Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 66
1.
Front Oncol ; 14: 1338098, 2024.
Article En | MEDLINE | ID: mdl-38406812

Background: Neoadjuvant chemoradiotherapy has emerged as the established treatment for locally advanced rectal cancer. Nevertheless, there remains a debate regarding the necessity of adjuvant chemotherapy for patients with locally advanced rectal cancer who exhibit a favorable tumor response (ypT0-2N0) after neoadjuvant chemoradiotherapy and surgery. Thus, the objective of this study is to investigate the impact of adjuvant chemotherapy on the oncological prognosis of rectal cancer patients who have a good response to neoadjuvant chemoradiotherapy. Materials and methods: The study was conducted following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses protocol. Articles were searched in the Web of Science, PubMed, and Cochrane Library databases. The primary outcomes assessed were 5-year overall survival, disease-free survival, cancer-specific survival, recurrence-free survival, local recurrence, and distant metastasis. The data was summarized using a random effects model. Results: A meta-analysis was conducted using 18 retrospective studies published between 2009 and 2023. The studies included 9 from China and 5 from Korea, involving a total of 6566 patients with ypT0-2N0 rectal cancer after neoadjuvant chemoradiotherapy. The pooled data revealed that adjuvant chemotherapy significantly improved 5-year overall survival (OR=1.75, 95% CI: 1.15-2.65, P=0.008), recurrence-free survival (OR=1.73, 95% CI: 1.20-2.48, P=0.003), and reduced distant metastasis (OR=0.68, 95% CI: 0.51-0.92, P=0.011). However, adjuvant chemotherapy did not have a significant effect on disease-free survival, cancer-specific survival, and local recurrence in ypT0-2N0 rectal cancer. Subgroup analysis indicated that adjuvant chemotherapy was beneficial in improving overall survival for ypT1-2N0 rectal cancer (OR=1.89, 95% CI: 1.13-3.19, P=0.003). Conclusion: The findings of the meta-analysis suggest that adjuvant chemotherapy may provide benefits in terms of oncological outcomes for rectal cancer patients with ypT0-2N0 after neoadjuvant chemoradiotherapy and radical surgery. However, further prospective clinical studies are needed to confirm these findings.

2.
Adv Clin Exp Med ; 2024 Jan 17.
Article En | MEDLINE | ID: mdl-38239081

BACKGROUND: Numerous studies have indicated the engagement of long non-coding RNA (lncRNA) in various cancer types, including colorectal cancer (CRC). However, the functional and mechanistic roles of lncRNAs in CRC remain largely elusive. OBJECTIVES: The aim of this study was to explore the function and mechanism of lncRNA BVES-AS1 in CRC. MATERIAL AND METHODS: The expression levels of BVES-AS1 were validated in CRC tissues and paired normal samples using quantitative real-time polymerase chain reaction (qPCR) Subsequently, the biological functions of BVES-AS1 in CRC cells were investigated both in vitro and in vivo. Various experimental techniques such as western blot, fluorescence in situ hybridization, RNA-sequencing (RNA-seq), biotin-labeled miRNA pulldown assay, dual-luciferase reporter gene assay, and RNA-protein immunoprecipitation (RIP) assay were employed to elucidate the potential mechanism of BVES-AS1. RESULTS: The findings of this study demonstrated that BVES-AS1 expression was downregulated in CRC tissues compared to normal tissues, and its expression level was associated with tumor infiltration and tumor-nodule-metastasis (TNM) stage. Furthermore, BVES-AS1 was found to suppress CRC cell proliferation, migration and metastasis both in vitro and in vivo. Mechanistically, BVES-AS1 acted as a sponge for miR-1269a and miR-1269b, thereby regulating SVEP1. Additionally, the silencing of SVEP1 activated the PI3K/AKT pathway. CONCLUSIONS: These results suggest that BVES-AS1 plays a crucial role in the progression of CRC through the miR-1269a/b-SVEP1-PI3K/AKT axis, providing new insights into the therapeutic strategies for CRC.

3.
World J Surg Oncol ; 22(1): 31, 2024 Jan 25.
Article En | MEDLINE | ID: mdl-38273352

BACKGROUND: Locally advanced rectal cancer is typically treated using a combination of neoadjuvant chemoradiotherapy and total mesorectal resection. While achieving pathological complete response following neoadjuvant chemoradiotherapy has been recognized as a positive prognostic factor in oncology, the necessity of adjuvant chemotherapy for locally advanced rectal cancer patients with pathological complete response after surgery remains uncertain. The objective of this meta-analysis was to examine the impact of adjuvant chemotherapy on the oncological outcomes of rectal cancer patients who attain pathological complete response after neoadjuvant chemoradiotherapy. METHODS: This meta-analysis followed the guidelines outlined in the preferred reporting items for systematic review and meta-analysis (PRISMA). The Web of Science, PubMed, and Cochrane Library databases were systematically searched to identify relevant literature. RESULTS: A total of 34 retrospective studies, including 9 studies from the NCBD database, involving 31,558 patients with pathological complete response rectal cancer, were included in the meta-analysis. The included studies were published between 2008 and 2023. The pooled analysis demonstrated that adjuvant chemotherapy significantly improved overall survival (HR = 0.803, 95% CI 0.678-0.952, P = 0.011), and no heterogeneity was observed (I2 = 0%). Locally advanced rectal cancer patients with pathological complete response who underwent adjuvant chemotherapy exhibited a higher 5-year overall survival rate compared to those who did not receive adjuvant chemotherapy (OR = 1.605, 95% CI 1.183-2.177, P = 0.002). However, the analysis also revealed that postoperative ACT did not lead to improvements in disease-free survival and recurrence-free survival within the same patient population. Subgroup analysis indicated that pathological complete response patients with clinical stage T3/T4, lymph node positivity, and younger than 70 years of age may benefit from adjuvant chemotherapy in terms of overall survival. CONCLUSIONS: The findings of this meta-analysis suggest that adjuvant chemotherapy has a beneficial effect on improving overall survival among rectal cancer patients with pathological complete response. However, no such association was observed in terms of disease-free survival and recurrence-free survival.


Chemoradiotherapy , Rectal Neoplasms , Humans , Retrospective Studies , Neoplasm Staging , Rectal Neoplasms/pathology , Chemotherapy, Adjuvant , Disease-Free Survival , Neoadjuvant Therapy , Pathologic Complete Response , Treatment Outcome , Chemoradiotherapy, Adjuvant
4.
Front Oncol ; 13: 1242193, 2023.
Article En | MEDLINE | ID: mdl-38074647

Aim: To investigate whether body composition parameters combined with systemic inflammatory markers and magnetic resonance imaging (MRI) can predict the pathological complete response (pCR) following neoadjuvant chemoradiotherapy (NCRT) in locally advanced rectal cancer (LARC). Methods: A retrospective analysis of data on LARC patients treated with NCTR and radical surgery between January 2013 and May 2023 was performed. Body composition parameters were assessed by measuring the skeletal muscle index (SMI), subcutaneous adipose index (SAI), and visceral adipose index (VAI) at the third lumbar vertebra level by computed tomography (CT). Inflammatory markers such as neutrophil to lymphocyte ratio (NLR) were obtained from laboratory tests performed prior to NCRT. MRI was conducted to evaluate MRI tumor regression grading (mrTRG). Logistic regression analyses were employed to identify factors affecting the pCR. The risk score of pCR was computed by a nomogram. The discrimination of the nomogram was determined using C-index and calibration curve. Results: Two hundred and ninety-one patients with LARC were enrolled in the study, 55 (18.9%) of whom achieved pCR after NCRT. Multivariate analysis suggested that pre-NCRT NLR≥2.6 (OR=0.378, 95% CI 0.164-0.868, P=0.022), mrTRG 3-5 (OR=0.256, 95%CI 0.121-0.54, P<0.001), and pre-NCRT L-SMI (OR=0.292, 95% CI 0.097-0.883, P=0.029) were independent risk factors for pCR. ROC curves analysis demonstrated that the performance of mrTRG combined with pre-NCRT NLR and pre-NCRT L-SMI in predicting pCR was significantly improved compared with mrTRG alone (AUC: 0.763 vs. 0.667). Additionally, mrTRG 3-5 (OR=0.375, 95% CI 0.219-0.641, P<0.001) was also an independent predictor for poor tumor regression. Conclusion: The pathological complete response of neoadjuvant chemoradiotherapy in locally advanced rectal cancer can be effectively predicted by combining the body composition parameters with blood biomarkers and magnetic resonance imaging.

