Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters











Database
Language
Publication year range
1.
Eur J Surg Oncol ; 50(12): 108647, 2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39217765

ABSTRACT

BACKGROUND: Breast cancer poses a significant threat to women's health worldwide. This study aimed to evaluate the association between various levels of physical activity and the incidence of breast cancer. METHODS: The data for this study were obtained from the National Health and Nutrition Examination Survey (NHANES), spanning 2011 to 2020. The study included female participants aged 20 years or older, who provided detailed data on breast cancer incidence, physical activity levels, and other pertinent variables. Adjusted odds ratios (ORs) and 95 % confidence intervals (CIs) were calculated using multivariate logistic regression analyses, alongside subgroup and sensitivity analyses. RESULTS: After adjusting for potential confounders, multivariate logistic regression analyses indicated that compared to individuals with low physical activity (<600 METs min/week), light physical activity (600 to < 1800 METs min/week), moderate physical activity (1800 to < 3000 METs min/week) and high physical activity (≥3000 METs min/week) were associated with breast cancer with adjusted ORs of 0.95 (95 % CI 0.68-1.34, P = 0.787), 0.92 (95 % CI 0.57-1.49, P = 0.747), and 0.56 (95 % CI 0.37-0.86, P = 0.009) respectively. These results were consistent across sensitivity and subgroup analyses. CONCLUSION: High-intensity physical activity may decrease the risk of breast cancer, highlighting the importance of proactively implementing healthy lifestyle interventions to protect the health of adult women.

2.
Aging (Albany NY) ; 13(18): 22164-22175, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34526411

ABSTRACT

Solute carrier organic anion transporter family member 1B3 (SLCO1B3) is a gene that encodes an organic anion-transporting polypeptide (OATP) 1B3, a membrane-bound multi-specific transporter in hepatocytes. SLCO1B3 was first reported in hepatocytes. Later, it was found that its expression is higher in colorectal cancer (CRC) than in the adjacent normal tissue. However, the role of SLCO1B3 in CRC is not well elucidated. In this study, the correlation between SLCO1B3 and the overall survival (OS) of CRC patients was evaluated using data from the GEO database. This study evaluated the relationship between SLCO1B3 and the clinicopathological characteristics and prognosis of CRC patients. The effects of SLCO1B3 knockdown, on human CRC cell proliferation, migration, and invasion in vitro and CRC tumorigenesis and metastasis in vivo were also examined. In addition, next-generation sequencing was used to identify SLCO1B3 mediators. The results confirmed the association between SLCO1B3 and poor OS of CRC patients, and SLCO1B3 was identified as the top hub gene associated with the OS. The study showed that high SLCO1B3 expression was associated with poor tumor differentiation, advanced disease stage, tumor invasion, lymph node metastasis, and poor OS. Next-generation sequencing revealed that SLCO1B3 knockdown affected the expression of several genes involved in cancer invasion, metastasis, and DNA repair. Moreover, the western blot analysis showed that SLCO1B3 knockdown downregulated p-STAT3, MMP-2, and MMP-9. In summary, we demonstrated that SLCO1B3 acts as a novel carcinogen in the CRC that drives the CRC tumorigenesis and metastasis. SLCO1B3 inhibitors, alone or in combination with current drugs, may have therapeutic benefits in CRC.


Subject(s)
Colorectal Neoplasms/metabolism , STAT3 Transcription Factor/metabolism , Solute Carrier Organic Anion Transporter Family Member 1B3/metabolism , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/physiopathology , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Neoplasm Metastasis , STAT3 Transcription Factor/genetics , Solute Carrier Organic Anion Transporter Family Member 1B3/genetics , Xenograft Model Antitumor Assays
3.
Carcinogenesis ; 42(1): 105-117, 2021 02 11.
Article in English | MEDLINE | ID: mdl-32531052

ABSTRACT

Dual oxidase 2 (DUOX2) is an important regulatory protein in the organic process of thyroid hormone iodine. Mounting evidence suggests that DUOX2 plays a crucial role in the occurrence and development of cancers. However, the function and mechanism of DUOX2 in colorectal cancer (CRC) have not been fully clarified. In the present study, the relationship between the expression of DUOX2 and the clinicopathological features and prognosis of CRC patients was analyzed. Furthermore, the effects of DUOX2 on proliferation and invasion in vitro and in vivo were examined. DUOX2-associated proteins were identified by immunoprecipitation (IP). Next-generation sequencing detection was performed to illustrate the mechanism of DUOX2 in CRC cells. It was found that the expression levels of DUOX2 in metastatic sites were significantly higher than those in primary tumor tissues, and this was demonstrated to be associated with poor prognosis. The knockdown of DUOX2 inhibited the invasion and migration of CRC cells. Furthermore, DUOX2 regulated the stability of ribosomal protein uL3 (RPL3) by affecting the ubiquitination status of RPL3, and the invasion and migration ability of DUOX2 can be reversed by the overexpression of RPL3. The downregulation of DUOX2 can affect the expression level of a large number of genes, and a number of these are enriched in the PI3K-AKT pathway. Some of the changes caused by DUOX2 can be reversed by RPL3. In summary, DUOX2 exhibits a significantly higher expression in CRC tumor samples, and facilitates the invasion and metastasis ability of CRC cells by interacting with RPL3.


Subject(s)
Colorectal Neoplasms/pathology , Dual Oxidases/metabolism , Liver Neoplasms/secondary , Lymphatic Metastasis/pathology , Ribosomal Proteins/metabolism , Animals , Carcinogenesis/pathology , Cell Movement , Cell Proliferation , Colectomy , Colon/pathology , Colon/surgery , Colorectal Neoplasms/surgery , Down-Regulation , Dual Oxidases/genetics , Female , Gene Knockdown Techniques , HCT116 Cells , HT29 Cells , Hepatectomy , Humans , Liver/pathology , Liver/surgery , Liver Neoplasms/surgery , Male , Mice , Middle Aged , Neoplasm Invasiveness/pathology , Prognosis , Protein Binding , Protein Stability , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein L3 , Signal Transduction , Ubiquitination , Xenograft Model Antitumor Assays
4.
Sci Rep ; 10(1): 11951, 2020 07 20.
Article in English | MEDLINE | ID: mdl-32686693

ABSTRACT

Transcobalamin (TCN1) is a vitamin B12 (cobalamin)-binding protein that regulates cobalamin homeostasis. Recent studies and bioinformatic analyses have found that TCN1 is highly expressed in cancer tissues and is associated with tumour aggressiveness and poor prognosis. The present study aimed to detect TCN1 as a novel biomarker for prognosis and chemosensitivity of colon cancer. Next-generation sequencing showed that TCN1 was one of several upregulated mRNAs in colon cancer, which was verified by further bioinformatics analyses. Western blotting (n = 9) and quantitative real time polymerase chain reaction (qRT-PCR, n = 30) revealed that TCN1 was highly expressed in colon cancer tissues at both the protein and mRNA level. A total of 194 cases of colon cancer were examined by immunohistochemistry and revealed that TCN1 expression level was related to advanced stages (P < 0.005). Kaplan-Meier analysis verified that patients with lower TCN1 expression usually had longer overall survival (P = 0.008). In addition, TCN1 was highly expressed in pulmonary metastatic tumour tissues (n = 37, P = 0.025) and exhibited higher levels in right-sided colon cancer than in left-sided colon cancer (P = 0.029). TCN1 expression in specimens that had received neoadjuvant chemotherapy decreased compared with that in colonoscopy biopsy tissues (n = 42, P = 0.009). Further bioinformatics analyses verified that apoptosis pathways might have a role in high TCN1 expression. All the studies revealed that TCN1 expression in colon cancer was significantly associated with malignant biological behaviour. Therefore, TCN1 could be used as a novel biomarker for colon cancer aggressiveness and prognosis and might also be a potential biomarker for predicting neoadjuvant chemosensitivity.


Subject(s)
Biomarkers, Tumor , Colonic Neoplasms/genetics , Colonic Neoplasms/mortality , Drug Resistance, Neoplasm/genetics , Gene Expression , Transcobalamins/genetics , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Computational Biology/methods , Female , Gene Expression Profiling , Gene Ontology , Humans , Immunohistochemistry , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Middle Aged , Neoadjuvant Therapy , Neoplasm Staging , Prognosis , Transcobalamins/metabolism
5.
J Cancer Res Clin Oncol ; 146(5): 1139-1152, 2020 May.
Article in English | MEDLINE | ID: mdl-32152662

ABSTRACT

OBJECTIVE: Increasing evidence has revealed that mechanical stress and elevated mechanical signals promote malignant tumor transformation and metastasis. This study aimed to explore the function of the mechanically activated ion-channel Piezo1 in the colon cancer metastasis and its potential regulatory mechanism. METHODS: First, we examined the expression levels of Piezo1 and mitochondrial calcium uniporter (MCU) both in colon cancer tissues and assessed the prognostic value of Piezo1 and MCU in a colon cancer cohort (n = 110). Second, functional assays were performed to investigate the effects of Piezo1 and MCU on colon cancer cell migration, invasion, and mitochondrial membrane potential. Third, we analyzed the expression of Piezo1, MCU, and HIF-1α by overexpressing/silencing each other's expression. RESULTS: We found that Piezo1 was up-regulated and MCU was down-regulated in colon cancer tissues. Piezo1 and MCU were both correlated with poor prognosis of patients with colon cancer. Overexpressing Piezo1 and silencing MCU could promote colon cancer cell migration and metastasis, reduce mitochondrial membrane potential, and promote each other's expression. We also found that HIF-1α was up-regulated in colon cancer tissues. Additionally, silencing Piezo1 inhibited the expression of HIF-1α and VEGF, which was contrary to MCU silencing. Intriguingly, Piezo1-overexpressing cells did not regain their migration behaviors when HIF-1α expression was inhibited, which was accompanied with the re-expression of MCU and VEGF. CONCLUSION: In our study, Piezo1 is involved in colon cancer cell metastasis. Furthermore, our findings indicated a possible Piezo1-MCU-HIF-1α-VEGF axis, which still need further exploration.


Subject(s)
Colonic Neoplasms/metabolism , Ion Channels/metabolism , Calcium Channels/metabolism , Cell Line, Tumor , Cell Movement/physiology , Cohort Studies , Colonic Neoplasms/pathology , HCT116 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunohistochemistry , Ion Channels/biosynthesis , Membrane Potential, Mitochondrial , Neoplasm Metastasis , Prognosis , Signal Transduction , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL