Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Acta Biochim Biophys Sin (Shanghai) ; 53(4): 481-491, 2021 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-33674820

RESUMO

In recent decades, the incidence of thyroid cancer (TC) has rapidly increased, leading us to explore the complex underlying mechanisms. We identified the gene Phospholipase C Delta 3 (PLCD3) as a potential oncogene in TC by conducting the whole transcriptome sequencing. Our study is to understand the oncogenic role of PLCD3 in TC. We verified the overexpression of PLCD3 in TC from The Cancer Genome Atlas, Gene Expression Omnibus databases, and a locally validated cohort. Clinical correlation analysis showed that PLCD3 expression was related to histological type, T stage, lymph node metastasis (LNM), and disease stage. The high expression of PLCD3 could be a distinguishing factor for TC and its LNM. The biological function was examined using small interfering RNA-transfected TC cell lines. Silenced PLCD3 could inhibit colony formation, migration, and invasion ability and promote apoptosis of TC cell lines. PLCD3 silencing reversed the epithelial-mesenchymal transition but induced the apoptotic progress. Further exploration revealed that PLCD3 might be associated with critical genes of the Hippo pathway. The expressions of RHOA, YAP1/TAZ, and their downstream targets were decreased significantly when PLCD3 was down-regulated. YAP1 overexpression rescued the tumor-suppressive effect caused by PLCD3 silencing. This study demonstrates that PLCD3 is an oncogene that supports tumorigenesis and progression in TC, and PLCD3 may be a potential target gene for TC treatment.


Assuntos
Apoptose , Movimento Celular , Proliferação de Células , Proteínas de Neoplasias/metabolismo , Fosfolipase C delta/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Neoplasias da Glândula Tireoide/metabolismo , Linhagem Celular Tumoral , Feminino , Via de Sinalização Hippo , Humanos , Metástase Linfática , Masculino , Invasividade Neoplásica , Proteínas de Neoplasias/genética , Fosfolipase C delta/genética , Proteínas Serina-Treonina Quinases/genética , Neoplasias da Glândula Tireoide/genética , Neoplasias da Glândula Tireoide/patologia
2.
J Cell Mol Med ; 24(22): 13070-13080, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32969138

RESUMO

Recently, the incidence of thyroid cancer is increasing worldwide. Papillary thyroid cancer (PTC) is the most common histological type of thyroid cancer. Whole-transcriptome sequence analysis was performed to further understand the primary molecular mechanisms of the occurrence and progression of PTC. Results showed that Eva-1 homolog A (EVA1A) may be a potential gene for the PTC-associated gene in thyroid cancer. In this work, the role of EVA1A expression in thyroid cancer was investigated. Real-time PCR was performed to detect the expression level of EVA1A in 43 pairs of PTC and four thyroid cancer cell lines. The Cancer Genome Atlas (TCGA) database was used to evaluate the relationship between the expression level of EVA1A and the pathological feature of PTC. The logistic regression analysis of the TCGA data set indicated that the expression of EVA1A was an independent risk factor for tumour, nde and metastasis (TNM) in PTC. This study shows the down-regulation of EVA1A inhibited the colony formation, proliferation, migration and invasion of PTC cell lines. In the protein level, knockdown of EVA1A can regulate the expression of N-cadherin, vimentin, Bcl-xL, Bax, YAP and TAZ. This study indicated that EVA1A was an oncogene associated with PTC.


Assuntos
Proteínas de Membrana/metabolismo , Proteínas Serina-Treonina Quinases , Transdução de Sinais , Câncer Papilífero da Tireoide/metabolismo , Neoplasias da Glândula Tireoide/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adulto , Idoso , Antígenos CD/metabolismo , Apoptose , Caderinas/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Transição Epitelial-Mesenquimal/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Via de Sinalização Hippo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica , Proteínas Serina-Treonina Quinases/metabolismo , Fatores de Risco , Software , Câncer Papilífero da Tireoide/patologia , Neoplasias da Glândula Tireoide/patologia , Fatores de Transcrição/metabolismo , Proteínas com Motivo de Ligação a PDZ com Coativador Transcricional , Vimentina/metabolismo , Proteínas de Sinalização YAP , Proteína X Associada a bcl-2/metabolismo , Proteína bcl-X/metabolismo
3.
J Cell Mol Med ; 24(23): 14059-14072, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33094920

