Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 147
Filter
1.
Proc Natl Acad Sci U S A ; 118(11)2021 03 16.
Article in English | MEDLINE | ID: mdl-33836600

ABSTRACT

The telomerase reverse transcriptase (TERT) has long been pursued as a direct therapeutic target in human cancer, which is currently hindered by the lack of effective specific inhibitors of TERT. The FOS/GABPB/(mutant) TERT cascade plays a critical role in the regulation of mutant TERT, in which FOS acts as a transcriptional factor for GABPB to up-regulate the expression of GABPB, which in turn activates mutant but not wild-type TERT promoter, driving TERT-promoted oncogenesis. In the present study, we demonstrated that inhibiting this cascade by targeting FOS using FOS inhibitor T-5224 suppressed mutant TERT cancer cells and tumors by inducing robust cell apoptosis; these did not occur in wild-type TERT cells and tumors. Mechanistically, among 35 apoptotic cascade-related proteins tested, the apoptosis induced in this process specifically involved the transcriptional activation of tumor necrosis factor-related apoptosis-inducing ligand receptor 2 (TRAIL-R2) and inactivation of survivin, two key players in the apoptotic cascade, which normally initiate and suppress the apoptotic cascade, respectively. These findings with suppression of FOS were reproduced by direct knockdown of TERT and prevented by prior knockdown of TRAIL-R2. Further experiments demonstrated that TERT acted as a direct transcriptional factor of survivin, up-regulating its expression. Thus, this study identifies a therapeutic strategy for TERT promoter mutation-driven cancers by targeting FOS in the FOS/GABPB/(mutant) TERT cascade, circumventing the current challenge in pharmacologically directly targeting TERT itself. This study also uncovers a mechanism through which TERT controls cell apoptosis by transcriptionally regulating two key players in the apoptotic cascade.


Subject(s)
Apoptosis/drug effects , Neoplasms/genetics , Proto-Oncogene Proteins c-fos/antagonists & inhibitors , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Survivin/genetics , Telomerase/genetics , Benzophenones/pharmacology , Benzophenones/therapeutic use , Carcinogenesis/drug effects , Carcinogenesis/genetics , Cell Line, Tumor , GA-Binding Protein Transcription Factor/genetics , GA-Binding Protein Transcription Factor/metabolism , Gene Expression Regulation, Neoplastic , Humans , Isoxazoles/pharmacology , Isoxazoles/therapeutic use , Mutation , Neoplasms/drug therapy , Neoplasms/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins c-fos/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Signal Transduction/drug effects , Survivin/metabolism , Telomerase/metabolism
2.
Proc Natl Acad Sci U S A ; 117(27): 15846-15851, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32561648

ABSTRACT

Combination use of BRAF V600E inhibitor dabrafenib and MEK inhibitor trametinib has become a standard treatment for human cancers harboring BRAF V600E. Its anticancer efficacies vary, however, with dramatic efficacy in some patients and drug resistance/tumor recurrence in others, which is poorly understood. Using thyroid cancer, melanoma, and colon cancer cell models, we showed that dabrafenib and trametinib induced robust apoptosis of cancer cells harboring both BRAF V600E and TERT promoter mutations but had little proapoptotic effect in cells harboring only BRAF V600E. Correspondingly, the inhibitors nearly completely abolished the growth of in vivo tumors harboring both mutations but had little effect on tumors harboring only BRAF V600E. Upon drug withdrawal, tumors harboring both mutations remained hardly measurable but tumors harboring only BRAF V600E regrew rapidly. BRAF V600E/MAP kinase pathway is known to robustly activate mutant promoter of TERT, a strong apoptosis suppressor. Thus, for survival, cancer cells harboring both mutations may have evolved to rely on BRAF V600E-promoted and high-TERT expression-mediated suppression of apoptosis. As such, inhibition of BRAF/MEK can trigger strong apoptosis-induced cell death and hence tumor abolishment. This does not happen in cells harboring only BRAF V600E as they have not developed reliance on TERT-mediated suppression of apoptosis due to the lack of mutant promoter-driven high-TERT expression. TERT promoter mutation governs BRAF-mutant cancer cells' apoptotic and hence therapeutic responses to BRAF/MEK inhibitors. Thus, the genetic duet of BRAF V600E and TERT promoter mutation represents an Achilles Heel for effective therapeutic targeting and response prediction in cancer.


Subject(s)
Apoptosis/drug effects , Mutation , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/drug effects , Proto-Oncogene Proteins B-raf/genetics , Telomerase/genetics , Animals , Cell Death , Cell Line, Tumor , Colonic Neoplasms , Drug Resistance, Neoplasm/drug effects , Female , Humans , Imidazoles/pharmacology , Melanoma/genetics , Mice, Nude , Mitogen-Activated Protein Kinases/metabolism , Oximes/pharmacology , Pyridones/pharmacology , Pyrimidinones/pharmacology , Thyroid Neoplasms/genetics , Xenograft Model Antitumor Assays
3.
Clin Infect Dis ; 73(2): 328-331, 2021 07 15.
Article in English | MEDLINE | ID: mdl-32750119

ABSTRACT

To explore any relationship between the ABO blood group and the coronavirus disease 2019 (COVID-19) susceptibility, we compared ABO blood group distributions in 2173 COVID-19 patients with local control populations, and found that blood group A was associated with an increased risk of infection, whereas group O was associated with a decreased risk.


Subject(s)
ABO Blood-Group System , COVID-19 , Disease Susceptibility , Humans , Retrospective Studies , SARS-CoV-2
4.
Clin Infect Dis ; 71(16): 2233-2235, 2020 11 19.
Article in English | MEDLINE | ID: mdl-32270178

ABSTRACT

The effect of host immune status on severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection remains unknown. Here, we report the first case of coronavirus disease 2019 (COVID-19) with human immunodeficiency virus type 1 (HIV-1)/hepatitis C virus coinfection, who showed a persistently negative SARS-CoV-2 RNA test but delayed antibody response in the plasma. This case highlights the influence of HIV-1-induced immune dysfunction on early SARS-CoV-2 clearance.


Subject(s)
Antibodies, Viral/blood , COVID-19/immunology , Coinfection/immunology , HIV Infections/complications , Hepatitis C/complications , Adult , Antibodies, Viral/immunology , Antibody Formation , COVID-19/blood , COVID-19/diagnosis , COVID-19 Serological Testing , Coinfection/virology , HIV Infections/immunology , HIV-1 , Hepacivirus , Hepatitis C/immunology , Humans , Immunocompromised Host , Male , Retrospective Studies , SARS-CoV-2 , Tomography, X-Ray Computed
5.
Clin Infect Dis ; 72(11): e920, 2021 06 01.
Article in English | MEDLINE | ID: mdl-32986816
6.
J Cell Biochem ; 117(7): 1491-6, 2016 07.
Article in English | MEDLINE | ID: mdl-26841328

ABSTRACT

Today the most common differentiated thyroid cancer in many countries is papillary thyroid microcarcinoma (PTMC). Although the majority of PTMCs exhibit an indolent clinical course, a few possess high risk for aggressiveness with tumor invasion, metastasis, and even patient mortality. This imposes significant confusion and often dilemma in the clinical management of PTMC. The present review summarizes the molecular pathogenesis, particularly the major genetic alterations, of PTMC that may have prognostic values in assisting risk stratification of this cancer and identification of the most aggressive cases from the many well-behaving cases. J. Cell. Biochem. 117: 1491-1496, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Carcinoma , Thyroid Neoplasms , Animals , Carcinoma/epidemiology , Carcinoma/genetics , Carcinoma/mortality , Carcinoma/therapy , Carcinoma, Papillary , Disease-Free Survival , Humans , Neoplasm Invasiveness , Neoplasm Metastasis , Survival Rate , Thyroid Cancer, Papillary , Thyroid Neoplasms/epidemiology , Thyroid Neoplasms/genetics , Thyroid Neoplasms/mortality , Thyroid Neoplasms/therapy
7.
BMC Med ; 14: 12, 2016 Jan 27.
Article in English | MEDLINE | ID: mdl-26817707

ABSTRACT

RAS mutations play an important role in thyroid tumorigenesis. Considerable effort has been made in the last decade to apply RAS mutations as molecular markers to the clinical management of thyroid nodules and thyroid cancer. Yet, for the low diagnostic sensitivities and specificities of RAS mutations, when used alone, and for their uncertain role in the clinical outcomes of thyroid cancer, it has been unclear how to appropriately use them to assist the management of thyroid nodules and thyroid cancer. Studies from recent years, now added from the Alexander group, have shed light on this issue, making a blurred clinical picture now emerge clearer-RAS mutations, when combined with other genetic markers, have high diagnostic negative predictive values for thyroid cancer; cytologically benign thyroid nodules, including those positive for RAS mutations, have long-term clinical stability when non-surgically managed; and differentiated thyroid cancers harboring RAS mutations alone have an excellent prognosis. This progress in understanding RAS mutations in thyroid cancer is showing a major impact on molecular-based practice in the management of thyroid cancer.Please see related research articles: http://dx.doi.org/10.1186/s12916-016-0554-1 and http://dx.doi.org/10.1186/s12916-015-0419-z.


Subject(s)
Thyroid Nodule , Female , Humans , Male
8.
Eur Radiol ; 26(8): 2611-22, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26560715

ABSTRACT

OBJECTIVES: To investigate the value of combined conventional ultrasound (US), strain elastography (SE) and acoustic radiation force impulse (ARFI) elastography for prediction of cervical lymph node metastasis (CLNM) in papillary thyroid cancer (PTC). METHODS: A consecutive series of 203 patients with 222 PTCs were preoperatively evaluated by US, SE, and ARFI including virtual touch tissue imaging (VTI) and virtual touch tissue quantification (VTQ). A multivariate analysis was performed to predict CLNM by 22 independent variables. Receiver operating characteristic (ROC) curve analysis was used to evaluate the diagnostic performance. RESULTS: Multivariate analysis demonstrated that VTI area ratio (VAR) > 1 was the best predictor for CLNM, followed by abnormal cervical lymph node (ACLN), capsule contact, microcalcification, capsule involvement, and multiple nodules (all P < 0.05). ROC analyses of these characteristics showed the areas under the curve (Az), sensitivity, and specificity were 0.600-0.630, 47.7 %-93.2 %, and 26.9 %-78.4 % for US, respectively; and they were 0.784, 83.0 %, and 73.9 %, respectively, for VAR > 1. As combination of US characteristics with and without VAR, the Az, sensitivity, and specificity were 0.803 and 0.556, 83.0 % and 100.0 %, and 77.6 % and 11.2 %, respectively (P < 0.001). CONCLUSIONS: ARFI elastography shows superior performance over conventional US, particularly when combined with US, in predicting CLNM in PTC patients. KEY POINTS: • Conventional ultrasound is useful in predicting cervical lymph node metastasis preoperatively. • Virtual touch tissue imaging area ratio is the strongest predicting factor. • Predictive performance is markedly improved by combining ultrasound characteristics with VAR. • Acoustic radiation force impulse elastography may be a promising complementary tool.


Subject(s)
Calcinosis/diagnostic imaging , Carcinoma/diagnostic imaging , Elasticity Imaging Techniques/methods , Lymph Nodes/diagnostic imaging , Neoplasms, Multiple Primary/diagnostic imaging , Thyroid Neoplasms/diagnostic imaging , Thyroid Nodule/diagnostic imaging , Adolescent , Adult , Aged , Calcinosis/pathology , Carcinoma/pathology , Carcinoma, Papillary , Female , Humans , Lymph Nodes/pathology , Lymphatic Metastasis , Male , Middle Aged , Multivariate Analysis , Neck , Neoplasms, Multiple Primary/pathology , ROC Curve , Reproducibility of Results , Thyroid Cancer, Papillary , Thyroid Neoplasms/pathology , Thyroid Nodule/pathology , Ultrasonography , Young Adult
9.
J Biol Chem ; 288(9): 6511-21, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23322780

ABSTRACT

Mammalian target of rapamycin (mTOR) is a serine/threonine protein kinase that regulates cell growth, proliferation, and survival. mTOR is frequently activated in human cancers and is a commonly sought anticancer therapeutic target. However, whether the human mTOR gene itself is a proto-oncogene possessing tumorigenicity has not been firmly established. To answer this question, we mutated evolutionarily conserved amino acids, generated eight mutants in the HEAT repeats (M938T) and the FAT (W1456R and G1479N) and kinase (P2273S, V2284M, V2291I, T2294I, and E2288K) domains of mTOR, and studied their oncogenicity. On transient expression in HEK293T cells, these mTOR mutants displayed elevated protein kinase activities accompanied by activated mTOR/p70S6K signaling at varying levels, demonstrating the gain of function of the mTOR gene with these mutations. We selected P2273S and E2288K, the two most catalytically active mutants, to further examine their oncogenicity and tumorigenicity. Stable expression of the two mTOR mutants in NIH3T3 cells strongly activated mTOR/p70S6K signaling, induced cell transformation and invasion, and remarkably, caused rapid tumor formation and growth in athymic nude mice after subcutaneous inoculation of the transfected cells. This study confirms the oncogenic potential of mTOR suggested previously and demonstrates for the first time its tumorigenicity. Thus, beyond the pivotal position of mTOR to relay the oncogenic signals from the upstream phosphatidylinositol 3-kinase/Akt pathway in human cancer, mTOR is capable potentially of playing a direct role in human tumorigenesis if mutated. These results also further support the conclusion that mTOR is a major therapeutic target in human cancers.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Mutation, Missense , Proto-Oncogene Proteins/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Amino Acid Substitution , Animals , Cell Line , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Humans , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Transplantation , Protein Structure, Tertiary , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , TOR Serine-Threonine Kinases/genetics , Transplantation, Heterologous
10.
Lancet ; 381(9871): 1058-69, 2013 Mar 23.
Article in English | MEDLINE | ID: mdl-23668556

ABSTRACT

Substantial developments have occurred in the past 5-10 years in clinical translational research of thyroid cancer. Diagnostic molecular markers, such as RET-PTC, RAS, and BRAF(V600E) mutations; galectin 3; and a new gene expression classifier, are outstanding examples that have improved diagnosis of thyroid nodules. BRAF mutation is a prognostic genetic marker that has improved risk stratification and hence tailored management of patients with thyroid cancer, including those with conventionally low risks. Novel molecular-targeted treatments hold great promise for radioiodine-refractory and surgically inoperable thyroid cancers as shown in clinical trials; such treatments are likely to become a component of the standard treatment regimen for patients with thyroid cancer in the near future. These novel molecular-based management strategies for thyroid nodules and thyroid cancer are the most exciting developments in this unprecedented era of molecular thyroid-cancer medicine.


Subject(s)
Biomarkers, Tumor/blood , Carcinoma/therapy , Molecular Targeted Therapy/trends , Thyroid Neoplasms/therapy , Carcinoma/diagnosis , Carcinoma/genetics , Carcinoma, Papillary , Early Detection of Cancer , Genes, Neoplasm/genetics , Genetic Markers , Genetic Testing/methods , Humans , Molecular Targeted Therapy/methods , Mutation/genetics , Prognosis , Thyroid Cancer, Papillary , Thyroid Neoplasms/diagnosis , Thyroid Neoplasms/genetics
11.
Tumour Biol ; 35(11): 11375-9, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25119588

ABSTRACT

This study demonstrates a strong association of high urinary iodine with thyroid nodules and papillary thyroid cancer as well as aggressive cancer features, suggesting that high urinary iodine is a risk factor for thyroid cancer. The risk of high iodine intake for thyroid cancer has been suggested but not established. The objective of the study was to evaluate the relationship between urine iodine levels and thyroid nodule and thyroid cancer. We preoperatively tested fasting urine iodine in 154 thyroid nodule patients and correlated the results with pathological diagnoses and compared with 306 subjects as normal control. The median urine iodine (MUI) was 331.33 µg/L in patients with benign thyroid nodules versus 466.23 µg/L in patients with papillary thyroid cancer (PTC) (P=0.003), both of which were in the excessive iodine state and higher than the MUI of 174.30 µg/L in the control group (P < 0.001), which was in the sufficient iodine state. Excessive iodine state (MUI>300 µg/L) was seen in 62.75% of patients with benign thyroid nodules and 66.99% of patients with PTC, both of which were significantly higher than the iodine excessive rate of 19.93% in the control group (P<0.001). Moreover, MUI in patients with PTC with lymph node metastasis was significantly higher than that of PTC patients without lymph node metastasis (P<0.001). Urine iodine of thyroid cancer patients with stage III and IV disease was significantly higher than that of patients with stage I and II diseases (P<0.001). Multivariable analyses showed that, like sand calcification of thyroid nodule and TSH, urine iodine was an independent risk factor for PTC. These data demonstrate a significant association between high urinary iodine and benign and malignant thyroid nodules and PTC aggressiveness, supporting high urinary iodine as a risk factor for thyroid malignancy. Further studies are warranted to confirm these findings.


Subject(s)
Biomarkers, Tumor/urine , Carcinoma, Papillary/urine , Iodine/urine , Thyroid Neoplasms/urine , Thyroid Nodule/urine , Adolescent , Carcinoma, Papillary/pathology , Case-Control Studies , Female , Follow-Up Studies , Humans , Male , Middle Aged , Neoplasm Staging , Prognosis , Thyroid Neoplasms/pathology , Thyroid Nodule/pathology
12.
J Clin Endocrinol Metab ; 109(5): 1222-1230, 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38064679

ABSTRACT

CONTEXT: The clinical relevance of the well-known association between thyroid cancer (TC) and breast cancer (BC) remains to be further defined. OBJECTIVE: This work aimed to investigate the effect of history of BC on the prognosis of TC. METHODS: This was a comparative cohort study of tumor behaviors and TC-specific mortality in 5598 patients with papillary thyroid cancer (PTC) and 604 patients with follicular thyroid cancer (FTC), all with a history of BC (TC-BC patients), and their propensity score-matched TC patients without a history of BC (TCnoBC patients) in Surveillance, Epidemiology and End Results (SEER) 18. The main outcome measure was TC-specific mortality. RESULTS: Lower TC distant metastasis rates of 2.4% vs 3.0% in PTC and 6.1% vs 9.1% in FTC and TC-specific mortality rates of 1.3% vs 2.6% in PTC and 5.8% vs 8.4% in FTC were found in TC-BC patients vs matched TCnoBC patients (all P < .05). Comparing TC-BC patients with matched TCnoBC patients, hazard ratios (HRs) for mortality were 0.472 (95% CI, 0.370-0.601) in PTC and 0.656 (95% CI, 0.461-0.934) in FTC (all P < .05). Such HRs for mortality in PTC were 0.397 (95% CI, 0.268-0.588; P < .001) when TC occurred before BC vs 0.607 (95% CI, 0.445-0.827; P = .002) when BC occurred before TC. CONCLUSION: This study demonstrates a robust protective effect of a history of BC on TC-specific patient survival, which has strong implications for more precise prognostication of TC in such patients.

13.
Article in English | MEDLINE | ID: mdl-38735658

ABSTRACT

PURPOSE: To investigate the role of coexisting RET/PTC rearrangement and TERT promoter mutation in the prognosis and therapeutic targeting in papillary thyroid cancer (PTC). METHODS: A total of 669 PTC patients with complete clinical follow-up and genetic data were pooled from thyroid cancer datasets TCGA, MSK MetTropism, and MSK-IMPACT, from whom 163 patients (112 women and 47 men, 4 unknown) with wild-type BRAF/RAS were identified, with median age (IQR) of 46.00 (33.00, 61.00) years and median follow-up time (IQR) of 16.13 (8.09, 27.91) months for comparative genotype cohort analysis of mortality. RESULTS: There was a significant concurrence index between RET/PTC and TERT promoter mutations, being 2.040 (95% CI 1.110-3.747, P = 0.023). Mortality occurred in 5/100 (5%) patients harboring neither mutation, 2/18 (11.1%) patients harboring TERT promoter mutation alone, 0/31 (0%) patients harboring RET/PTC alone, and 7/14 (50%) patients harboring both genetic alterations, corresponding to HRs (95% CI) of 1 (Reference), 2.469 (0.405-14.02), 3.296e-09 (0-inf), and 9.019 (2.635-30.87), respectively, which remained essentially unchanged after adjustment for patient race, sex, and age. Similar results were observed with BRAF/RAS and TERT promoter mutations. Mechanistically, RET/PTC used the MAP kinase pathway to upregulate the mutated TERT, but not the wild-type TERT, and, correspondingly, targeting RET and MEK could suppress mutated TERT but not the wild-type TERT. CONCLUSION: Coexisting RET/PTC and TERT promoter mutation identify PTC as a unique clinical entity with high mortality, providing new implications for genetic-based prognostication and potential therapeutic targeting of RET and MEK guided by RET/PTC and TERT status.

14.
J Natl Cancer Inst ; 116(5): 694-701, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38113409

ABSTRACT

BACKGROUND: BRAF V600E and TERT promoter alterations are core components in current genetics-based risk assessment for precision management of papillary thyroid cancer. It remains unknown whether this approach could achieve even better precision through a widely recognized prognostic single-nucleotide variation (SNV, formerly SNP), rs2853669T>C, in the TERT promoter. METHODS: The genetic status of alterations and SNV were examined by sequencing genomic DNA from papillary thyroid cancer in 608 patients (427 women and 181 men) aged 47 years (interquartile range = 37-57), with a median follow-up time of 75 months (interquartile range = 36-123), and their relationship with clinical outcomes was analyzed. A luciferase reporter assay was performed to examine TERT promoter activities. RESULTS: TERT promoter alterations showed a strong association with papillary thyroid cancer recurrence in the presence of genotype TT of rs2853669 (adjusted hazard ratio [HR] = 2.12, 95% confidence interval [CI] = 1.10 to 4.12) but not TC/CC (adjusted HR = 1.17, 95% CI = 0.56 to 2.41). TERT and BRAF alterations commonly coexisted and synergistically promoted papillary thyroid cancer recurrence. With this genetic duet, TT of rs2853669 showed a robustly higher disease recurrence than TC/CC (adjusted HR = 14.26, 95% CI = 2.86 to 71.25). Patients with the genetic trio of BRAF V600E, TERT alteration, and TT of rs2853669 had a recurrence of 76.5% vs recurrence of 8.4% with neither variation and with TC/CC (HR = 13.48, 95% CI = 6.44 to 28.21). T allele of rs2853669 strongly increased TERT promoter activities, particularly the variant promoters. CONCLUSIONS: The SNV rs2853669T>C dramatically refines the prognostic power of BRAF V600E and TERT promoter alterations to a higher precision, suggesting the need for including this SNV in the current genetics-based risk prognostication of papillary thyroid cancer.


Subject(s)
Proto-Oncogene Proteins B-raf , Telomerase , Thyroid Cancer, Papillary , Thyroid Neoplasms , Adult , Female , Humans , Male , Middle Aged , Carcinoma, Papillary/genetics , Carcinoma, Papillary/pathology , Genotype , Mutation , Neoplasm Recurrence, Local/genetics , Polymorphism, Single Nucleotide , Prognosis , Promoter Regions, Genetic , Proto-Oncogene Proteins B-raf/genetics , Telomerase/genetics , Thyroid Cancer, Papillary/genetics , Thyroid Cancer, Papillary/pathology , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology
15.
JAMA ; 309(14): 1493-501, 2013 Apr 10.
Article in English | MEDLINE | ID: mdl-23571588

ABSTRACT

IMPORTANCE: BRAF V600E is a prominent oncogene in papillary thyroid cancer (PTC), but its role in PTC-related patient mortality has not been established. OBJECTIVE: To investigate the relationship between BRAF V600E mutation and PTC-related mortality. DESIGN, SETTING, AND PARTICIPANTS: Retrospective study of 1849 patients (1411 women and 438 men) with a median age of 46 years (interquartile range, 34-58 years) and an overall median follow-up time of 33 months (interquartile range, 13-67 months) after initial treatment at 13 centers in 7 countries between 1978 and 2011. MAIN OUTCOMES AND MEASURES: Patient deaths specifically caused by PTC. RESULTS: Overall, mortality was 5.3% (45/845; 95% CI, 3.9%-7.1%) vs 1.1% (11/1004; 95% CI, 0.5%-2.0%) (P < .001) in BRAF V600E-positive vs mutation-negative patients. Deaths per 1000 person-years in the analysis of all PTC were 12.87 (95% CI, 9.61-17.24) vs 2.52 (95% CI, 1.40-4.55) in BRAF V600E-positive vs mutation-negative patients; the hazard ratio (HR) was 2.66 (95% CI, 1.30-5.43) after adjustment for age at diagnosis, sex, and medical center. Deaths per 1000 person-years in the analysis of the conventional variant of PTC were 11.80 (95% CI, 8.39-16.60) vs 2.25 (95% CI, 1.01-5.00) in BRAF V600E-positive vs mutation-negative patients; the adjusted HR was 3.53 (95% CI, 1.25-9.98). When lymph node metastasis, extrathyroidal invasion, and distant metastasis were also included in the model, the association of BRAF V600E with mortality for all PTC was no longer significant (HR, 1.21; 95% CI, 0.53-2.76). A higher BRAF V600E-associated patient mortality was also observed in several clinicopathological subcategories, but statistical significance was lost with adjustment for patient age, sex, and medical center. For example, in patients with lymph node metastasis, the deaths per 1000 person-years were 26.26 (95% CI, 19.18-35.94) vs 5.93 (95% CI, 2.96-11.86) in BRAF V600E-positive vs mutation-negative patients (unadjusted HR, 4.43 [95% CI, 2.06-9.51]; adjusted HR, 1.46 [95% CI, 0.62-3.47]). In patients with distant tumor metastasis, deaths per 1000 person-years were 87.72 (95% CI, 62.68-122.77) vs 32.28 (95% CI, 16.14-64.55) in BRAF V600E-positive vs mutation-negative patients (unadjusted HR, 2.63 [95% CI, 1.21-5.72]; adjusted HR, 0.84 [95% CI, 0.27-2.62]). CONCLUSIONS AND RELEVANCE: In this retrospective multicenter study, the presence of the BRAF V600E mutation was significantly associated with increased cancer-related mortality among patients with PTC. Because overall mortality in PTC is low and the association was not independent of tumor features, how to use BRAF V600E to manage mortality risk in patients with PTC is unclear. These findings support further investigation of the prognostic and therapeutic implications of BRAF V600E status in PTC.


Subject(s)
Carcinoma/genetics , Carcinoma/mortality , DNA Mutational Analysis , Proto-Oncogene Proteins B-raf/genetics , Thyroid Neoplasms/genetics , Thyroid Neoplasms/mortality , Adult , Aged , Carcinoma/pathology , Carcinoma, Papillary , Female , Humans , Lymphatic Metastasis , Male , Middle Aged , Mutation , Neoplasm Invasiveness , Retrospective Studies , Risk , Thyroid Cancer, Papillary , Thyroid Neoplasms/pathology
16.
Oncogene ; 42(20): 1634-1648, 2023 05.
Article in English | MEDLINE | ID: mdl-37020036

ABSTRACT

The classical oxidizing enzymatic activity of Ten Eleven Translocation 1 (TET1) and its tumor suppressor role are well known. Here, we find that high TET1 expression is associated with poor patient survival in solid cancers often having hypoxia, which is inconsistent with its tumor suppressor role. Through a series of in vitro and in vivo studies, using thyroid cancer as a model, we demonstrate that TET1 plays a tumor suppressor function in normoxia and, surprisingly, an oncogenic function in hypoxia. Mechanistically, TET1 mediates HIF1α-p300 interaction by acting as a co-activator of HIF1α to promote CK2B transcription under hypoxia, which is independent of its enzymatic activity; CK2 activates the AKT/GSK3ß signaling pathway to promote oncogenesis. Activated AKT/GSK3ß signaling in turn maintains HIF1α at elevated levels by preventing its K48-linked ubiquitination and degradation, creating a feedback loop to enhance the oncogenicity of TET1 in hypoxia. Thus, this study uncovers a novel oncogenic mechanism in which TET1 promotes oncogenesis and cancer progression through a non-enzymatic interaction between TET1 and HIF1α in hypoxia, providing novel therapeutic targeting implications for cancer.


Subject(s)
Carcinogenesis , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins , Humans , Carcinogenesis/genetics , Cell Hypoxia/genetics , Cell Line, Tumor , Glycogen Synthase Kinase 3 beta/metabolism , Hypoxia/genetics , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism
17.
Thyroid ; 33(9): 1064-1077, 2023 09.
Article in English | MEDLINE | ID: mdl-37416988

ABSTRACT

Background: Thyrotropin receptor (TSHR) plays a central role in maintaining thyroid function and TSHR impairment causes hypothyroidism, which is often associated with metabolic disarrangement. The most common type of hypothyroidism is autoimmune disease-related and the mechanism, particularly with respect to the role of microRNAs (miRNAs), has not been delineated. Methods: Serum from 30 patients with subclinical hypothyroidism (SCH) and 30 healthy individuals were collected and exosomal miR-146a (exo-miR-146a) was examined, followed by extensive mechanistic investigation using various molecular and cellular experimental approaches and genetic-knockout mouse models. Results: Our clinical investigation showed that exo-miR-146a was systemically elevated in the serum of patients with SCH (p = 0.04) compared with healthy individuals, prompting us to investigate the biological effects of miR-146a in cells. We found that miR-146a could target and down-regulate neuron-glial antigen 2 (Ng2), with consequent down-regulation of TSHR. We next generated a thyroid-specific Ng2 knockout (Thy-Ng2-/-) mouse model and found a significant down-regulation of TSHR in Thy-Ng2-/- mice, accompanied by the development of hypothyroidism and metabolic disorders. We further found that a decrease in NG2 resulted in decreased receptor tyrosine kinase-linked downstream signaling and down-regulation of c-Myc, consequently resulting in up-regulation of miR-142 and miR-146a in thyroid cells. Up-regulated miR-142 targeted the 3'-untranslated region (UTR) of TSHR messenger RNA (mRNA) and post-transcriptionally down-regulated TSHR, explaining the development of hypothyroidism above. Local up-regulation of miR-146a in thyroid cells augments the earlier cited processes initiated by systemically elevated miR-146a, thereby forming a feedback loop to propel the development and progression of hypothyroidism. Conclusions: This study has uncovered a self-augmenting molecular loop initiated by elevated exo-miR-146a to suppress TSHR through targeting and down-regulating NG2, thereby initiating and propelling the development and progression of hypothyroidism.


Subject(s)
Hypothyroidism , MicroRNAs , Animals , Mice , Feedback , Hypothyroidism/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Receptors, Thyrotropin/genetics
18.
J Clin Oncol ; 41(6): 1296-1306, 2023 02 20.
Article in English | MEDLINE | ID: mdl-36378996

ABSTRACT

PURPOSE: To explore the novel diagnostic value of epigenetic imprinting biomarkers in thyroid nodules. PATIENTS AND METHODS: A total of 550 patients with fine-needle aspiration (FNA)-evaluated and histopathologically confirmed thyroid nodules were consecutively recruited from eight medical centers. Quantitative chromogenic imprinted gene in situ hybridization (QCIGISH) was used to assess the allelic expression of imprinted genes SNRPN and HM13, on the basis of which a diagnostic grading model for thyroid nodules was developed. The model was retrospectively trained on 124 postsurgical thyroid samples, optimized on 32 presurgical FNA samples, and prospectively validated on 394 presurgical FNA samples. Blinded central review-based cytopathologic and histopathologic diagnoses were used as the reference standard. RESULTS: For thyroid malignancy, the QCIGISH test achieved an overall diagnostic sensitivity of 100% (277/277), a specificity of 91.5% (107/117; 95% CI, 86.4 to 96.5), a positive predictive value (PPV) of 96.5% (95% CI, 94.4 to 98.6), and a negative predictive value (NPV) of 100% in the prospective validation, with a diagnostic accuracy of 97.5% (384/394; 95% CI, 95.9 to 99.0). QCIGISH demonstrated a PPV of 97.8% (95% CI, 94.7 to 100) and NPV of 100%, with a diagnostic accuracy of 98.2% (111/113; 95% CI, 95.8 to 100), for indeterminate Bethesda III-V thyroid nodules. QCIGISH demonstrated a PPV of 96.6% (95% CI, 91.9 to 100) and a NPV of 100%, with a diagnostic accuracy of 97.5% (79/81; 95% CI, 94.2 to 100), for Bethesda III-IV. For Bethesda VI, QCIGISH showed a 100% (184/184) accuracy. CONCLUSION: This imprinting biomarker-based test can effectively distinguish malignant from benign thyroid nodules. The high PPV and NPV make the test both an excellent rule-in and rule-out diagnostic tool. With such a diagnostic performance and its technical simplicity, this novel thyroid molecular test is clinically widely applicable.


Subject(s)
Thyroid Neoplasms , Thyroid Nodule , Humans , Thyroid Nodule/diagnosis , Thyroid Nodule/genetics , Thyroid Nodule/pathology , Retrospective Studies , Thyroid Neoplasms/diagnosis , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Biomarkers , Epigenesis, Genetic
19.
Biochem Biophys Res Commun ; 425(1): 45-50, 2012 Aug 17.
Article in English | MEDLINE | ID: mdl-22820187

ABSTRACT

We have previously reported that the BRAFV600E signaling causes genome-wide aberrations in gene methylation in melanoma cells. To explore the potential molecular mechanisms for this epigenetic effect of BRAFV600E, in this in silico study we analyzed 11 microarray datasets retrieved from NCBI GEO database and examined the relationship of the expression of the epigenetic genes (genes involved in epigenetic regulation) with BRAFV600E signaling, methylation and expression of tumor-suppressor genes (TSGs) in melanoma, and patient survival with this cancer. Among 273 epigenetic genes examined, 12 genes were down-regulated (named DD genes) and 16 were up-regulated (UU genes) by suppression of the BRAFV600E signaling using inhibitors. While the expression of 245 non-DD/UU genes overall had no correlation with the expression and methylation of a set of potential TSGs, the expression of DD genes was significantly correlated negatively with the TSG expression and positively with TSG methylation. Expression of UU genes was positively, albeit weakly, associated with the TSG expression. Overall, no correlation was found between UU gene expression and TSG methylation. Importantly, the expression of DD genes, but not UU genes, was significantly associated with decreased survival of patients with melanoma. Interestingly, the promoters of DD genes contain more binding motifs of c-fos and myc, two BRAFV600E signaling-related transcription factors, than those of UU and non-DD/UU genes. Thus, these results link epigenetic genes to methylation and suppression of tumor suppressor genes as a mechanism involved in BRAFV600E-promoted melanoma tumorigenesis and uncover a novel molecular signature that predicts a poor prognosis of melanoma.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Melanoma/genetics , Melanoma/mortality , Proto-Oncogene Proteins B-raf/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/mortality , DNA Methylation , Down-Regulation , Genes, Tumor Suppressor , Humans , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction , Up-Regulation
20.
Endocr Rev ; 28(7): 742-62, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17940185

ABSTRACT

In recent years, the T1799A B-type Raf kinase (BRAF) mutation in thyroid cancer has received enthusiastic investigation, and significant progress has been made toward understanding its tumorigenic role and clinical significance. Among various thyroid tumors, this mutation occurs uniquely in papillary thyroid cancer (PTC), the most common endocrine malignancy, and some apparently PTC-derived anaplastic thyroid cancers. Many studies have found this mutation to be associated with those clinicopathological characteristics of PTC that are conventionally known to predict tumor progression and recurrence, including, for example, old patient age, extrathyroidal invasion, lymph node metastasis, and advanced tumor stages. Direct association of BRAF mutation with the clinical progression, recurrence, and treatment failure of PTC has also been demonstrated. The BRAF mutation has even been correlated with PTC recurrence in patients with conventionally low-risk clinicopathological factors. Some molecular mechanisms determining BRAF mutation-promoted progression and the aggressiveness of PTC have recently been uncovered. These include the down-regulation of major tumor suppressor genes and thyroid iodide-metabolizing genes and the up-regulation of cancer-promoting molecules, such as vascular endothelial growth factor, matrix metalloproteinases, nuclear transcription factor kappaB, and c-Met. Thus, BRAF mutation represents a novel indicator of the progression and aggressiveness of PTC. Significant advances have also occurred in the preclinical testing of new therapeutic strategies targeting the MAPK pathway aberrantly activated by BRAF mutation and other related mutations. New mitogen extracellular kinase (MEK) inhibitors developed recently are particularly promising therapeutic agents for thyroid cancer. With these advances, it has become clearer that BRAF mutation will likely have significant impact on the clinical management of PTC.


Subject(s)
Carcinoma, Papillary/genetics , Carcinoma, Papillary/pathology , Proto-Oncogene Proteins B-raf/genetics , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Papillary/drug therapy , Humans , Mutation , Neoplasm Invasiveness , Neoplasm Recurrence, Local/genetics , Prognosis , Risk , Signal Transduction/drug effects , Thyroid Neoplasms/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL