1
|
Jiang M, Ma S, Xuan Y, Chen K. Synthetic approaches and clinical application of KRAS inhibitors for cancer therapy. Eur J Med Chem 2025; 291:117626. [PMID: 40252381 DOI: 10.1016/j.ejmech.2025.117626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 04/08/2025] [Accepted: 04/09/2025] [Indexed: 04/21/2025]
Abstract
Kirsten rat sarcoma viral oncogene homolog (KRAS) mutations are among the most common oncogenic alterations in various cancers, including pancreatic, colorectal, and non-small cell lung cancer (NSCLC). Targeting KRAS has long been considered a difficult challenge due to its high affinity for guanosine triphosphate (GTP) and the lack of a druggable binding site. However, recent advancements in small-molecule inhibitor design have led to the development of targeted therapies aimed at KRAS mutations, particularly the KRASG12C mutation. Inhibitors such as Sotorasib and Adagrasib have shown promise in preclinical and clinical studies by irreversibly binding to the mutant KRAS protein, locking it in an inactive state and disrupting downstream signaling pathways critical for tumor growth and survival. These inhibitors have demonstrated clinical efficacy in treating patients with KRASG12C-mutated cancers, leading to tumor regression, prolonged progression-free survival, and improved patient outcomes. This review discusses the synthetic strategies employed to develop these KRAS inhibitor and also examines the clinical application of these inhibitors, highlighting the challenges and successes encountered during clinical trials. Ultimately, KRAS inhibitors represent a breakthrough in cancer therapy, offering a promising new treatment option for patients with KRAS-driven tumors.
Collapse
Affiliation(s)
- Min Jiang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Shaowei Ma
- Department of Interventional Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ying Xuan
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| | - Kuanbing Chen
- Department of Thoracic Surgery, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
2
|
Liang C, Wu Y, Wang L, Bai J, Wang D, Zhao F. Adoption of enhanced multislice spiral computed tomography combined with magnetic resonance imaging in staging and preoperative assessment of colon cancer patients. Biomed Mater Eng 2025:9592989251335125. [PMID: 40259556 DOI: 10.1177/09592989251335125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2025]
Abstract
BackgroundColon cancer (CC) refers to malignant tumor of the digestive tract worldwide and is also among the cancers with high mortality rates.ObjectiveThe aim of this work was to evaluate the diagnostic performance of multislice spiral CT (MSCT), magnetic resonance imaging (MRI), and MSCT + MRI in different stages of colon cancer (CC) (T1-T2, T3, T4). This work compared the differences in sensitivity (Sen), specificity (Spe), accuracy (Acc), and area under the curve (AUC) values among these methods and explored the optimal diagnostic strategy.MethodsA total of 120 patients with CC confirmed by pathological biopsy and 30 individuals suspected of CC but without detected tumors (as controls) were selected. All subjects underwent MSCT, MRI, and combined MSCT + MRI examinations. Statistical analyses of Sen, Spe, Acc, and AUC values were performed.ResultsIn the T1-T2 stage, MSCT had a Sen of 85.2%, Acc of 86.8%, and an AUC value of 0.878; MRI had a Spe of 91.0%, Sen of 81.6%, and an AUC value of 0.865; the combined MSCT + MRI examination had a Sen of 90.6% and an AUC of 0.903. In the T3 stage, MRI had a significantly higher Sen (91.7%) than MSCT (80.0%), with an AUC of 0.887, while the combined MSCT + MRI examination had a Sen of 98.3% and an AUC of 0.942. In the T4 stage, the combined MSCT + MRI examination performed the best, with a Sen of 100% and an AUC of 0.933, and compared with MSCT or MRI alone, the differences were statistically significant (P < 0.05).ConclusionMSCT and MRI each have their own advantages in the diagnosis of different stages of CC. MSCT is suitable for initial screening in the T1-T2 stage, while MRI is more effective in assessing tumor invasiveness in the T3 and higher stages. The combined MSCT + MRI examination can provide more comprehensive diagnostic information, especially in the T4 stage, where it shows the highest Sen and Acc. Selecting the appropriate examination method based on the patient's specific condition and staging needs is of great significance in improving the diagnostic Acc of CC.
Collapse
Affiliation(s)
- Cong Liang
- Department of Gastroenterology, Shaanxi Second People's Hospital, Xi'an, Shaanxi Province, China
| | - Ying Wu
- Department of Gastroenterology, Shaanxi Second People's Hospital, Xi'an, Shaanxi Province, China
| | - Limei Wang
- Department of Gastroenterology, Shaanxi Second People's Hospital, Xi'an, Shaanxi Province, China
| | - Junfang Bai
- Department of Gastroenterology, Shaanxi Second People's Hospital, Xi'an, Shaanxi Province, China
| | - Dan Wang
- Department of Gastroenterology, Shaanxi Second People's Hospital, Xi'an, Shaanxi Province, China
| | - Fei Zhao
- Department of Medical Imaging, Norinco General Hospital, Xi'an, Shaanxi Province, China
| |
Collapse
|
3
|
Han B, Zhang Y, Feng X, Yang J, Wang B, Fang J, Wang Z, Zhu J, Niu G, Guo Y. The power of microbes: the key role of gut microbiota in the initiation and progression of colorectal cancer. Front Oncol 2025; 15:1563886. [PMID: 40297806 PMCID: PMC12034544 DOI: 10.3389/fonc.2025.1563886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
Colorectal cancer (CRC) is ranked as the third most prevalent malignancy and is a leading cause of cancer-related mortality globally, significantly affecting the health and longevity of middle-aged individuals and the elderly. The primary risk factors for CRC are mainly due to unhealthy dietary habits and lifestyle choices, and they have been shown to profoundly influence the composition of the gut microbiota. Given that dietary patterns are critical determinants of gut microbial diversity, a compelling association exists between gut microbiota and the pathogenesis of CRC. Recent research has increasingly focused on the intricate interplay between gut microbiota and CRC, exploring its role in disease initiation, progression, and the modulation of host immune responses. Investigations have demonstrated that certain specific microbial communities can promote inflammation, disrupt metabolic pathways, and produce carcinogenic compounds, thereby contributing to the development of CRC. Conversely, a diverse and balanced gut microbiome may confer protective effects against cancer through mechanisms such as the production of short-chain fatty acids and the enhancement of intestinal barrier integrity. This article provides a comprehensive overview of the characteristics of the gut microbial community and its complex relationship with CRC. It highlights potential mechanisms through which gut microbiota may influence CRC pathogenesis, including chronic inflammation, toxins, metabolites, epigenetic dysregulation, and immune regulatory dysfunction. Additionally, this review summarizes innovative strategies for CRC prevention and treatment, emphasizing the therapeutic potential of probiotics and natural plant extracts. By elucidating these connections, this work aims to enhance the understanding of the gut microbiome's role in CRC.
Collapse
Affiliation(s)
- Bo Han
- Department of General Surgery, 63650 Military Hospital, Urumqi, China
| | - Yongfeng Zhang
- Department of General Surgery, 63650 Military Hospital, Urumqi, China
| | - Xue Feng
- Department of Cardiology, 63650 Military Hospital, Urumqi, China
| | - Jun Yang
- Department of General Surgery, 63650 Military Hospital, Urumqi, China
| | - Baolin Wang
- Department of General Surgery, 63650 Military Hospital, Urumqi, China
| | - Jiang Fang
- Department of General Surgery, 63650 Military Hospital, Urumqi, China
| | - Zhigang Wang
- Department of General Surgery, 63650 Military Hospital, Urumqi, China
| | - Jun Zhu
- Department of General Surgery, 63650 Military Hospital, Urumqi, China
| | - Ge Niu
- Department of General Surgery, 63650 Military Hospital, Urumqi, China
| | - Youxiang Guo
- Department of General Surgery, 63650 Military Hospital, Urumqi, China
| |
Collapse
|
4
|
Samanta S, Roy J, Debnath B, Ljungman M, Neamati N. PSP205, a Novel Phenyl Sulfonyl Piperidine, Induces Apoptotic Cell Death in Colon Cancer by Modulating Coat Protein Complex-Mediated Vesicle Trafficking. ACS Pharmacol Transl Sci 2025; 8:1072-1086. [PMID: 40242573 PMCID: PMC11997887 DOI: 10.1021/acsptsci.4c00617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/20/2024] [Accepted: 12/26/2024] [Indexed: 04/18/2025]
Abstract
The endoplasmic reticulum (ER) stress and autophagic pathways offer attractive targets for the development of new cancer drugs. Here, we identified a novel phenyl sulfonyl piperidine, PSP205, that induces prolonged ER-stress-mediated autophagy and apoptosis in colon cancer cells. Transcriptome analysis of cells exposed to PSP205 unveiled transcriptional upregulation of genes associated with the ER stress response or unfolded protein response (UPR), in addition to vesicle transport. Among the top upregulated genes, DNAJB9, XBP1, PDIA4, HSPA5, SEC24D, and SEC11C are implicated in ER stress. Gene set enrichment analysis revealed the enrichment of gene sets involved in the UPR, mTORC1 signaling, hypoxia, the P53 pathway, apoptosis, and the ER-Golgi-vesicle-mediated transport pathway. Mechanistic studies showed that PSP205 acts on the IRE1-TRAF2-JNK pathway to modulate autophagic flux, leading to macroautophagy, ER-phagy, and deformation of Golgi. Our study also demonstrated that PSP205 decreases the expression of the COPI coat complex subunit beta 2 (COPB2) in the presence of COPB2 siRNA. Furthermore, PSP205 synergistically killed colon cancer cells in combination with proteasome and topoisomerase inhibitors. Cumulatively, our findings suggest that PSP205 targets cancer cells via a novel mechanism, specifically by decreasing the level of COPB2, which has not been extensively studied in the context of cancer therapy development and warrants further investigation.
Collapse
Affiliation(s)
- Soma Samanta
- Department
of Medicinal Chemistry, College of Pharmacy, Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Joyeeta Roy
- Department
of Medicinal Chemistry, College of Pharmacy, Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Bikash Debnath
- Department
of Medicinal Chemistry, College of Pharmacy, Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Mats Ljungman
- Department
of Radiation Oncology, Rogel Cancer Center, and Center for RNA Biomedicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Nouri Neamati
- Department
of Medicinal Chemistry, College of Pharmacy, Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
5
|
Duan Y, Wu Y, Tian J, Yin Y, Yuan Z, Zhu W, Zhou S, Li C, Feng S. Elucidation of the mechanism Underlying the promotion of ferroptosis and enhanced antitumor immunity by citrus polymethoxyflavones in CRC cells. Front Pharmacol 2025; 16:1571178. [PMID: 40290432 PMCID: PMC12021823 DOI: 10.3389/fphar.2025.1571178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 04/01/2025] [Indexed: 04/30/2025] Open
Abstract
Background Colon cancer is a prevalent condition with a high mortality rate on a global scale. Research has indicated that Citrus polymethoxyflavones (PMFs), a class of flavonoids found in Citrus, possess the potential to demonstrate anti-tumor efficacy. Ferroptosis a form of cell death that is dependent on iron accumulation and lipid peroxidation. Immunotherapy is one of the most commonly used anti-tumor modalities in a clinical setting. Consequently, studies on the pharmacodynamic mechanism of Citrus to determine whether it can modulate tumor immunity through ferroptosis provide new ideas for the clinical treatment of colon cancer. Purpose The objective of this study is to ascertain whether Citrus inhibits PD-L1 through ferroptosis and promotes tumor immunity among patients with colon cancer. Methods The inhibitory effect of PMFs on colon cancer was proved by in vitro experiment and in vivo model. In addition, the occurrence of ferroptosis was detected by measuring key ferroptosis indicators. Bioinformatics analysis was then performed to identify the crossover genes for Citrus polymethoxylflavonoids, colon cancer, and ferroptosis. Finally, key genes were identified by immunocorrelation analysis including WB, Q-PCR and flow cytometry. These experiments were designed to reveal the potential mechanisms of PMFs on ferroptosis and anti-tumor immunity. Results In vitro cell proliferation experiment and the growth of transplanted tumor mice showed that PMFs had inhibitory effect on colon cancer. In addition, the change of ferroptosis index showed that PMFs promoted the occurrence of ferroptosis, followed by Q-PCR and WB detection of NOX4 and TIMP1, the key genes screened by bioinformatics, found that PMFs inhibited PD-L1 by down-regulating TIMP1, thus affecting colon cancer. Flow cytometry showed that CD4+ T expression increased and CD8+ T cell expression decreased after treatment, suggesting that anti-tumor immunity was activated. Conclusion It is conceivable that the tumor immune microenvironment may be subject to regulation during the inhibition of colon cancer through ferroptosis in PMFs. The ferroptosis-related gene TIMP1 has been observed to regulate PD-L1, thereby promoting anti-tumor immunity in colon cancer. However, further investigation is required to ascertain the underlyingprecise mechanisms.
Collapse
Affiliation(s)
- Yingying Duan
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Department of Pharmacy, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yu Wu
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Department of Pharmacy, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jiaqi Tian
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Department of Pharmacy, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yuqin Yin
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Department of Pharmacy, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Zhongwen Yuan
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Department of Pharmacy, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Wenting Zhu
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Department of Pharmacy, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Suyue Zhou
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Department of Pharmacy, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Chen Li
- Department of Pharmacy, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Senling Feng
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Department of Pharmacy, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
6
|
Zhou H, Gao Z, Wu X, Wang Y, Zhang L. CSF2 promotes chemoresistance in colorectal cancer by regulating Notch pathway. Discov Oncol 2025; 16:495. [PMID: 40202620 PMCID: PMC11981976 DOI: 10.1007/s12672-025-02285-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 04/01/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Increasing evidence suggests that resistance to 5-fluorouracil (5FU) and oxaliplatin (OXP) in colorectal cancer (CRC) is linked to poor prognosis. This study aimed to probe the effect of colony-stimulating factor 2 (CSF2) on the resistance of CRC to 5FU and OXP. METHODS The expression of CSF2 in CRC and the impact of abnormal CSF2 expression on the prognosis of CRC patients were analyzed using bioinformatics. The half-maximal inhibitory concentrations (IC50) of 5FU and OXP on CRC cells were determined using the CCK-8 assay. Apoptosis in CRC cells was assessed through flow cytometry. mRNA and protein levels were measured using qRT-PCR and western blot, respectively. Gene Set Enrichment Analysis (GSEA) was conduced to investigate the signaling pathways regulated by CSF2 in CRC. The Notch pathway activator Jagged-1 (JAG) was employed to verify whether CSF2 influences the resistance of CRC cells to 5-FU and OXP by modulating the Notch signaling pathway. RESULTS High expression of CSF2 is associated with poor prognosis in CRC patients. CSF2 is downregulated in CRC cells that resistance to 5-FU and OXP. Silencing CSF2 inhibits resistance to 5FU and OXP, reduces the survival of resistant CRC cells, and promotes apoptosis. CSF2 activates the Notch signaling pathway, which is highly expressed in CRC resistant cells; conversely, silencing CSF2 inhibits the activation of this pathway. Treatment with JAG reversed the effects of CSF2 silencing on resistance to 5FU and OXP in CRC cells. CONCLUSION The silencing of CSF2 inhibited the resistance of CRC cells to 5FU and OXP by regulating the Notch signaling pathway.
Collapse
Affiliation(s)
- Hairong Zhou
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical University, No.287 Changhuai Road, Bengbu, 233004, Anhui, China
| | - Zhenyuan Gao
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical University, No.287 Changhuai Road, Bengbu, 233004, Anhui, China
| | - Xiao Wu
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical University, No.287 Changhuai Road, Bengbu, 233004, Anhui, China
| | - Yaping Wang
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical University, No.287 Changhuai Road, Bengbu, 233004, Anhui, China
| | - Lu Zhang
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical University, No.287 Changhuai Road, Bengbu, 233004, Anhui, China.
| |
Collapse
|
7
|
Han Y, Zhou Z, Li R, Wang H. Tumor-Derived Exosomal circ_0020095 Promotes Colon Cancer Cell Proliferation and Metastasis by Inhibiting M1 Macrophage Polarization. J Biochem Mol Toxicol 2025; 39:e70225. [PMID: 40165503 DOI: 10.1002/jbt.70225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 01/20/2025] [Accepted: 03/03/2025] [Indexed: 04/02/2025]
Abstract
Tumor-associated macrophages (TAM) have been shown to regulate colon cancer (CC) progression. However, it is not clear whether tumor-derived exosomal circular RNA (circRNA) regulates TAM to influence CC progression. The expression levels of circ_0020095, M1 macrophage markers, M2 macrophage markers, and interleukin-1 receptor-associated kinase 1 (IRAK1) were determined by qRT-PCR. Cell proliferation, migration and invasion were examined by EdU assay, wound healing assay and transwell assay. Exosomes derived from CC cells were used to treat M0 macrophages. M1 macrophage surface marker CD86 was detected by flow cytometry, and protein expression was examined by western blot. Then, the medium of exosome-treated M0 macrophages was used to culture CC cells to determine CC cell functions. RNA pull-down assay, RIP assay and dual-luciferase reporter assay were performed to validate interaction. Circ_0020095 had elevated expression in CC tissues and cells, and its knockdown repressed CC cell proliferation and metastasis. M0 macrophages could take by CC cell-derived exosomes to regulate circ_0020095 expression. Exosomal circ_0020095 restrained M1 macrophage polarization and increased M2 macrophage polarization to enhance CC cell progression. Besides, IRAK1 silencing could promote CC cell proliferation and metastasis by inhibiting M1 macrophage polarization, and its overexpression also abolished the effect of exosomal circ_0020095. Mechanistically, circ_0020095 could competitively bind to IGF2BP1 and then reduced the binding ability of IGF2BP1 and IRAK1 3'UTR. Tumor-derived exosomal circ_0020095 promoted CC cell progression via inhibiting M1 macrophage polarization through IGF2BP1/IRAK1 axis.
Collapse
Affiliation(s)
- Yue Han
- The Second Department of Gastrointestinal Surgery, Shandong Provincial Third Hospital, Jinan City, China
| | - Zhe Zhou
- The Second Department of Gastrointestinal Surgery, Shandong Provincial Third Hospital, Jinan City, China
| | - Rudong Li
- The Second Department of Gastrointestinal Surgery, Shandong Provincial Third Hospital, Jinan City, China
| | - Hong Wang
- The Second Department of Gastrointestinal Surgery, Shandong Provincial Third Hospital, Jinan City, China
| |
Collapse
|
8
|
Jiang B, Li J, Wang J. Exploration of the Prognostic Value of m5C Methylation Protein NOP2 and NSUN6 in Colon Cancer. Genet Test Mol Biomarkers 2025; 29:74-84. [PMID: 40067737 DOI: 10.1089/gtmb.2024.0416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025] Open
Abstract
Background: Colorectal cancer is a prevalent malignancy with high incidence and poor prognosis. This study explores the clinical significance of 5-methylcytosine RNA modification factors, specifically the NOP2/Sun RNA Methyltransferase (NSUN) family, in colorectal cancer. Methods: Utilizing data from The Cancer Genome Atlas database, we analyzed the expression levels of NSUN family members in tumor tissues, their prognostic relevance, and their relationship with immune cell infiltration. To further investigate, paraffin-embedded cancer tissue microarrays were used to assess the expression of NOP2 Nucleolar Protein (NOP2) and NSUN6 in colorectal cancer tissues and adjacent normal tissues. The correlation between the expression of these genes and patient prognosis was also examined. Results: Bioinformatic analysis revealed that NOP2 is highly expressed in tumors, whereas NOP2/Sun RNA Methyltransferase 6 (NSUN6) is linked to poor prognosis. Immune infiltration analysis demonstrated that NOP2 expression is significantly correlated with varying levels of immune cell infiltration, including a positive association with myeloid-derived suppressor cells (MDSCs), M1 macrophages, and natural killer cells and a negative correlation with regulatory T cells and M2 macrophages. NSUN6 expression showed a significant positive correlation with MDSC infiltration. Clinical sample analysis indicated that NOP2 expression is strongly associated with tumor grade and nerve invasion, whereas NSUN6 is significantly related to nerve invasion. Survival analyses revealed that high levels of NOP2 and NSUN6 are linked to shorter overall survival. Notably, NSUN6 expression, vascular invasion, and T stage emerged as key predictors of colorectal cancer patient survival. Conclusions: These findings suggest that NOP2 and NSUN6 may serve as valuable molecular markers for predicting poor prognosis in colorectal cancer, with potential applications in clinical decision-making.
Collapse
Affiliation(s)
- Bin Jiang
- Tonglu District, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, People's Republic of China
| | - Jie Li
- Department of Hepatobiliary Surgery, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, People's Republic of China
| | - Jianguo Wang
- Department of Hepatobiliary Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, People's Republic of China
| |
Collapse
|
9
|
Liu ZY, Liu RT, Cheng WH, Zhang BY, Zhang XY, Zhou Y, Ye XQ, Zhou CY, Wang XJ, Sun Q, Ji J. Neratinib derivative 7A induces apoptosis in colon cancer cells via the p53 pathway. Bioorg Med Chem Lett 2025; 117:130069. [PMID: 39674380 DOI: 10.1016/j.bmcl.2024.130069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 12/05/2024] [Accepted: 12/07/2024] [Indexed: 12/16/2024]
Abstract
Colorectal cancer remains a significant health threat, with its incidence continuously rising, underscoring the urgent need for the development of new therapeutic agents. In our previous research, we identified 7A, a derivative of Neratinib, as having pronounced antitumor activity. However, its specific effects and mechanisms in colorectal cancer have not been thoroughly investigated. Therefore, this study employed in vivo and in vitro experiments, utilizing techniques such as RNA sequencing, Western blotting, and PCR, to provide a comprehensive analysis of 7A's mechanism of action in colorectal cancer. The results indicate that 7A induces DNA damage and activates the P53 pathway, thereby promoting apoptosis in colorectal cancer cells. Additionally, 7A treatment significantly reduced angiogenesis and tumor weight. Our findings suggest that 7A, a Neratinib derivative, holds promise as a novel candidate for colorectal cancer therapy.
Collapse
Affiliation(s)
- Zhi-Yu Liu
- College of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, China
| | - Ruo-Tong Liu
- College of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, China
| | - Wen-Hao Cheng
- College of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, China
| | - Bo-Yu Zhang
- College of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, China
| | - Xing-Yu Zhang
- College of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, China
| | - Ying Zhou
- College of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, China
| | - Xiao-Qing Ye
- College of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, China
| | - Chun-Yun Zhou
- College of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, China
| | - Xiu-Jun Wang
- College of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, China.
| | - Qian Sun
- The First People's Hospital of Lianyungang, The First Affiliated Hospital of Kangda College of Nanjing Medical University. Lianyungang 222000, China.
| | - Jing Ji
- College of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, China; College of Pharmacy and Chemistry and Chemical Engineering, Taizhou University, Taizhou, 225 300, China.
| |
Collapse
|
10
|
Wu H, Zhang W, Chang J, Wu J, Zhang X, Jia F, Li L, Liu M, Zhu J. Comprehensive analysis of mitochondrial-related gene signature for prognosis, tumor immune microenvironment evaluation, and candidate drug development in colon cancer. Sci Rep 2025; 15:6173. [PMID: 39979377 PMCID: PMC11842742 DOI: 10.1038/s41598-024-85035-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 12/30/2024] [Indexed: 02/22/2025] Open
Abstract
Colon adenocarcinoma (COAD), a common digestive system malignancy, involves crucial alterations in mitochondria-related genes influencing tumor growth, metastasis, and immune evasion. Despite limited studies on prognostic models for these genes in COAD, we established a mitochondrial-related risk prognostic model, including nine genes based on available TCGA and MitoCarta 3.0 databases, and validated its predictive power. We investigated the tumor microenvironment (TME), immune cell infiltration, complex cell communication, tumor mutation burden, and drug sensitivity of COAD patients using R language, CellChat, and additional bioinformatic tools from single-cell and bulk-tissue sequencing data. The risk model revealed significant differences in immune cell infiltration between high-risk and low-risk groups, with the strongest correlation found between tissue stem cells and macrophages in COAD. The risk score exhibited a robust correlation with TME signature genes and immune checkpoint molecules. Integrating the risk score with the immune score, microsatellite status, or TMB through TIDE analysis enhanced the accuracy of predicting immunotherapy benefits. Predicted drug efficacy offered options for both high- and low-risk group patients. Our study established a novel mitochondrial-related nine-gene prognostic signature, providing insights for prognostic assessment and clinical decision-making in COAD patients.
Collapse
Affiliation(s)
- Hao Wu
- Department of Medical Cell Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China
| | - Wentao Zhang
- Department of Medical Cell Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China
| | - Jingjia Chang
- Department of Medical Cell Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China
| | - Jin Wu
- Department of Molecular & Cellular Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | - Xintong Zhang
- Department of Medical Cell Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China
| | - Fengfeng Jia
- Taiyuan Technology Transfer Promotion Center, Taiyuan, 030006, China
| | - Li Li
- Department of Medical Cell Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China
| | - Ming Liu
- Department of Medical Cell Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China.
| | - Jianjun Zhu
- Department of Medical Cell Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China.
| |
Collapse
|
11
|
Zhu Q, Wang M, Wang Y, Li B, Zheng J, Hu Y, Shi C, Wang D, Cao D, Liu Z, Zheng X, Wang K. Discovery of novel xanthohumol C derivatives regulating XRCC2 transcription and expression for the treatment of colorectal cancer. Bioorg Med Chem 2025; 118:118048. [PMID: 39731930 DOI: 10.1016/j.bmc.2024.118048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 12/13/2024] [Accepted: 12/18/2024] [Indexed: 12/30/2024]
Abstract
X-ray repair cross-complementing 2 (XRCC2), a critical protein in homologous recombination (HR), plays a significant role in the occurrence, progression, and drug resistance of colorectal cancer (CRC). In this study, a series of xanthohumol C derivatives were synthesized, and their anticancer activity was evaluated. The results revealed that A33 demonstrated the potent anticancer activity and effectively inhibited the proliferation of CRC cells in vitro. Mechanistic investigations revealed that A33 suppressed the transcription and expression of XRCC2, resulting in cell cycle delay and the accumulation of DNA damage, ultimately leading to cell proliferation inhibition. Furthermore, A33 displayed high safety, favorable bioavailability (F = 37.51 %), and potent tumor growth inhibition in vivo, which highlighting its potential as a candidate for the development of novel anti-CRC therapies.
Collapse
Affiliation(s)
- Qianqian Zhu
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China; Taizhou Hospital of Zhejiang Province, Taizhou, Zhejiang 317000, China
| | - Mengying Wang
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China
| | - Yan Wang
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China
| | - Bin Li
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China
| | - Jiahao Zheng
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China
| | - Yina Hu
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China
| | - Changgui Shi
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China
| | - Dalong Wang
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China
| | - Di Cao
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China
| | - Zhiguo Liu
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China.
| | - Xiaohui Zheng
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China; The Key Laboratory of Pediatric Hematology and Oncology Diseases of Wenzhou, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Kun Wang
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China.
| |
Collapse
|
12
|
Wang M, Qi J, Tan Z, Zhou R, Zhuo Q, Deng X, Wang Z, Zhou R, Li F, Xu Y. GABPα targeted by miR-378a-5p inhibits the growth and angiogenesis of colorectal carcinoma. Int J Biochem Cell Biol 2025; 179:106729. [PMID: 39710138 DOI: 10.1016/j.biocel.2024.106729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 11/26/2024] [Accepted: 12/19/2024] [Indexed: 12/24/2024]
Abstract
Considering the high degree of malignancy, recurrence rate and poor prognosis, exploring promising targets is an imperious strategy for colorectal carcinoma therapy. Recent studies have indicated that GABPα plays a role in cancer aggressiveness, but its exact function and regulatory mechanisms in colorectal cancer progression remain unclear. This study aims to explore the biological role of GABPα and its upstream regulator, miR-378a-5p, in modulating cancer progression. The expression levels of GABPα and miR-378a-5p were analyzed through comprehensive data mining and qPCR assays. The functional effects of GABPα were assessed using CCK-8, wound healing, transwell invasion assay, tube formation and xenograft model in nude mice. A co-transfection assay was also performed to investigate the regulatory relationship between miR-378a-5p and GABPα. We found that GABPα expression was significantly downregulated in human colorectal cancer tissues and cell lines. Functional assays revealed that GABPα overexpression suppressed the proliferation, migration, invasion and angiogenesis of colorectal cancer cells, and in vivo experiments further confirmed the inhibitory role of GABPα. Additionally, miR-378a-5p was upregulated in colorectal cancer, and GABPα was identified as a direct target of miR-378a-5p, as confirmed by luciferase reporter assays. Furthermore, overexpression of GABPα partially counteracted the enhanced malignant behaviors of cancer cells induced by miR-378a-5p. Our findings suggest that miR-378a-5p promotes the aggressive progression of colorectal cancer by directly targeting GABPα, highlighting this regulatory axis as a potential therapeutic target for colorectal carcinoma.
Collapse
Affiliation(s)
- Mengyi Wang
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei 430081, China
| | - Jiangfa Qi
- College of Biomedicine and Health, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Zhenlin Tan
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei 430081, China
| | - Runlong Zhou
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei 430081, China
| | - Qing Zhuo
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei 430081, China
| | - Xiaotong Deng
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei 430081, China
| | - Zhenrong Wang
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei 430081, China
| | - Ruijie Zhou
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei 430081, China
| | - Fan Li
- Wuhan Bio-Raid Biotechnology Co., Ltd., Wuhan, Hubei 430075, China
| | - Yao Xu
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei 430081, China.
| |
Collapse
|
13
|
Liu F, Wang Y, Xia L, Sun C, Li Y, Xia Y. Immunological characterization and prognostic of colon cancer evaluated by angiogenesis-related features: a computational analysis and in vitro experiments. Discov Oncol 2025; 16:101. [PMID: 39881026 PMCID: PMC11780071 DOI: 10.1007/s12672-025-01835-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 01/20/2025] [Indexed: 01/31/2025] Open
Abstract
BACKGROUND Diseases are often caused by multiple factors, angiogenesis-related genes (ARGs) have been shown to be associated with cancer, however, their role in colon cancer had not been fully explored. This study investigated potential biomarkers based on ARGs to improve prognosis and treatment effect in colon cancer. METHODS ARGs associated with colon cancer prognosis were identified using Cox regression analysis and LASSO analysis. Furthermore, a prognostic model was constructed in colon cancer based on the 3 ARGs, and its biological function were analyzed. We evaluated the differences in tumor immune microenvironment based on prognostic signature. Finally, cell experiments confirmed the function of genes in colon cancer. RESULTS The prognostic value of ARGs in colon cancer patients has been comprehensively analyzed for the first time and identified 3 ARGs with prognostic values. A prognosis risk model was constructed based on 3 ARGs and its prognostic value was validated on an independent external colon cancer dataset. In colon cancer patients, this prognostic feature was an independent risk factor and was significantly correlated with clinical feature information of colon cancer patients. This feature was also related to the immune microenvironment of colon cancer. Cell experiments showed that high expression of TNF Receptor Superfamily Member 1B (TNFRSF1B) significantly promoted apoptosis and inhibited proliferation of colon cancer cells. Therefore, TNFRSF1B may become an important regulatory factor in the progression of colon cancer by participating in intracellular functional regulation. CONCLUSIONS This study constructed a prognostic risk model based on three ARGs and for the first time discovered that TNFRSF1B may become an important regulatory factor in cancer progression by participating in intracellular functional regulation.
Collapse
Affiliation(s)
- Fei Liu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Oncology, Anhui Public Health Clinical Center, Hefei, China
| | - Yi Wang
- Department of Oncology, The Third Affiliated Hospital of Anhui Medical University, Anhui, China
| | - Leiming Xia
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Hematology, Anhui Public Health Clinical Center, Hefei, China
| | - Chen Sun
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Oncology, Anhui Public Health Clinical Center, Hefei, China
| | - Yun Li
- School of Biology and Pharmaceutical Engineering, Wuhan Polytechnic University, Wuhan, China
| | - Yunhong Xia
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
- Department of Oncology, Anhui Public Health Clinical Center, Hefei, China.
| |
Collapse
|
14
|
Li R, He S, Qin T, Ma Y, Xu K, Liu S, Zhan W. Glycosylation gene expression profiles enable prognosis prediction for colorectal cancer. Sci Rep 2025; 15:798. [PMID: 39755729 PMCID: PMC11700200 DOI: 10.1038/s41598-024-84300-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 12/23/2024] [Indexed: 01/06/2025] Open
Abstract
This study developed a prognostic model for patients with colon adenocarcinoma (COAD) based on glycosylation-associated genes. By analyzing TCGA-COAD data, 110 key genes were identified, and a prognostic model incorporating five glycosylation-related genes was constructed. The model exhibits good predictive performance and is significantly associated with clinical features such as age, N stage, M stage, and lymph node count. The prognostic genes are involved in various biological processes and pathways, influence T cell differentiation, and may contribute to CRC development. High-risk patients show a higher degree of immune cell infiltration. This model aids in the early diagnosis, prognosis assessment, and treatment planning for CRC, and offers a direction for further research.
Collapse
Affiliation(s)
- Rui Li
- Department of Rehabilitation, Beijing Jishuitan Hospital Guizhou Hospital, Guiyang, 550014, Guizhou, China
| | - Sha He
- Department of Rehabilitation, Beijing Jishuitan Hospital Guizhou Hospital, Guiyang, 550014, Guizhou, China
| | - Ting Qin
- Department of Rehabilitation, Beijing Jishuitan Hospital Guizhou Hospital, Guiyang, 550014, Guizhou, China
| | - Yanyan Ma
- Department of Rehabilitation, Beijing Jishuitan Hospital Guizhou Hospital, Guiyang, 550014, Guizhou, China
| | - Kunyao Xu
- Department of Geriatrics, The Second Affiliated Hospital of Guizhou, University of Traditional Chinese Medicine, Guiyang, 550003, Guizhou, China
| | - Shan Liu
- The Second Clinical School of Guizhou, University of Traditional Chinese Medicine, Guiyang, 550003, Guizhou, China
| | - Wei Zhan
- Department of Anus and Intestine Surgery, The Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Street, Yunyan District, Guiyang City, 550004, Guizhou Province, China.
| |
Collapse
|
15
|
Dong L, Liu S, Sun W, Liu S, Zhang N, Zhang S. Mitochondrial Deoxyguanosine Kinase Induces 5-Fluorouracil Chemotherapy Sensitivity through Autophagy. Curr Cancer Drug Targets 2025; 25:306-316. [PMID: 39171468 DOI: 10.2174/0115680096337375240801080008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/15/2024] [Accepted: 07/19/2024] [Indexed: 08/23/2024]
Abstract
AIMS The purpose of this study was to investigate the role of DGUOK in the progression of colorectal cancer (CRC) and its impact on the sensitivity of CRC cells to 5-FU treatment. METHODS We conducted bioinformatics analysis and qRT-PCR to evaluate DGUOK expression in CRC tissues/cells. Cell viability of CRC cells treated with 5-FU was assessed using CCK-8 and colony formation assays. Autophagy levels were determined through immunofluorescence assays and Western blot analysis. Additionally, the influence of p-p38 on autophagy was investigated via Western blotting. A rescue assay was performed to confirm whether DGUOK/p38 affects 5-FU sensitivity in CRC cells through autophagy. RESULTS Our findings indicate that DGUOK is upregulated in CRC tissues compared to normal tissues, correlating with increased cell proliferation and migration. Functionally, inhibition of DGUOK enhances autophagy, thereby decreasing the sensitivity of CRC cells to 5-FU. This effect is partly mediated by DGUOK's impact on the mitogen-activated protein kinase (MAPK) pathway, specifically promoting the phosphorylation of p38 MAPK, a crucial regulator in autophagy pathways. CONCLUSION These results suggest that DGUOK could serve as a novel marker for predicting the efficacy of 5-FU in CRC treatment.
Collapse
Affiliation(s)
- Lu Dong
- School of Clinical Medicine, Shandong Second Medical University, Weifang, 261000, China
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, State Key Laboratory for Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Diseases, Beijing, 100050, China
| | - Sifan Liu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, State Key Laboratory for Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Diseases, Beijing, 100050, China
| | - Wenjing Sun
- School of Clinical Medicine, Shandong Second Medical University, Weifang, 261000, China
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, State Key Laboratory for Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Diseases, Beijing, 100050, China
| | - Siying Liu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, State Key Laboratory for Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Diseases, Beijing, 100050, China
| | - Nan Zhang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, State Key Laboratory for Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Diseases, Beijing, 100050, China
| | - Shutian Zhang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, State Key Laboratory for Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Diseases, Beijing, 100050, China
| |
Collapse
|
16
|
Lee DE, Lee HM, Jun Y, Choi SY, Lee SJ, Kwon OS. Metformin induces apoptosis in TRAIL-resistant colorectal cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119873. [PMID: 39500444 DOI: 10.1016/j.bbamcr.2024.119873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 10/10/2024] [Accepted: 10/28/2024] [Indexed: 11/11/2024]
Abstract
Resistance to chemotherapy drugs, which commonly occurs during the treatment of colorectal cancer (CRC), can lead to tumor recurrence and metastasis, so combinational treatment strategies according to the cancer cell type are urgently needed to overcome drug resistance and increase therapeutic efficiency. To this end, the tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a promising anticancer strategy. Some CRC cell lines such as SW620 have low sensitivity to TRAIL, so additional sensitizers are required to make the strategy effective. Therefore, we focused on the apoptotic effect of combinational metformin and TRAIL treatment on TRAIL-resistant SW620 cells. Treatment with TRAIL alone did not induce apoptosis whereas combined treatment with metformin and TRAIL significantly increased it. TRAIL activated caspases through an extrinsic pathway but increased resistance to apoptosis through the protein kinase B or AKT (PKB/AKT)/mammalian target of rapamycin (mTOR) pathway. On the other hand, metformin reduced the inhibitory effect of X-linked inhibitor of apoptosis (XIAP) by blocking the AKT and nuclear factor kappa B (NF-κB) pathways and activated CCAAT-enhancer-binding protein homologous protein (CHOP) via endoplasmic reticulum (ER) stress but without inducing apoptosis. In addition, metformin induced cell-cycle arrest, thereby blocking cell proliferation and growth. These results were also confirmed through an in vivo mouse xenograft CRC model, in which combined treatment with metformin and TRAIL induced tumor cell death, thus demonstrating the anticancer effect of their coadministration. Therefore, cotreatment of metformin and TRAIL could be an effective anticancer treatment strategy for TRAIL-resistant CRC.
Collapse
Affiliation(s)
- Da Eun Lee
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Hae Min Lee
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Yunhyeok Jun
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Soo Young Choi
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea.
| | - Su Jin Lee
- Department of Biochemistry, Chungbuk National University College of Medicine and Medical Research Center, Cheongju 28644, Republic of Korea
| | - Oh-Shin Kwon
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea.
| |
Collapse
|
17
|
Jin L, Lin Z, Jin A. Sterol regulatory element binding transcription factor 1 is an important prognostic factor for colon adenocarcinoma and closely related to immune infiltration. Cytojournal 2024; 21:67. [PMID: 39917010 PMCID: PMC11801665 DOI: 10.25259/cytojournal_43_2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 11/18/2024] [Indexed: 02/09/2025] Open
Abstract
Objective Sterol Regulatory Element Binding Transcription Factor 1 (SREBF1) encodes a core protein that has a crucial function in the metabolism of cholesterol and lipids. This transcription factor is a member of the family of transcription factors and highly expressed in a variety of cancer types. As of now, there are few reports on the relationship between the expression of SREBF1 and colon adenocarcinoma (COAD). Hence, this study utilizes databases and a range of experiments to explore the relationship between the expression of SREBF1 and tumor immune infiltration, as well as the occurrence and development of tumors. Material and Methods The expression of SREBF1 in pan-cancers was retrieved through databases such as TIMER, Gene Expression Profiling Interactive Analysis (GEPIA), and UALCAN. The expression of SREBF1 in HCT-116 and SW480 cells was detected using western blot. Furthermore, we also found that knockdown SREBF1 can inhibit the proliferation and migration of COAD cells. The correlation between SREBF1 and autophagy in COAD cells was detected using acridine orange (AO) staining, western blot, and immunofluorescence (IF). Results The databases of TIMER, GEPIA and UALCAN revealed that SREBF1 is overexpressed in pan-cancer tissues, and closely associated with the prognosis of the patients with cancer. Further immunohistochemical staining showed that SREBF1 was overexpressed in COAD, and closely related to the clinical stage and lymph node metastasis. Western blot revealed that SREBF1 was significantly expressed in both HCT-116 and SW480 COAD cells; knockdown of SREBF1 could inhibit the proliferation, DNA replication, and migration of COAD cells. The AO staining, western blot, and IF experiments also showed that silencing SREBF1 could promote the autophagy of COAD cell. Meanwhile, the TIMER database indicates a significant positive correlation between the presence of immune cells in COAD and variations in copy number alteration of SREBF1. Conclusion SREBF1 might serve as a potential prognostic marker for COAD and be associated with immune cell infiltration.
Collapse
Affiliation(s)
- Liying Jin
- Central Laboratory, The Affiliated Hospital of Yanbian University, Yanji, Jilin, China
- Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji, Jilin, China
| | - Zhenhua Lin
- Central Laboratory, The Affiliated Hospital of Yanbian University, Yanji, Jilin, China
- Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji, Jilin, China
| | - Aihua Jin
- Central Laboratory, The Affiliated Hospital of Yanbian University, Yanji, Jilin, China
| |
Collapse
|
18
|
Guo L, Li X, Kang Y, Sun H. The acceleration of cisplatin resistance in colorectal cancer by lncRNA NORAD through regulation of miR-106a-5p/Cyclin D1 axis. J Chemother 2024:1-12. [PMID: 39648415 DOI: 10.1080/1120009x.2024.2436808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 10/30/2024] [Accepted: 11/18/2024] [Indexed: 12/10/2024]
Abstract
Colorectal cancer (CRC) is one of the leading causes of cancer-related mortality. LncRNA NORAD is frequently upregulated and positively associated with various cancer progressions. We discovered NORAD was significantly upregulated in CRC tissues and cells. NORAD sponged miR-106a-5p to form a ceRNA complex. MiR-106a-5p was remarkedly downregulated in CRC tumors and cells. Silencing NORAD or overexpression of miR-106a-5p effectively increased cisplatin sensitivity. In the established cisplatin resistant cell line, NORAD was upregulated and miR-106a-5p was downregulated. Furthermore, we disclosed miR-106a-5p directly targeted 3'UTR of CCND1, which is an important cell cycle regulator and is frequently overexpressed in human cancers. Rescue experiments showed restoration of CCND1 in miR-106a-5p-overexpressing CRC cells successfully recovered cisplatin resistance. Finally, restoration of miR-106a-5p in NORAD-overexpressing CRC cells re-sensitized cisplatin resistance by targeting CCND1. Summarily, this study uncovered a NORAD-promoted cisplatin resistance through modulating the miR-106a-5p-CCND1 axis, contributing to developing novel therapy for treating chemoresistant CRC.
Collapse
Affiliation(s)
- Liping Guo
- Department of Interventional Radiology, The First Medical Center of the PLA General Hospital, Beijing, China
| | - Xianmei Li
- Department of Interventional Radiology, The First Medical Center of the PLA General Hospital, Beijing, China
| | - Yujuan Kang
- Department of Interventional Radiology, The First Medical Center of the PLA General Hospital, Beijing, China
| | - Hui Sun
- Department of Diagnostic Radiology, The First Medical Center of the PLA General Hospital, Beijing, China
| |
Collapse
|
19
|
Jiang Q, Yao F, An Y, Lai X, Li X, Yu Z, Yang XD. Novel nanotherapeutics for cancer immunotherapy by albumin nanoparticles functionalized with PD-1 and PD-L1 aptamers. Cancer Nanotechnol 2024; 15:3. [DOI: 10.1186/s12645-023-00239-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 12/12/2023] [Indexed: 01/05/2025] Open
Abstract
Abstract
Background
PD-1/PD-L1 blockade plays a crucial role in cancer immunotherapy. Exploration of new technologies to further enhance the efficacy of PD-1/PD-L1 blockade is therefore of potential medical importance. Nanotherapeutics can accumulate in tumor tissues due to enhanced permeability and retention (EPR) effects. In this study, a novel nanotherapeutic for cancer immunotherapy was implemented with albumin nanoparticles functionalized by both PD-1 and PD-L1 aptamers.
Results
Albumin nanoparticles (NP) were functionalized with either PD-1 aptamers (PD1-NP), PD-L1 aptamers (PDL1-NP), or both types of aptamers (PD1-NP-PDL1). Average sizes of PD1-NP, PDL1-NP, and PD1-NP-PDL1 were 141.8 nm, 141.8 nm, and 164.2 nm, respectively. PD1-NP had good affinity for activated T cells that expresses PD-1. Similarly, PDL1-NP could bind with MDA-MB-231 or CT26 tumor cells that express PD-L1. Moreover, the bispecific PD1-NP-PDL1 could bind with both the activated T cells and the PD-L1-expressing tumor cells, and tether the two type of cells together. Functionally, aptamer-modified nanoparticles exhibited stronger immune-stimulating effects vs. free aptamers. Specifically, PD1-NP or PDL1-NP induced stronger lymphocyte-mediated cytotoxicity against PD-L1-expressing tumor cells in vitro vs. free PD-1 or PD-L1 aptamers. Animal studies also showed that PD1-NP or PDL1-NP significantly improved antitumor efficacy against CT26 colon cancer in vivo vs. free PD-1 or PD-L1 aptamers. Importantly, the bispecific PD1-NP-PDL1 further boosted the in vivo antitumor efficacy compared with PD1-NP or PDL1-NP, without raising systemic toxicity.
Conclusion
The results suggest that the bispecific PD1-NP-PDL1 is a promising nanotherapeutic to improve the efficacy of PD-1/PD-L1 blockade, and may have application potential in colon cancer treatment.
Collapse
|
20
|
Antunes FTT, Gandini MA, Gadotti VM, Quintão NLM, Santin JR, Souza IA, David LS, Snutch TP, Hildebrand M, Zamponi GW. Contribution of T-type calcium channel isoforms to cold and mechanical sensitivity in naïve and oxaliplatin-treated mice of both sexes. Br J Pharmacol 2024; 181:5062-5078. [PMID: 39295452 DOI: 10.1111/bph.17337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/01/2024] [Accepted: 08/12/2024] [Indexed: 09/21/2024] Open
Abstract
BACKGROUND AND PURPOSE The chemotherapy agent oxaliplatin can give rise to oxaliplatin-induced peripheral neuropathy (OIPN). Here, we investigated whether T-type calcium channels (Cav3) contribute to OIPN. EXPERIMENTAL APPROACH We chronically treated mice with oxaliplatin and assessed pain responses and changes in expression of Cav3.2 calcium channels. We also tested the effects of T-type channel blockers on cold sensitivity in wild-type and Cav3.2 null mice. KEY RESULTS Oxaliplatin treatment led to mechanical and cold hypersensitivity in male and female mice. Mechanical hypersensitivity persisted in Cav3.2 null mice of both sexes. Intraperitoneal or intrathecal delivery of pan T-type channel inhibitors attenuated mechanical hypersensitivity in wild-type but not Cav3.2 null mice. Remarkably cold hypersensitivity occurred in female but not male Cav3.2 null mice even without oxaliplatin treatment. Unexpectedly, intrathecal, intraplantar or intraperitoneal delivery of T-type channel inhibitors Z944 or TTA-P2 transiently induced cold hypersensitivity in both male and female wild-type mice. Acute knockdown of specific Cav3 isoforms revealed that the depletion of Cav3.1 in males and depletion of either Cav3.1 or Cav3.2 in females triggered cold hypersensitivity. Finally, reducing Cav3.2 expression by disrupting the interactions between Cav3.2 and the deubiquitinase USP5 with the small organic molecule II-2 reversed oxaliplatin-induced mechanical and cold hypersensitivity and importantly did not trigger cold allodynia. CONCLUSION AND IMPLICATIONS Altogether, our data indicate that T-type channels differentially contribute to the regulation of cold and mechanical hypersensitivity, and raise the possibility that T-type channel blockers could promote cold allodynia.
Collapse
Affiliation(s)
- Flavia T T Antunes
- Department of Clinical Neurosciences, Cumming School of Medicine, Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Canada
| | - Maria A Gandini
- Department of Clinical Neurosciences, Cumming School of Medicine, Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Canada
| | - Vinicius M Gadotti
- Department of Clinical Neurosciences, Cumming School of Medicine, Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Canada
- School of Health Sciences, Postgraduate Program in Pharmaceutical Sciences, Universidade do Vale do Itajaí (UNIVALI), Itajaí, Brazil
| | - Nara Lins Meira Quintão
- School of Health Sciences, Postgraduate Program in Pharmaceutical Sciences, Universidade do Vale do Itajaí (UNIVALI), Itajaí, Brazil
| | - José Roberto Santin
- School of Health Sciences, Postgraduate Program in Pharmaceutical Sciences, Universidade do Vale do Itajaí (UNIVALI), Itajaí, Brazil
| | - Ivana A Souza
- Department of Clinical Neurosciences, Cumming School of Medicine, Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Canada
| | | | - Terrance P Snutch
- Michael Smith Laboratories and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | | | - Gerald W Zamponi
- Department of Clinical Neurosciences, Cumming School of Medicine, Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Canada
| |
Collapse
|
21
|
Shalata W, Gluzman A, Man S, Cohen AY, Abu Jama A, Gothelf I, Tourkey L, Neime AE, Abu Juma’a A, Peri-Hanania K, Machluf O, Shoham Levin G, Shalata S, Hayadri A, Abu Zeid EED, Abu Yasin N, Meirovitz A, Yakobson A. Colorectal Cancer in Elderly Patients: Insights into Presentations, Prognosis, and Patient Outcomes. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1951. [PMID: 39768833 PMCID: PMC11678490 DOI: 10.3390/medicina60121951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/04/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025]
Abstract
Background and Objectives: Colorectal cancer (CRC) ranks as the third most prevalent cancer globally and is the third leading cause of cancer-related deaths. In 2020 alone, there were over 1.9 million new cases of CRC and nearly 0.9 million deaths worldwide. The incidence and outcomes of CRC exhibit significant geographical and temporal variations, largely influenced by diverse risk factors among populations. Recognizing the prognostic factors and the presenting symptoms of CRC, a leading global cancer with high mortality, can enhance early detection and thereby improve clinical outcomes. Materials and Methods: This retrospective, observational study analyzed 724 CRC elderly patients aged 70 and over (median age 80, 53.17% male), treated at a single center. Data on demographics, clinical characteristics, and outcomes were collected. Overall survival was analyzed using Kaplan-Meier curves, with stratification based on tumor location, disease staging, lymph node involvement, and family history. Results: Our study encompassed all CRC cases treated with surgery and systemic therapies (chemotherapy or biological agents) from July 2002 to September 2020. We focused on comparing prognosis between left-sided and right-sided CRC, as well as rectal cancer. We found that left-sided CRC demonstrated a superior prognosis compared to rectal cancer (p = 0.0022). Furthermore, among patients with CRC, tumors originating in the rectum were associated with worse outcomes compared to those arising in both the right and left colon, regardless of disease stage (p = 0.0049). Additionally, a family history of CRC was associated with poorer prognosis, impacting both metastatic (p = 0.0022) and localized disease (p = 0.035). The main symptoms prompting patients to start an investigation of CRC were abdominal pain (31.49%), anemia (18.08%), rectal bleeding (hematochezia) (17.82%), change in bowel habits (9.94%), and weight loss (7.60%). Conclusions: This study provides valuable insights into the symptoms prompting initial investigation and the prognostic factors associated with CRC in an elderly population with varied characteristics. It underscores the need for increased vigilance in recognizing key symptoms and the importance of personalized treatment strategies tailored to these prognostic factors.
Collapse
Affiliation(s)
- Walid Shalata
- The Legacy Heritage Cancer Center, Dr. Larry Norton Institute, Soroka Medical Center, Beer-Sheva 84105, Israel
- Medical School for International Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel (E.E.D.A.Z.)
| | - Alexander Gluzman
- The Legacy Heritage Cancer Center, Dr. Larry Norton Institute, Soroka Medical Center, Beer-Sheva 84105, Israel
- Medical School for International Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel (E.E.D.A.Z.)
| | - Sofia Man
- The Legacy Heritage Cancer Center, Dr. Larry Norton Institute, Soroka Medical Center, Beer-Sheva 84105, Israel
- Medical School for International Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel (E.E.D.A.Z.)
| | - Ahron Yehonatan Cohen
- The Legacy Heritage Cancer Center, Dr. Larry Norton Institute, Soroka Medical Center, Beer-Sheva 84105, Israel
- Medical School for International Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel (E.E.D.A.Z.)
| | - Ashraf Abu Jama
- The Legacy Heritage Cancer Center, Dr. Larry Norton Institute, Soroka Medical Center, Beer-Sheva 84105, Israel
- Medical School for International Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel (E.E.D.A.Z.)
| | - Itamar Gothelf
- Goldman Medical School, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel;
| | - Lena Tourkey
- The Legacy Heritage Cancer Center, Dr. Larry Norton Institute, Soroka Medical Center, Beer-Sheva 84105, Israel
| | - Ala Eddin Neime
- Medical School for International Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel (E.E.D.A.Z.)
| | - Ali Abu Juma’a
- Medical School for International Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel (E.E.D.A.Z.)
| | | | - Oshri Machluf
- PhaseV Trials Ltd., Tel Aviv 67443, Israel; (K.P.-H.)
| | | | - Sondos Shalata
- Nutrition Unit, Galilee Medical Center, Nahariya 22000, Israel
| | - Ahab Hayadri
- Medical School for International Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel (E.E.D.A.Z.)
| | - Ez El Din Abu Zeid
- Medical School for International Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel (E.E.D.A.Z.)
| | - Nashat Abu Yasin
- The Legacy Heritage Cancer Center, Dr. Larry Norton Institute, Soroka Medical Center, Beer-Sheva 84105, Israel
- Medical School for International Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel (E.E.D.A.Z.)
| | - Amichay Meirovitz
- The Legacy Heritage Cancer Center, Dr. Larry Norton Institute, Soroka Medical Center, Beer-Sheva 84105, Israel
- Medical School for International Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel (E.E.D.A.Z.)
| | - Alexander Yakobson
- The Legacy Heritage Cancer Center, Dr. Larry Norton Institute, Soroka Medical Center, Beer-Sheva 84105, Israel
- Medical School for International Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel (E.E.D.A.Z.)
| |
Collapse
|
22
|
Yan H, Su Y, Wang L. Impact of Ahmadi Continuing Nursing Model on self-care ability, stoma complications and quality of life of colostomy patients. BMC Gastroenterol 2024; 24:421. [PMID: 39573999 PMCID: PMC11583414 DOI: 10.1186/s12876-024-03497-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 11/07/2024] [Indexed: 11/24/2024] Open
Abstract
OBJECTIVE To analyze the effects of Ahmadi Continuing Nursing Model (ACNM) on the self-care ability, stoma complications and life quality in colostomy patients. METHODS The clinical data of 120 patients who underwent postoperative colostomy in our hospital from June 2020 to June 2023 were retrospectively analyzed. The patients were divided into control group (n = 60, treated with routine nursing) and observation group (n = 60, treated with the ACNM on the basis of routine nursing) according to different nursing methods. Postoperative recovery of gastrointestinal function, ostomy adaptability, self-care ability, and life quality before and after nursing were compared. The probability of complications before and after nursing was recorded between the two groups. RESULTS The time of first exhaust was 3.65 ± 0.82 d, the time of first meal was 1.83 ± 0.65 d, and the first bowel sound recovery was 1.47 ± 0.53 d in the observation group, which were shorter than those in the control group (4.38 ± 1.20 d, 3.12 ± 1.15 d, 2.39 ± 1.06 d, P < 0.001). After intervention, the positive emotions in the ostomy adaptation score were 32.09 ± 5.03 points, negative emotions were 31.41 ± 5.70 points, social life adaptation were 27.12 ± 4.98 points, and the total score was 90.78 ± 5.98 points in the observation group, which were significantly higher than those in the control group (26.32 ± 4.52 points, 24.25 ± 6.02 points, 20.25 ± 4.02 points, 67.25 ± 6.09 points, P < 0.001). The self-willingness was 34.18 ± 4.02 points, self-care skill was 10.57 ± 2.23 points, self-care knowledge was 18.59 ± 3.10 points, and the total score was 63.18 ± 4.98 points in the observation group, which were significantly higher than those in the control group (25.25 ± 3.08 points, 8.72 ± 2.13 points, 15.26 ± 2.70 points, 45.69 ± 4.09 points, P < 0.001). The physical function was 79.74 ± 2.81 points, psychological function was 76.71 ± 3.05 points, social function was 78.11 ± 3.50 points, and material life status was 60.06 ± 2.98 points in the quality of life in the observation group, which were significantly higher than those in the control group (75.36 ± 2.68 points, 69.72 ± 2.93 points, 72.33 ± 3.42 points, 51.23 ± 3.08 points, P < 0.001). CONCLUSION ACNM effectively promoted the recovery of gastrointestinal function after surgery in colostomy patients by improving patients' stoma adaptability, self-care ability and life quality and reducing the occurrence of complications, which was worthy of promotion.
Collapse
Affiliation(s)
- Huiming Yan
- Department of Nursing, Shengjing Hospital of China Medical University, 39 Huaxiang Road, Tiexi District, Shenyang City, Liaoning Province, 110000, China
| | - Ying Su
- Department of Nursing, Shengjing Hospital of China Medical University, 39 Huaxiang Road, Tiexi District, Shenyang City, Liaoning Province, 110000, China
| | - Lina Wang
- Department of Nursing, Shengjing Hospital of China Medical University, 39 Huaxiang Road, Tiexi District, Shenyang City, Liaoning Province, 110000, China.
| |
Collapse
|
23
|
Shahpari M, Hashemi M, Younesirad T, Hasanzadeh A, Mosanne MM, Ahmadifard M. The functional roles of competitive endogenous RNA (ceRNA) networks in apoptosis in human cancers: The circRNA/miRNA/mRNA regulatory axis and cell signaling pathways. Heliyon 2024; 10:e37089. [PMID: 39524849 PMCID: PMC11546195 DOI: 10.1016/j.heliyon.2024.e37089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 08/27/2024] [Indexed: 11/16/2024] Open
Abstract
Circular RNAs are noncoding RNAs with circular conformation mainly due to backsplicing event. CircRNAs can potentially impact cell biological processes by interacting with cell signaling pathways. Numerous circRNAs have been found to be aberrantly expressed in a variety of cancers. These RNAs can act as ceRNA (competitive endogenous RNA) by sponging certain miRNAs to form circRNA/miRNA/mRNA networks. Dysregulation of ceRNA networks may lead to dysfunctions in various cell pathways, which modulate apoptosis-associated genes and ultimately result in cancer progression. Since disruption of apoptosis is one of the leading causes of cancer development, one approach for cancer treatment is to drive cells toward apoptosis. In this review, we present a summary of studies on the role of ceRNA networks in cellular signaling pathways that regulate apoptosis; these networks are suggested to be potential biomarkers for cancer treatment.
Collapse
Affiliation(s)
| | | | - Tayebeh Younesirad
- Department of Medical Genetics, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Aida Hasanzadeh
- Department of Medical Genetics, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Mohammad mahdi Mosanne
- Department of Medical Genetics, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Mohamadreza Ahmadifard
- Department of Medical Genetics, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
24
|
Kuzminska J, Szyk P, Mlynarczyk DT, Bakun P, Muszalska-Kolos I, Dettlaff K, Sobczak A, Goslinski T, Jelinska A. Curcumin Derivatives in Medicinal Chemistry: Potential Applications in Cancer Treatment. Molecules 2024; 29:5321. [PMID: 39598712 PMCID: PMC11596437 DOI: 10.3390/molecules29225321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/20/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024] Open
Abstract
Curcumin, a naturally occurring compound found in the rhizome of Curcuma plants, particularly in turmeric (Curcuma longa L.), exhibits a broad range of biological activities, including anti-inflammatory, antioxidant, and anticancer properties. Curcumin has demonstrated effectiveness in inhibiting tumor growth, arousing interest for its potential in treating various cancers, such as breast, lung, prostate, and brain cancers. However, the clinical application of curcumin is limited due to its low chemical stability, poor water solubility, and low bioavailability. In response to these challenges, structural modifications of curcumin have been explored to improve its pharmacological properties, including enhanced anticancer selectivity index and bioavailability. This review highlights promising chemical modifications of curcumin that could lead to the development of more effective anticancer therapies. By functionalizing the parent curcumin molecule, researchers aim to create more stable and bioavailable compounds with enhanced therapeutic potential, making curcumin derivatives promising candidates for medical applications.
Collapse
Affiliation(s)
- Joanna Kuzminska
- Chair and Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland; (I.M.-K.); (K.D.); (A.S.); (A.J.)
- Doctoral School, Poznan University of Medical Sciences, Bukowska 70, 60-812 Poznan, Poland;
| | - Piotr Szyk
- Doctoral School, Poznan University of Medical Sciences, Bukowska 70, 60-812 Poznan, Poland;
- Chair and Department of Chemical Technology of Drugs, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland; (D.T.M.); (P.B.)
| | - Dariusz T. Mlynarczyk
- Chair and Department of Chemical Technology of Drugs, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland; (D.T.M.); (P.B.)
| | - Pawel Bakun
- Chair and Department of Chemical Technology of Drugs, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland; (D.T.M.); (P.B.)
| | - Izabela Muszalska-Kolos
- Chair and Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland; (I.M.-K.); (K.D.); (A.S.); (A.J.)
| | - Katarzyna Dettlaff
- Chair and Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland; (I.M.-K.); (K.D.); (A.S.); (A.J.)
| | - Agnieszka Sobczak
- Chair and Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland; (I.M.-K.); (K.D.); (A.S.); (A.J.)
| | - Tomasz Goslinski
- Chair and Department of Chemical Technology of Drugs, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland; (D.T.M.); (P.B.)
| | - Anna Jelinska
- Chair and Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland; (I.M.-K.); (K.D.); (A.S.); (A.J.)
| |
Collapse
|
25
|
Yenigun VB, Kocyigit A, Kanimdan E, Balkan E, Gul AZ. Copper (II) increases anti-Proliferative activity of thymoquinone in colon cancer cells by increasing genotoxic, apoptotic, and reactive oxygen species generating effects. Toxicon 2024; 250:108103. [PMID: 39278473 DOI: 10.1016/j.toxicon.2024.108103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 09/12/2024] [Accepted: 09/12/2024] [Indexed: 09/18/2024]
Abstract
Thymoquinone is the main active compound derived from the essential oil of the Nigella sativa plant seed. While thymoquinone is an antioxidant, it has been reported in several studies that thymoquinone has dose-dependent pro-oxidant activity with the Fenton reaction in the presence of transition elements such as iron and copper. This study aimed to investigate cytotoxic, apoptotic, genotoxic, and reactive oxygen species (ROS) generating effects of thymoquinone treated with copper in colon cancer cells. HT-29 cells were treated with pro-oxidant-acting doses of thymoquinone alone and together with the non-toxic dose of Copper (II) Sulfate for 24 h. Cytotoxic, apoptotic, genotoxic, and ROS production activities were analyzed by MTT viability test, Acridine Orange/Ethidium Bromide (AO/EB) staining, alkaline single cell gel electrophoresis and H2DCF-DA assay, respectively. Viability results showed that thymoquinone and copper synergistically affect cancer cells, and DNA damage was increased with the synergic effect. The intracellular ROS was increased when thymoquinone and copper were applied together. Applying redox-active copper (II) with thymoquinone increases DNA damage, apoptosis, and cell death by increasing the amount of intracellular ROS through pro-oxidant activity. Treatments targeting copper-related pathways may open new therapeutic avenues for cancer treatment.
Collapse
Affiliation(s)
- Vildan Betul Yenigun
- Bezmialem Vakıf University, Faculty of Medicine, Department of Medical Biochemistry, Istanbul, Turkey; Bezmialem Vakif University, Vocational School of Health Services, Istanbul, Turkey
| | - Abdurrahim Kocyigit
- Bezmialem Vakıf University, Faculty of Medicine, Department of Medical Biochemistry, Istanbul, Turkey.
| | - Ebru Kanimdan
- Bezmialem Vakıf University, Faculty of Medicine, Department of Medical Biochemistry, Istanbul, Turkey; Bezmialem Vakif University, Vocational School of Health Services, Istanbul, Turkey
| | - Ezgi Balkan
- Bezmialem Vakıf University, Faculty of Medicine, Department of Medical Biochemistry, Istanbul, Turkey
| | - Ayse Zehra Gul
- Bezmialem Vakıf University, Faculty of Medicine, Department of Medical Biochemistry, Istanbul, Turkey
| |
Collapse
|
26
|
Li GG, Chu XF, Xing YM, Xue X, Ihtisham B, Liang XF, Xu JX, Mi Y, Zheng PY. Baicalin Prevents Colon Cancer by Suppressing CDKN2A Protein Expression. Chin J Integr Med 2024; 30:1007-1017. [PMID: 38941045 DOI: 10.1007/s11655-024-4109-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2024] [Indexed: 06/29/2024]
Abstract
OBJECTIVE To observe the therapeutic effects and underlying mechanism of baicalin against colon cancer. METHODS The effects of baicalin on the proliferation and growth of colon cancer cells MC38 and CT26. WT were observed and predicted potential molecular targets of baicalin for colon cancer therapy were studied by network pharmacology. Furthermore, molecular docking and drug affinity responsive target stability (DARTS) analysis were performed to confirm the interaction between potential targets and baicalin. Finally, the mechanisms predicted by in silico analyses were experimentally verified in-vitro and in-vivo. RESULTS Baicalin significantly inhibited proliferation, invasion, migration, and induced apoptosis in MC38 and CT26 cells (all P<0.01). Additionally, baicalin caused cell cycle arrest at the S phase, while the G0/G1 phase was detected in the tiny portion of the cells. Subsequent network pharmacology analysis identified 6 therapeutic targets associated with baicalin, which potentially affect various pathways including 39 biological processes and 99 signaling pathways. In addition, molecular docking and DARTS predicted the potential binding of baicalin with cyclin dependent kinase inhibitor 2A (CDKN2A), protein kinase B (AKT), caspase 3, and mitogen-activated protein kinase (MAPK). In vitro, the expressions of CDKN2A, MAPK, and p-AKT were suppressed by baicalin in MC38 and CT26 cells. In vivo, baicalin significantly reduced the tumor size and weight (all P<0.01) in the colon cancer mouse model via inactivating p-AKT, CDKN2A, cyclin dependent kinase 4, cyclin dependent kinase 2, interleukin-1, tumor necrosis factor α, and activating caspase 3 and mouse double minute 2 homolog signaling (all P<0.05). CONCLUSION Baicalin suppressed the CDKN2A protein level to prevent colon cancer and could be used as a therapeutic target for colon cancer.
Collapse
Affiliation(s)
- Gang-Gang Li
- Henan Key Laboratory of Helicobacter pylori, Microbiota and Gastrointestinal Cancers, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 400015, China
| | - Xiu-Feng Chu
- Henan Key Laboratory of Helicobacter pylori, Microbiota and Gastrointestinal Cancers, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 400015, China
| | - Ya-Min Xing
- Henan Key Laboratory of Helicobacter pylori, Microbiota and Gastrointestinal Cancers, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 400015, China
| | - Xia Xue
- Henan Key Laboratory of Helicobacter pylori, Microbiota and Gastrointestinal Cancers, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 400015, China
| | - Bukhari Ihtisham
- Henan Key Laboratory of Helicobacter pylori, Microbiota and Gastrointestinal Cancers, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 400015, China
| | - Xin-Feng Liang
- Henan Key Laboratory of Helicobacter pylori, Microbiota and Gastrointestinal Cancers, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 400015, China
| | - Ji-Xuan Xu
- Henan Key Laboratory of Helicobacter pylori, Microbiota and Gastrointestinal Cancers, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 400015, China
| | - Yang Mi
- Henan Key Laboratory of Helicobacter pylori, Microbiota and Gastrointestinal Cancers, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 400015, China
| | - Peng-Yuan Zheng
- Henan Key Laboratory of Helicobacter pylori, Microbiota and Gastrointestinal Cancers, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 400015, China.
| |
Collapse
|
27
|
Tian Y, Zhao Q, Wu H, Guo J, Wu H. VWA2 protein molecular mechanism predicts colorectal cancer: Promoting cell invasion and migration by inhibiting NK cell activation. Int J Biol Macromol 2024; 279:135394. [PMID: 39245093 DOI: 10.1016/j.ijbiomac.2024.135394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/30/2024] [Accepted: 09/05/2024] [Indexed: 09/10/2024]
Abstract
The onset and progression of colorectal cancer is intricately linked to a multitude of factors. Among these, immune cells present within the tumor microenvironment play a pivotal role, particularly natural killer (NK) cells, which are essential for mediating anti-tumor immunity. This study aims to elucidate the mechanism by which the VWA2 protein facilitates the invasion and migration of colorectal cancer cells through the inhibition of NK cell activation. Understanding this molecular mechanism is crucial for deciphering the underlying processes involved in colorectal cancer. To achieve the study's objectives, various methodologies were employed, including cell culture techniques, transgenic technology, and assessments of NK cell functionality. The "limma" bioinformatics tool was utilised to identify differentially expressed genes (DEGs) between samples of colon cancer or polyps and normal tissue through transcriptome sequencing. Subsequent Wien analysis was conducted to pinpoint overlapping genes of interest. The impact of VWA2 on both the invasion and migration of colorectal cancer cell lines was assessed through experiments designed for the overexpression and knockout of VWA2.In addition, flow cytometry was employed to evaluate the activation status of NK cells, enabling an analysis of how VWA2 modulates relevant signaling pathways. The findings revealed that overexpression of VWA2 led to a marked inhibition of NK cell activation, which corresponded with reduced cytotoxic activity against tumor cells. Further examination indicated that VWA2 significantly amplified the migration and invasion capabilities of colorectal cancer cells by upregulating immunosuppressive factors while simultaneously downregulating pro-inflammatory factors. Conversely, the reduction of VWA2 expression was shown to markedly enhance NK cell functionality and decrease the invasive potential of colorectal cancer cells. Thus, the evidence suggests that the VWA2 protein actively promotes the migration and invasion of colorectal cancer cells primarily by suppressing NK cell activation, highlighting its potential role as a significant contributor to tumor progression in colorectal cancer.
Collapse
Affiliation(s)
- Yang Tian
- Department of Endoscope, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
| | - Qi Zhao
- Department of Endoscope, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
| | - Haowa Wu
- Department of Gastroenterology, Aviation General Hospital, Chaoyang District, Beijing 100012, China
| | - Jiaqi Guo
- Department of Gastroenterology, The Affiliated Hospital of Northwest University Xi'an No. 3 Hospital, Xi'an 710000, Shaanxi Province, China
| | - Huaxing Wu
- Department of Endoscope, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China.
| |
Collapse
|
28
|
Yu C, Li H, Zhang C, Tang Y, Huang Y, Lu H, Jin K, Zhou J, Yang J. Solute carrier family 4 member 4 (SLC4A4) is associated with cell proliferation, migration and immune cell infiltration in colon cancer. Discov Oncol 2024; 15:597. [PMID: 39467887 PMCID: PMC11519258 DOI: 10.1007/s12672-024-01488-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 10/22/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Solute Carrier Family 4 Member 4 (SLC4A4) is a membrane protein-coding gene for a Na+/HCO3- cotransporter and plays a crucial role in regulating pH, bicarbonate secretion and homeostasis. However, the prognostic and immunological role of SLC4A4 in colon cancer remains unknown. METHOD In this study, expression profiles of SLC4A4 were retrieved from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases, to which a variety of bioinformatic analyses were performed. Sangerbox, Xiantao, ESTIMATE and TIMER online tools were used to delve into the relationship between SLC4A4 expression and immune cell infiltration. The role of SLC4A4 in the proliferation and migration of colon cancer cells was verified by CCK8, EdU and wound healing assays. The related molecules and pathways that SLC4A4 may affect were validated by bioinformatic prediction and western blotting analysis. RESULTS The expression levels of SLC4A4 were significantly lower in colon cancer tissues than in normal tissues and its low expression was positively correlated with poor prognosis. TIMER and ESTIMATE showed that SLC4A4 broadly influenced immune cell infiltration. Experiments in vitro demonstrated that SLC4A4 inhibited partial epithelial-mesenchymal transition (EMT) phenotypes. CONCLUSIONS To conclude, our study revealed that SLC4A4 is lowly expressed in colon cancer tissues, and SLC4A4 may inhibit the progression of colon cancer via regulating partial EMT phenotypes and immune cell infiltration, which may provide new perspectives for the development of more precise and personalized immune anti-tumor therapies.
Collapse
Affiliation(s)
- Chengqing Yu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu, China
| | - Haoran Li
- Department of General Surgery, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu, China
| | - Chen Zhang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu, China
| | - Yuchen Tang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu, China
| | - Yujie Huang
- Department of Emergency Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Haodong Lu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu, China
| | - Kanghui Jin
- Department of General Surgery, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu, China
| | - Jian Zhou
- Department of General Surgery, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu, China.
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, China.
| | - Jian Yang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
29
|
Fu Y, Zhang X, Qiao Q. PLXDC1 serves as a potential prognostic marker and involves in malignant progression and macrophage polarization in colon cancer. J Biochem Mol Toxicol 2024; 38:e23832. [PMID: 39267413 DOI: 10.1002/jbt.23832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/22/2024] [Accepted: 08/22/2024] [Indexed: 09/17/2024]
Abstract
The malignant behavior and immune escape ability of cancer cells lead to therapeutic failure and poor prognosis for patients with various cancers, including colon cancer. Plexin domain containing 1 (PLXDC1) was initially identified to exert key roles in tumor by regulating angiogenesis and has recently proved to be involved in cell proliferation and migration of glioblastoma and gastric cancer cells. However, its roles in colon cancer remain unclear. In this study, the online bioinformatics databases confirmed high expression of PLXDC1 in colon cancer specimens, which was associated with cancer stages and nodal metastasis. Similarly, the increased expression of PLXDC1 was also validated in our collected samples and colon cancer cells. Moreover, patients with high expression of PLXDC1 had shorter survival, indicating that PLXDC1 might be a potential prognostic predictor for colon cancer patients. Notably, targeting PLXDC1 inhibited cancer cell viability and invasion, and enhanced cell apoptosis. Intriguingly, Tumor Immune Estimation Resource database confirmed that PLXDC1 expression was related to various tumor-infiltrating immune cells in colon adenocarcinoma including macrophages, and its expression was also correlated with M2-like macrophage markers. In vitro, colon cancer cells with PLXDC1 downregulation had a reduced ability to recruit and polarize macrophage towards M2 phenotype by decreasing the percentage of CD206+ cells and M2-like markers (CD206, CD163, arginase1, and interleukin 10 [IL-10]). Moreover, PLXDC1 knockdown attenuated M2 macrophage-mediated promotion in cancer cell viability and invasion. Mechanically, inhibition of PLXDC1 suppressed activation of the IL-6/Signal transducer and activator of transcription 3 (STAT3) signaling. Reactivating the above pathway by transfection with IL-6 plasmids reversed the suppressive effects of PLXDC1 knockdown on cancer cell malignant behaviors, macrophage recruitment and M2-like polarization. Thus, PLXDC1 downregulation may inhibit the malignancy of colon cancer cells and their ability to recruit and polarize macrophages towards M2 phenotype by blocking the IL-6/STAT3 pathway. Together, targeting PLXDC1 may attenuate the progression of colon cancer by direct roles in cancer cells and indirect roles in macrophage polarization, representing a promising therapeutic target for colon cancer patients.
Collapse
Affiliation(s)
- Yu Fu
- Department of Oncology and Hematology, People's Hospital of Leshan, Leshan, Sichuan, China
| | - Xuan Zhang
- Department of Oncology and Hematology, People's Hospital of Leshan, Leshan, Sichuan, China
| | - Qing Qiao
- Department of Oncology and Hematology, People's Hospital of Leshan, Leshan, Sichuan, China
| |
Collapse
|
30
|
Sueyoshi S, Vitor Silva J, Guizze F, Giarolla J. Dendrimers as drug delivery systems for oncotherapy: Current status of promising applications. Int J Pharm 2024; 663:124573. [PMID: 39134292 DOI: 10.1016/j.ijpharm.2024.124573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 08/08/2024] [Accepted: 08/08/2024] [Indexed: 08/20/2024]
Abstract
Cancer affects millions of people worldwide, causing death and serious health problems. Despite significant investment in the development of new anticancer compounds, there are still several limitations that can still be found. Many compounds exhibit high levels of toxicity and low bioavailability. Therefore, it is urgent to design safer, more effective, and particularly more selective compounds for oncological treatment. Dendrimers are polymeric structures that have been shown to be potential drug nanocarriers to overcome physicochemical, pharmacokinetic, and indirect pharmacodynamic issues. Due to their versatility, they can be used in the design of nanovaccines, lipophilic complexes, amphiphilic complexes, smart nanocomplexes, and others. This work targets the use of dendrimers in oncological treatment and their importance and effectiveness as drug delivery systems for the development of new therapies. For this review, only publications from the last two years are considered in this review.
Collapse
Affiliation(s)
- Sophia Sueyoshi
- Department of Pharmacy, School of Pharmaceutical Sciences, University of São Paulo, Av Prof Lineu Prestes, 580, Bl. 13, CEP 05508-900 São Paulo, SP, Brazil
| | - João Vitor Silva
- Department of Pharmacy, School of Pharmaceutical Sciences, University of São Paulo, Av Prof Lineu Prestes, 580, Bl. 13, CEP 05508-900 São Paulo, SP, Brazil
| | - Felipe Guizze
- Department of Pharmacy, School of Pharmaceutical Sciences, University of São Paulo, Av Prof Lineu Prestes, 580, Bl. 13, CEP 05508-900 São Paulo, SP, Brazil
| | - Jeanine Giarolla
- Department of Pharmacy, School of Pharmaceutical Sciences, University of São Paulo, Av Prof Lineu Prestes, 580, Bl. 13, CEP 05508-900 São Paulo, SP, Brazil.
| |
Collapse
|
31
|
Tang J, Wang W, Tang G. ENO2 in progression and treatment of colon adenocarcinoma: integrative bioinformatics analysis on non-apoptotic cell death. Discov Oncol 2024; 15:478. [PMID: 39331182 PMCID: PMC11436658 DOI: 10.1007/s12672-024-01208-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 07/30/2024] [Indexed: 09/28/2024] Open
Abstract
Colon adenocarcinoma (COAD) is one of the most common types of cancer. The interconnection between non-apoptotic cell death and COAD has not been adequately addressed. In our study, an integrative bioinformatics analysis was performed to explore non-apoptotic cell death-related biomarkers in COAD. ENO2 was determined as a potent biomarker for prognosis, drug response, immunity, and immunotherapy prediction. We used EdU and RT-qPCR assays to test our hypothesis and investigate how the ENO2 gene may influence or regulate cancer-related processes. ENO2 was expected to be a potential target in COAD.
Collapse
Affiliation(s)
- Jia Tang
- Department of Gastroenterology, The Seventh People's Hospital of Chongqing, Chongqing, 401320, People's Republic of China
| | - Weiqiang Wang
- Department of Gastroenterology, The Seventh People's Hospital of Chongqing, Chongqing, 401320, People's Republic of China
| | - Guangming Tang
- Department of Gastroenterology, The Seventh People's Hospital of Chongqing, Chongqing, 401320, People's Republic of China.
| |
Collapse
|
32
|
Malik S, Sikander M, Wahid M, Dhasmana A, Sarwat M, Khan S, Cobos E, Yallapu MM, Jaggi M, Chauhan SC. Deciphering cellular and molecular mechanism of MUC13 mucin involved in cancer cell plasticity and drug resistance. Cancer Metastasis Rev 2024; 43:981-999. [PMID: 38498072 DOI: 10.1007/s10555-024-10177-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 02/26/2024] [Indexed: 03/19/2024]
Abstract
There has been a surge of interest in recent years in understanding the intricate mechanisms underlying cancer progression and treatment resistance. One molecule that has recently emerged in these mechanisms is MUC13 mucin, a transmembrane glycoprotein. Researchers have begun to unravel the molecular complexity of MUC13 and its impact on cancer biology. Studies have shown that MUC13 overexpression can disrupt normal cellular polarity, leading to the acquisition of malignant traits. Furthermore, MUC13 has been associated with increased cancer plasticity, allowing cells to undergo epithelial-mesenchymal transition (EMT) and metastasize. Notably, MUC13 has also been implicated in the development of chemoresistance, rendering cancer cells less responsive to traditional treatment options. Understanding the precise role of MUC13 in cellular plasticity, and chemoresistance could pave the way for the development of targeted therapies to combat cancer progression and enhance treatment efficacy.
Collapse
Affiliation(s)
- Shabnam Malik
- Department of Immunology and Microbiology, School of Medicine, Biomedical Research Building, University of Texas Rio Grande Valley, 5300 North L Street, McAllen, TX, 78504, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Mohammed Sikander
- Department of Immunology and Microbiology, School of Medicine, Biomedical Research Building, University of Texas Rio Grande Valley, 5300 North L Street, McAllen, TX, 78504, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Mohd Wahid
- Unit of Research and Scientific Studies, College of Nursing and Allied Health Sciences, University of Jazan, Jizan, Saudi Arabia
| | - Anupam Dhasmana
- Department of Immunology and Microbiology, School of Medicine, Biomedical Research Building, University of Texas Rio Grande Valley, 5300 North L Street, McAllen, TX, 78504, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Maryam Sarwat
- Amity Institute of Pharmacy, Amity University, Uttar Pradesh, Noida, India
| | - Sheema Khan
- Department of Immunology and Microbiology, School of Medicine, Biomedical Research Building, University of Texas Rio Grande Valley, 5300 North L Street, McAllen, TX, 78504, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Everardo Cobos
- Department of Medicine, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Murali M Yallapu
- Department of Immunology and Microbiology, School of Medicine, Biomedical Research Building, University of Texas Rio Grande Valley, 5300 North L Street, McAllen, TX, 78504, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Meena Jaggi
- Department of Immunology and Microbiology, School of Medicine, Biomedical Research Building, University of Texas Rio Grande Valley, 5300 North L Street, McAllen, TX, 78504, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Subhash C Chauhan
- Department of Immunology and Microbiology, School of Medicine, Biomedical Research Building, University of Texas Rio Grande Valley, 5300 North L Street, McAllen, TX, 78504, USA.
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA.
| |
Collapse
|
33
|
Bai H, Xian N, Zhao F, Zhou Y, Qin S. The dual role of SUSD2 in cancer development. Eur J Pharmacol 2024; 977:176754. [PMID: 38897441 DOI: 10.1016/j.ejphar.2024.176754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 06/04/2024] [Accepted: 06/16/2024] [Indexed: 06/21/2024]
Abstract
Sushi domain-containing protein 2 (SUSD2, also known as the complement control protein domain) is a representative and vital protein in the SUSD protein family involved in many physiological and pathological processes beyond complement regulation. Cancer is one of the leading causes of death worldwide. The complex role of SUSD2 in tumorigenesis and cancer progression has raised increasing concerns. Studies suggest that SUSD2 has different regulatory tendencies among different tumors and exerts its biological effects in a cancer type-specific manner; for instance, it has oncogenic effects on breast cancer, gastric cancer, and glioma and has tumor-suppression effects on lung cancer, bladder cancer, and colon cancer. Moreover, SUSD2 can be regulated by noncoding RNAs, its promoter methylation and other molecules, such as Galectin-1 (Gal-1), tropomyosin alpha-4 chain (TPM4), and p63. The therapeutic implications of targeting SUSD2 have already been preliminarily revealed in some malignancies, including melanoma, colon cancer, and breast cancer. This article reviews the role and regulatory mechanisms of SUSD2 in cancer development, as well as its structure and distribution. We hope that this review will advance the understanding of SUSD2 as a diagnostic and/or prognostic biomarker and provide new avenues for the development of novel cancer therapies.
Collapse
Affiliation(s)
- Han Bai
- The MED-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Building 21, Western China Science and Technology Innovation Harbor, Xi'an, 710000, China
| | - Ningyi Xian
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Fengyu Zhao
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yikun Zhou
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Sida Qin
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
34
|
Jovandaric MZ. Importance of diet and intestinal microbiota in the prevention of colorectal cancer - colonoscopy early screening diagnosis. World J Gastrointest Oncol 2024; 16:3428-3435. [PMID: 39171174 PMCID: PMC11334020 DOI: 10.4251/wjgo.v16.i8.3428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 05/01/2024] [Accepted: 05/24/2024] [Indexed: 08/07/2024] Open
Abstract
Colorectal cancer is a term used to describe colon and rectal cancer, which is the third most common type of cancer. A MEDLINE and PubMed search resulted in the inclusion of manuscripts written in the last 10 years, using keywords relevant to the topic of the manuscript. By analyzing the aim of the searched studies and manuscripts, adequate articles were included that described the stated problem. The frequency of colorectal cancer varies with climate, nutrition, and many other factors, primarily endogenous, hereditary, intestinal microbiome, as well as external factors, such as exposure of the individual to stress, and bad eating habits. Colon cancer and rectal cancer or colorectal cancer in general in the early stages of the disease, may not show symptoms or are barely noticeable. Colorectal cancer symptoms will most often not develop until the disease has progressed to stage 2 or beyond. Regular screening tests for colon or rectal cancer, especially colonoscopy, are recommended as part of a regular checkup for people aged 50 years or younger who are at high risk due to a family history of the disease or other cancers. Diet and colonoscopy as an early screening method play an important role in the prevention of colorectal cancer.
Collapse
Affiliation(s)
- Miljana Z Jovandaric
- Department of Neonatology, Clinic for Gynecology and Obstetrics, University Clinical Center of Serbia, Belgrade 11070, Serbia
| |
Collapse
|
35
|
Awad H, Elshebli S, Hasan K, Eid Y, Obeidat F, Alzyoud M, Alakhras B, AlShammas F. Comparing Clinicopathological and Immunohistochemical Features of Colorectal Carcinoma between Young and Old Age Groups. Diagnostics (Basel) 2024; 14:1743. [PMID: 39202231 PMCID: PMC11353569 DOI: 10.3390/diagnostics14161743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 09/03/2024] Open
Abstract
The incidence of colorectal carcinoma (CRC) is increasing among individuals younger than 50, and some studies suggest the presence of differences in CRC among old and young individuals regarding clinical and histopathological features. The aim of this study was to compare clinicopathological features, mismatch repair protein status, and expression of certain immunohistochemical stains between young and old groups. The study included 180 cases and found significant histological and immunohistochemical differences between the two groups. CRC in the young tends to be more right-sided and has a higher percentage of dMMR proteins, but less expression of p53 mutations. These features are commoner in Lynch syndrome, and more investigations to study the relationship between young-onset CRC and hereditary syndromes are needed. Young-onset CRC also tends to show higher expression of tumor cell PD-L1, which is an expected finding, as dMMR cases are more likely to be immunogenic. Two other significant differences are the higher percentage of mucinous carcinoma and the higher tumor grade in young-onset CRC. These two features suggest a more advanced disease with possibly worse outcomes; however, there is no difference in disease stage between the two age groups.
Collapse
Affiliation(s)
- Heyam Awad
- Department of Histopathology, Microbiology and Forensic Medicine, University of Jordan, Amman 11942, Jordan; (S.E.); (K.H.); (Y.E.); (F.O.)
- Department of Lab Medicine, Jordan University Hospital, Amman 11942, Jordan; (M.A.); (B.A.); (F.A.)
| | - Sanad Elshebli
- Department of Histopathology, Microbiology and Forensic Medicine, University of Jordan, Amman 11942, Jordan; (S.E.); (K.H.); (Y.E.); (F.O.)
| | - Khaled Hasan
- Department of Histopathology, Microbiology and Forensic Medicine, University of Jordan, Amman 11942, Jordan; (S.E.); (K.H.); (Y.E.); (F.O.)
| | - Yousef Eid
- Department of Histopathology, Microbiology and Forensic Medicine, University of Jordan, Amman 11942, Jordan; (S.E.); (K.H.); (Y.E.); (F.O.)
| | - Fatima Obeidat
- Department of Histopathology, Microbiology and Forensic Medicine, University of Jordan, Amman 11942, Jordan; (S.E.); (K.H.); (Y.E.); (F.O.)
- Department of Lab Medicine, Jordan University Hospital, Amman 11942, Jordan; (M.A.); (B.A.); (F.A.)
| | - Mohammad Alzyoud
- Department of Lab Medicine, Jordan University Hospital, Amman 11942, Jordan; (M.A.); (B.A.); (F.A.)
| | - Basheer Alakhras
- Department of Lab Medicine, Jordan University Hospital, Amman 11942, Jordan; (M.A.); (B.A.); (F.A.)
| | - Faris AlShammas
- Department of Lab Medicine, Jordan University Hospital, Amman 11942, Jordan; (M.A.); (B.A.); (F.A.)
| |
Collapse
|
36
|
Zhang J, Huang X, Zhang T, Gu C, Zuo W, Fu L, Dong Y, Liu H. Antitumorigenic potential of Lactobacillus-derived extracellular vesicles: p53 succinylation and glycolytic reprogramming in intestinal epithelial cells via SIRT5 modulation. Cell Biol Toxicol 2024; 40:66. [PMID: 39110260 PMCID: PMC11306434 DOI: 10.1007/s10565-024-09897-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 06/21/2024] [Indexed: 08/10/2024]
Abstract
OBJECTIVE Colorectal cancer progression involves complex cellular mechanisms. This study examines the effects of Lactobacillus plantarum-derived extracellular vesicles (LEVs) on the SIRT5/p53 axis, focusing on glycolytic metabolic reprogramming and abnormal proliferation in intestinal epithelial cells. METHODS LEVs were isolated from Lactobacillus plantarum and incubated with Caco-2 cells. Differential gene expression was analyzed through RNA sequencing and compared with TCGA-COAD data. Key target genes and pathways were identified using PPI network and pathway enrichment analysis. Various assays, including RT-qPCR, EdU staining, colony formation, flow cytometry, and Western blotting, were used to assess gene expression, cell proliferation, and metabolic changes. Co-immunoprecipitation confirmed the interaction between SIRT5 and p53, and animal models were employed to validate in vivo effects. RESULTS Bioinformatics analysis indicated the SIRT5/p53 axis as a critical pathway in LEVs' modulation of colorectal cancer. LEVs were found to inhibit colorectal cancer cell proliferation and glycolytic metabolism by downregulating SIRT5, influencing p53 desuccinylation. In vivo, LEVs regulated this axis, reducing tumor formation in mice. Clinical sample analysis showed that SIRT5 and p53 succinylation levels correlated with patient prognosis. CONCLUSION Lactobacillus-derived extracellular vesicles play a pivotal role in suppressing colonic tumor formation by modulating the SIRT5/p53 axis. This results in decreased glycolytic metabolic reprogramming and reduced proliferation in intestinal epithelial cells.
Collapse
Affiliation(s)
- Jingbo Zhang
- Department of Spleen and Stomach Disease, Yubei District Hospital of Traditional Chinese Medicine, Chongqing, 401120, China
| | - Xiumei Huang
- Department of Digestion, Rongchang District People's Hospital of Chongqing, No.3, North Guangchang Road, Changyuan Street, Rongchang District, Chongqing, 402460, China
| | - Tingting Zhang
- Department of Digestion, Rongchang District People's Hospital of Chongqing, No.3, North Guangchang Road, Changyuan Street, Rongchang District, Chongqing, 402460, China
| | - Chongqi Gu
- Department of Pediatrics, Rongchang District People's Hospital, Chongqing, 402460, China
| | - Wei Zuo
- Department of Herbal Medicine, School of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, 400016, China
- Department of Pharmacology, Academician Workstation, Changsha Medical University, Changsha, 410219, China
| | - Lijuan Fu
- Department of Herbal Medicine, School of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, 400016, China
- Department of Pharmacology, Academician Workstation, Changsha Medical University, Changsha, 410219, China
| | - Yiping Dong
- Department of Digital Medicine, Department of Bioengineering and Imaging, Army Medical University, Chongqing, 400038, China
| | - Hao Liu
- Department of Pediatrics, Rongchang District People's Hospital, Chongqing, 402460, China.
| |
Collapse
|
37
|
Elias MG, Aputen AD, Fatima S, Mann TJ, Karan S, Mikhael M, de Souza P, Gordon CP, Scott KF, Aldrich-Wright JR. Chemotherapeutic Potential of Chlorambucil-Platinum(IV) Prodrugs against Cisplatin-Resistant Colorectal Cancer Cells. Int J Mol Sci 2024; 25:8252. [PMID: 39125821 PMCID: PMC11312340 DOI: 10.3390/ijms25158252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
Chlorambucil-platinum(IV) prodrugs exhibit multi-mechanistic chemotherapeutic activity with promising anticancer potential. The platinum(II) precursors of the prodrugs have been previously found to induce changes in the microtubule cytoskeleton, specifically actin and tubulin of HT29 colon cells, while chlorambucil alkylates the DNA. These prodrugs demonstrate significant anticancer activity in 2D cell and 3D spheroid viability assays. A notable production of reactive oxygen species has been observed in HT29 cells 72 h post treatment with prodrugs of this type, while the mitochondrial membrane potential was substantially reduced. The cellular uptake of the chlorambucil-platinum(IV) prodrugs, assessed by ICP-MS, confirmed that active transport was the primary uptake mechanism, with platinum localisation identified primarily in the cytoskeletal fraction. Apoptosis and necrosis were observed at 72 h of treatment as demonstrated by Annexin V-FITC/PI assay using flow cytometry. Immunofluorescence measured via confocal microscopy showed significant changes in actin and tubulin intensity and in architecture. Western blot analysis of intrinsic and extrinsic pathway apoptotic markers, microtubule cytoskeleton markers, cell proliferation markers, as well as autophagy markers were studied post 72 h of treatment. The proteomic profile was also studied with a total of 1859 HT29 proteins quantified by mass spectroscopy, with several dysregulated proteins. Network analysis revealed dysregulation in transcription, MAPK markers, microtubule-associated proteins and mitochondrial transport dysfunction. This study confirms that chlorambucil-platinum(IV) prodrugs are candidates with promising anticancer potential that act as multi-mechanistic chemotherapeutics.
Collapse
Affiliation(s)
- Maria George Elias
- School of Science, Western Sydney University, Sydney, NSW 2751, Australia; (M.G.E.); (A.D.A.); (S.K.); (C.P.G.)
- Medical Oncology, Ingham Institute for Applied Medical Research, Liverpool Hospital, Liverpool, NSW 2170, Australia; (S.F.); (T.J.M.); (K.F.S.)
| | - Angelico D. Aputen
- School of Science, Western Sydney University, Sydney, NSW 2751, Australia; (M.G.E.); (A.D.A.); (S.K.); (C.P.G.)
| | - Shadma Fatima
- Medical Oncology, Ingham Institute for Applied Medical Research, Liverpool Hospital, Liverpool, NSW 2170, Australia; (S.F.); (T.J.M.); (K.F.S.)
- School of Medicine, Western Sydney University, Sydney, NSW 2751, Australia
| | - Timothy J. Mann
- Medical Oncology, Ingham Institute for Applied Medical Research, Liverpool Hospital, Liverpool, NSW 2170, Australia; (S.F.); (T.J.M.); (K.F.S.)
- School of Medicine, Western Sydney University, Sydney, NSW 2751, Australia
| | - Shawan Karan
- School of Science, Western Sydney University, Sydney, NSW 2751, Australia; (M.G.E.); (A.D.A.); (S.K.); (C.P.G.)
| | - Meena Mikhael
- Mass Spectrometry Facility, Western Sydney University, Sydney, NSW 2751, Australia;
| | - Paul de Souza
- Nepean Clinical School, Faculty of Medicine and Health, University of Sydney, Kingswood, NSW 2747, Australia;
| | - Christopher P. Gordon
- School of Science, Western Sydney University, Sydney, NSW 2751, Australia; (M.G.E.); (A.D.A.); (S.K.); (C.P.G.)
| | - Kieran F. Scott
- Medical Oncology, Ingham Institute for Applied Medical Research, Liverpool Hospital, Liverpool, NSW 2170, Australia; (S.F.); (T.J.M.); (K.F.S.)
- School of Medicine, Western Sydney University, Sydney, NSW 2751, Australia
| | - Janice R. Aldrich-Wright
- School of Science, Western Sydney University, Sydney, NSW 2751, Australia; (M.G.E.); (A.D.A.); (S.K.); (C.P.G.)
- Medical Oncology, Ingham Institute for Applied Medical Research, Liverpool Hospital, Liverpool, NSW 2170, Australia; (S.F.); (T.J.M.); (K.F.S.)
| |
Collapse
|
38
|
Tâlvan CD, Tâlvan ET, Mohor CI, Budișan L, Grecu V, Mihalache M, Zănoagă O, Chira S, Berindan-Neagoe I, Cristea V, Mohor CI. Exploring miRNA Profiles in Colon Cancer: A Focus on miR101-3p, miR106a-5p, and miR326. Cancers (Basel) 2024; 16:2285. [PMID: 38927989 PMCID: PMC11201595 DOI: 10.3390/cancers16122285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/08/2024] [Accepted: 06/19/2024] [Indexed: 06/28/2024] Open
Abstract
Early diagnosis and prognosis of cancer progression through biomarker profiling are crucial in managing colon cancer patients. Our research aimed to investigate the expression of miR-101-3p, miR-106a-5p, and miR-326 in tumor and adjacent healthy tissues of colon cancer patients and determine their potential diagnostic utility. This study included 40 patients divided into four groups according to the TNM staging classification. MiRNA expression was analyzed using qRT-PCR. The results showed that miR-101-3p, miR-106a-5p, and miR-326 are overexpressed in adjacent healthy tissues but decrease in advanced cancer stages. MiR-106a-5p and miR-326 are strongly correlated with colon cancer severity. These findings suggest that miRNA profiling could be useful for early diagnosis and prognosis in colon cancer management.
Collapse
Affiliation(s)
- Constantin-Dan Tâlvan
- Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (C.-D.T.); (C.I.M.); (M.M.); (C.I.M.)
| | - Elena-Teodora Tâlvan
- Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (C.-D.T.); (C.I.M.); (M.M.); (C.I.M.)
| | - Călin Ilie Mohor
- Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (C.-D.T.); (C.I.M.); (M.M.); (C.I.M.)
| | - Liviuța Budișan
- Research Center for Functional Genomic, Biomedicine and Translational Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy Cluj-Napoca, 400012 Cluj-Napoca, Romania; (L.B.); (O.Z.); (S.C.); (I.B.-N.); (V.C.)
| | - Valentin Grecu
- Faculty of Engineering, “Lucian Blaga” University of Sibiu, 550025 Sibiu, Romania;
| | - Manuela Mihalache
- Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (C.-D.T.); (C.I.M.); (M.M.); (C.I.M.)
| | - Oana Zănoagă
- Research Center for Functional Genomic, Biomedicine and Translational Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy Cluj-Napoca, 400012 Cluj-Napoca, Romania; (L.B.); (O.Z.); (S.C.); (I.B.-N.); (V.C.)
| | - Sergiu Chira
- Research Center for Functional Genomic, Biomedicine and Translational Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy Cluj-Napoca, 400012 Cluj-Napoca, Romania; (L.B.); (O.Z.); (S.C.); (I.B.-N.); (V.C.)
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomic, Biomedicine and Translational Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy Cluj-Napoca, 400012 Cluj-Napoca, Romania; (L.B.); (O.Z.); (S.C.); (I.B.-N.); (V.C.)
| | - Victor Cristea
- Research Center for Functional Genomic, Biomedicine and Translational Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy Cluj-Napoca, 400012 Cluj-Napoca, Romania; (L.B.); (O.Z.); (S.C.); (I.B.-N.); (V.C.)
| | - Cosmin Ioan Mohor
- Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (C.-D.T.); (C.I.M.); (M.M.); (C.I.M.)
| |
Collapse
|
39
|
Li J, Jiang Z, He J, Yang K, Chen J, Deng Q, Li X, Wu F, Xu S, Jiang Z. Effect of CHRDL1 on angiogenesis and metastasis of colorectal cancer cells via TGF-β/VEGF pathway. Mol Carcinog 2024; 63:1092-1105. [PMID: 38415870 DOI: 10.1002/mc.23711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 01/17/2024] [Accepted: 02/19/2024] [Indexed: 02/29/2024]
Abstract
Colorectal cancer (CRC) is a common digestive tract tumor with the third incidence and death in the world. There is still an urgent need for effective therapeutic targets and prognostic markers for CRC. Herein, we report a novel potential target and marker, Chordin like-1 (CHRDL1). The function of CHRDL1 has been reported in gastric cancer, breast cancer, and oral squamous cell carcinoma. However, the biological effect of CHRDL1 in CRC remains unrevealed. Transwell and tube formation experiments were used to determine the biological function of CHRDL1. Western blot and rescue experiments were used to determine the specific mechanisms of CHRDL1. Results showed CHRDL1 is significantly downregulated in CRC cell lines and tissues. In vitro, experiments confirmed that CHRDL1 can inhibit cell growth, migration, invasion, angiogenesis and reverse epithelial-mesenchymal transformation. In vivo, experiments proved that it can inhibit tumor growth and metastasis. Mechanistically, we newly find that CHRDL1 exerts biological functions through the transforming growth factor-beta (TGF-β)/vascular endothelial growth factor signaling axis in vitro and in vivo. Therefore, we concluded that CHRDL1 reduces the growth, migration, and angiogenesis of CRC cells by downregulating TGF-β signaling. Our new findings on CHRDL1 may provide a basis for clinical antiangiogenesis therapy and the prognosis of CRC.
Collapse
Affiliation(s)
- Junfeng Li
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhongxiang Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jin He
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Kun Yang
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jun Chen
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qianxi Deng
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoqing Li
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fan Wu
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shuman Xu
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zheng Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
40
|
Kaimuangpak K, Rosalina R, Thumanu K, Weerapreeyakul N. Macromolecules with predominant β-pleated sheet proteins in extracellular vesicles released from Raphanus sativus L. var. caudatus Alef microgreens induce DNA damage-mediated apoptosis in HCT116 colon cancer cells. Int J Biol Macromol 2024; 269:132001. [PMID: 38702007 DOI: 10.1016/j.ijbiomac.2024.132001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 03/14/2024] [Accepted: 04/23/2024] [Indexed: 05/06/2024]
Abstract
Plant-derived bioactive macromolecules (i.e., proteins, lipids, and nucleic acids) were prepared as extracellular vesicles (EVs). Plant-derived EVs are gaining pharmaceutical research interest because of their bioactive components and delivery properties. The spherical nanosized EVs derived from Raphanus sativus L. var. caudatus Alef microgreens previously showed antiproliferative activity in HCT116 colon cancer cells from macromolecular compositions (predominantly proteins). To understand the mechanism of action, the biological activity studies, i.e., antiproliferation, cellular biochemical changes, DNA conformational changes, DNA damage, apoptotic nuclear morphological changes, apoptosis induction, and apoptotic pathways, were determined by neutral red uptake assay, synchrotron radiation-based Fourier transform infrared microspectroscopy, circular dichroism spectroscopy, comet assay, 4',6-diamidino-2-phenylindole (DAPI) staining, flow cytometry, and caspase activity assay, respectively. EVs inhibited HCT116 cell growth in concentration- and time-dependent manners, with a half-maximal inhibitory concentration of 675.4 ± 33.8 μg/ml at 48 h and a selectivity index of 1.5 ± 0.076. HCT116 treated with EVs mainly changed the cellular biochemical compositions in the nucleic acids and carbohydrates region. The DNA damage caused no changes in DNA conformation. The apoptotic nuclear morphological changes were associated with the increased apoptotic cell population. The apoptotic cell death was induced by both extrinsic and intrinsic pathways. EVs have potential as antiproliferative bioparticles.
Collapse
Affiliation(s)
- Karnchanok Kaimuangpak
- Graduate School (Research and Development in Pharmaceuticals Program), Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand.
| | - Reny Rosalina
- Graduate School (Biomedical Sciences Program), Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand.
| | - Kanjana Thumanu
- Synchrotron Light Research Institute (Public Organization), Nakhon Ratchasima 30000, Thailand.
| | - Natthida Weerapreeyakul
- Research Institute for Human High Performance and Health Promotion, Khon Kaen University, Khon Kaen 40002, Thailand; Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand.
| |
Collapse
|
41
|
Cui Y, Lv P, Zhang C. NSUN6 mediates 5-methylcytosine modification of METTL3 and promotes colon adenocarcinoma progression. J Biochem Mol Toxicol 2024; 38:e23749. [PMID: 38800929 DOI: 10.1002/jbt.23749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 04/12/2024] [Accepted: 05/16/2024] [Indexed: 05/29/2024]
Abstract
Colon adenocarcinoma (COAD) is a common and fatal malignant tumor of digestive system with complex etiology. 5-Methylcytosine (m5C) modification of RNA by the NSUN gene family (NSUN1-NSUN7) and DNMT2 reshape cell biology and regulate tumor development. However, the expression profile, prognostic significance and function of these m5C modifiers in COAD remain largely unclear. By mining multiple integrated tumor databases, we found that NSUN1, NSUN2, NSUN5, and NSUN6 were overexpressed in COAD tumor samples relative to normal samples. Clinically, high expression of NSUN6 was significantly associated with shorter survival (including both disease-free survival and overall survival) in COAD patients. NSUN6 was further confirmed to be upregulated at both tissue and cellular levels of COAD, suggesting that NSUN6 plays a critical role in disease progression. Through comprehensive gene enrichment analysis and cell-based functional validation, it was revealed that NSUN6 promoted the cell cycle progression and cell proliferation of COAD. Mechanistically, NSUN6 upregulates the expression of oncogenic METTL3 and catalyzes its m5C modification in COAD cells. Overexpression of METTL3 significantly relieved the cell cycle inhibition of COAD caused by NSUN6 deficiency. Furthermore, NSUN6 was negatively associated with the abundance of infiltrating immune cells in COAD tumors, such as activated B cells, natural killer cells, effector memory CD8 T cells, and regulatory T cells. Importantly, pan-cancer analysis further uncovered that NSUN6 was dysregulated and heterogeneous in various tumors. Thus our findings extend the role of m5C transferase in COAD and suggest that NSUN6 is a potential biomarker and target for this malignancy.
Collapse
Affiliation(s)
- Yuanbo Cui
- Department of Translational Medicine Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan, China
| | - Pengju Lv
- Department of Clinical Laboratory, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan, China
| | - Chunyan Zhang
- Department of Clinical Laboratory, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
42
|
Mei Y, Gu L, Chen Y, Zhang P, Cheng Y, Yuan R, Li X, Wang X, Guo P, He D, Zeng J. A Novel Photosensitizer Based 450-nm Blue Laser-Mediated Photodynamic Therapy Induces Apoptosis in Colorectal Cancer - in Vitro and in Vivo Study. FRONT BIOSCI-LANDMRK 2024; 29:199. [PMID: 38812322 DOI: 10.31083/j.fbl2905199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/22/2024] [Accepted: 04/16/2024] [Indexed: 05/31/2024]
Abstract
BACKGROUND Due to its non-invasive and widely applicable features, photodynamic therapy (PDT) has been a prominent treatment approach against cancer in recent years. However, its widespread application in clinical practice is limited by the dark toxicity of photosensitizers and insufficient penetration of light sources. This study assessed the anticancer effects of a novel photosensitizer 5-(4-amino-phenyl)-10,15,20-triphenylporphyrin with diethylene-triaminopentaacetic acid (ATPP-DTPA)-mediated PDT (hereinafter referred to as ATPP-PDT) under the irradiation of a 450-nm blue laser on colorectal cancer (CRC) in vivo and in vitro. METHODS After 450-nm blue laser-mediated ATPP-PDT and the traditional photosensitizer 5-aminolevulinic acid (5-ALA)-PDT treatment, cell viability was detected through Cell Counting Kit-8 (CCK-8) and 5-ethynyl-2'-deoxyuridine (EdU) assays. Reactive oxygen species (ROS) generation was quantified by flow cytometry and fluorescence microscopy. Western blotting and transcriptome RNA sequencing and functional experiments were used to evaluate cell apoptosis and its potential mechanism. Anti-tumor experiment in vivo was performed in nude mice with subcutaneous tumors. RESULTS ATPP-DTPA had a marvelous absorption in the blue spectrum. Compared with 5-ALA, ATPP-DTPA could achieve significant killing effects at a lower dose. Owing to generating an excessive amount of ROS, 450-nm blue laser-mediated PDT based on ATPP-DTPA resulted in evident growth inhibition and apoptosis in CRC cells in vitro. After transcriptome RNA sequencing and functional experiments, p38 MAPK signaling pathway was confirmed to be involved in the regulation of apoptosis induced by 450-nm blue laser-mediated ATPP-PDT. Additionally, animal studies using xenograft model confirmed that ATPP-PDT had excellent anti-tumor effect and reasonable biosafety in vivo. CONCLUSIONS PDT mediated by 450-nm blue laser combined with ATPP-DTPA may be a novel and effective method for the treatment of CRC.
Collapse
Affiliation(s)
- Yibo Mei
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, 710061 Xi'an, Shaanxi, China
| | - Lijiang Gu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, 710061 Xi'an, Shaanxi, China
| | - Yuhang Chen
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, 710061 Xi'an, Shaanxi, China
| | - Pan Zhang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, 710061 Xi'an, Shaanxi, China
| | - Yifan Cheng
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, 710061 Xi'an, Shaanxi, China
| | - Renfei Yuan
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, 710061 Xi'an, Shaanxi, China
| | - Xing Li
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, 710038 Xi'an, Shaanxi, China
| | - Xinyang Wang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, 710061 Xi'an, Shaanxi, China
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, 710061 Xi'an, Shaanxi, China
- Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, 710061 Xi'an, Shaanxi, China
| | - Peng Guo
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, 710061 Xi'an, Shaanxi, China
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, 710061 Xi'an, Shaanxi, China
- Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, 710061 Xi'an, Shaanxi, China
| | - Dalin He
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, 710061 Xi'an, Shaanxi, China
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, 710061 Xi'an, Shaanxi, China
- Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, 710061 Xi'an, Shaanxi, China
| | - Jin Zeng
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, 710061 Xi'an, Shaanxi, China
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, 710061 Xi'an, Shaanxi, China
- Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, 710061 Xi'an, Shaanxi, China
| |
Collapse
|
43
|
Feng X, He S, Chen Y, Zhang L. Deubiquitinase BRCC3 promotes the migration, invasion and EMT progression of colon adenocarcinoma by stabilizing MET expression. Genes Genomics 2024; 46:637-646. [PMID: 38470543 DOI: 10.1007/s13258-024-01508-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 02/24/2024] [Indexed: 03/14/2024]
Abstract
BACKGROUND Breast cancer type 1 susceptibility protein/breast cancer type 2 susceptibility protein-containing complex subunit 3 (BRCC3), a deubiquitinase (DUBs), is overexpressed in various cancers. However, the underlying biological roles of BRCC3 in adenocarcinoma colon (COAD) have yet to be decrypted. OBJECTIVE In this work, we explored the potential biological function of BRCC3 in the natural process of COAD cells. METHODS The expression levels of BRCC3 in COAD tissues and cell lines were investigated via quantitative real time polymerase chain reaction and western blotting analyses. Meanwhile, short hairpin RNAs targeting BRCC3 (sh-BRCC3) or mesenchymal-epithelial transition factor (MET) (sh-MET) were used to investigate the biological function, including proliferation, apoptosis, migration, invasion, and epithelial-mesenchymal transition (EMT) progression in COAD cells. Furthermore, the expression levels of EMT-related biomarkers were detected with western blotting analysis. Furthermore, we also performed Co-IP assay to identify the correlation between BRCC3 and MET. RESULTS BRCC3 expression was increased in COAD tissues and cell lines. ShRNA-mediated downmodulation of BRCC3 in COAD cell lines induced EMT progression. BRCC3 knockdown resulted in decreased migration as well as invasion and increased apoptosis of SW480 and Lovo cells. Besides, MET was regulated by BRCC3 and involved in the migration, invasion, and EMT in SW480 and Lovo cells. Finally, we uncovered that the overexpressed MET reversed the effects of BRCC3 knockdown in COAD cell development. CONCLUSIONS BRCC3 acted as a critical factor in the development of COAD by deubiquitinating and stabilizing MET, which might provide an emerging biomarker for the therapeutic and diagnosis strategy of COAD.
Collapse
Affiliation(s)
- Xiu Feng
- Department of Oncology, Nantong First People's Hospital and Affiliated Hospital 2 of Nantong University, No.666 Shengli Road, Chongchuan District, Nantong, 226000, Jiangsu, China
| | - Shengnan He
- Department of Oncology, Nantong First People's Hospital and Affiliated Hospital 2 of Nantong University, No.666 Shengli Road, Chongchuan District, Nantong, 226000, Jiangsu, China
| | - Ying Chen
- Department of Oncology, Nantong First People's Hospital and Affiliated Hospital 2 of Nantong University, No.666 Shengli Road, Chongchuan District, Nantong, 226000, Jiangsu, China.
| | - Liang Zhang
- Department of Oncology, Nantong First People's Hospital and Affiliated Hospital 2 of Nantong University, No.666 Shengli Road, Chongchuan District, Nantong, 226000, Jiangsu, China.
| |
Collapse
|
44
|
Ahmed R, Zaitone SA, Abdelmaogood AKK, Atef HM, Soliman MFM, Badawy AM, Ali HS, Zaid A, Mokhtar HI, Elabbasy LM, Kandil E, Yosef AM, Mahran RI. Chemotherapeutic potential of betanin/capecitabine combination targeting colon cancer: experimental and bioinformatic studies exploring NFκB and cyclin D1 interplay. Front Pharmacol 2024; 15:1362739. [PMID: 38645563 PMCID: PMC11026609 DOI: 10.3389/fphar.2024.1362739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 03/13/2024] [Indexed: 04/23/2024] Open
Abstract
Introduction: Betanin (C₂₄H₂₆N₂O₁₃) is safe to use as food additives approved by the FDA with anti-inflammatory and anticancer effects in many types of cancer cell lines. The current experiment was designed to test the chemotherapeutic effect of the combination of betanin with the standard chemotherapeutic agent, capecitabine, against chemically induced colon cancer in mice. Methods: Bioinformatic approach was designed to get information about the possible mechanisms through which the drugs may control cancer development. Five groups of mice were assigned as, (i) saline, (ii) colon cancer, (iii) betanin, (iv) capecitabine and (v) betanin/capecitabine. Drugs were given orally for a period of six weeks. Colon tissues were separated and used for biological assays and histopathology. Results: In addition, the mRNA expression of TNF-α (4.58-fold), NFκB (5.33-fold), IL-1β (4.99-fold), cyclin D1 (4.07-fold), and IL-6 (3.55-fold) and protein levels showed several folds increases versus the saline group. Tumor histopathology scores in the colon cancer group (including cryptic distortion and hyperplasia) and immunostaining for NFκB (2.94-fold) were high while periodic-acid Schiff staining demonstrated poor mucin content (33% of the saline group). These pathologic manifestations were reduced remarkably in betanin/capecitabine group. Conclusion: Collectively, our findings demonstrated the usefulness of betanin/capecitabine combination in targeting colon cancer and highlighted that betanin is a promising adjuvant therapy to capecitabine in treating colon cancer patients.
Collapse
Affiliation(s)
- Rehab Ahmed
- Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
- Department of Pharmaceutics, Faculty of Pharmacy, University of Khartoum, Khartoum, Sudan
| | - Sawsan A. Zaitone
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
| | | | - Huda M. Atef
- Department of Histology and Cell Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Mona F. M. Soliman
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Horus University, New Damiettta, Egypt
| | - Alaa M. Badawy
- Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Howaida S. Ali
- Department of Pharmacology, Faculty of Medicine, University of Tabuk, Tabuk, Saudi Arabia
- Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - AbdelNaser Zaid
- Department of Surgery, Faculty of Medicine, Jazan University, Jazan, Saudi Arabia
- Department of General Surgery, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Hatem I. Mokhtar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Sinai University-Kantara Branch, Ismailia, Egypt
| | - Lamiaa M. Elabbasy
- Department of Medical Biochemistry & Molecular Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
- Department of Basic Medical Sciences, College of Medicine, Almaarefa University, Riyadh, Saudi Arabia
| | - Emad Kandil
- Department of Basic Medical Sciences, College of Medicine, Almaarefa University, Riyadh, Saudi Arabia
| | - Asmaa Mokhtar Yosef
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Rama I. Mahran
- Department of Pharmacology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
45
|
Peng Y, Zhang Z, Yang G, Dai Z, Cai X, Liu Z, Yun Q, Xu L. N6-methyladenosine reader protein IGF2BP1 suppresses CD8 + T cells-mediated tumor cytotoxicity and apoptosis in colon cancer. Apoptosis 2024; 29:331-343. [PMID: 37848671 DOI: 10.1007/s10495-023-01893-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2023] [Indexed: 10/19/2023]
Abstract
Tumor immune escape is an important manner for colon cancer to escape effective killing by immune system. Currently, the immune checkpoint PD-1/PD-L1-targeted immunotherapy has emerged as a promising therapeutic strategy in colon cancer. Here, present work aims to investigate the biological function of N6-methyladenosine (m6A) reader insulin-like growth factor 2 mRNA binding protein 1 (IGF2BP1) in regulating colon cancer's immune escape and CD8 + T cells-mediated tumor cytotoxicity and apoptosis. Results illustrated that IGF2BP1 was closely correlated to the colon cancer patients' poor clinical outcome. Functionally, upregulation of IGF2BP1 suppressed the CD8+ T-cells mediated antitumor immunity through reducing their tumor cytotoxicity. Mechanistically, MeRIP-Seq revealed that programmed death ligand 1 (PD-L1) mRNA had a remarkable m6A modified site on 3'-UTR genomic. Moreover, PD-L1 acted as the target of IGF2BP1, which enhanced the stability of PD-L1 mRNA. Overall, these results indicated that IGF2BP1 targeted PD-L1 to accelerate the immune escape in colon cancer by reducing CD8 + T cells-mediated tumor cytotoxicity in m6A-dependent manner. The findings demonstrate the potential of m6A-targeted immune checkpoint blockade in colon cancer, providing a novel insight for colon cancer immune escape and antitumor immunity in further precise treatment.
Collapse
Affiliation(s)
- Yao Peng
- Department of Gastroenterology and Hepatology, Shenzhen University General Hospital, Shenzhen, 518055, China
- Shenzhen University Clinical Medical Academy, Shenzhen, 518055, China
- Shenzhen University International Cancer Center, Shenzhen, 518055, China
| | - Zhili Zhang
- Department of Gastroenterology and Hepatology, Shenzhen University General Hospital, Shenzhen, 518055, China
- Shenzhen University Clinical Medical Academy, Shenzhen, 518055, China
- Shenzhen University International Cancer Center, Shenzhen, 518055, China
| | - Gongli Yang
- Department of Gastroenterology and Hepatology, Shenzhen University General Hospital, Shenzhen, 518055, China
| | - Zhongming Dai
- Department of Gastroenterology and Hepatology, Shenzhen University General Hospital, Shenzhen, 518055, China
| | - Xunchao Cai
- Department of Gastroenterology and Hepatology, Shenzhen University General Hospital, Shenzhen, 518055, China
- Shenzhen University Clinical Medical Academy, Shenzhen, 518055, China
| | - Zhenyu Liu
- Department of Gastroenterology and Hepatology, Shenzhen University General Hospital, Shenzhen, 518055, China
| | - Qian Yun
- Department of Gastroenterology and Hepatology, Shenzhen University General Hospital, Shenzhen, 518055, China
| | - Long Xu
- Department of Gastroenterology and Hepatology, Shenzhen University General Hospital, Shenzhen, 518055, China.
- Shenzhen University Clinical Medical Academy, Shenzhen, 518055, China.
- Shenzhen University International Cancer Center, Shenzhen, 518055, China.
| |
Collapse
|
46
|
McDonald HG, Kerekes DM, Kim J, Khan SA. Precision Oncology in Gastrointestinal and Colorectal Cancer Surgery. Surg Oncol Clin N Am 2024; 33:321-341. [PMID: 38401913 DOI: 10.1016/j.soc.2023.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2024]
Abstract
Precision medicine is used to treat gastrointestinal malignancies including esophageal, gastric, small bowel, colorectal, and pancreatic cancers. Cutting-edge assays to detect and treat these cancers are active areas of research and will soon become standard of care. Colorectal cancer is a prime example of precision oncology as disease site is no longer the final determinate of treatment. Here, the authors describe how leveraging an understanding of tumor biology translates to individualized patient care using evidence-based practices.
Collapse
Affiliation(s)
- Hannah G McDonald
- Department of General Surgery, Division of Surgical Oncology, The University of Kentucky, 800 Rose Street, Lexington, KY 40508, USA
| | - Daniel M Kerekes
- Department of General Surgery, Division of Surgical Oncology, Yale University, 15 York Street, New Haven, CT 06510, USA
| | - Joseph Kim
- Department of General Surgery, Division of Surgical Oncology, The University of Kentucky, 800 Rose Street, Lexington, KY 40508, USA
| | - Sajid A Khan
- Department of Surgery, Yale University, 15 York Street, New Haven, CT 06510, USA.
| |
Collapse
|
47
|
He L, Li H, Wang Y, Li W, Gao L, Xu B, Hu J, He P, Pu W, Sun G, Wang Z, Han Q, Liu B, Chen H. Complete remission in a pretreated, microsatellite-stable, KRAS-mutated colon cancer patient after treatment with sintilimab and bevacizumab and platinum-based chemotherapy: a case report and literature review. Front Immunol 2024; 15:1354613. [PMID: 38617840 PMCID: PMC11010642 DOI: 10.3389/fimmu.2024.1354613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 03/01/2024] [Indexed: 04/16/2024] Open
Abstract
Metastatic colon cancer remains an incurable disease, and it is difficult for existing treatments to achieve the desired clinical outcome, especially for colon cancer patients who have received first-line treatment. Although immune checkpoint inhibitors (ICIs) have demonstrated durable clinical efficacy in a variety of solid tumors, their response requires an inflammatory tumor microenvironment. However, microsatellite-stable (MSS) colon cancer, which accounts for the majority of colorectal cancers, is a cold tumor that does not respond well to ICIs. Combination regimens open the door to the utility of ICIs in cold tumors. Although combination therapies have shown their advantage even for MSS colon cancer, it remains unclear whether combination therapies show their advantage in patients with pretreated metastatic colon cancer. We report a patient who has achieved complete remission and good tolerance with sintilimab plus bevacizumab and platinum-based chemotherapy after postoperative recurrence. The patient had KRAS mutation and MSS-type colon cancer, and his PD-1+CD8+ and CD3-CD19-CD14+CD16-HLA-DR were both positive. He has achieved a progression-free survival of 43 months and is still being followed up at our center. The above results suggest that this therapeutic regimen is a promising treatment modality for the management of pretreated, MSS-type and KRAS-mutated metastatic colorectal cancer although its application to the general public still needs to be validated in clinical trials.
Collapse
Affiliation(s)
- Lijuan He
- Lanzhou University Second Hospital, Lanzhou, China
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, China
| | - Haiyuan Li
- Lanzhou University Second Hospital, Lanzhou, China
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, China
| | - Yunpeng Wang
- Lanzhou University Second Hospital, Lanzhou, China
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, China
| | - Weidong Li
- Lanzhou University Second Hospital, Lanzhou, China
- Department of Pathology, Lanzhou University Second Hospital, Lanzhou, China
| | - Lei Gao
- Lanzhou University Second Hospital, Lanzhou, China
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, China
| | - Bo Xu
- Lanzhou University Second Hospital, Lanzhou, China
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, China
| | - Jike Hu
- Lanzhou University Second Hospital, Lanzhou, China
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, China
| | - Puyi He
- Lanzhou University Second Hospital, Lanzhou, China
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, China
| | - Weigao Pu
- Lanzhou University Second Hospital, Lanzhou, China
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, China
| | - Guodong Sun
- Lanzhou University Second Hospital, Lanzhou, China
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, China
| | - Zhuanfang Wang
- Lanzhou University Second Hospital, Lanzhou, China
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, China
| | - Qinying Han
- Lanzhou University Second Hospital, Lanzhou, China
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, China
| | - Ben Liu
- Lanzhou University Second Hospital, Lanzhou, China
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, China
| | - Hao Chen
- Lanzhou University Second Hospital, Lanzhou, China
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, China
- Gansu Provincial Key Laboratory Of Environmental Oncology, Lanzhou, China
| |
Collapse
|
48
|
Zhang Y, Wang Y, Xin E, Zhang Z, Ma D, Liu T, Gao F, Bian T, Sun Y, Wang M, Wang Z, Yan X, Li Y. Network pharmacology and experimental verification reveal the mechanism of Hedysari Radix and Curcumae Rhizoma with the optimal compatibility ratio against colitis-associated colorectal cancer. JOURNAL OF ETHNOPHARMACOLOGY 2024; 322:117555. [PMID: 38110130 DOI: 10.1016/j.jep.2023.117555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 12/01/2023] [Accepted: 12/04/2023] [Indexed: 12/20/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The herb pair Astragali Radix (AR) and Curcumae Rhizoma (vinegar-processed, VPCR), derived from the traditional Chinese medicine (TCM) text 'Yixuezhongzhongcanxilu', have long been used to treat gastrointestinal diseases, notably colitis-associated colorectal cancer (CAC). Hedysari Radix (HR), belonging to the same Leguminosae family as AR but from a different genus, is traditionally used as a substitute for AR when paired with VPCR in the treatment of CAC. However, the optimal compatibility ratio for HR-VPCR against CAC and the underlying mechanisms remain unclear. AIM OF THE STUDY To investigate the optimal compatibility ratio and underlying mechanisms of HR-VPCR against CAC using a combination of comparative pharmacodynamics, network pharmacology, and experimental verification. MATERIALS AND METHODS The efficacy of different compatibility ratios of HR-VPCR against CAC was evaluated using various indicators, including the body weight, colon length, tumor count, survival rate, disease activity index (DAI) score, Haemotoxylin and Eosin (H&E) pathological sections, inflammation cytokines (IL-1β, IL-6, IL-10, TNF-α), tumor markers (K-Ras, p53), and intestinal permeability proteins (claudin-1, E-cadherin, mucin-2). Then, the optimal compatibility ratio of HR-VPCR against CAC was determined based on the fuzzy matter-element analysis by integrating the above indicators. After high-performance liquid chromatography (HPLC) analysis for the optimal compatibility ratio of HR-VPCR, potential active components of HR-VPCR were identified by TCMSP and the previous bibliographies. Swiss Targets and GeneCards were adopted to predict the targets of the active components and the targets of CAC, respectively. Then, the common targets of HR-VPCR against CAC were obtained by Venn analysis. PPI networks were constructed in STRING. GO and KEGG enrichments were visualized by the David database. Finally, the predicted pathway was experimentally validated via Western blot. RESULTS Various compatibility ratios of HR-VPCR demonstrated notable therapeutic effects to some extent, evidenced by improvements in body weight, colon length, tumor count, pathological symptoms (DAI score), colon and organ indexes, survival rate, and modulation of inflammation factors (IL-1β, IL-6, IL-10, TNF-α), as well as tumor markers (K-Ras, p53), and down-regulation of intestinal permeability proteins (claudin-1, E-cadherin, mucin-2) in CAC mice. Among these ratios, the ratio 4:1 represents the optimal compatibility ratio by the fuzzy matter-element analysis. Thirty active components of HR-VPCR were carefully selected, targeting 553 specific genes. Simultaneously, 2022 targets associated with CAC were identified. 88 common targets were identified after generating a Venn plot. Following PPI network analysis, 29 core targets were established, with AKT1 ranking highest among them. Further analysis via GO and KEGG enrichment identified the PI3K-AKT signaling pathway as a potential mechanism. Experimental validation confirmed that HR-VPCR intervention effectively reversed the activated PI3K-AKT signaling pathway. CONCLUSIONS The optimal compatibility ratio for the HR-VPCR herb pair in alleviating CAC is 4:1. HR-VPCR exerts its effects by alleviating intestinal inflammation, improving intestinal permeability, and regulating the PI3K-AKT signaling pathway.
Collapse
Affiliation(s)
- Yugui Zhang
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China; Key Laboratory of Standard and Quality of Chinese Medicine Research of Gansu, Engineering Research Center of Chinese Medicine Pharmaceutical Process of Gansu, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China.
| | - Yanjun Wang
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China; Key Laboratory of Standard and Quality of Chinese Medicine Research of Gansu, Engineering Research Center of Chinese Medicine Pharmaceutical Process of Gansu, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China.
| | - Erdan Xin
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China; Key Laboratory of Standard and Quality of Chinese Medicine Research of Gansu, Engineering Research Center of Chinese Medicine Pharmaceutical Process of Gansu, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China.
| | - Zhuanhong Zhang
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China; Key Laboratory of Standard and Quality of Chinese Medicine Research of Gansu, Engineering Research Center of Chinese Medicine Pharmaceutical Process of Gansu, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China.
| | - Dingcai Ma
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China; Key Laboratory of Standard and Quality of Chinese Medicine Research of Gansu, Engineering Research Center of Chinese Medicine Pharmaceutical Process of Gansu, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China.
| | - Ting Liu
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China; Key Laboratory of Standard and Quality of Chinese Medicine Research of Gansu, Engineering Research Center of Chinese Medicine Pharmaceutical Process of Gansu, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China.
| | - Feiyun Gao
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China; Key Laboratory of Standard and Quality of Chinese Medicine Research of Gansu, Engineering Research Center of Chinese Medicine Pharmaceutical Process of Gansu, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China.
| | - Tiantian Bian
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China; Key Laboratory of Standard and Quality of Chinese Medicine Research of Gansu, Engineering Research Center of Chinese Medicine Pharmaceutical Process of Gansu, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China.
| | - Yujing Sun
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China; Key Laboratory of Standard and Quality of Chinese Medicine Research of Gansu, Engineering Research Center of Chinese Medicine Pharmaceutical Process of Gansu, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China; Scientific Research and Experimental Center, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China.
| | - Maomao Wang
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China; Key Laboratory of Standard and Quality of Chinese Medicine Research of Gansu, Engineering Research Center of Chinese Medicine Pharmaceutical Process of Gansu, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China.
| | - Zhe Wang
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China; Key Laboratory of Standard and Quality of Chinese Medicine Research of Gansu, Engineering Research Center of Chinese Medicine Pharmaceutical Process of Gansu, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China.
| | - Xingke Yan
- College of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China.
| | - Yuefeng Li
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China; Key Laboratory of Standard and Quality of Chinese Medicine Research of Gansu, Engineering Research Center of Chinese Medicine Pharmaceutical Process of Gansu, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China; Scientific Research and Experimental Center, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China.
| |
Collapse
|
49
|
Xu M, Xi S, Li H, Xia Y, Mei G, Cheng Z. Prognosis significance and potential association between ALDOA and AKT expression in colorectal cancer. Sci Rep 2024; 14:6488. [PMID: 38499636 PMCID: PMC10948905 DOI: 10.1038/s41598-024-57209-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Accepted: 03/15/2024] [Indexed: 03/20/2024] Open
Abstract
Colorectal cancer (CRC) is one of the most common malignant tumors of the digestive tract and a leading cause of cancer-related death worldwide. Since many CRC patients are diagnosed already in the advanced stage, and traditional chemoradiotherapy is prone to drug resistance, it is important to find new therapeutic targets. In this study, the expression levels of ALDOA and p-AKT were detected in cancer tissues and paired normal tissues, and it was found that they were significantly increased in CRC tissues, and their high expression indicated poor prognosis. Moreover, a positive correlation between the expression of ALDOA and p-AKT was found in CRC tissues and paired normal tissues. In addition, the Kaplan-Meier analysis revealed that the group with both negative of ALDOA/p-AKT expression had longer five-year survival rates compared with the other group. Besides, the group with both high expression of ALDOA/p-AKT had a worse prognosis compared with the other group. Based on the expression of ALDOA and p-AKT in tumor tissues, we can effectively distinguish tumor tissues from normal tissues through cluster analysis. Furthermore, we constructed nomograms to predict 3-year and 5-year overall survival, showing that the expression of ALDOA/p-AKT plays a crucial role in predicting the prognosis of CRC patients. Therefore, ALDOA/p-AKT may act as a crucial role in CRC, which may provide new horizons for targeted therapies for CRC.
Collapse
Affiliation(s)
- Menglin Xu
- Department of Oncology, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241000, China
| | - Shihang Xi
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241000, China
| | - Haoran Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, No.2 Zheshan West Road, Jinghu, Wuhu, 241000, Anhui, China
| | - Yong Xia
- Department of Education Affairs, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241000, China
| | - Guangliang Mei
- Department of Party Affairs, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241000, China
| | - Zhengwu Cheng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, No.2 Zheshan West Road, Jinghu, Wuhu, 241000, Anhui, China.
| |
Collapse
|
50
|
Ma M, Zheng Z, Li J, He Y, Kang W, Ye X. Association between the gut microbiota, inflammatory factors, and colorectal cancer: evidence from Mendelian randomization analysis. Front Microbiol 2024; 15:1309111. [PMID: 38562480 PMCID: PMC10982360 DOI: 10.3389/fmicb.2024.1309111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 03/06/2024] [Indexed: 04/04/2024] Open
Abstract
Background Colorectal cancer (CRC) is one of the most common malignant tumors primarily affecting individuals over the age of 50 years. Recent studies have suggested that the dysbiosis of the gut microbiota, a community of microorganisms in the human gut, is closely associated with the occurrence and development of CRC. Additionally, inflammatory factors (IFs) have also been reported to play a significant role in the development of CRC. However, the causal relationships between the gut microbiota, IFs, and CRC remain unclear. Methods In this study, we performed Mendelian randomization (MR) analysis using publicly available genome-wide association study (GWAS) data to explore the causal relationship between the gut microbiota, IFs, and CRC. The gut microbiota GWAS data were obtained from the MiBioGen study, while the IFs GWAS data were derived from the comprehensive analysis of three independent cohorts. Causal relationship analysis was conducted using appropriate instrumental variables (IVs) and statistical models. Results MR analysis of the gut microbiota and CRC revealed a negative correlation between the Lachnospiraceae species in the gut and CRC risk, while a positive correlation was observed between Porphyromonadaceae species, Lachnospiraceae UCG010 genus, Lachnospira genus, and Sellimonas genus in the gut, and CRC risk. Additionally, we observed a causal relationship between IL-10 and CRC risk. These findings suggest that the dysbiosis of the gut microbiota might be associated with an increased risk of CRC and that specific bacterial groups may play a crucial role in the occurrence and development of CRC. Conclusion Using MR analysis, this study revealed the causal relationships between the gut microbiota, IFs, and CRC. The negative correlation between the Lachnospiraceae species in the gut and CRC risk, as well as the causal relationship between IL-10 and CRC, provide important clues for the potential roles of gut microbiota regulation and inflammatory factor control in the prevention and treatment of CRC.
Collapse
Affiliation(s)
| | | | | | | | - Weiming Kang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xin Ye
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| |
Collapse
|