RESUMO

The incidence of thyroid cancer is increasing in recent years worldwide, but the underlying mechanisms await further exploration. We utilized the bioinformatic analysis to discover that Immortalization up-regulated protein (IMUP) could be a potential oncogene in the papillary thyroid cancer (PTC). We verified this finding in several databases and locally validated cohorts. Clinicopathological features analyses showed that high expression of IMUP is positively related to malignant clinicopathological features in PTC. Braf-like PTC patients with higher IMUP expression had shorter disease-free survival. The biological function of IMUP in PTC cell lines (KTC-1 and TPC-1) was investigated using small interfering RNA. Our results showed that silencing IMUP suppresses proliferation, migration and invasion while inducing apoptosis in PTC cell lines. Changes of the expression of apoptosis-related molecules were identified by real-time quantitative polymerase chain reaction and Western blotting. We also found that YAP1 and TAZ, the critical effectors in the Hippo pathway, were down-regulated when the IMUP is silenced. Rescue experiments showed that overexpression of YAP1 reverses the tumour inhibitory effect caused by IMUP knockdown. Our study demonstrated that IMUP has an oncogenic function in PTC and might be a new target gene in the treatment of PTC.


Assuntos
Apoptose , Biomarcadores Tumorais , Transformação Celular Neoplásica/metabolismo , Câncer Papilífero da Tireoide/etiologia , Câncer Papilífero da Tireoide/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adulto , Idoso , Apoptose/genética , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Transformação Celular Neoplásica/genética , Biologia Computacional/métodos , Bases de Dados Genéticas , Suscetibilidade a Doenças , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Metástase Neoplásica , Estadiamento de Neoplasias , Prognóstico , Curva ROC , Câncer Papilífero da Tireoide/mortalidade , Câncer Papilífero da Tireoide/patologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcriptoma , Proteínas de Sinalização YAP
4.
Clin Exp Pharmacol Physiol ; 47(4): 696-702, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31855284

RESUMO

The incidence of thyroid cancer has increased in recent decades. The potential molecular mechanisms of papillary thyroid cancer (PTC) are still to be uncovered. In recent years, a number of studies reported that LRRC super family members are up-regulated in cancer cells. Cancer cells can experience a feature change from an epithelial to a mesenchymal phenotype, which is called epithelial-mesenchymal transition (EMT) during cancer progression. We found that LRRC52-AS1 is highly expressed in PTC cell lines rather than normal tissues and this observation was consistent with The Cancer Genome Atlas (TCGA) cohort. In a word, LRRC52-AS1 is associated with clinical progression in papillary thyroid cancer. In vitro experiments showed that knocking down LRRC52-AS1 significantly decreased the migration and invasion of the PTC cell lines (BCPAP and TPC). Meanwhile, LRRC52-AS1 may influence the progress of papillary thyroid cancer via mesenchymal markers N-cadherin, vimentin and TAZ. The LRRC52-AS1 gene is up-regulated in papillary thyroid cancer, and knockdown of LRRC52-AS1 could restrain cellular migration, and invasion in vitro. This study indicated that LRRC52-AS1 is a gene associated with PTC and might become a potential therapeutic target in PTC.


Assuntos
Movimento Celular/genética , Progressão da Doença , Proteínas de Membrana/genética , RNA Antissenso/genética , Câncer Papilífero da Tireoide/patologia , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal/genética , Humanos , Invasividade Neoplásica/genética , Fenótipo , Câncer Papilífero da Tireoide/genética
5.
J Cell Mol Med ; 23(12): 7974-7984, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31568662

