver, renal, pancreatic, endometrial
FUNDC-1Pro-tumorigenicCervical, hepatocarcinoma, breast, colorectal, leukemia, ovarian, pancreatic, Prostate, lung, and breast adenocarcinoma; and Glioblastoma[157,158,159]
NIXPro-tumorigenicPancreatic[160]
Anti-tumorigenicOsteosarcoma[161]
BNIP3Pro-tumorigenicLung, prostate, glioblastoma multiforme, cervical tumors, endometrial, breast carcinomas and gastric adenocarcinomas[162,163,164]
Anti-tumorigenicBreast,
Pancreatic
[165,166]
AMBRAPro-tumorigenicHepatocellular carcinoma, metastatic breast cancer, and medulloblastoma[167]
Anti-tumorigenicColorectal cancer cell, melanoma, and squamous cell carcinoma[167]
FKBP8Pro-tumorigenicHepatocellular Carcinoma[168]
Anti-tumorigenicSchwannoma, melanoma[169]
NLRX1Pro-tumorigenicBreast[170]
Anti-tumorigenicPancreatic[171]
Prohibitin-2Pro-tumorigenicLung, Cervical[172,173]
1 This table presents only mitochondrial proteins involved in mitophagy, not proteins involved in the phagophore or autophagosome formation.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Cabané, P.; Correa, C.; Bode, I.; Aguilar, R.; Elorza, A.A. Biomarkers in Thyroid Cancer: Emerging Opportunities from Non-Coding RNAs and Mitochondrial Space. Int. J. Mol. Sci. 2024, 25, 6719. https://doi.org/10.3390/ijms25126719

AMA Style

Cabané P, Correa C, Bode I, Aguilar R, Elorza AA. Biomarkers in Thyroid Cancer: Emerging Opportunities from Non-Coding RNAs and Mitochondrial Space. International Journal of Molecular Sciences. 2024; 25(12):6719. https://doi.org/10.3390/ijms25126719

Chicago/Turabian Style

Cabané, Patricio, Claudio Correa, Ignacio Bode, Rodrigo Aguilar, and Alvaro A. Elorza. 2024. "Biomarkers in Thyroid Cancer: Emerging Opportunities from Non-Coding RNAs and Mitochondrial Space" International Journal of Molecular Sciences 25, no. 12: 6719. https://doi.org/10.3390/ijms25126719

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

Article Metrics

Back to TopTop