5.
J Exp Clin Cancer Res ; 42(1): 334, 2023 Dec 06.
Article En | MEDLINE | ID: mdl-38057879

BACKGROUND: With the advancements in bioinformatic technology, an increasing number of circular RNAs (circRNAs) have been discovered and their crucial roles in the development and progression of various malignancies have been confirmed through multiple pathways. However, the specific mechanisms involving protein-binding circRNAs in colorectal cancer (CRC) remain largely unexplored. METHODS: Differential circRNA expression was assessed using a human circRNA microarray in five CRC tissue and paired normal samples. CircGPRC5A expression was then confirmed in the CRC tissues and paired normal samples using qRT-PCR. The biological function of circGPRC5A in CRC were studied in vitro and in vivo. Western blotting, fluorescence in situ hybridization, immunofluorescence, RNA pulldown, mass spectrometry, immunoprecipitation, quantitative phosphoproteomics, and RNA-binding protein immunoprecipitation assays were used to study circGPRC5A. RESULTS: Our analysis revealed that circGPRC5A expression was higher in CRC tissues compared to normal tissues and was associated with tumor size, tumor stage and lymph node status. CircGPRC5A promoted CRC cell proliferation, migration, and metastasis in vitro and in vivo. CircGPRC5A could stabilize PPP1CA protein by inhibiting the binding between UBA1 and PPP1CA, and increasing YAP dephosphorylation. CONCLUSIONS: Our study revealed that circGPRC5A plays an essential function in CRC progression by stabilizing PPP1CA protein and enhancing YAP dephosphorylation. CircGPRC5A could act as a novel and potential target for CRC.


Colorectal Neoplasms , MicroRNAs , Humans , Cell Proliferation/genetics , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , In Situ Hybridization, Fluorescence , MicroRNAs/genetics , Protein Phosphatase 1/metabolism , RNA/genetics , RNA, Circular/genetics , RNA, Circular/metabolism
6.
Sci Rep ; 13(1): 16408, 2023 09 29.
Article En | MEDLINE | ID: mdl-37775715

Cancer-associated fibroblasts (CAFs) influence many aspects of pancreatic adenocarcinoma (PAAD) carcinogenesis, including tumor cell proliferation, angiogenesis, invasion, and metastasis. A six-gene prognostic signature was constructed for PAAD based on the 189 CAF marker genes identified in single-cell RNA-sequencing data. Multivariate analyses showed that the risk score was independently prognostic for survival in the TCGA (P < 0.001) and ICGC (P = 0.004) cohorts. Tumor infiltration of CD8 T (P = 0.005) cells and naïve B cells (P = 0.001) was greater in the low-risk than in the high-risk group, with infiltration of these cells negatively correlated with risk score. Moreover, the TMB score was lower in the low-risk than in the high-risk group (P = 0.0051). Importantly, patients in low-risk group had better immunotherapy responses than in the high-risk group in an independent immunotherapy cohort (IMvigor210) (P = 0.039). The CAV1 and SOD3 were highly expressed in CAFs of PAAD tissues, which revealed by immunohistochemical staining. In summary, this comprehensive analysis resulted in the development of a novel prognostic signature, which was associated with immune cell infiltration, drug sensitivity, and TMB, and could predict the prognosis and immunotherapy response of patients with PAAD.


Adenocarcinoma , Cancer-Associated Fibroblasts , Pancreatic Neoplasms , Humans , Adenocarcinoma/genetics , Adenocarcinoma/therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/therapy , Prognosis , Immunotherapy , RNA , Tumor Microenvironment/genetics , Pancreatic Neoplasms
7.
BMC Cancer ; 23(1): 246, 2023 Mar 14.
Article En | MEDLINE | ID: mdl-36918834

BACKGROUND: The purpose of this study was to explore the short-term efficacy and safety of neoadjuvant chemotherapy combined with arterial chemoembolization for locally advanced gastric cancer (LAGC). METHODS: We retrospectively analyzed the clinical data of 203 patients with LAGC who received neoadjuvant therapy from June 2019 to December 2021. The patients were divided into a neoadjuvant chemotherapy combined with arterial chemoembolization group (combined group, n = 102) and a neoadjuvant chemotherapy group (conventional group, n = 101). The adverse events of chemotherapy, postoperative complications and pathological complete response (pCR) rate were compared between the two groups. Univariate and multivariate analyses were performed to evaluate the potential factors affecting pCR. RESULTS: A total of 78.8% of the patients were in clinical stage III before neoadjuvant therapy. A total of 52.2% of the patients underwent surgery after receiving two cycles of neoadjuvant therapy. There were 21.2% patients with ≥ grade 3 (CTCAE 4.0) adverse events of chemotherapy and 11.3% patients with Clavien-Dindo classification ≥ grade 3 postoperative complications. Compared with the conventional group, the combination group did not experience an increase in the adverse events of chemotherapy or postoperative complications. The pCR rate in the combined group was significantly higher than that in the conventional group (16.7% vs. 4.95%, P = 0.012). The multivariate analysis showed that arterial chemoembolization, pre-treatment neutrophil-to-lymphocyte ratio (NLR) and pre-treatment platelet-to-lymphocyte ratio (PLR) were independent factors affecting pCR. CONCLUSION: Neoadjuvant chemotherapy combined with arterial chemoembolization contributed to improving the pCR rate of LAGC patients. Arterial chemoembolization, pre-treatment NLR and pre-treatment PLR were also predictors of pCR.


Neoadjuvant Therapy , Stomach Neoplasms , Humans , Retrospective Studies , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Lymphocytes/pathology , Postoperative Complications/pathology , Antineoplastic Combined Chemotherapy Protocols/adverse effects
8.
Biotechnol Genet Eng Rev ; : 1-19, 2023 Mar 23.
Article En | MEDLINE | ID: mdl-36951606

Long noncoding RNAs (lncRNA) have a critical role in colorectal cancer (CRC) development and progression. However, the role of the lncRNA HOXB-AS4 in CRC remains unclear. In this study, we found that HOXB-AS4 was markedly upregulated in tumor tissues compared to precancerous tissues. Loss-of-function assays in HT29 and SW480 cells confirmed that knockdown of HOXB-AS4 inhibited proliferation, migration, and promoted apoptosis. In addition, HOXB-AS4 was shown to regulate histone deacetylase 7 (HDAC7) expression by acting as a molecular sponge to bind to and adsorb miR-140-5p. These findings were confirmed by the dual-luciferase reporter assay. Functional recovery experiments further demonstrated the crucial role of the HOXB-AS4/miR-140-5p/HDAC7 axis in modulating the malignant phenotype of CRC cells. Collectively, our data suggested that HOXB-AS4 regulated the malignant tumor aggression of HT29 and SW480 cells through the miR-140-5p/HDAC7 axis and PI3K/AKT signaling pathway. Our study provides novel insights into the mechanism of action of HOXB-AS4 in CRC and highlights its potential use as a targeted therapy.

9.
Biomol Biomed ; 23(3): 483-495, 2023 May 01.
Article En | MEDLINE | ID: mdl-36373629

Colorectal cancer (CRC) is regarded as one of the most prevalent neoplasms worldwide, and ubiquitination and N6-methyladenosine (m6A) modification regulate the outgrowth of multiple cancers. This study attempted to explore the effect of ubiquitin-specific peptidases 29 (USP29) on the malignant proliferation of CRC cells via stabilizing Vir-like m6A methyltransferase associated (VIRMA/KIAA1429). First, upregulations of USP29, KIAA1429, and SRY-box transcription factor 8 (SOX8) were found in CRC tissues and cells through real-time quantitative polymerase chain reaction and Western blotting. After transfection of si-USP29, the proliferation of CRC cells was evaluated by the cell counting kit-8, colony formation and 5-ethynyl-2'-deoxyuridine assays, and we observed that depletion of USP29 inhibited the proliferation of CRC cells. Co-immunoprecipitation confirmed the binding of USP29 to KIAA1429. Mechanically, USP29 mediated deubiquitination to stabilize the protein levels of KIAA1429, and KIAA1429 promoted the stability of SOX8 mRNA through m6A modification. Moreover, overexpression of KIAA1429 or SOX8 reversed the inhibitory effects of USP29 depletion on CRC cell proliferation. Finally, the xenograft tumor model revealed the promotive role of USP29 in the proliferation of CRC cells in vivo. Altogether, USP29 facilitates the malignant proliferation of CRC cells via upregulating the KIAA1429/SOX8 axis.


Colorectal Neoplasms , Animals , Humans , Cell Line, Tumor , Disease Models, Animal , Up-Regulation , Cell Proliferation/genetics , Colorectal Neoplasms/genetics , SOXE Transcription Factors/genetics , Ubiquitin-Specific Proteases/genetics
10.
World J Gastrointest Oncol ; 15(12): 2185-2196, 2023 Dec 15.
Article En | MEDLINE | ID: mdl-38173429