RESUMO

Thyroid cancer is maintaining at a high incidence level and its carcinogenesis is mainly affected by a complex gene interaction. By analysis of the next-generation resequencing of paired papillary thyroid cancer (PTC) and adjacent thyroid tissues, we found that Growth Associated Protein 43 (GAP43), a phosphoprotein activated by protein kinase C, might be novel markers associated with PTC. However, its function in thyroid carcinoma has been poorly understood. We discovered that GAP43 was significantly overexpressed in thyroid carcinoma and these results were consistent with that in The Cancer Genome Atlas (TCGA) cohort. In addition, some clinicopathological features of GAP43 in TCGA database showed that up-regulated GAP43 is significantly connected to lymph node metastasis (P < 0.001) and tumour size (P = 0.038). In vitro experiments, loss of function experiments was performed to investigate GAP43 in PTC cell lines (TPC-1 and BCPAP). The results proved that GAP43 knockdown in PTC cell significantly decreased the function of cell proliferation, colony formation, migration, and invasion and induced cell apoptosis. Furthermore, we also indicated that GAP43 could modulate the expression of epithelial-mesenchymal transition-related proteins, which could influence invasion and migration. Put those results together, GAP43 is a gene which was associated with PTC and might be a potential therapeutic target.


Assuntos
Proliferação de Células/genética , Transição Epitelial-Mesenquimal/genética , Proteína GAP-43/metabolismo , Metástase Linfática , Câncer Papilífero da Tireoide/metabolismo , Neoplasias da Glândula Tireoide/metabolismo , Adulto , Apoptose/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Estudos de Coortes , Bases de Dados Genéticas , Feminino , Proteína GAP-43/genética , Regulação Neoplásica da Expressão Gênica/genética , Técnicas de Silenciamento de Genes , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica/genética , Estadiamento de Neoplasias , RNA Interferente Pequeno , Fatores de Risco , Câncer Papilífero da Tireoide/genética , Câncer Papilífero da Tireoide/patologia , Neoplasias da Glândula Tireoide/genética , Neoplasias da Glândula Tireoide/patologia , Neoplasias da Glândula Tireoide/secundário
6.
Histol Histopathol ; : 18786, 2024 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-39005145

RESUMO

PURPOSE: Our previous study demonstrated that NRF3 (NFE2L3, Nuclear Factor-erythroid 2-related factor 3) could suppress cell metastasis and proliferation in breast cancer. In this study, we investigated the mechanisms underlying its function in breast cancer. METHODS: In the present study, NRF3 expression and its clinical characteristics in breast cancer were analyzed using public datasets and clinical specimens. After breast cancer cells were overexpressed NRF3, FACS was used to detect the intracellular ROS levels. The migration and invasion activities of NRF3-ectopic expressed breast cancer cells were determined by transwell assay. To validate the role of ROS/ERK axis in the inhibitory effect of NRF3 in cell metastasis, ROS scavenger NAC was also included. RESULTS: We found that NRF3 mRNA was highly expressed, while NRF3 protein was extremely lowly expressed in breast cancer tissues compared with their normal counterparts, and low level NRF3 was associated with poorer prognosis in patients with triple negative breast cancer (TNBC). More interestingly, overexpression of NRF3 protein significantly increased cellular ROS production and dramatically decreased p-ERK level and cell migration in TNBC cells. Mechanistically, NRF3 protein was found to be mutually regulated by valosin-containing protein (VCP). Strikingly, VCP-knockdown dramatically increased NRF3 protein expression, but NRF3-knockin also decreased VCP expression in return. Moreover, antioxidant NAC treatment effectively increased the level of p-ERK and VCP expression, as well as cell migration and invasion abilities of TNBC cells. CONCLUSION: NRF3, a tumor suppressor downregulated by VCP, could attenuate cell metastasis in TNBC cells by increasing cellular ROS accumulation and subsequently inhibiting the ERK phosphorylation.

