Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Cancer Imaging ; 24(1): 75, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38886866

ABSTRACT

OBJECTIVE: The aim of our study is to find a better way to identify a group of papillary thyroid carcinoma (PTC) with more aggressive behaviors and to provide a prediction model for lymph node metastasis to assist in clinic practice. METHODS: Targeted sequencing of DNA/RNA was used to detect genetic alterations. Gene expression level was measured by quantitative real-time PCR, western blotting or immunohistochemistry. CCK8, transwell assay and flow cytometry were used to investigate the effects of concomitant gene alterations in PTC. LASSO-logistics regression algorithm was used to construct a nomogram model integrating radiomic features, mutated genes and clinical characteristics. RESULTS: 172 high-risk variants and 7 fusion types were detected. The mutation frequencies in BRAF, TERT, RET, ATM and GGT1 were significantly higher in cancer tissues than benign nodules. Gene fusions were detected in 16 samples (2 at the DNA level and 14 at the RNA level). ATM mutation (ATMMUT) was frequently accompanied by BRAFMUT, TERTMUT or gene fusions. ATMMUT alone or ATM co-mutations were significantly positively correlated with lymph node metastasis. Accordingly, ATM knock-down PTC cells bearing BRAFV600E, KRASG12R or CCDC6-RET had higher proliferative ability and more aggressive potency than cells without ATM knock-down in vitro. Furthermore, combining gene alterations and clinical features significantly improved the predictive efficacy for lymph node metastasis of radiomic features, from 71.5 to 87.0%. CONCLUSIONS: Targeted sequencing of comprehensive genetic alterations in PTC has high prognostic value. These alterations, in combination with clinical and radiomic features, may aid in predicting invasive PTC with higher accuracy.


Subject(s)
Lymphatic Metastasis , Thyroid Cancer, Papillary , Thyroid Neoplasms , Humans , Lymphatic Metastasis/diagnostic imaging , Thyroid Cancer, Papillary/genetics , Thyroid Cancer, Papillary/pathology , Thyroid Cancer, Papillary/diagnostic imaging , Male , Female , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Thyroid Neoplasms/diagnostic imaging , Middle Aged , Mutation , Adult , Proto-Oncogene Proteins B-raf/genetics , Ataxia Telangiectasia Mutated Proteins/genetics , Nomograms , Biomarkers, Tumor/genetics , Telomerase/genetics , Radiomics
2.
Med Oncol ; 41(6): 160, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38763968

ABSTRACT

Papillary thyroid carcinoma (PTC) is a common endocrine malignancy. The pathology of PTC is far from clear. As a kinase that can be targeted, the role of TNIK in PTC has not been investigated. This study was focused on the effects and molecular mechanisms of TNIK in PTC. Both public datasets and clinical specimens were used to verify TNIK expression. The effects of TNIK were investigated in both cell lines and mice models. Transcriptome analysis was used to explore the underlying mechanism of TNIK. Immunofluorescence, wound healing, and qRT-PCR assays were used to validate the mechanism of TNIK in PTC. The therapeutic effects of TNIK inhibitor NCB-0846 were evaluated by flow cytometry, western blot, and subcutaneous xenografts mice. TNIK expression was upregulated in PTC tissues. TNIK knockdown could suppress cell proliferation and tumor growth in no matter cell models or nude mice. The transcriptome analysis, GO enrichment analysis, and GSEA analysis results indicated TNIK was highly correlated with cytoskeleton, cell motility, and Wnt pathways. The mechanistic studies demonstrated that TNIK regulated cytoskeleton remodeling and promoted cell migration. NCB-0846 significantly inhibited TNIK kinase activity, induced cell apoptosis, and activated apoptosis-related proteins in a dose-dependent manner. In addition, NCB-0846 inhibited tumor growth in tumor-bearing mice. In summary, we proposed a novel regulatory mechanism in which TNIK-mediated cytoskeleton remodeling and cell migration to regulate tumor progression in PTC. TNIK is a therapeutic target in PTC and NCB-0846 would act as a novel targeted drug for PTC therapy.


Subject(s)
Cell Proliferation , Thyroid Cancer, Papillary , Thyroid Neoplasms , Animals , Female , Humans , Male , Mice , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Mice, Inbred BALB C , Mice, Nude , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Thyroid Cancer, Papillary/pathology , Thyroid Cancer, Papillary/drug therapy , Thyroid Cancer, Papillary/genetics , Thyroid Cancer, Papillary/metabolism , Thyroid Neoplasms/pathology , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/genetics , Xenograft Model Antitumor Assays
3.
Small Methods ; : e2301310, 2024 Jan 02.
Article in English | MEDLINE | ID: mdl-38164884

ABSTRACT

Gene therapy has been adapted for improving malignant tumor treatment. However, pharmacotherapies targeting cancer remain limited and are generally inapplicable for rare disease patients. Oleanolic acid (OA) is a plant-derived triterpenoid that is frequently used in Chinese medicine as a safe but slow-acting treatment for many disorders. Here, the congruent pharmacological activities of OA and CRISPR-dCas9 in targeting AURKA or KDM1A and improving disease-specific prognosis and used a synthetic-biology-inspired design principle to engineer a therapeutic gene circuit that enables a concerted action of both drugs are utilized. In particular, the OA-triggered CRISPR-dCas9 transcriptional repression system rapidly and simultaneously attenuated lung and thyroid cancer. Collectively, this work shows that rationally engineered synthetic gene circuits are capable of treating multifactorial diseases in a synergistic manner by multiplexing the targeting efficiencies of single therapeutics.

4.
Int J Biol Sci ; 19(12): 3970-3986, 2023.
Article in English | MEDLINE | ID: mdl-37564214

ABSTRACT

Thyroid cancer has become the most frequent endocrine-related malignancy. Currently, a mounting body of evidences support the clinical strategies for extending the benefit of PARP inhibitors beyond BRCA-mutant cancers. However, the functions and molecular mechanisms of PARP inhibitors in thyroid cancers (TCs) are not fully understood. Here, on the one hand, we revealed that niraparib promotes the accumulation of DNA damage in TCs. On the other hand, we indicated that niraparib inhibits the transcription of DIMT1 through promoting Pol II pausing in a PAR-dependent manner, subsequently leading to a global translation inhibition in TCs. Meanwhile, we found that niraparib activates the NF-κB signaling pathway by inhibiting the PARylation of p65, which decreases its ubiquitination and degradation level through E3 ubiquitin ligase RNF146. Moreover, bortezomib (a small molecule inhibitor of the NF-κB signaling pathway) could significantly enhance the anti-tumor effect of niraparib on TCs in vitro and in vivo. Our findings provide mechanistic supports for the efficacy of PARP inhibitors in cancer cells lacking BRCA-mutant.


Subject(s)
Antineoplastic Agents , Thyroid Neoplasms , Humans , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , NF-kappa B/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/genetics , Signal Transduction
5.
J Cancer ; 14(9): 1592-1604, 2023.
Article in English | MEDLINE | ID: mdl-37325052

ABSTRACT

The BRAFV600E mutation is the most common oncogenic mutation in thyroid cancer, suggesting an aggressive subtype of thyroid cancer and poor prognosis. Vemurafenib, a selective inhibitor of BRAFV600E, may provide therapeutic benefit in various cancers including thyroid cancer. However, the prevalence of drug resistance remains a challenge because of the feedback activation of the MAPK/ERK and PI3K/AKT pathways. In treating thyroid cancer cells with vemurafenib, we have detected reactivation of the MAPK/ERK signaling pathway as a result of the release of multiple receptor tyrosine kinases (RTKs) from the negative feedback of ERK phosphorylation. SHP2 is an important target protein downstream of the RTK signaling pathway. Decreasing it through SHP2 knockdown or the use of an inhibitor of SHP2 (SHP099) was found to significantly increase the early sensitivity and reverse the late resistance to vemurafenib in BRAFV600E mutant thyroid cancer cells. Overall, our findings suggest that blocking SHP2 reverses the reactivation of the MAPK/ERK signaling pathway caused by the activation of RTKs and improves the sensitivity of thyroid cancer to vemurafenib, which has potential implications for mechanism-based early combination strategies to treat thyroid cancer.

6.
Front Endocrinol (Lausanne) ; 14: 964074, 2023.
Article in English | MEDLINE | ID: mdl-36896175

ABSTRACT

Objective: Central lymph node metastasis (CLNM) is a predictor of poor prognosis for papillary thyroid carcinoma (PTC) patients. The options for surgeon operation or follow-up depend on the state of CLNM while accurate prediction is a challenge for radiologists. The present study aimed to develop and validate an effective preoperative nomogram combining deep learning, clinical characteristics and ultrasound features for predicting CLNM. Materials and methods: In this study, 3359 PTC patients who had undergone total thyroidectomy or thyroid lobectomy from two medical centers were enrolled. The patients were divided into three datasets for training, internal validation and external validation. We constructed an integrated nomogram combining deep learning, clinical characteristics and ultrasound features using multivariable logistic regression to predict CLNM in PTC patients. Results: Multivariate analysis indicated that the AI model-predicted value, multiple, position, microcalcification, abutment/perimeter ratio and US-reported LN status were independent risk factors predicting CLNM. The area under the curve (AUC) for the nomogram to predict CLNM was 0.812 (95% CI, 0.794-0.830) in the training cohort, 0.809 (95% CI, 0.780-0.837) in the internal validation cohort and 0.829(95%CI, 0.785-0.872) in the external validation cohort. Based on the analysis of the decision curve, our integrated nomogram was superior to other models in terms of clinical predictive ability. Conclusion: Our proposed thyroid cancer lymph node metastasis nomogram shows favorable predictive value to assist surgeons in making appropriate surgical decisions in PTC treatment.


Subject(s)
Deep Learning , Thyroid Neoplasms , Humans , Thyroid Cancer, Papillary/diagnostic imaging , Thyroid Cancer, Papillary/surgery , Thyroid Cancer, Papillary/pathology , Nomograms , Lymphatic Metastasis/diagnostic imaging , Lymphatic Metastasis/pathology , Lymph Nodes/diagnostic imaging , Lymph Nodes/surgery , Lymph Nodes/pathology , Thyroid Neoplasms/diagnostic imaging , Thyroid Neoplasms/surgery , Thyroid Neoplasms/pathology
7.
Dis Markers ; 2022: 9541080, 2022.
Article in English | MEDLINE | ID: mdl-36510497

ABSTRACT

Quercetin, a flavonoid with anti-inflammatory and anticancer properties, is expected to be an innovative anticancer therapeutic agent for papillary thyroid carcinoma (PTC). However, the downstream signaling pathways that mediate quercetin-dependent anticancer properties remain to be deciphered. Herein, potential targets of quercetin were screened with several bioinformatic avenues including PharmMapper, Gene Expression Omnibus (GEO) database, protein-protein interaction (PPI) network, and molecular docking. Besides, western blot, CCK-8 transwell analysis of migration and invasion, flow cytometric analysis, and colony formation assays were performed to investigate the underlying mechanism. We found four core nodes (MMP9, JUN, SPP1, and HMOX1) by constructing a PPI network with 23 common targets. Through functional enrichment analysis, we confirmed that the above four target genes are enriched in the TNF, PI3K-AKT, and NF-κB signaling pathways, which are involved in the inflammatory microenvironment and inhibit the development and progression of tumors. Furthermore, molecular docking results demonstrated that quercetin shows strong binding efficiency with the proteins encoded by these 4 key proteins. Finally, quercetin displayed strong antitumor efficacy in PTC cell lines. In this research, we demonstrated the application of network pharmacology in evaluating the mechanisms of action and molecular targets of quercetin, which regulates a variety of proteins and signaling pathways in PTC. These data might explain the mechanism underlying the anticancer effects of quercetin in PTC.


Subject(s)
Carcinoma, Papillary , Drugs, Chinese Herbal , Thyroid Neoplasms , Humans , Quercetin/pharmacology , Thyroid Cancer, Papillary/drug therapy , Network Pharmacology , Molecular Docking Simulation , Phosphatidylinositol 3-Kinases , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/genetics , Tumor Microenvironment
8.
Cell Biosci ; 12(1): 195, 2022 Dec 05.
Article in English | MEDLINE | ID: mdl-36471438

ABSTRACT

BACKGROUND: Treatment failure is the main cause of death from papillary thyroid carcinoma (PTC). It is urgent to look for new intervention targets and to develop new therapies for treating PTC. Aurora-A kinase (AURKA) functionally regulates cell mitosis and is closely related to the occurrence and development of a variety of tumours. However, the expression and potential functions of AURKA in PTC remain largely elusive. RESULTS: Clinicopathologically, AURKA is highly expressed in PTC tissues compared to normal tissues and is correlated with lymph node metastasis, TNM stage and patient prognosis. Biologically, AURKA functions as an oncoprotein to promote the proliferation and migration of PTC cells. Mechanistically, AURKA directly binds to SIN1 and compromises CUL4B-based E3 ligase-mediated ubiquitination and subsequent degradation of SIN1, leading to hyperactivation of the mTORC2-AKT pathway in PTC cells. CONCLUSIONS: We found that AURKA plays critical roles in regulating the progression of PTC by activating the mTORC2-AKT pathway, highlighting the potential of targeting AURKA to treat PTC.