BACKGROUND: It is unclear that paired-related homeobox 1 (PRRX1) induces epithelial-mesenchymal transition (EMT) in oesophageal cancer and the specific function of PRRX1 in oesophageal cancer metastasis. AIM: To assess the significance of PRRX1 expression and investigate the mechanism of EMT in oesophageal cancer metastasis. METHODS: Detect the expression of PRRX1 by immunohistochemistry in oesophageal tumour tissues and adjacent normal oesophageal tissues; the PRRX1 short hairpin RNA (shRNA) or blank vector lentiviral gene delivery system was transfected into cells; cell proliferation assay, soft agar colony formation assays, cell invasion and migration assays and animal studies were used to observe cells biological characteristics In vitro and in vivo; XAV939 and LiCl were used to alter the activity of Wnt/ß-catenin pathway. Immunofluorescence staining and western blot analysis were used to detect protein expression of EMT markers and Wnt/ß-catenin pathway. RESULTS: PRRX1 is expressed at high levels in oesophageal cancer specimens and is closely related to tumour metastasis in patients with oesophageal cancer. Regulation of PRRX1 expression might exert obvious effects on cell proliferation, especially the migration and invasion of oesophageal cancer cells. Moreover, silencing PRRX1 expression using a shRNA produced the opposite effects. In addition, when PRRX1 was overexpressed, inhibition of the Wnt/ß-catenin pathway with XAV939 negated the effect of PRRX1 on EMT, whereas when PRRX1 was downregulated, activation of the Wnt/ß-catenin pathway with LiCl impaired the effect on EMT. CONCLUSION: PRRX1 is upregulated in oesophageal cancer is closely correlated with cancer metastasis. Additionally, PRRX1 induces EMT in oesophageal cancer metastasis through activation of Wnt/ß-catenin signalling.

11.
Zhongguo Zhong Yao Za Zhi ; 47(22): 6183-6190, 2022 Nov.
Article Zh | MEDLINE | ID: mdl-36471943

Taking lipophagy as the breakthrough point, we explored the mechanism of Zexie Decoction(ZXD) in improving lipid metabolism in the hepatocyte model induced by palmitic acid(PA) and in the animal model induced by high-fat diet(HFD) on the basis of protein kinase B(Akt)/transcription factor EB(TFEB) signaling pathway. Co-localization was carried out for the microtubule-associated protein light chain 3(LC3) plasmid labeled with green fluorescent protein(GFP) and lipid droplets(LDs), and immunofluorescence co-localization for liver LC3 of HFD mice and perilipin 2(PLIN2). The results showed that ZXD up-regulated the expression of LC3, reduced lipid accumulation in hepatocytes, and increased the co-localization of LC3 and LDs, thereby activating lipo-phagy. Western blot results confirmed that ZXD increased autophagy-related protein LC3Ⅱ/LC3Ⅰ transformation ratio and lysosome-associated membrane protein 2(LAMP2) in vivo and in vitro and promoted the degradation of sequestosome-1(SQSTM1/p62)(P<0.05). The results above jointly explained that ZXD regulated lipophagy. Furthermore, ZXD activated TFEB expression(P<0.05) and reversed the PA-and HFD-induced decrease of TFEB nuclear localization in hepatocytes(P<0.05). Meanwhile, ZXD activated liver TFEB to up-regulate the expression of the targets Lamp2, Lc3 B, Bcl2, and Atg5(P<0.05). Additionally, ZXD down-regulated the protein level of p-Akt upstream of TFEB in vivo and in vitro. In conclusion, ZXD may promote lipophagy by regulating the Akt/TFEB pathway.


Autophagy , Drugs, Chinese Herbal , Hepatocytes , Proto-Oncogene Proteins c-akt , Animals , Mice , Autophagy/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Microtubule-Associated Proteins/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Drugs, Chinese Herbal/pharmacology
12.
Clin Transl Med ; 12(4): e752, 2022 04.
Article En | MEDLINE | ID: mdl-35485210

BACKGROUND: Ferroptosis is principally caused by iron catalytic activity and intracellular lipid peroxidation. Long noncoding RNAs (lncRNAs) play crucial roles in tumorigenesis. However, the potential interplay between lncRNA LINC01606 and ferroptosis in colon cancer remains elusive. METHODS: The expression level of LNC01606 in colon cancer tissue was detected by quantitative real-time polymerase chain reaction. The functional role of LNC01606 was investigated by gain- and loss-of-function assays both in vitro and in vivo. The LINC01606-SCD1-Wnt/ß-catenin-TFE3 axis were screened and validated by DNA/RNA pull down, gas chromatography-mass spectrometry, RNA immunoprecipitation and dual-luciferase reporter. RESULTS: The expression of lncRNA LINC01606 was frequently upregulated in human colon cancer and strongly associated with a poor prognosis. LINC01606 functioned as an oncogene and promotes colon cancer cell growth, invasion and stemness both in vitro and in vivo. Moreover, LINC01606 protected colon cancer cells from ferroptosis by decreasing the concentration of iron, lipid reactive oxygen species, mitochondrial superoxide and increasing mitochondrial membrane potential. Mechanistically, LINC01606 enhanced the expression of stearoyl-CoA desaturase 1 (SCD1), serving as a competing endogenous RNA to modulate miR-423-5p expression, subsequently activating the canonical Wnt/ß-catenin signaling, and transcription factor binding to IGHM enhancer 3 (TFE3) increased LINC01606 transcription after recruitment to the promoter regions of LINC01606. Furthermore, we confirmed that upregulated LINC01606 and Wnt/ß-catenin formed a positive feedback regulatory loop, further inhibiting ferroptosis and enhancing stemness. CONCLUSIONS: LINC01606 functions as an oncogene to facilitate tumor cell stemness, proliferation and inhibit ferroptosis and is a promising therapeutic target for colon cancer.


Colonic Neoplasms , MicroRNAs , RNA, Long Noncoding , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Death , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Feedback , Gene Expression Regulation, Neoplastic/genetics , Humans , Iron/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Stearoyl-CoA Desaturase/genetics , Stearoyl-CoA Desaturase/metabolism , beta Catenin/genetics , beta Catenin/metabolism
13.
Cancer Cell Int ; 22(1): 75, 2022 Feb 11.
Article En | MEDLINE | ID: mdl-35148781

BACKGROUND: Colorectal cancer (CRC) is one of the most commonly diagnosed cancers in both men and women in China. In previous studies, Sestrin2 was demonstrated to have functions in CRC. However, the relationship between Sestrin2 and cancer stemness has not been reported. METHODS AND RESULTS: To investigate the contribution of Sestrin2 in CRC, we performed bioinformatics analysis of The Cancer Genome Atlas datasets and found that Sestrin2 was downregulated in CRC. Using a lentivirus vector, we verified that Sestrin2 suppressed CRC cell proliferation, migration, and colony formation. Furthermore, sphere formation, flow cytometry, quantitative PCR, and western blot analysis verified the influence of Sestrin2 on cancer stemness, including the expression of cluster of differentiation 44, octamer-binding transcription factor 4, sex-determining region Y-Box 2, CXC chemokine receptor 4, and the Wnt pathway downstream factors ß-catenin and c-Myc. Consistently, the Wnt pathway activator BML-284 partially rescued the effects of Sestrin2 on the expression of proteins related to cancer stemness. Furthermore, in a mouse xenoplant model, tumors expressing Sestrin2 were significantly reduced in size with corresponding changes in cancer stemness. CONCLUSIONS: Collectively, our results suggest that Sestrin2 inhibits CRC cell progression by downregulating the Wnt signaling pathway. Thus, Sestrin2 may be a promising therapeutic target for CRC.

14.
J Oncol ; 2022: 8197701, 2022.
Article En | MEDLINE | ID: mdl-35035483

OBJECTIVE: Neoadjuvant radiotherapy (nRT) is an important treatment approach for rectal cancer. The relationship, however, between nRT and postoperative complications is still controversial. Here, we conducted a meta-analysis to evaluate such concerns. METHODS: The electronic literature from 1983 to 2021 was searched in PubMed, Embase, and Web of Science. Postoperative complications after nRT were included in the meta-analysis. The pooled odds ratio (OR) was calculated by the random-effects model. Statistical analysis was conducted by Review Manager 5.3 and STATA 14. RESULTS: A total of 23,723 patients from 49 studies were included in the meta-analysis. The pooled results showed that nRT increased the risk of anastomotic leakage (AL) compared to upfront surgery (OR = 1.23; 95% CI, 1.07-1.41; p=0.004). Subgroup analysis suggested that both long-course (OR = 1.20, 95% CI 1.03-1.40; p=0.02) and short-course radiotherapy (OR = 1.25, 95% CI, 1.02-1.53; p=0.04) increased the incidence of AL. In addition, nRT was the main risk factor for wound infection and pelvic abscess. The pooled data in randomized controlled trials, however, indicated that nRT was not associated with AL (OR = 1.01; 95% CI 0.82-1.26; p=0.91). CONCLUSIONS: nRT may increase the risk of AL, wound infection, and pelvic abscess compared to upfront surgery among patients with rectal cancer.

15.
Cell Death Dis ; 12(6): 605, 2021 06 11.
Article En | MEDLINE | ID: mdl-34117220

Colorectal cancer is the most common gastrointestinal cancer and causes severe damage to human health. PRDX2 is a member of the peroxiredoxin family reported to have a high level of expression in colorectal cancer. However, the mechanisms by which PRDX2 promotes the proliferation of colorectal cancer are still unclear. Here, the results indicated that PRDX2 expression was upregulated in colorectal cancer and closely correlated with poor prognosis. Functionally, PRDX2 promoted the proliferation of colorectal cancer cells. Mechanistically, PRDX2 could bind RPL4, reducing the interaction between RPL4 and MDM2. These findings demonstrate that the oncogenic property of PRDX2 may be attributed to its regulation of the RPL4-MDM2-p53 pathway, leading to p53 ubiquitinated degradation.


Adenocarcinoma/pathology , Colorectal Neoplasms/pathology , Peroxiredoxins/physiology , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Animals , Cell Proliferation/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Female , HCT116 Cells , HEK293 Cells , Humans , Mice , Mice, Nude , Peroxiredoxins/genetics , Proteolysis , Signal Transduction/genetics , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism , Ubiquitin/metabolism , Ubiquitination
16.
Biochem Biophys Res Commun ; 557: 228-235, 2021 06 11.
Article En | MEDLINE | ID: mdl-33887588

LncRNA-cCSC1 is highly expressed in colorectal cancer (CRC). The study was designed to evaluate the function and mechanism of lncRNA-cCSC1 in cell proliferation of CRC. RT-PCR was used to measure the expression levels of lncRNA-cCSC1 in CRC cell lines. CCK-8, colony formation, EdU staining, flow cytometry and Western blot were performed to examine the effect of interference with lncRNA-cCSC1 expression on cell proliferation. miR-124-3p and the target genes of miR-124-3p were investigated using bioinformatics analysis and verified by dual-luciferase reporter, RT-PCR and Western blot. Rescue experiments were carried out to confirm the role of miR-124-3p in cell proliferation of CRC. Our results showed that cell proliferation of CRC was promoted by lncRNA-cCSC1 upregulation and inhibited by lncRNA-cCSC1 downregulation. In addition, miR-124-3p is predicted to be the target of lncRNA-cCSC1 and is negatively correlated with lncRNA-cCSC1. Moreover, the addition of miR-124-3p mimics or inhibitor reversed the effects induced by lncRNA-cCSC1 overexpression or silencing on cell proliferation of CRC. Additionally, lncRNA-cCSC1 regulated the expression level of CD44, a target gene of miR-124-3p. Finally, we studied the effects of the lncRNA-cCSC1/miR-124-3p axis on CD44. These results indicate that lncRNA-cCSC1 promotes cell proliferation of CRC through sponging miR-124-3p and upregulating CD44.


Adenocarcinoma/metabolism , Cell Proliferation/genetics , Colorectal Neoplasms/metabolism , Hyaluronan Receptors/metabolism , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Databases, Genetic , Down-Regulation , Gene Expression Regulation, Neoplastic/genetics , Humans , Hyaluronan Receptors/genetics , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Up-Regulation
17.
Aging (Albany NY) ; 13(8): 11170-11187, 2021 03 26.
Article En | MEDLINE | ID: mdl-33819194

Colon cancer stem cells (CCSCs) play an important role in facilitating colon cancer occurrence, metastasis and drug resistance. The results of our previous studies confirmed that the well-studied antioxidant gene peroxiredoxin-2 (PRDX2) promotes colon cancer progression. However, the underlying function and mechanisms associated with PRDX2 remodeling in the context of CCSCs have remained poorly studied. In our present study, we demonstrated that PRDX2 is highly expressed in CD133/CD44-positive colon cancer tissues and spheroid CD133+CD44+ CCSCs. PRDX2 overexpression was shown to be closely correlated with CD133+CD44+ CCSCs in colon cancer. Furthermore, PRDX2 depletion markedly suppressed CD133+CD44+ CCSC stemness maintenance, tumor initiation, migration and invasion and liver metastasis. Furthermore, the expression of various EMT markers and Wnt/ß-catenin signaling proteins was altered after PRDX2 inhibition. In addition, PRDX2 knockdown led to increased ROS production in CD133+CD44+ CCSCs, sensitizing CCSCs to oxidative stress and chemotherapy. These results suggest that PRDX2 could be a possible therapeutic target in CCSCs.


Colonic Neoplasms/pathology , Liver Neoplasms/genetics , Neoplastic Stem Cells/pathology , Peroxiredoxins/metabolism , Animals , Colon/pathology , Colonic Neoplasms/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , HCT116 Cells , HT29 Cells , Humans , Liver Neoplasms/secondary , Mice , Peroxiredoxins/genetics , Spheroids, Cellular , Xenograft Model Antitumor Assays
18.
Aging (Albany NY) ; 12(16): 16390-16409, 2020 07 20.
Article En | MEDLINE | ID: mdl-32692719

Colorectal cancer (CRC) is a prevalent worldwide disease in which the antioxidant enzyme peroxiredoxin 2 (PRDX2) plays an important role. To investigate the molecular mechanism of PRDX2 in CRC, we performed bioinformatics analysis of The Cancer Genome Atlas (TCGA) datasets and Gene Expression Omnibus (GEO) DataSets (accession no. GSE81429). Our results suggest that PRDX2 is associated with cell-cycle progression and autophagy activated by the P38 MAPK/FOXO signaling pathway. Using a short-hairpin RNA vector, we verified that PRDX2 is essential for CRC cell proliferation and S-phase progression. Immunostaining, electron microscopy and western blotting assays verified the effect of PRDX2 knockdown on autophagy flux and p38 activation. The P38 activator dehydrocorydaline chloride partially rescued the effects of sh-PRDX2 on the expression of proteins related to cell-cycle progression and autophagy. We verified the correlation between PRDX2 expression and the expression of an array of cell-cycle and autophagy-related genes using data from an independent set of 222 CRC patient samples. A mouse xenoplast model was consistent with in vitro results. Our results suggest that PRDX2 promotes CRC cell-cycle progression via activation of the p38 MAPK pathway.


Autophagy , Cell Cycle , Cell Proliferation , Colorectal Neoplasms/enzymology , Peroxiredoxins/metabolism , Animals , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Databases, Genetic , Enzyme Activation , Female , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , HCT116 Cells , HT29 Cells , Humans , Mice, Inbred BALB C , Mice, Nude , Peroxiredoxins/genetics , Phosphorylation , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism
19.
J Int Med Res ; 48(6): 300060520925941, 2020 Jun.
Article En | MEDLINE | ID: mdl-32495673

OBJECTIVE: To investigate the impact of enhanced recovery after surgery (ERAS) on the postoperative neutrophil-lymphocyte ratio (NLR) in patients with colorectal cancer. METHODS: A total of 200 patients with colorectal cancer who underwent surgery between January 2015 and November 2018 were enrolled in the study. They were divided into a traditional treatment group (n=100) and an ERAS group (n=100). The traditional treatment group underwent radical laparoscopic colorectal surgery, and the ERAS group underwent traditional treatment plus the ERAS protocol (preoperative improvement of glucose tolerance, unconventional indwelling stomach and urinary tubes, intraoperative body temperature management, fluid management, postoperative pain management, early oral feeding, and early activities). Clinical data were collected for all patients. NLR levels before and after surgery, and complications were compared between the two groups. RESULTS: Postoperative NLR was significantly lower in the ERAS compared with the traditional treatment group. The incidence of complications, including anastomotic leakage, pulmonary infection, urinary tract infection, and cardiopulmonary dysfunction were also significantly lower in the ERAS group. CONCLUSION: Enhanced recovery after surgery can reduce the increase in postoperative NLR and reduce the occurrence of postoperative complications, which results will be of clinical value.


Colorectal Neoplasms/surgery , Enhanced Recovery After Surgery , Laparoscopy/adverse effects , Lymphocytes/immunology , Neutrophils/immunology , Postoperative Complications/epidemiology , Aged , Female , Humans , Incidence , Laparoscopy/rehabilitation , Length of Stay , Lymphocyte Count , Male , Middle Aged , Postoperative Complications/blood , Postoperative Complications/immunology , Postoperative Complications/prevention & control , Postoperative Period , Treatment Outcome
20.
Transl Cancer Res ; 9(12): 7440-7449, 2020 Dec.
Article En | MEDLINE | ID: mdl-35117344

BACKGROUND: Annexin A3 (ANXA3) is overexpressed in various cancers and is a potential target for cancer treatment. However, clinical implication and biological function of ANXA3 in colon cancer remain unknown. This study aimed to investigate the relationship between hypoxia-inducible factor 1-alpha (HIF-1α) and ANXA3, and explore the function of ANXA3 in colon carcinoma. METHODS: Expression levels of HIF-1α and ANXA3 in human colon carcinoma specimens and colon cancer cell lines were detected by immunohistochemistry, real-time PCR and Western blot analysis. The proliferation of colon cancer cells was examined. Nude mice were used for xenograft tumor model, and HIF-1α siRNA or control adenovirus was injected into the tumor. RESULTS: HIF-1α and ANXA3 expression levels were higher in colon cancer tissues than their expression levels in normal colon tissues. In addition, HIF-1α and ANXA3 expression increased in colon cancer cells under hypoxic condition. Knockdown of HIF-1α decreased HIF-1α and ANXA3 expression, and inhibited the proliferation and growth of colon cancer cells. In nude mouse model, silencing HIF-1α decreased volume of xenograft tumor and ANXA3 expression. CONCLUSIONS: ANXA3 expression is upregulated by HIF-1α in colon cancer in response to hypoxic stress and contributes to colon tumor growth. ANXA3 may represent a new therapeutic target for colon carcinoma.

...