7.
Discov Med ; 35(174): 45-56, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-37024441

RESUMO

BACKGROUND: The aberrant expression of adipocyte enhancer binding protein 1 (AEBP1) has been observed in many cancers and it seems to be involved in the tumorigenesis, progression, and metastasis in numerous tumor types. However, the contribution of AEBP1 in breast cancer (BCa) remains inexplicable. METHODS: Information related to the diagnostic significance and expression of AEBP1 in BCa was obtained from the public dataset Kaplan-Meier Plotter (http://kmplot.com/analysis/) and the dataset UALCAN (https://ualcan.path.uab.edu/index.html). The MTT (methyl thiazolyl tetrazolium) assay, colony formation assay, Transwell® assay, and FACS (fluorescence-activated cell sorting) assay were used to detect the proliferation, invasive and apoptotic ability of cells before and after treatment. In addition, we constructed an AEBP1 overexpression vector and silenced AEBP1, combined with Real-Time Quantitative Reverse Transcription PCR (qRT-PCR), western blot, immunohistochemistry and TUNEL (terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling) assay to investigate the prognostic significance, biological functions and potential mechanisms of AEBP1 in BCa. RESULTS: Higher expression of AEBP1 mRNA (message RNA) was observed in BCa patients with later-stage, who obtained poorer overall survival. Meanwhile, compared with adjacent noncancerous tissues, AEBP1 protein expression was dramatically upregulated in the BCa ones. Furthermore, overexpressed AEBP1 enhanced cell proliferation, migration, invasion, and blocked cell apoptosis in BCa cells. Moreover, the research certificated that AEBP1 upregulated the expression of MMP (matrix metalloproteinase)-2, 9, vimentin, N-cadherin (neural-cadherin), phosphorylation of ERK (extracellular signal-regulated kinase), Smad2/3 (Abbreviated from Sma for nematode and Mad for Drosophila) and AKT (V-akt murine thymoma viral oncogene homolog), while down-regulated the expression of E-cadherin (epithelial cadherin) and PTEN (phosphatase and tensin homolog deleted on chromosome 10). To inhibit cell apoptosis, enforced expression of AEBP1 effectively blocked the cleavage of caspase 9 and p53 (protein 53) and promoted the expression of anti-apoptotic protein Bcl-2 (B-cell lymphoma-2). Finally, AEBP1 accelerated subcutaneously transplanted tumor growth in nude mice by increasing the expression of the cell proliferation biomarker ki67, the phosphorylation of AKT, and blocked apoptosis in vivo. CONCLUSIONS: In summary, these data suggested the important role of AEBP1 in the BCa progression, which could be used as a potential biomarker for prognostic hallmark and a novel therapeutic strategy.


Assuntos
Apoptose , Proteínas Proto-Oncogênicas c-akt , Animais , Camundongos , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Transição Epitelial-Mesenquimal , Regulação Neoplásica da Expressão Gênica , Camundongos Nus , Invasividade Neoplásica/genética , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo
8.
Artigo em Inglês | MEDLINE | ID: mdl-36089788

RESUMO

AIMS: AHNAK2 may be used as a candidate marker for TC diagnosis and treatment. BACKGROUND: Thyroid cancer [TC] is the most frequent malignancy in endocrine carcinoma, and the incidence has been increasing for decades. OBJECTIVE: To understand the molecular mechanism of DTC, we performed next-generation sequencing [NGS] on 79 paired DTC tissues and normal thyroid tissues. The RNA-sequencing [RNA-seq] data analysis results indicated that AHNAK nucleoprotein 2 [AHNAK2] was significantly upregulated in the thyroid cancer patient's tissue. METHOD: We also analyzed AHNAK2 mRNA levels of DTC tissues and normal tissues from The Cancer Genome Atlas [TCGA]. The association between the expression level of AHNAK2 and clinicopathological features was evaluated in the TCGA cohort. Furthermore, AHNAK2 gene expression was analyzed by quantitative real-time polymerase chain reaction [qRT-PCR] in 40 paired DTC tissues and adjacent normal thyroid tissues. The receiver operating characteristic [ROC] curve was performed to evaluate the diagnostic value of AHNAK2. For cell experiments in vitro, AHNAK2 was knocked down using small interfering RNA [siRNA], and the biological function of AHNAK2 in TC cell lines was investigated. The expression of AHNAK2 was significantly upregulated in both the TCGA cohort and the local cohort. RESULT: The analysis results of the TCGA cohort indicated that the upregulation of AHNAK2 was associated with tumor size [P<0.001], lymph node metastasis [P<0.001], and disease stage [P<0.001]. The area under the curve [AUC] [TCGA: P<0.0001; local validated cohort: P<0.0001.] in the ROC curve revealed that AHNAK2 might be considered a diagnostic biomarker for TC. The knockdown of AHNAK2 reduced TC cell proliferation, colony formation, migration, invasion, cell cycle, and induced cell apoptosis. CONCLUSION: Furthermore, the protein levels of phospho-PI3 Kinase p85 and phospho-AKT were down-regulated in the transfected TC cell. Our study results indicate that AHNAK2 may promote metastasis and proliferation of thyroid cancer through PI3K/AKT signaling pathway. Thus, AHNAK2 may be used as a candidate marker for TC diagnosis and treatment.

9.
J Cancer ; 13(2): 508-516, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35069898

RESUMO

Thyroid cancer is a disease with an extremely high incidence rate and is divided into papillary, follicular, medullary, and undifferentiated thyroid cancers. Among them, papillary carcinoma is the most common subtype. We assessed expression of ETNK2 in public databases and found that ETNK2 is upregulated in PTC. Cohort and RNA sequencing data were used to verify this discovery. To further determine the relationship between ETNK2 and papillary thyroid carcinoma, we performed an in vitro experiment. In a PTC cell line, silencing ETNK2 inhibited cell proliferation, weakened cell migration and invasion ability, promoted apoptosis, and blocked the cell cycle. In addition, western blotting suggested that ETNK2 is related to the HIPPO pathway and may activate the EMT pathway through the HIPPO pathway to promote the development of thyroid cancer. These results revealed that ETNK2 is related to the occurrence and development of papillary thyroid carcinoma, suggesting that ETNK2 may be an oncogene associated with PTC.

10.
Pathol Res Pract ; 238: 154091, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36057192

RESUMO

Thyroid cancer is a common malignant tumor for the adult and the potential molecular mechanism of papillary thyroid cancer cell metastasis is still unclear. We used sequencing techniques to analyze paired papillary thyroid carcinoma (PTC) and adjacent thyroid tissue and identified a gene, PDZK1IP1, that was significantly overexpressed in thyroid cancer. We found It has been detected to play an important role in many malignant tumors. But the role in papillary thyroid cancer was still unknown, we decided to find a new marker and therapeutic target for the disease. The present study shows that PDZK1IP1 may be a potential gene that leads to thyroid cancer. In our study, silencing PDZK1IP1 can inhibit PTC cell proliferation, migration, invasion, apoptosis, and cell cycle arrest. This study surmised that PDZK1IP1 was an oncogene that correlated with tumor development.

11.
Transl Cancer Res ; 10(9): 4033-4046, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35116701

RESUMO

BACKGROUND: Long non-coding RNAs (lncRNAs) play an important part in tumorigenesis and cancer metastasis and can serve as a potential biosignature for cancer prognosis. However, the use of lncRNA signatures to predict survival in breast carcinoma is yet unreported. METHODS: The lncRNA expression profiles and homologous clinical data of 913 breast carcinoma samples from the Cancer Genome Atlas (TCGA), were analyzed to obtain 2,547 differentially expressed lncRNAs. Univariate Cox proportional risk regression was applied to both the training and testing datasets to screen the common prognostic lncRNAs. Potential prognostic LncRNAs were screened by multivariate Cox proportional risk regression in the training data set of the selected LncRNAs. RESULTS: Seven lncRNAs (LINC02037, MAPT-AS1, RP1-37C10.3, RP11-344E13.4, RP11-454P21.1, RP11-616M22.1, SPACA6P-AS) were prominently associated with overall survival. Kaplan-Meier analysis and receiver operating characteristic (ROC) curves indicated that these indicators were sensitive and specific for survival prediction. The areas under the ROC curve of the seven-lncRNA signature in predicting 3- and 5-year survival rates were 0.771 and 0.780 respectively in the combined cohort. Furthermore, enrichment analysis revealed that these seven lncRNAs might participate multiple pathways related to tumorigenesis and prognosis. CONCLUSIONS: The proposed seven-lncRNA signature could serve as a latent prognostic biomarker for survival prediction in patients with breast carcinoma.