9.
Front Immunol ; 13: 991656, 2022.
Article in English | MEDLINE | ID: mdl-36211409

ABSTRACT

Glucose metabolism-related genes play an important role in the development and immunotherapy of many tumours, but their role in thyroid cancer is ambiguous. To investigate the role of glucose metabolism-related genes in the development of papillary thyroid cancer (PTC) and their correlation with the clinical outcome of PTC, we collected transcriptomic data from 501 PTC patients in the Cancer Genome Atlas (TCGA). We performed nonnegative matrix decomposition clustering of 2752 glucose metabolism-related genes from transcriptome data and classified PTC patients into three subgroups (C1 for high activation of glucose metabolism, C2 for low activation of glucose metabolism and C3 for moderate activation of glucose metabolism) based on the activation of different glucose metabolism-related genes in 10 glucose metabolism-related pathways. We found a positive correlation between the activation level of glucose metabolism and the tumour mutation burden (TMB), neoantigen number, mRNA stemness index (mRNAsi), age, and tumour stage in PTC patients. Next, we constructed a prognostic prediction model for PTC using six glucose metabolism-related genes (PGBD5, TPO, IGFBPL1, TMEM171, SOD3, TDRD9) and constructed a nomogram based on the risk score and clinical parameters of PTC patients. Both the prognostic risk prediction model and nomogram had high stability and accuracy for predicting the progression-free interval (PFI) in PTC patients. Patients were then divided into high-risk and low-risk groups by risk score. The high-risk group was sensitive to paclitaxel and anti-PD-1 treatment, and the low-risk group was sensitive to sorafenib treatment. We found that the high-risk group was enriched in inflammatory response pathways and associated with high level of immune cell infiltration. To verify the accuracy of the prognostic prediction model, we knocked down PGBD5 in PTC cells and found that the proliferation ability of PTC cells was significantly reduced. This suggests that PGBD5 may be a relatively important oncogene in PTC. Our study constructed a prognostic prediction model and classification of PTC by glucose metabolism-related genes, which provides a new perspective on the role of glucose metabolism in the development and immune microenvironment of PTC and in guiding chemotherapy, targeted therapy and immune checkpoint blockade therapy of PTC.


Subject(s)
Glucose , Thyroid Neoplasms , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Immunity , Paclitaxel , RNA, Messenger , Sorafenib , Thyroid Cancer, Papillary/drug therapy , Thyroid Cancer, Papillary/genetics , Thyroid Cancer, Papillary/metabolism , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Tumor Microenvironment
10.
Am J Cancer Res ; 12(1): 247-264, 2022.
Article in English | MEDLINE | ID: mdl-35141016

ABSTRACT

Pharmacologic targeting of components of the MAPK/ERK pathway in differentiated thyroid carcinoma (DTC) is often limited due to the development of adaptive resistance. However, the detailed mechanism of MEK inhibitor (MEKi) resistance is not fully understood. Here, MEKi-resistant models were constructed successfully, in which multiple receptor tyrosine kinases (RTKs) signaling pathways and Src-homology 2 domain-containing phosphatase 2 (SHP2) were activated in MEKi-resistant cells. Given the physiological role of SHP2 as the downstream target of many RTKs, we first found blockade of SHP2 enhanced the sensitivity to MEKi in constructed MEKi-resistant models. Interestingly, we also found that compared with MEKi treatment alone, MEKi in combination with an SHP2 inhibitor markedly suppressed the reactivation of the MEK/ERK pathway; thus, the addition of the SHP2 inhibitor significantly improved the antitumor effects of MEKi. The synergistic suppression of DTC upon treatment with both inhibitors was further confirmed in xenograft models and transgenic models. Thus, our data suggest that RTKs activation leads to reactivation of the MAPK pathway and resistance to MEKi in DTC, which is reversed by SHP2 blockade. As a novel active inhibitor of SHP2, SHP099 in combination with MEKi is a promising therapeutic approach for advanced DTC and MEKi-resistant one.

11.
Cell Death Dis ; 13(2): 124, 2022 02 08.
Article in English | MEDLINE | ID: mdl-35136031

ABSTRACT

The use of the BRAF inhibitor vemurafenib exhibits drug resistance in the treatment of thyroid cancer (TC), and finding more effective multitarget combination therapies may be an important solution. In the present study, we found strong correlations between Ref-1 high expression and BRAF mutation, lymph node metastasis, and TNM stage. The oxidative stress environment induced by structural activation of BRAF upregulates the expression of Ref-1, which caused intrinsic resistance of PTC to vemurafenib. Combination inhibition of the Ref-1 redox function and BRAF could enhance the antitumor effects of vemurafenib, which was achieved by blocking the action of Ref-1 on BRAF proteins. Furthermore, combination treatment could cause an overload of autophagic flux via excessive AMPK protein activation, causing cell senescence and cell death in vitro. And combined administration of Ref-1 and vemurafenib in vivo suppressed PTC cell growth and metastasis in a cell-based lung metastatic tumor model and xenogeneic subcutaneous tumor model. Collectively, our study provides evidence that Ref-1 upregulation via constitutive activation of BRAF in PTC contributes to intrinsic resistance to vemurafenib. Combined treatment with a Ref-1 redox inhibitor and a BRAF inhibitor could make PTC more sensitive to vemurafenib and enhance the antitumor effects of vemurafenib by further inhibiting the MAPK pathway and activating the excessive autophagy and related senescence process.


Subject(s)
DNA-(Apurinic or Apyrimidinic Site) Lyase , Proto-Oncogene Proteins B-raf , Thyroid Neoplasms , Vemurafenib , Animals , Cell Line, Tumor , DNA-(Apurinic or Apyrimidinic Site) Lyase/antagonists & inhibitors , Drug Resistance, Neoplasm/genetics , Humans , Mutation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/metabolism , Thyroid Cancer, Papillary/drug therapy , Thyroid Cancer, Papillary/genetics , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Vemurafenib/pharmacology
12.
J Healthc Eng ; 2022: 4348396, 2022.
Article in English | MEDLINE | ID: mdl-35178227

ABSTRACT

PURPOSE: The purpose of the current study was to determine whether older patients with differentiated thyroid cancer (DTC) who received surgical treatment had a better cause-specific survival (CSS) than patients who were recommended surgery, but declined, and whether patients who underwent postoperative RAI-131 therapy had an impact on CSS based on TNM staging and number of lymph node metastases for all total or near-total thyroidectomy patients. Patients and Methods: This retrospective, population-based study analyzed the clinical data of 162 DTC patients from signal institution in China and 26,487 cases from the Surveillance, Epidemiology, and End Results (SEER) program registry. The patients were divided into two groups (underwent surgery and surgery recommended, but not performed) in the SEER cohort. Furthermore, patients were grouped as follows: T4; N1b; M1; T1-3N0-1a; specific number of lymph node metastases; and total or near-total thyroidectomy. Results: The 120-month cause-specific survival (CSS) rate of women and men showed a gradual declining trend from 60-64 to ≥80 years of age in the group that underwent surgery. The CSS rate of women and men showed a marked downward and irregular trend with an increase in age in the recommended, but no surgery group in the SEER cohort. Univariate analysis indicated that the surgery group had a higher 120-month CSS in women in most stages and men, compared with the no surgery group in the SEER cohort. The analysis of the SEER cohort showed that RAI-131 therapy was associated with an improved 80-month CSS in T4/N1b/M1 women (P < 0.0183) and men (P < 0.0011). However, there were no CSS differences between the RAI-131 therapy and the no-RAI-131 group for the patients with T4/N1b/M1 (AJCC 7th) thyroid cancer in the Chinese cohort. There was no CSS difference in women or men between the T1-3N0 and T1-3N1a patients in the SEER cohort. And similar findings were observed in T1-3N1a patients in the Chinese cohort. There was no statistical difference between the two subgroups. Conclusions: Surgical treatment should be recommended for elderly DTC patients because surgery can lead to a better CSS. High-risk patients achieve a higher benefit-to-risk ratio with RAI-131 therapy. To avoid the adverse effects associated with RAI-131 therapy, a multidisciplinary discussion should be arranged for intermediate- and low-risk patients.


Subject(s)
Thyroid Neoplasms , Aged , Female , Humans , Iodine Radioisotopes/therapeutic use , Lymphatic Metastasis/drug therapy , Male , Retrospective Studies , Thyroid Neoplasms/radiotherapy , Thyroid Neoplasms/surgery , Thyroidectomy
13.
Theranostics ; 12(4): 1500-1517, 2022.
Article in English | MEDLINE | ID: mdl-35198054

ABSTRACT

Background: Cancer stem cells (CSCs) are highly tumorigenic, chemotherapy-resistant, tumor growth-sustaining, and are implicated in tumor recurrence. Previous studies have shown that lysine-specific histone demethylase 1A (KDM1A) is highly expressed in several human malignancies and CSCs. However, the role of KDM1A in CSCs and the therapeutic potential of KDM1A inhibitors for the treatment of the advanced thyroid cancer are poorly understood. Methods: Firstly, KDM1A was identified as an important epigenetic modifier that maintained the stemness of thyroid cancer through a mini histone methylation modifier screen and confirmed in thyroid cancer tissues and cell lines. RNA sequence was performed to discover the downstream genes of KDM1A. The underlying mechanisms were further investigated by ChIP, IP and dual luciferase reporter assays, gain and loss of function assays. Results: Here we report that KDM1A regulates the stemness of thyroid cancer and promotes thyroid cancer progression via the Wnt/ß-catenin pathway. Mechanistically, KDM1A down-regulates two antagonists of the canonical Wnt pathway, APC2 and DKK1, by demethylating H3K4me1/2 of the APC2 promoter region and the nonhistone substrate HIF-1α, resulting in the inhibition of APC2 transcription and the activation of the HIF-1α/microRNA-146a/DKK1 axis. Importantly, we also demonstrate that GSK-LSD1, a highly selective inhibitor of KDM1A, significantly inhibits thyroid cancer progression and enhances the sensitivity of thyroid cancer to chemotherapy. Conclusions: KDM1A plays an important role in thyroid cancer progression and maintains stemness, our study provides a new strategy for the therapy of advanced thyroid cancer.


Subject(s)
Thyroid Neoplasms , Wnt Signaling Pathway , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Histone Demethylases/metabolism , Humans , Neoplasm Recurrence, Local/genetics , Neoplastic Stem Cells/metabolism , Thyroid Neoplasms/metabolism , Wnt Signaling Pathway/genetics , beta Catenin/genetics , beta Catenin/metabolism
14.
Bioengineered ; 13(1): 190-205, 2022 01.
Article in English | MEDLINE | ID: mdl-34964699

ABSTRACT

The phosphoprotein phosphatase catalytic subunit (PPPCs) family has been shown to play an important role in the development and progression of various malignancies, but its expression patterns and biological functions in breast cancer (BC) remain unclear. Therefore, we aimed to investigate the clinical significance and biological functions of the PPPCs family to understand its possible significance in the diagnosis, prognosis and treatment of breast cancer. We comprehensively investigated the expression levels, diagnostic accuracy, prognostic outcomes, biological functions and effects on immune cell infiltration of the PPPCs family in breast cancer using online databases. Except for PPP1CB, PPP1CC, PPP5C and PPEF1, the mRNA expression levels of the PPPCs family in breast cancer tissues were significantly different from those in paracancerous tissues. The differentially expressed genes (DEGs) were associated with the clinicopathological parameters and prognosis of breast cancer. The DEGs were mainly associated with the WNT signaling pathway, antigen presentation and DNA repair. In addition, the DEGs significantly affected the infiltration of immune cells in breast cancer tissues. Among the PPPCs family, PPP1CA and PPP4C played a prominent role in the progression of breast cancer, and inhibition of PPP1CA and PPP4C expression by siRNA can significantly inhibit breast cancer cells proliferation and migration. In conclusion, the PPPCs family, especially PPP1CA and PPP4C, could be used as new biomarkers to improve diagnostic accuracy, predict prognosis and novel targets for the treatment of breast cancer.


Subject(s)
Breast Neoplasms/genetics , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/metabolism , Protein Phosphatase 1/genetics , Protein Phosphatase 1/metabolism , Up-Regulation , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Databases, Genetic , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Prognosis , Survival Analysis
15.
Endocr Relat Cancer ; 29(2): 87-98, 2022 01 20.
Article in English | MEDLINE | ID: mdl-34874277

ABSTRACT

Increasing body of recent studies determining the expression of tumor-specific major histocompatibility complex (MHC) class II protein supports its potential role in several malignancies, but little is known in human medullary thyroid cancer (MTC). Here, we report the expression of MHC-II and its clinicopathologic and prognostic relevance in MTC patients. Immunohistochemistry staining revealed a significant reduction in tumor cell-specific MHC-II expression in a higher AJCC stage and its poor prognostic correlation with human MTC development. Further statistical analysis identified the low MHC-II expression as a significant and independent risk factor for MTC recurrence and patient survival. Moreover, in vitro studies showed that the MHC-II expression was remarkably increased by RET inhibitors, which were prescribed to treat advanced MTC. Similarly, inhibitors blocking the MAPK/ERK and AKT/mTOR pathways also augmented MHC-II expression, suggesting their implications in RET-MHC-II signaling axis. Importantly, in vitro assays manifested enhanced peripheral blood leukocytes-mediated cytotoxicity in MTC cells treated with RET inhibitors, which were partially alleviated by HLA knock-down. Together, our study demonstrates that low MHC-II expression levels may serve as a prognostic biomarker for aggressive diseases in MTC patients and indicates that RET activation may promote MTC immune escape through downregulating MHC-II expression.


Subject(s)
Carcinoma, Neuroendocrine , Thyroid Neoplasms , Carcinoma, Neuroendocrine/metabolism , Humans , Prognosis , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogene Proteins c-ret/metabolism , Thyroid Neoplasms/metabolism
16.
Am J Cancer Res ; 11(11): 5402-5414, 2021.
Article in English | MEDLINE | ID: mdl-34873468

ABSTRACT

m6A methylation has been demonstrated to be one of the most important epigenetic regulation mechanisms in cell differentiation and cancer development especially m6A derived diagnostic and prognostic biomarkers have been identified in the past several years. However, systemic investigation to the interaction between germline single-nucleotide polymorphisms (SNPs) and m6A has not been conducted yet. In this study, we collected previous identified significant thyroid cancer associated SNPs from UKB cohort (358 cases and 407,399 controls) and ICR cohort (3,001 patients and 287,550 controls) and thyroid eQTL (sample size = 574 from GTEx project) and m6A-SNP (N = 1,678,126) were applied to prioritize the candidate SNPs. Finally, five candidate genes (PLEKHA8, SMUG1, CDC123, RMI2, ACSM5) were identified to be thyroid cancer associated m6A-related genetic susceptibility. Loss and gain function studies of m6A writer proteins confirm that ACSM5 is regulated by m6A methylation of mRNA. Moreover, ACSM5 is downregulated in thyroid cancer and inversely correlated with PTC malignancy and patient survival. Together, our study highlight mRNA-seq and m6A-seq double analysis provided a novel approach to identify cancer biomarkers and understanding the heterogeneity of human cancers.

17.
Ann Transl Med ; 9(10): 866, 2021 May.
Article in English | MEDLINE | ID: mdl-34164500

ABSTRACT

BACKGROUND: Celastrol, a triterpene present in the traditional Chinese medicine (TCM) Triptergium wilfordii, has been demonstrated to have remarkable anticancer activity. However, its specific mechanism on papillary thyroid carcinoma (PTC) remains to be elucidated. METHODS: Potential targets of celastrol were screened from public databases. Through the Gene Expression Omnibus (GEO) online database, we obtained the bioinformatics analysis profile of PTC, GSE33630, and analyzed the differentially expressed genes (DEGs). Then, a protein-protein interaction (PPI) network was constructed by utilizing the STRING database. Furthermore, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis were conducted. Finally, drug interactions between hub genes and celastrol were verified by molecular docking. RESULTS: Four core nodes (MMP9, JUN, ICAM1, and VCAM1) were discerned via constructing a PPI network of 47 common targets. Through functional enrichment analysis, it was confirmed that the above target genes were basically enriched in the interleukin-17 (IL-17), nuclear factor kappa-B (NF-κB), and tumor necrosis factor (TNF) signaling pathways, which are involved in the inflammatory microenvironment to inhibit the development and progression of tumors. Molecular docking results demonstrated that celastrol has a strong binding efficiency with the 4 key proteins. CONCLUSIONS: In this research, it was demonstrated that celastrol can regulate a variety of proteins and signaling pathways against PTC, providing a theoretical basis for future clinical applications.

18.
Cell Death Dis ; 12(4): 347, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33795650

ABSTRACT

Papillary thyroid carcinoma (PTC) is one of the most common kinds of endocrine-related cancer and has a heterogeneous prognosis. Metabolic reprogramming is one of the hallmarks of cancers. Aberrant glucose metabolism is associated with malignant biological behavior. However, the functions and mechanisms of glucose metabolism genes in PTC are not fully understood. Thus, data from The Cancer Genome Atlas database were analyzed, and lactate dehydrogenase A (LDHA) was determined to be a potential novel diagnostic and therapeutic target for PTCs. The research objective was to investigate the expression of LDHA in PTCs and to explore the main functions and relative mechanisms of LDHA in PTCs. Higher expression levels of LDHA were found in PTC tissues than in normal thyroid tissues at both the mRNA and protein levels. Higher expression levels of LDHA were correlated with aggressive clinicopathological features and poor prognosis. Moreover, we found that LDHA not only promoted PTC migration and invasion but also enhanced tumor growth both in vitro and in vivo. In addition, we revealed that the metabolic products of LDHA catalyzed induced the epithelial-mesenchymal transition process by increasing the relative gene H3K27 acetylation. Moreover, LDHA knockdown activated the AMPK pathway and induced protective autophagy. An autophagy inhibitor significantly enhanced the antitumor effect of FX11. These results suggested that LDHA enhanced the cell metastasis and proliferation of PTCs and may therefore become a potential therapeutic target for PTCs.


Subject(s)
Autophagy/drug effects , Cell Movement/drug effects , Lactate Dehydrogenase 5/pharmacology , Neoplasm Metastasis/drug therapy , Thyroid Cancer, Papillary/genetics , Carcinogenesis/drug effects , Carcinogenesis/genetics , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Lactate Dehydrogenase 5/metabolism , Thyroid Cancer, Papillary/drug therapy , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/metabolism
19.
Anticancer Agents Med Chem ; 21(16): 2198-2203, 2021 10 28.
Article in English | MEDLINE | ID: mdl-33653254

ABSTRACT

BACKGROUND: Hepatitis B virus X protein (HBx) is an indispensable progression factor in Hepatocellular Carcinoma (HCC). CCL15 could be a peculiar proteomic biomarker of HCC with tumorigenesis and tumor invasion. OBJECTIVE: The aim of the study was to explore the relationship between HBx and CCL15 expression in HCC. METHODS: HBV-positive HCC pathological tissue samples and corresponding adjacent non-tumor liver tissues were collected. The expression of HBx and CCL15 was analyzed by immunohistochemistry, real-time Polymerase Chain Reaction (PCR), and western blot analysis in tissues or in vitro. RESULTS: The levels of CCL15 mRNA and protein expression in HCC samples were observably higher than those of adjacent non-tumor liver tissues. The CCL15 was significantly associated with the expression of HBx in HBV-positive HCC samples. The up-regulation of HBx induced CCL15 expression in vitro. The high expression score of CCL15 was significant associated with the poor prognosis of HCC patients. CONCLUSION: The CCL15 expression was observably associated with HBx in HCC patients. The CCL15 may be considered as an indicator in the clinical management of HBV-associated HCC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Chemokines, CC/metabolism , Liver Neoplasms/metabolism , Macrophage Inflammatory Proteins/metabolism , Trans-Activators/metabolism , Up-Regulation , Viral Regulatory and Accessory Proteins/metabolism , Carcinoma, Hepatocellular/pathology , Humans , Liver Neoplasms/pathology , Tumor Cells, Cultured
20.
ACS Appl Mater Interfaces ; 13(5): 6043-6052, 2021 Feb 10.
Article in English | MEDLINE | ID: mdl-33525876

ABSTRACT

DNA methylation is a kind of a crucial epigenetic marker orchestrating gene expression, molecular function, and cellular phenotype. However, manipulating the methylation status of specific genes remains challenging. Here, a clustered regularly interspaced palindromic repeats-Cas9-based near-infrared upconversion-activated DNA methylation editing system (CNAMS) was designed for the optogenetic editing of DNA methylation. The fusion proteins of photosensitive CRY2PHR, the catalytic domain of DNMT3A or TET1, and the fusion proteins for CIBN and catalytically inactive Cas9 (dCas9) were engineered. The CNAMS could control DNA methylation editing in response to blue light, thus allowing methylation editing in a spatiotemporal manner. Furthermore, after combination with upconversion nanoparticles, the spectral sensitivity of DNA methylation editing was extended from the blue light to near-infrared (NIR) light, providing the possibility for remote DNA methylation editing. These results demonstrated a meaningful step forward toward realizing the specific editing of DNA methylation, suggesting the wide utility of our CNAMS for functional studies on epigenetic regulation and potential therapeutic strategies for related diseases.


Subject(s)
CRISPR-Associated Protein 9/genetics , CRISPR-Cas Systems/genetics , Gene Editing , Genetic Techniques , Infrared Rays , Thyroid Neoplasms/genetics , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis/genetics , CRISPR-Associated Protein 9/metabolism , Cell Survival , Cells, Cultured , DNA Methylation/genetics , Female , HEK293 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Particle Size , Surface Properties , Thyroid Neoplasms/pathology , Thyroid Neoplasms/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...