12.
Oncol Lett ; 21(6): 442, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33868480

RESUMO

Thyroid cancer (TC) is the most common type of endocrine malignancy in humans, and its relative incidence has increased continuously in recent years. However, the primary molecular mechanisms of thyroid tumorigenesis and progression remain unclear. Papillary TC (PTC) is the most common subtype of TC. Recent studies have reported that one of the tumorigenesis and progression mechanisms is driven by genetic alterations that regulate the TC cell signaling pathway. In the present study, RNA sequencing (RNA-seq) was performed on 79 paired PTC and adjacent normal thyroid tissues to further study the molecular mechanisms of TC. Reverse transcription-quantitative PCR was used to detect the expression levels of LEM domain containing 1 (LEMD1) in 47 paired PTC and adjacent normal thyroid tissue samples. Initial analysis revealed that LEMD1 expression was significantly upregulated in TC tissues compared with that in normal tissues. The results of the thyroid RNA-seq datasets from The Cancer Genome Atlas were consistent with the RNA-seq analysis results of the present study. High LEMD1 expression increased the risk of lymph node metastasis in patients with TC. The biological function of LEMD1 on cell proliferation, migration, invasion and apoptosis was investigated in vitro via small interfering RNA and overexpression vector. Gene set enrichment analysis indicated that high LEMD1 expression was associated with epithelial-mesenchymal transition (EMT) and the Wnt/ß-catenin signaling pathway. Western blotting revealed that LEMD1 modulated the protein expression levels of E-cadherin, N-cadherin, vimentin, ß-catenin and cleaved-caspase 3. In conclusion, the present results indicated that LEMD1 may drive TC cell tumorigenesis and progression by activating the Wnt/ß-catenin signaling pathway and EMT.

13.
Hum Cell ; 34(3): 901-917, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33665786

RESUMO

Breast cancer (BC) threatened the life health of a tremendous amount of the population, and the estimated number of death is still rising nowadays. We found that stress-induced phosphoprotein 1 (STIP1) is overexpressed in BC tissues compared to non-tumorous breast tissues. Our study is to validate the prognostic value of STIP1 and investigate its biological role in BC. We verified the upregulation of STIP1 in multiple databases, proved that STIP1 is upregulated in BC tissues and cell lines using real-time quantitative PCR (qRT-PCR). We used small interfering RNA to examine the function of STIP1 in BC cell lines (BT-549, MDA-MB-231, Hs-578 T) and explored the mechanism of function of STIP1 in BC cells using Western blotting and qRT-PCR. Analyses of multiple databases indicated that high STIP1 expression is a marker that effectively distinguishes BC patients from healthy control and predicts worse clinical outcomes in BC. The loss-of-function experiments showed that STIP1 silencing results in inhibition of cell proliferation and migration, inducing cell apoptosis, and S-phase arrest in vitro. Our study also showed that STIP1 downregulation inhibited the JAK2/STAT3 pathway and epithelial-mesenchymal transition process. Rescue experiments demonstrated that the oncogenic effect of STIP1 is partially dependent on mediating JAK2 expression. This study verified that STIP1 is an oncogenic gene that promotes BC progression and serves as a valuable diagnostic and outcome-related marker of BC.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Regulação Neoplásica da Expressão Gênica/genética , Expressão Gênica/genética , Proteínas de Choque Térmico/fisiologia , Apoproteínas/genética , Biomarcadores Tumorais , Neoplasias da Mama/diagnóstico , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Progressão da Doença , Feminino , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Janus Quinase 2/metabolismo , Prognóstico , Fase S/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
14.
Aging (Albany NY) ; 12(16): 16437-16456, 2020 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-32857728

RESUMO

The incidence of thyroid cancer remains high worldwide, and papillary thyroid cancer (PTC) is the most common type. Potassium Calcium-Activated Channel Subfamily N Member 4 (KCNN4) has been reported as an oncogene in various cancers. We examined expression of KCNN4 in public databases and discovered that it is upregulated in PTC. We verified this finding using our own validated cohort and RNA sequencing data. We also found that KCNN4 is a diagnostic and prognostic biomarker that is associated with disease-free survival, immune infiltration, and several other clinicopathological features of PTC. Gene Set Enrichment Analysis indicated that apoptotic and epithelial-mesenchymal transition gene sets are both upregulated in PTC patients with higher KCNN4 levels. In PTC cell lines, silencing KCNN4 inhibited cell proliferation, migration and invasion. Moreover, quantitative real-time PCR and Western blotting indicated that silencing KCNN4 increased expression of apoptotic genes in PTC cells and reduced the expression of genes involved in their epithelial-mesenchymal transition. These results suggest that KCNN4 promotes PTC progression by inducing epithelial-mesenchymal transition and suppressing apoptosis, which suggests KCNN4 may be a useful diagnostic and prognostic biomarker of PTC.


Assuntos
Biomarcadores Tumorais/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Câncer Papilífero da Tireoide/metabolismo , Neoplasias da Glândula Tireoide/metabolismo , Apoptose , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Biomarcadores Tumorais/genética , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Bases de Dados Genéticas , Progressão da Doença , Intervalo Livre de Doença , Transição Epitelial-Mesenquimal , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/genética , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica , Transdução de Sinais , Câncer Papilífero da Tireoide/genética , Câncer Papilífero da Tireoide/patologia , Câncer Papilífero da Tireoide/terapia , Neoplasias da Glândula Tireoide/genética , Neoplasias da Glândula Tireoide/patologia , Neoplasias da Glândula Tireoide/terapia , Regulação para Cima
15.
Cancer Manag Res ; 12: 8631-8642, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32982454

RESUMO

INTRODUCTION: Breast cancer (BC) is one of the most prevalent malignancies in women and its incidence has increased steadily over recent years (0.3% per year). However, the mechanism of BC tumorigenesis remains elaborate elucidation. With the aid of RNA sequencing technology, we discovered that immortalization-upregulated protein (IMUP) is overexpressed in BC tissues compared to normal breast tissues. Our study is to understand the role of IMUP in BC. METHODS: We validated the upregulation of IMUP from multiple public databases. By using quantitative real-time polymerase chain reaction (qRT-PCR), we proved that IMUP is overexpressed in BC tissues and cell lines. We performed proliferation, migration, invasion and apoptosis assays to explore the function of IMUP in BC cell lines (MCF-7 and MDA-MB-231). Besides, we investigated the effect of IMUP silencing on epithelial-mesenchymal transition using Western blotting and qRT-PCR. RESULTS AND DISCUSSION: We validated that IMUP expression in BC tissues and cell lines is higher than that in the normal control group. The clinical analysis showed that IMUP is associated with lymph node metastasis and the outcome of neoadjuvant taxol-based therapy. The loss of function assay demonstrated that, with silencing IMUP, the capacities of proliferation, migration, and invasion of BC cell lines were impaired, while the apoptosis rate of cells increased. Meanwhile, the downregulation of IMUP could hinder the procession of epithelial-mesenchymal transition. CONCLUSION: Our study proved that IMUP plays a vital role in BC and acts as a potential target and marker in future therapy.

16.
Biomark Med ; 14(11): 969-980, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32940082

RESUMO

Aim: Thyroid cancer (TC) is one of the most common types of endocrine malignancy and poses a significant challenge to human health. The long noncoding RNA 389641 (LOC389641) has been found to be associated with many types of cancer. However, the function of LOC389641 in papillary TC (PTC) remains unknown. Our aim is to explore LOC389641 expression and its role in TC. Materials & methods: The function of LOC389641 was determined by colony formation, migration and invasion assays in PTC. Western blot assays were performed to determine the biomarker of epithelial-mesenchymal transition. Results: In this study, we show that LOC389641 is involved in PTC, which suggests that it may be a target for TC therapies.


Assuntos
Progressão da Doença , Transição Epitelial-Mesenquimal/genética , RNA Longo não Codificante/genética , Câncer Papilífero da Tireoide/genética , Câncer Papilífero da Tireoide/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA