51
|
Chang YF, Wang HH, Shu CW, Tsai WL, Lee CH, Chen CL, Liu PF. TMEM211 Promotes Tumor Progression and Metastasis in Colon Cancer. Curr Issues Mol Biol 2023; 45:4529-4543. [PMID: 37367036 DOI: 10.3390/cimb45060287] [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: 04/24/2023] [Revised: 05/19/2023] [Accepted: 05/22/2023] [Indexed: 06/28/2023] Open
Abstract
Colon cancer is the third most important cancer type, leading to a remarkable number of deaths, indicating the necessity of new biomarkers and therapeutic targets for colon cancer patients. Several transmembrane proteins (TMEMs) are associated with tumor progression and cancer malignancy. However, the clinical significance and biological roles of TMEM211 in cancer, especially in colon cancer, are still unknown. In this study, we found that TMEM211 was highly expressed in tumor tissues and the increased TMEM211 was associated with poor prognosis in colon cancer patients from The Cancer Genome Atlas (TCGA) database. We also showed that abilities regarding migration and invasion were reduced in TMEM211-silenced colon cancer cells (HCT116 and DLD-1). Moreover, TMEM211-silenced colon cancer cells showed decreased levels of Twist1, N-cadherin, Snail and Slug but increased levels of E-cadherin. Levels of phosphorylated ERK, AKT and RelA (NF-κB p65) were also decreased in TMEM211-silenced colon cancer cells. Our findings indicate that TMEM211 regulates epithelial-mesenchymal transition for metastasis through coactivating the ERK, AKT and NF-κB signaling pathways, which might provide a potential prognostic biomarker or therapeutic target for colon cancer patients in the future.
Collapse
Affiliation(s)
- Yung-Fu Chang
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Translational Research Center of Neuromuscular Diseases, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - Hsing-Hsang Wang
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Chih-Wen Shu
- Institute of BioPharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Wei-Lun Tsai
- Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 81362, Taiwan
| | - Cheng-Hsin Lee
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Chun-Lin Chen
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Pei-Feng Liu
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| |
Collapse
|
52
|
Chang H, Sun W, Zhao Y, Sun T, Zhao Z. Targeting Pokemon is a novel strategy to suppress cancer aggressiveness of non-small cell lung cancer: Identification of Pokemon as ideal target for developing anti-NSCLC drugs. Arch Biochem Biophys 2023; 742:109637. [PMID: 37182800 DOI: 10.1016/j.abb.2023.109637] [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: 02/07/2023] [Revised: 05/10/2023] [Accepted: 05/11/2023] [Indexed: 05/16/2023]
Abstract
Although it is widely reported that Pokemon acts as an oncogene in the pathogenesis of multiple cancers, but its role and detailed molecular mechanisms in regulating non-small cell lung cancer (NSCLC) progression have not been fully delineated. Here, by performing Real-Time qPCR analysis, we verified that Pokemon was high-expressed in NSCLC tissues and cells, compared to the corresponding normal lung tissues and epithelial cells. Then, the small interfering RNA (siRNA) for Pokemon was transfected into the NSCLC cells to verify its biological functions, and our results suggested that silencing of Pokemon suppressed the malignant phenotypes, including cell viability, mitosis, colony formation, epithelial-mesenchymal transition (EMT), mobility and cancer stem cell (CSC) properties in NSCLC cells. Mechanistically, we confirmed that knockdown of Pokemon decreased the expression levels of phosphorylated Akt (p-Akt), phosphorylated GSK-3β (p-GSK-3β) and Snail to inactivate the oncogenic Akt/GSK-3β/Snail signal pathway, and deletion of Snail also had similar effects to hamper the development of NSCLC. Next, our rescuing experiments validated that Pokemon ablation-induced suppressing effects on NSCLC cell malignancy were all abrogated by overexpressing Snail. Finally, the in vivo experiments confirmed that silencing of Pokemon downregulated Snail to hamper tumorigenesis of NSCLC cells in xenograft tumor-bearing mice models. Taken together, we firstly uncovered the underlying mechanisms by which the Pokemon/Akt/GSK-3β/Snail signal pathway contributed to the development of NSCLC, and this signal pathway could be potentially used as therapeutic targets for the development of personalized anti-NSCLC drugs.
Collapse
Affiliation(s)
- Hao Chang
- Department of Thoracic Surgery, The First Affiliated Hospital of Harbin Medical University, Youzheng Street No. 23, Nangang District, Harbin, 150001, Heilongjiang, China.
| | - Wenxue Sun
- Department of Respiratory Medicine, The Third People's Hospital of Shenzhen City, Shenzhen, 518000, Guangdong, China.
| | - Yiming Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Harbin Medical University, Youzheng Street No. 23, Nangang District, Harbin, 150001, Heilongjiang, China.
| | - Tianhao Sun
- Department of Thoracic Surgery, The First Affiliated Hospital of Harbin Medical University, Youzheng Street No. 23, Nangang District, Harbin, 150001, Heilongjiang, China.
| | - Zhihong Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Harbin Medical University, Youzheng Street No. 23, Nangang District, Harbin, 150001, Heilongjiang, China.
| |
Collapse
|
53
|
Huang F, Wang Z, Zhu L, Lin C, Wang JX. Comprehensive Analysis of the Expression, Prognostic Value, and Immune Infiltration Activities of GABRD in Colon Adenocarcinoma. Mediators Inflamm 2023; 2023:8709458. [PMID: 37181811 PMCID: PMC10169248 DOI: 10.1155/2023/8709458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 04/03/2023] [Accepted: 04/16/2023] [Indexed: 05/16/2023] Open
Abstract
Colon adenocarcinoma (COAD) is one of the tumors with the highest mortality rates. It is of the utmost significance to make an accurate prognostic assessment and to tailor one's treatment to the specific needs of the patient. Multiple lines of evidence point to the possibility that genetic variables and clinicopathological traits are connected to the onset and development of cancer. In the past, a number of studies have revealed that gamma-aminobutyric acid type A receptor subunit delta (GABRD) plays a role in the advancement of a number of different cancers. However, its function in COAD was rarely reported. In this study, we analyzed TCGA datasets and identified 29 survival-related differentially expressed genes (DEGs) in COAD patients. In particular, GABRD expression was noticeably elevated in COAD specimens. There was a correlation between high GABRD expression and an advanced clinical stage. According to the results of the survival tests, patients whose GABRD expression was high had a lower overall survival time and progression-free survival time than those whose GABRD expression was low. GABRD expression was found to be an independent predictive predictor for overall survival, as determined by multivariate COX regression analysis. Additionally, the predictive nomogram model can accurately predict the fate of individuals with COAD. In addition, we observed that GABRD expressions were positively associated with the expression of T cells regulatory (Tregs), macrophages M0, while negatively associated with the expression of T cells CD8, T cells follicular helper, macrophages M1, dendritic cells activated, eosinophils, and T cells CD4 memory activated. The IC50 of BI-2536, bleomycin, embelin, FR-180204, GW843682X, LY317615, NSC-207895, rTRAIL, and VX-11e was higher in the GABRD high-expression group. In conclusion, we have shown evidence that GABRD is a novel biomarker that is connected with immune cell infiltration in COAD and may be utilized to predict the prognosis of COAD patients.
Collapse
Affiliation(s)
- Fakun Huang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fujian, China
| | | | - Liyue Zhu
- Fujian Medical University, Fuzhou, Fujian, China
| | | | - Jia-xing Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fujian, China
| |
Collapse
|
54
|
Wang MY, Wang XW, Zhao WX, Li Y, Cai ML, Wang KX, Xi XM, Zhao C, Zhou HM, Shao RG, Xia GM, Zhang YF, Zhao WL. Enhanced binding of β-catenin and β-TrCP mediates LMPt's anti-CSCs activity in colorectal cancer. Biochem Pharmacol 2023; 212:115577. [PMID: 37137416 DOI: 10.1016/j.bcp.2023.115577] [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: 01/14/2023] [Revised: 04/25/2023] [Accepted: 04/25/2023] [Indexed: 05/05/2023]
Abstract
Cancer stem cells (CSCs), a subpopulation of tumor cells with the features of self-renewal, tumor initiation, and insensitivity to common physical and chemical agents, are the key to cancer relapses, metastasis, and resistance. Accessible CSCs inhibitory strategies are primarily based on small molecule drugs, yet toxicity limits their application. Here, we report a liposome loaded with low toxicity and high effectiveness of miriplatin, lipo-miriplatin (LMPt) with high miriplatin loading, and robust stability, exhibiting a superior inhibitory effect on CSCs and non-CSCs. LMPt predominantly inhibits the survival of oxaliplatin-resistant (OXA-resistant) cells composed of CSCs. Furthermore, LMPt directly blocks stemness features of self-renewal, tumor initiation, unlimited proliferation, metastasis, and insensitivity. In mechanistic exploration, RNA sequencing (RNA-seq) revealed that LMPt downregulates the levels of pro-stemness proteins and that the β-catenin-mediated stemness pathway is enriched. Further research shows that either in adherent cells or 3D-spheres, the β-catenin-OCT4/NANOG axis, the vital pathway to maintain stemness, is depressed by LMPt. The consecutive activation of the β-catenin pathway induced by mutant β-catenin (S33Y) and OCT4/NANOG overexpression restores LMPt's anti-CSCs effect, elucidating the key role of the β-catenin-OCT4/NANOG axis. Further studies revealed that the strengthened binding of β-catenin and β-TrCP initiates ubiquitination and degradation of β-catenin induced by LMPt. In addition,the ApcMin/+transgenicmouse model, in which colon tumors are spontaneously formed, demonstrates LMPt's potent anti-non-CSCs activity in vivo.
Collapse
Affiliation(s)
- Meng-Yan Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences/Peking Union Medical College, Tiantanxili #1, Beijing 100050, P.R. China
| | - Xiao-Wei Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences/Peking Union Medical College, Tiantanxili #1, Beijing 100050, P.R. China
| | - Wen-Xia Zhao
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences/Peking Union Medical College, Tiantanxili #1, Beijing 100050, P.R. China
| | - Yang Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences/Peking Union Medical College, Tiantanxili #1, Beijing 100050, P.R. China
| | - Mei-Lian Cai
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences/Peking Union Medical College, Tiantanxili #1, Beijing 100050, P.R. China
| | - Ke-Xin Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences/Peking Union Medical College, Tiantanxili #1, Beijing 100050, P.R. China
| | - Xiao-Ming Xi
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences/Peking Union Medical College, Tiantanxili #1, Beijing 100050, P.R. China
| | - Cong Zhao
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences/Peking Union Medical College, Tiantanxili #1, Beijing 100050, P.R. China
| | - Hui-Min Zhou
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences/Peking Union Medical College, Tiantanxili #1, Beijing 100050, P.R. China
| | - Rong-Guang Shao
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences/Peking Union Medical College, Tiantanxili #1, Beijing 100050, P.R. China.
| | - Gui-Min Xia
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences/Peking Union Medical College, Tiantanxili #1, Beijing 100050, P.R. China.
| | - Ye-Fan Zhang
- Department of Hepatobiliary Surgery/National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Wu-Li Zhao
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences/Peking Union Medical College, Tiantanxili #1, Beijing 100050, P.R. China.
| |
Collapse
|
55
|
Zhang H, Xie L, Zhang N, Qi X, Lu T, Xing J, Akhtar MF, Li L, Liu G. Donkey Oil-Based Ketogenic Diet Prevents Tumor Progression by Regulating Intratumor Inflammation, Metastasis and Angiogenesis in CT26 Tumor-Bearing Mice. Genes (Basel) 2023; 14:genes14051024. [PMID: 37239383 DOI: 10.3390/genes14051024] [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: 03/04/2023] [Revised: 04/25/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
Colon cancer is one of the typical malignant tumors, and its prevalence has increased yearly. The ketogenic diet (KD) is a low-carbohydrate and high-fat dietary regimen that inhibits tumor growth. Donkey oil (DO) is a product with a high nutrient content and a high bioavailability of unsaturated fatty acids. Current research investigated the impact of the DO-based KD (DOKD) on CT26 colon cancer in vivo. Our findings revealed that DOKD administration significantly lowered CT26+ tumor cell growth in mice, and the blood β-hydroxybutyrate levels in the DOKD group was significantly higher than those in the natural diet group. Western blot results showed that DOKD significantly down-regulated Src, hypoxia inducible factor-1α (HIF-1α), extracellular signal-related kinases 1 and 2 (Erk1/2), snail, neural cadherin (N-cadherin), vimentin, matrix metallopeptidase 9 (MMP9), signal transducer and activator of transcription 3 (STAT3), and vascular endothelial growth factor A (VEGFA), and it significantly up-regulated the expressions of Sirt3, S100a9, interleukin (IL)-17, nuclear factor-kappaB (NF-κB) p65, Toll-like receptor 4 (TLR4), MyD88, and tumor necrosis factor-α. Meanwhile, in vitro validation results showed that LW6 (a HIF-1α inhibitor) significantly down-regulated the expressions of HIF-1α, N-cadherin, vimentin, MMP9, and VEGFA, which supported those of the in vivo findings. Furthermore, we found that DOKD inhibited CT26+ tumor cell growth by regulating inflammation, metastasis, and angiogenesis by activating the IL-17/TLR4/NF-κB p65 pathway and inhibiting the activation of the Src/HIF-1α/Erk1/2/Snail/N-cadherin/Vimentin/MMP9 and Erk1/2/HIF-1α/STAT3/VEGFA pathways. Our findings suggest that DOKD may suppress colon cancer progression and help prevent colon cancer cachexia.
Collapse
Affiliation(s)
- Huachen Zhang
- College of Agronomy, Shandong Engineering Technology Research Center for Efficient Breeding and Ecological Feeding of Black Donkey, Shandong Donkey Industry Technology Collaborative Innovation Center, Liaocheng University, Liaocheng 252000, China
| | - Lan Xie
- College of Agronomy, Shandong Engineering Technology Research Center for Efficient Breeding and Ecological Feeding of Black Donkey, Shandong Donkey Industry Technology Collaborative Innovation Center, Liaocheng University, Liaocheng 252000, China
| | - Ning Zhang
- Biopharmaceutical Research Institute, Liaocheng University, Liaocheng 252000, China
| | - Xingzhen Qi
- College of Agronomy, Shandong Engineering Technology Research Center for Efficient Breeding and Ecological Feeding of Black Donkey, Shandong Donkey Industry Technology Collaborative Innovation Center, Liaocheng University, Liaocheng 252000, China
| | - Ting Lu
- College of Agronomy, Shandong Engineering Technology Research Center for Efficient Breeding and Ecological Feeding of Black Donkey, Shandong Donkey Industry Technology Collaborative Innovation Center, Liaocheng University, Liaocheng 252000, China
| | - Jingya Xing
- Inner Mongolia Key Laboratory of Equine Genetics, Breeding and Reproduction, Equine Research Center, College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Muhammad Faheem Akhtar
- College of Agronomy, Shandong Engineering Technology Research Center for Efficient Breeding and Ecological Feeding of Black Donkey, Shandong Donkey Industry Technology Collaborative Innovation Center, Liaocheng University, Liaocheng 252000, China
| | - Lanjie Li
- Office of International Programs, Liaocheng University, Liaocheng 252000, China
| | - Guiqin Liu
- College of Agronomy, Shandong Engineering Technology Research Center for Efficient Breeding and Ecological Feeding of Black Donkey, Shandong Donkey Industry Technology Collaborative Innovation Center, Liaocheng University, Liaocheng 252000, China
| |
Collapse
|
56
|
Park SM, Jee W, Park YR, Kim H, Na YC, Jung JH, Jang HJ. Euonymus sachalinensis Induces Apoptosis by Inhibiting the Expression of c-Myc in Colon Cancer Cells. Molecules 2023; 28:molecules28083473. [PMID: 37110707 PMCID: PMC10140968 DOI: 10.3390/molecules28083473] [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: 03/09/2023] [Revised: 04/09/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
We hypothesized that Euonymus sachalinensis (ES) induces apoptosis by inhibiting the expression of c-Myc in colon cancer cells, and this study proved that the methanol extract of ES has anticancer effects in colon cancer cells. ES belongs to the Celastraceae family and is well known for its medicinal properties. Extracts of species belonging to this family have been used to treat diverse diseases, including rheumatoid arthritis, chronic nephritis, allergic conjunctivitis, rhinitis, and asthma. However, ES has been targeted because there are currently few studies on the efficacy of ES for various diseases, including cancer. ES lowers cell viability in colon cancer cells and reduces the expression of c-Myc protein. We confirm that the protein level of apoptotic factors such as PARP and Caspase 3 decrease when ES is treated with Western blot, and confirm that DNA fragments occur through TUNEL assay. In addition, it is confirmed that the protein level of oncogenes CNOT2 and MID1IP1 decrease when ES is treated. We have also found that ES enhances the chemo-sensitivity of 5-FU in 5-FU-resistant cells. Therefore, we confirm that ES has anticancer effects by inducing apoptotic cell death and regulating the oncogenes CNOT2 and MID1IP1, suggesting its potential for use in the treatment of colon cancer.
Collapse
Affiliation(s)
- So-Mi Park
- College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Wona Jee
- College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Ye-Rin Park
- College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Hyungsuk Kim
- Department of Korean Rehabilitation Medicine, Kyung Hee University Medical Center, Seoul 02447, Republic of Korea
| | - Yun-Cheol Na
- Western Seoul Center, Korea Basic Science Institute, 150 Bugahyeon-ro, Seodaemun-gu, Seoul 03759, Republic of Korea
| | - Ji Hoon Jung
- College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Hyeung-Jin Jang
- College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
57
|
Kang KT, Shin MJ, Moon HJ, Choi KU, Suh DS, Kim JH. TRRAP Enhances Cancer Stem Cell Characteristics by Regulating NANOG Protein Stability in Colon Cancer Cells. Int J Mol Sci 2023; 24:ijms24076260. [PMID: 37047234 PMCID: PMC10094283 DOI: 10.3390/ijms24076260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
NANOG, a stemness-associated transcription factor, is highly expressed in many cancers and plays a critical role in regulating tumorigenicity. Transformation/transcription domain-associated protein (TRRAP) has been reported to stimulate the tumorigenic potential of cancer cells and induce the gene transcription of NANOG. This study aimed to investigate the role of the TRRAP-NANOG signaling pathway in the tumorigenicity of cancer stem cells. We found that TRRAP overexpression specifically increases NANOG protein stability by interfering with NANOG ubiquitination mediated by FBXW8, an E3 ubiquitin ligase. Mapping of NANOG-binding sites using deletion mutants of TRRAP revealed that a domain of TRRAP (amino acids 1898–2400) is responsible for binding to NANOG and that the overexpression of this TRRAP domain abrogated the FBXW8-mediated ubiquitination of NANOG. TRRAP knockdown decreased the expression of CD44, a cancer stem cell marker, and increased the expression of P53, a tumor suppressor gene, in HCT-15 colon cancer cells. TRRAP depletion attenuated spheroid-forming ability and cisplatin resistance in HCT-15 cells, which could be rescued by NANOG overexpression. Furthermore, TRRAP knockdown significantly reduced tumor growth in a murine xenograft transplantation model, which could be reversed by NANOG overexpression. Together, these results suggest that TRRAP plays a pivotal role in the regulation of the tumorigenic potential of colon cancer cells by modulating NANOG protein stability.
Collapse
Affiliation(s)
- Kyung-Taek Kang
- Department of Physiology, College of Medicine, Pusan National University, Yangsan 50612, Gyeongsangnam-do, Republic of Korea
| | - Min-Joo Shin
- Department of Physiology, College of Medicine, Pusan National University, Yangsan 50612, Gyeongsangnam-do, Republic of Korea
| | - Hye-Ji Moon
- Department of Physiology, College of Medicine, Pusan National University, Yangsan 50612, Gyeongsangnam-do, Republic of Korea
| | - Kyung-Un Choi
- Department of Pathology, College of Medicine, Pusan National University, Yangsan 50612, Gyeongsangnam-do, Republic of Korea
| | - Dong-Soo Suh
- Department of Obstetrics and Gynecology, School of Medicine, Pusan National University, Yangsan 50612, Gyeongsangnam-do, Republic of Korea
| | - Jae-Ho Kim
- Department of Physiology, College of Medicine, Pusan National University, Yangsan 50612, Gyeongsangnam-do, Republic of Korea
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Gyeongsangnam-do, Republic of Korea
- Correspondence: ; Tel.: +82-51-510-8073; Fax: +82-51-510-8076
| |
Collapse
|
58
|
Deng Z, Xu Y, Cai Y, Lin W, Zhang L, Jiang A, Zhou Y, Zhao R, Zhao H, Liu Z, Yan T. Inhibition of Ribosomal RNA Processing 15 Homolog (RRP15) Suppressed Tumor Growth, Invasion and Epithelial to Mesenchymal Transition (EMT) of Colon Cancer. Int J Mol Sci 2023; 24:3528. [PMID: 36834940 PMCID: PMC9965612 DOI: 10.3390/ijms24043528] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 01/31/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
Although ribosomal RNA processing 15 Homolog (RRP15) has been implicated in the occurrence of various cancers and is considered a potential target for cancer treatment, its significance in colon cancer (CC) is unclear. Thus, this present study aims to determine RRP15 expression and biological function in CC. The results demonstrated a strong expression of RRP15 in CC compared to normal colon specimens, which was correlated with poorer overall survival (OS) and disease-free survival (DFS) of the patients. Among the nine investigated CC cell lines, RRP15 demonstrated the highest and lowest expression in HCT15 and HCT116 cells, respectively. In vitro assays demonstrated that the knockdown of RRP15 inhibited the growth, colony-forming ability and invasive ability of the CC cells whereas its overexpression enhanced the above oncogenic function. Moreover, subcutaneous tumors in nude mice showed that RRP15 knockdown inhibited the CC growth while its overexpression enhanced their growth. Additionally, the knockdown of RRP15 inhibited the epithelial-mesenchymal transition (EMT), whereas overexpression of RRP15 promoted the EMT process in CC. Collectively, inhibition of RRP15 suppressed tumor growth, invasion and EMT of CC, and might be considered a promising therapeutic target for treating CC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Zhaoguo Liu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Tingdong Yan
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, China
| |
Collapse
|
59
|
Zhu S, Yin J, Ye Q, Xiang J, Zhang Z, Yan B. Combined preoperative prognostic nutritional index and D-dimer score predicts outcome in colorectal cancer. BMC Surg 2023; 23:30. [PMID: 36750842 PMCID: PMC9903491 DOI: 10.1186/s12893-023-01925-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/27/2023] [Indexed: 02/09/2023] Open
Abstract
BACKGROUND The prognostic nutritional index (PNI) and D-dimer (DD) levels represent useful prognostic indicators in colorectal cancer (CRC); however, a combination of these indicators, namely, the PNI and DD score (PDS) was less addressed. METHODS A retrospective study with 183 patients after curative surgery was conducted. Patients were divided into 3 subgroups: PDS 0, decreased PNI and increased DD levels; PDS 1, decreased or increased PNI and DD levels; PDS 2, increased PNI and decreased DD levels. The differences in disease-free survival (DFS) and overall survival (OS) were compared among these subgroups, and risk factors for outcome were determined. RESULTS A total of 56, 65 and 62 patients were assigned to the PDS 0, 1 and 2 subgroups, respectively. PDS was significant in predicting both the DFS (area under the curve (AUC) = 0.68, P < 0.001) and OS (AUC = 0.74, P < 0.001). PDS 0 patients were more likely to be associated with old age (P = 0.032), laparotomy (P < 0.001), elevated CEA (P = 0.001), T3 + T4 (P = 0.001) and advanced TNM stage (P = 0.031). PDS 0 patients had significantly inferior DFS (log rank = 18.35, P < 0.001) and OS (log rank = 28.34, P < 0.001) than PDS 1 or 2 patients. PDS was identified as an independent risk factor for both DFS (PDS 1: HR = 0.54, 95% CI: 0.30-1.00, P = 0.049; PDS 2: HR = 0.40, 95% CI: 0.20-0.79, P = 0.009) and OS (PDS 1: HR = 0.44, 95% CI: 0.22-0.88, P = 0.020; PDS 2: HR = 0.17, 95% CI: 0.06-0.45, P < 0.001). CONCLUSION The PDS is a useful prognostic indicator for CRC patients after curative surgery, and PDS 0 patients have inferior survival. Additional future studies are needed to validate these findings.
Collapse
Affiliation(s)
- Shibin Zhu
- Department of Clinical Laboratory, Hainan Hospital of Chinese PLA General Hospital, Sanya, Hainan, 572000, People's Republic of China
| | - Jianyuan Yin
- Department of Critical Care Medicine, Hainan Hospital of Chinese PLA General Hospital, Sanya, Hainan, 572000, People's Republic of China
| | - Qianwen Ye
- Department of Oncology, Hainan Hospital of Chinese PLA General Hospital, No. 80 of Jianglin Road, Haitang District, Sanya, Hainan, 572000, People's Republic of China
| | - Jia Xiang
- Department of Oncology, Hainan Hospital of Chinese PLA General Hospital, No. 80 of Jianglin Road, Haitang District, Sanya, Hainan, 572000, People's Republic of China
| | - Zihao Zhang
- Department of Clinical Laboratory, Hainan Hospital of Chinese PLA General Hospital, Sanya, Hainan, 572000, People's Republic of China
| | - Bing Yan
- Department of Oncology, Hainan Hospital of Chinese PLA General Hospital, No. 80 of Jianglin Road, Haitang District, Sanya, Hainan, 572000, People's Republic of China.
| |
Collapse
|
60
|
Feng X, Li Z, Guo W, Hu Y. The effects of traditional Chinese medicine and dietary compounds on digestive cancer immunotherapy and gut microbiota modulation: A review. Front Immunol 2023; 14:1087755. [PMID: 36845103 PMCID: PMC9945322 DOI: 10.3389/fimmu.2023.1087755] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 01/17/2023] [Indexed: 02/09/2023] Open
Abstract
Digestive tract-related cancers account for four of the top ten high-risk cancers worldwide. In recent years, cancer immunotherapy, which exploits the innate immune system to attack tumors, has led to a paradigm shifts in cancer treatment. Gut microbiota modification has been widely used to regulate cancer immunotherapy. Dietary compounds and traditional Chinese medicine (TCM) can alter the gut microbiota and its influence on toxic metabolite production, such as the effect of iprindole on lipopolysaccharide (LPS), and involvement in various metabolic pathways that are closely associated with immune reactions. Therefore, it is an effective strategy to explore new immunotherapies for gastrointestinal cancer to clarify the immunoregulatory effects of different dietary compounds/TCMs on intestinal microbiota. In this review, we have summarized recent progress regarding the effects of dietary compounds/TCMs on gut microbiota and their metabolites, as well as the relationship between digestive cancer immunotherapy and gut microbiota. We hope that this review will act as reference, providing a theoretical basis for the clinical immunotherapy of digestive cancer via gut microbiota modulation.
Collapse
Affiliation(s)
- Xiaoli Feng
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Zhenhao Li
- Department of General Surgery, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Weihong Guo
- Department of General Surgery, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China,*Correspondence: Weihong Guo, ; Yanfeng Hu,
| | - Yanfeng Hu
- Department of General Surgery, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China,*Correspondence: Weihong Guo, ; Yanfeng Hu,
| |
Collapse
|
61
|
Zhou G, Lv X, Zhong X, Ying W, Li W, Feng Y, Xia Q, Li J, Jian S, Leng Z. Suspension culture strategies to enrich colon cancer stem cells. Oncol Lett 2023; 25:116. [PMID: 36844615 PMCID: PMC9950343 DOI: 10.3892/ol.2023.13702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 11/16/2021] [Indexed: 02/09/2023] Open
Abstract
How to efficiently obtain high-purity cancer stem cells (CSCs) has been the basis of CSC research, but the optimal conditions for serum-free suspension culture of CSCs are still unclear. The present study aimed to define the optimal culture medium composition and culture time for the enrichment of colon CSCs via suspension culture. Suspension cell cultures of colon cancer DLD-1 cells were prepared using serum-free medium (SFM) containing variable concentrations of epidermal growth factor (EGF) and basic fibroblast growth factor (bFGF) to produce spheroids. Culture times were set at 10, 20 and 30 days. A total of nine different concentrations of EGF and bFGF were added to SFM to generate nine experimental groups. The proportions of CD44+, CD133+, and CD44+CD133+ double-positive spheroid cells were detected via flow cytometry. mRNA expression of stemness-, epithelial-mesenchymal transition- and Wnt/β-catenin pathway-associated genes was determined via reverse transcription-quantitative PCR. Self-renewal ability was evaluated by a sphere-forming assay. Tumorigenesis was studied in vitro using a colony formation assay and in vivo via subcutaneous cell injection in nude mice. It was found that the highest expression proportions of CD133+ and CD44+ spheroid cells were observed in group (G)9 (20 ng/ml EGF + 20 ng/ml bFGF) at 30 days (F=123.554 and 99.528, respectively, P<0.001), CD133+CD44+ cells were also observed in G9 at 30 days (and at 10 days in G3 and 20 days in G6; F=57.897, P<0.001). G9 at 30 days also displayed the highest expression of Krüppel-like factor 4, leucine-rich repeat-containing G protein-coupled receptor 5, CD44, CD133, Vimentin and Wnt-3a (F=22.682, 25.401, 3.272, 7.852, 13.331 and 17.445, respectively, P<0.001) and the lowest expression of E-cadherin (F=10.851, P<0.001). G9 at 30 days produced the highest yield of cell spheroids, as determined by a sphere forming assay (F=19.147, P<0.001); colony formation assays also exhibited the greatest number of colonies derived from G9 spheroids at 30 days (F=60.767, P<0.01), which also generated the largest mean tumor volume in the subcutaneous tumorigenesis xenograft model (F=12.539, P<0.01). In conclusion, 20 ng/ml EGF + 20 ng/ml bFGF effectively enriched colon CSCs when added to suspension culture for 30 days, and conferred the highest efficiency compared with other combinations.
Collapse
Affiliation(s)
- Guojun Zhou
- Department of Hepatobiliary Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China,Cancer Stem Cells Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Xiaojiang Lv
- Cancer Stem Cells Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Xiaorong Zhong
- Department of Hepatobiliary Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China,Cancer Stem Cells Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Wei Ying
- Department of Hepatobiliary Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China,Cancer Stem Cells Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Wenbo Li
- Department of Hepatobiliary Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China,Cancer Stem Cells Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Yanchao Feng
- Department of Hepatobiliary Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China,Cancer Stem Cells Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Qinghua Xia
- Department of General Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430074, P.R. China
| | - Jianshui Li
- Department of Hepatobiliary Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China,Cancer Stem Cells Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Shunhai Jian
- Department of Pathology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China,Professor Shunhai Jian, Department of Pathology, Affiliated Hospital of North Sichuan Medical College, 63 Wenhua Road, Nanchong, Sichuan 637000, P.R. China, E-mail:
| | - Zhengwei Leng
- Department of Hepatobiliary Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China,Cancer Stem Cells Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China,Correspondence to: Professor Zhengwei Leng, Department of Hepatobiliary Surgery, Affiliated Hospital of North Sichuan Medical College, 234, Fujiang Road, Nanchong, Sichuan 637000, P.R. China, E-mail:
| |
Collapse
|
62
|
Clark AG, Bertrand FE, Sigounas G. A potential requirement for Smad3 phosphorylation in Notch-mediated EMT in colon cancer. Adv Biol Regul 2023; 88:100957. [PMID: 36739740 DOI: 10.1016/j.jbior.2023.100957] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/04/2023] [Accepted: 01/27/2023] [Indexed: 01/30/2023]
Abstract
Colorectal cancer (CRC) remains a challenging disease to treat due to several factors including stemness and epithelial to mesenchymal transition (EMT). Dysfunctional signaling pathways such as Notch and TGF-β contribute to these phenomena. We previously found that cells expressing constitutively active Notch1 also had increased expression of Smad3, an important member of the TGF-β signaling pathway. We hypothesized that Smad3, mediates the Notch-induced stemness and EMT observed in CRC cells. The human colorectal carcinoma cell line HCT-116, stably transduced with constitutively active Notch-1 (ICN) or a GFP-vector control was treated with different combinations of TGF-β1, DAPT (a Notch inhibitor), or SIS3 (a Smad3 inhibitor). Western blot analysis was performed to determine the effects of Smad3 stimulation and inhibition on Notch and potential downstream EMT-related targets, CD44, Slug and Snail. Smad3 inhibition induced a decrease in Notch1 and Notch3 receptor expression and effectively inhibited CD44, Slug, and Snail expression. Colosphere forming ability was also reduced in cells with inhibited Smad3. These results indicate a key role of TGF-β signaling in Notch1-induced tumorigenesis, and suggest a potential use for Smad3 inhibitors in combination with Notch1 inhibitors that are already in use for CRC treatments.
Collapse
Affiliation(s)
- Alexander G Clark
- Department of Internal Medicine, Division of Hematology/Oncology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - Fred E Bertrand
- Department of Clinical and Diagnostic Sciences, School of Health Professions, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - George Sigounas
- Department of Internal Medicine, Division of Hematology/Oncology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA.
| |
Collapse
|
63
|
Miao Z, Zhao X, Liu X. Hypoxia induced β-catenin lactylation promotes the cell proliferation and stemness of colorectal cancer through the wnt signaling pathway. Exp Cell Res 2023; 422:113439. [PMID: 36464122 DOI: 10.1016/j.yexcr.2022.113439] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 10/21/2022] [Accepted: 11/27/2022] [Indexed: 12/03/2022]
Abstract
Colorectal cancer (CRC) is a common malignant tumor of digestive system. Its incidence rate and mortality rate ranks the third among all the malignant tumors. The objective of this study was to explore the role of β-catenin in the CRC progression. The CRC tissues were collected to analyze the β-catenin levels. The CRC cells (SW620 and RRKO) were treated with hypoxia to simulate the hypoxic microenvironment of tumor in vitro. The β-catenin levels in the CRC cells were assessed with RT-qPCR, Western blot and Immunofluorescence. The cell biological behaviors were determined with CCK-8, flow cytometry and sphere formation assays. Besides, the glucose uptake, lactate production, ECAR and OCR was detected by seahorse. For the β-catenin lactylation determination, the IP and Western blot assay was performed. Then the protein stability of β-catenin was measured after cycloheximide treatment. The results showed that β-catenin was highly expressed in the CRC tissues and cells. Hypoxia treatment dramatically increased the protein levels and lactylation of β-catenin in the CRC cells. In addition, β-catenin knockdown dramatically inhibited the cell growth and stemness of the CRC cells. Besides, activation of Wnt signaling pathway neutralized the role of sh-β-catenin in the hypoxia treated CRC cells. In conclusion, this study confirmed that hypoxia induced the glycolysis promoted the β-catenin lactylation, which further enhanced the protein stability and expression of β-catenin, thus aggravating the malignant behaviors of CRC cells.
Collapse
Affiliation(s)
- Zhi Miao
- Frontier Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, 300072, China; Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, China.
| | - Xiaomeng Zhao
- Frontier Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, 300072, China; Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, China
| | - Xiang Liu
- Department of Laboratory Medicine, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430016, China; The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| |
Collapse
|
64
|
Investigation of Three Morchella Species for Anticancer Activity Against Colon Cancer Cell Lines by UPLC-MS-Based Chemical Analysis. Appl Biochem Biotechnol 2023; 195:486-504. [PMID: 36094647 DOI: 10.1007/s12010-022-04131-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2022] [Indexed: 01/13/2023]
Abstract
In search of new anticancer agents, natural products including fungal compounds had been used as potential anticancer agents. The aim of this study was to investigate the anticancer activity of Morchella extracts against colon cancer cell line and UPLC-DAD-MS/MS analysis for the identification of compounds. The cytotoxic activity of the three Morchella species was examined for their anti-carcinogenic properties against the colon cancer cell lines. Phytochemical analyses were performed to screen Morchella for the presence of anti-cancerous compounds. All the fungal extracts inhibited the viability of colon cancer cells in a dose-dependent manner. Major compounds identified in Morchella included amino acid, fatty acid, sterol, flavonoid, peptide, glutamic acid, alkaloid, terpenoid, cyclopyrrolones, and coumarin. Several new compounds were detected among all the three Morchella extracts. In conclusion, all the fungal extracts showed potential inhibition of colon cancer cells and actively arrested the cell viability. It was concluded that the identified bioactive compounds might be the main constituents contributing to the anticancer activity of Morchella against human colon cancer cell lines. Thus, Morchella extracts are a potential source of bioactive compounds with cytotoxicity and could potentially be used as functional food supplements. Due to the nature of impressive findings, this investigation should be undertaken further to allow the studies to explore and develop a potential cytotoxic agents against colon cancer.
Collapse
|
65
|
Tao W, Liu F, Cheng YX, Zhang B, Liu XY, Zhang W, Peng D. Comparison of Postoperative Outcome and Prognosis Among Laparoscopic Left Colectomy and Laparoscopic Sigmoidectomy in Sigmoid Colon Cancer Patients: A Propensity Score Matching Study. Cancer Control 2023; 30:10732748231210676. [PMID: 37982606 PMCID: PMC10664434 DOI: 10.1177/10732748231210676] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 10/06/2023] [Accepted: 10/09/2023] [Indexed: 11/21/2023] Open
Abstract
PURPOSE The purpose of this study was to investigate the effect of laparoscopic left colectomy (LLC) and laparoscopic sigmoidectomy (LSD) on short-term outcomes and prognosis of sigmoid colon cancer (SCC) patients using propensity score matching (PSM). METHODS In this retrospective study, the SCC patients who underwent LLC or LSD surgery were collected from a single clinical center from Jan 2011 to Dec 2019. Short-term outcomes and prognosis were compared between patients who received LSD surgery and LLC surgery. RESULTS A total of 356 patients were included in this study. After 1:1 PSM analysis, there were 50 patients who underwent LLC surgery and 50 patients who underwent LSD surgery left in this study. No significant difference was found in baseline characteristics after PSM (P > .05). In comparison with the LLC surgery group, the LSD surgery group had shorter operation time (P = .003) after PSM. Moreover, the surgical procedure was not an independent predictor for overall survival (OS) (P = .918, 95% CI = .333-2.688) and disease-free survival DFS (P = .730, 95% CI = .335-2.150), but age (OS: P = .009, 95% CI = 1.010-1.075; DFS: P = .014, 95% CI = 1.007-1.061) and tumor stage (OS: P = .004, 95% CI = 1.302-3.844; DFS: P < .01, 95% CI = 1.572-4.171) were the independent risk factors for OS and DFS in SCC patients. CONCLUSION There was no significant difference between the two surgical procedures for prognosis of SCC patients. However, the possible reasons for changing the surgical procedures should be cautious by surgeons.
Collapse
Affiliation(s)
- Wei Tao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Fei Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yu-Xi Cheng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Bin Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiao-Yu Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wei Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dong Peng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
66
|
Dai X, Xie Y, Dong M. Cancer-associated fibroblasts derived extracellular vesicles promote angiogenesis of colorectal adenocarcinoma cells through miR-135b-5p/FOXO1 axis. Cancer Biol Ther 2022; 23:76-88. [PMID: 35100092 PMCID: PMC8812748 DOI: 10.1080/15384047.2021.2017222] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Colorectal adenocarcinoma (COAD) is a prevalent malignant tumor. Cancer-associated fibroblasts (CAFs)-derived extracellular vesicles (EVs) (CAFs-EVs) are implicated in COAD treatment. This study explored the mechanism of CAFs-EVs in COAD. CAFs and normal fibroblast (NFs) were isolated from COAD tissues and adjacent normal tissues. Vimentin, α-SMA, and FAP expressions were detected. EVs were isolated from CAFs and identified. SW480 and HCT116 cells were co-incubated with EVs. The EV uptake and COAD cell malignant behaviors were assessed. EV-treated SW480 and HCT116 cells were co-cultured with human umbilical vein endothelial cells (HUVECs). Extensive analyses were conducted to examine HUVEC proliferation, migration, and angiogenesis, and miR-135b-5p expression in COAD cells, and SW480 and HCT116 cells. CAFs were transfected with the miR-135b-5p inhibitor. miR-135b-5p downstream targets were predicted. FOXO1 expression in the co-culture system was determined and then overexpressed to evaluate its role in HUVECs mediated by COAD cells. COAD mouse model was established by transplanting SW480 cells into nude mice and injecting with EVs. Tumor growth rate, volume, and weight were examined. Ki67, VEGF, CD34, FOXO1 expressions, and VEGF content were detected. CAFs-EVs promoted COAD cell malignant behaviors and COAD cells-mediated HUVEC proliferation, migration, and angiogenesis. CAFs-EVs delivered miR-135b-5p into COAD cells. miR-135b-5p targeted FOXO1. Inhibition of miR-135b-5p in EVs or overexpression of FOXO1 partially reversed the effect of EVs on promoting COAD-induced angiogenesis. CAFs-EVs promoted tumor proliferation and angiogenesis of COAD in vivo. CAFs-EVs delivered miR-135b-5p into COAD cells to downregulate FOXO1 and promote HUVECs proliferation, migration, and angiogenesis.
Collapse
Affiliation(s)
- Xiaoyu Dai
- Department of Anus & Intestine Sugery, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo City, Zhejiang, China
| | - Yangyang Xie
- Pharmacy Department, Ningbo Eye Hospital, Ningbo City, Zhejiang, China
| | - Mingjun Dong
- Department of Anus & Intestine Sugery, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo City, Zhejiang, China
| |
Collapse
|
67
|
Singh AK, Shuaib M, Prajapati KS, Kumar S. Rutin Potentially Binds the Gamma Secretase Catalytic Site, Down Regulates the Notch Signaling Pathway and Reduces Sphere Formation in Colonospheres. Metabolites 2022; 12:926. [PMID: 36295828 PMCID: PMC9610901 DOI: 10.3390/metabo12100926] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 11/16/2022] Open
Abstract
Rutin, a natural flavonol, can modulate molecular signaling pathways and has considerable potential in cancer treatment. However, little is known about the effect of rutin on the notch signaling pathway (NSP) in cancer and cancer stem-like cells. In this study, we explored the effect of rutin on gamma secretase (GS, a putative notch signaling target) inhibition mediated NICD (Notch Intracellular Domain) production in colon cancer cells. Molecular docking, MM-GBSA, and Molecular dynamics (MD) simulation experiments were performed to check rutin's GS catalytic site binding potential. The HCT-116 colon cancer and cancer stem-like cells (colonospheres) were utilized to validate the in silico findings. The NICD production, notch promoter assay, expression of notch target genes, and cancer stemness/self-renewal markers were studied at molecular levels. The results were compared with the Notch-1 siRNA transfected test cells. The in silico study revealed GS catalytic site binding potential in rutin. The in vitro results showed a decreased NICD formation, an altered notch target gene (E-cad, Hes-1, and Hey-1) expression, and a reduction in stemness/self-renewal markers (CD44, c-Myc, Nanog, and Sox2) in test cells in a time and dose-dependent manner. In conclusion, rutin inhibits the notch signaling pathway and reduces the stemness/self-renewal property in colon cancer cells and the colonospheres by targeting gamma secretase. The clinical efficacy of rutin in combination therapy in colon cancer may be studied in the future.
Collapse
Affiliation(s)
| | | | | | - Shashank Kumar
- Molecular Signaling & Drug Discovery Laboratory, Department of Biochemistry, Central University of Punjab, Guddha, Bathinda 151401, Punjab, India
| |
Collapse
|
68
|
Huang X, Huan Y, Liu L, Ye Q, Guo J, Yan B. Preoperative low absolute lymphocyte count to fibrinogen ratio correlated with poor survival in nonmetastatic colorectal cancer. World J Surg Oncol 2022; 20:309. [PMID: 36153540 PMCID: PMC9508774 DOI: 10.1186/s12957-022-02775-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 08/27/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Preoperative absolute lymphocyte count (LC) and fibrinogen (FIB) are useful prognostic indicators in colorectal cancer (CRC). However, the prognostic value of the LC to FIB ratio (LFR) has never been addressed. METHODS A total of 189 nonmetastatic CRC patients after resection were enrolled retrospectively. The significance of the LFR in predicting disease-free survival (DFS) and overall survival (OS) was estimated by receiver operating characteristic curve analysis, and the prognostic efficacy was compared with individual LC and FIB. Patients were assigned to LFR low or high subgroups. Differences in clinicopathological features among these subgroups were calculated, and the survival differences of these subgroups were determined by the Kaplan-Meier analysis. A Cox proportional hazards model was applied to test the risk factors for survival. RESULTS Taking 0.54 as the optimal cutoff point, the LFR had sensitivities of 79.70% and 86.40% and specificities of 52.30% and 51.00% in predicting the DFS and OS, respectively. A total of 109/189 (57.67%) patients were assigned to the LFR low group, and these patients were more likely to be characterized by criteria such as T3 + T4 (P < 0.01), stage 3 (P < 0.01), tumor deposits (P = 0.01), high CEA (P < 0.01), or CA19-9 levels (P = 0.04). And they also displayed worse DFS (log rank = 18.57, P < 0.01) and OS (log rank = 20.40, P < 0.01) than the high LFR group. Finally, the LFR was independently associated with inferior DFS (HR = 0.32, 95% CI: 0.16-0.61, P < 0.01) and OS (HR = 0.23, 95% CI: 0.09-0.55, P < 0.01). CONCLUSIONS The LFR is a useful prognostic indicator in nonmetastatic CRC, and patients with a relatively low LFR had poor survival.
Collapse
Affiliation(s)
- Xiang Huang
- Department of Pulmonary Function Test, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200443, People's Republic of China
| | - Yu Huan
- Department of Clinic Laboratory, Hainan Hospital of Chinese PLA General Hospital, Sanya, Hainan Province, 572000, People's Republic of China
| | - Long Liu
- Department Traditional Chinese Medicine, Tianyou Hospital of Tongji University, Shanghai, 200331, People's Republic of China
| | - Qianwen Ye
- Department of Oncology, Hainan Hospital of Chinese PLA General Hospital, Sanya, Hainan Province, 572000, People's Republic of China
| | - Jian Guo
- Department of Pulmonary Function Test, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200443, People's Republic of China.
| | - Bing Yan
- Department of Oncology, Hainan Hospital of Chinese PLA General Hospital, Sanya, Hainan Province, 572000, People's Republic of China.
| |
Collapse
|
69
|
Liu H, Hu Y, Qi B, Yan C, Wang L, Zhang Y, Chen L. Network pharmacology and molecular docking to elucidate the mechanism of pulsatilla decoction in the treatment of colon cancer. Front Pharmacol 2022; 13:940508. [PMID: 36003525 PMCID: PMC9393233 DOI: 10.3389/fphar.2022.940508] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 06/28/2022] [Indexed: 01/01/2023] Open
Abstract
Objective: Colon cancer is a malignant neoplastic disease that seriously endangers the health of patients. Pulsatilla decoction (PD) has some therapeutic effects on colon cancer. This study is based on the analytical methods of network pharmacology and molecular docking to study the mechanism of PD in the treatment of colon cancer. Methods: Based on the Traditional Chinese Medicine Systems Pharmacology Database, the main targets and active ingredients in PD were filtered, and then, the colon cancer-related targets were screened using Genecards, OMIM, PharmGKB, and Drugbank databases. Then, the screened drug and disease targets were Venn analyzed to obtain the intersection targets. Cytoscape software was used to construct the “Components–Targets–Pathway” map, and the String database was used to analyze the protein interaction network of the intersecting targets and screen the core targets, and then, the core targets were analyzed using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses. Molecular docking was implemented using AutoDockTools to predict the binding capacity for the core targets and the active components in PD. Results: Sixty-five ingredients containing 188 nonrepetitive targets were screened and 180 potential targets of PD anticolon cancer were identified, including 10 core targets, namely, MAPK1, JUN, AKT1, TP53, TNF, RELA, MAPK14, CXCL8, ESR1, and FOS. The results of GO analysis showed that PD anticolon cancer may be related to cell proliferation, apoptosis, energy metabolism, immune regulation, signal transduction, and other biological processes. The results of KEGG analysis indicated that the PI3K-Akt signaling pathway, MAPK signaling pathway, proteoglycans in cancer, IL-17 signaling pathway, cellular senescence, and TNF signaling pathway were mainly involved in the regulation of tumor cells. We further selected core targets with high degree values as receptor proteins for molecular docking with the main active ingredients of the drug, including MAPK1, JUN, and AKT1. The docking results showed good affinity, especially quercetin. Conclusion: This study preliminarily verified that PD may exert its effect on the treatment of colon cancer through multi-ingredients, multitargets, and multipathways. This will deepen our understanding of the potential mechanisms of PD anticolon cancer and establish a foundation for further basic experimental research.
Collapse
Affiliation(s)
- Huan Liu
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Yuting Hu
- College of Integration Science, Yanbian University, Yanji, China
- *Correspondence: Yuting Hu, ; Liang Chen,
| | - Baoyu Qi
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Chengqiu Yan
- Anorectal Diagnosis and Treatment Center, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Lin Wang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Yiwen Zhang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Liang Chen
- Anorectal Diagnosis and Treatment Center, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
- *Correspondence: Yuting Hu, ; Liang Chen,
| |
Collapse
|
70
|
Safa AR. Drug and apoptosis resistance in cancer stem cells: a puzzle with many pieces. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2022; 5:850-872. [PMID: 36627897 PMCID: PMC9771762 DOI: 10.20517/cdr.2022.20] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 05/10/2022] [Accepted: 05/26/2022] [Indexed: 01/13/2023]
Abstract
Resistance to anticancer agents and apoptosis results in cancer relapse and is associated with cancer mortality. Substantial data have provided convincing evidence establishing that human cancers emerge from cancer stem cells (CSCs), which display self-renewal and are resistant to anticancer drugs, radiation, and apoptosis, and express enhanced epithelial to mesenchymal progression. CSCs represent a heterogeneous tumor cell population and lack specific cellular targets, which makes it a great challenge to target and eradicate them. Similarly, their close relationship with the tumor microenvironment creates greater complexity in developing novel treatment strategies targeting CSCs. Several mechanisms participate in the drug and apoptosis resistance phenotype in CSCs in various cancers. These include enhanced expression of ATP-binding cassette membrane transporters, activation of various cytoprotective and survival signaling pathways, dysregulation of stemness signaling pathways, aberrant DNA repair mechanisms, increased quiescence, autophagy, increased immune evasion, deficiency of mitochondrial-mediated apoptosis, upregulation of anti-apoptotic proteins including c-FLIP [cellular FLICE (FADD-like IL-1β-converting enzyme)-inhibitory protein], Bcl-2 family members, inhibitors of apoptosis proteins, and PI3K/AKT signaling. Studying such mechanisms not only provides mechanistic insights into these cells that are unresponsive to drugs, but may lead to the development of targeted and effective therapeutics to eradicate CSCs. Several studies have identified promising strategies to target CSCs. These emerging strategies may help target CSC-associated drug resistance and metastasis in clinical settings. This article will review the CSCs drug and apoptosis resistance mechanisms and how to target CSCs.
Collapse
Affiliation(s)
- Ahmad R. Safa
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
71
|
Sokolov D, Sharda N, Giri B, Hassan MS, Singh D, Tarasiewicz A, Lohr C, von Holzen U, Kristian T, Waddell J, Reiter RJ, Ahmed H, Banerjee A. Melatonin and andrographolide synergize to inhibit the colospheroid phenotype by targeting Wnt/beta-catenin signaling. J Pineal Res 2022; 73:e12808. [PMID: 35619550 PMCID: PMC9288490 DOI: 10.1111/jpi.12808] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 04/19/2022] [Accepted: 05/23/2022] [Indexed: 11/30/2022]
Abstract
β-catenin signaling, and angiogenesis are associated with colospheroid (CSC), development. CSCs, spheroids derived from colon cancer cells, are responsible for metastasis, drug resistance, and disease recurrence. Whether dysregulating β-catenin and inhibiting angiogenesis reduce CSC growth is unknown. In this study, the molecular mechanism of CSC growth inhibition was evaluated using a novel combination of melatonin (MLT) and andrographolide (AGP). These drugs have anticarcinogenic, antioxidant, and antimetastatic properties. CSCs were obtained from two metastatic colon cancer cell lines (HT29 and HCT-15). The viability and stemness were monitored (FDA propidium iodide staining and immunoblot for CD44, CD133, Nanog, Sox2, and Oct4). The drug combination synergistically diminished stemness via increased reactive oxygen species (ROS) levels, reduced mitochondrial membrane potential and ATP level. MLT + AGP induced cell death by inhibiting β-catenin expression and its downregulatory signals, Cyclin D1, c-Myc. MLT + AGP treated cells exhibited translocation of phospho-β-catenin to the nucleus and dephosphorylated-β-catenin. Downregulation of β-catenin activation and its transcription factors (TCF4 and LEF1) and GTP binding/G-protein related activity were found in the dual therapy. Angiogenic inhibition is consistent with downregulation of VEGF messenger RNA transcripts (VEGF189), phosphorylated VEGF receptor protein expression, matrigel invasion, and capillary tube inhibition. In vivo, the intravenous injection of MLT + AGP slowed HT29 metastatic colon cancer. Histopathology indicated significant reduction in microvascular density and tumor index. Immunohistochemistry for caspase 7, and β-catenin found increased apoptosis and downregulation of β-catenin signals. The mechanism(s) of decreased colospheroids growth were the inhibition of the Wnt/β-catenin pathway. Our results provide a rationale for using MLT in combination with AGP for the inhibition of CRCs.
Collapse
Affiliation(s)
- Daniil Sokolov
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, U.S.A
| | - Neha Sharda
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, U.S.A
| | - Banabihari Giri
- Division of Virology and Immunology, Maryland Department of Health, Baltimore, Maryland, U.S.A
| | - Md Sazzad Hassan
- Department of Surgery, Indiana University School of Medicine, South Bend, IN, U.S.A
- Harper Cancer Research Institute, South Bend, IN, U.S.A
| | - Damandeep Singh
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, U.S.A
| | - Agnieszka Tarasiewicz
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, U.S.A
| | - Charity Lohr
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, U.S.A
| | - Urs von Holzen
- Department of Surgery, Indiana University School of Medicine, South Bend, IN, U.S.A
- Harper Cancer Research Institute, South Bend, IN, U.S.A
- Goshen Center for Cancer Care, Goshen, Goshen, IN, U.S.A
- University of Basel, Basel, Switzerland
| | - Tibor Kristian
- VAMHCS, University of Maryland School of Medicine, Baltimore, Maryland, U.S.A
- Department of Anesthesiology and the Center for Shock, Trauma, and Anesthesiology Research (STAR)
| | - Jaylyn Waddell
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, U.S.A
| | - Russel J. Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX 78229, U.S.A
| | | | - Aditi Banerjee
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, U.S.A
- Corresponding author: Department of Pediatrics, University of Maryland School of Medicine, Bressler Research Building, 13-043, 655 W. Baltimore Street, Baltimore, MD 21201, Voice: (410) 706-1772, Fax: (410) 328-1072,
| |
Collapse
|
72
|
Cui G, Li G, Pang Z, Florholmen J, Goll R. The presentation and regulation of the IL-8 network in the epithelial cancer stem-like cell niche in patients with colorectal cancer. Biomed Pharmacother 2022; 152:113252. [PMID: 35687912 DOI: 10.1016/j.biopha.2022.113252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/02/2022] [Accepted: 06/02/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Accumulative evidence suggests that the biological behavior of cancer stem-like cells (CSCs) is regulated by their surrounding niche, in which cytokines function as one of the main mediators for the interaction between CSCs and their microenvironment in the colorectal cancer (CRC). METHODS We characterized the presentation of CSCs and the interleukin (IL)- 8 network in the adenoma/CRC epithelium using quantitative real-time PCR (q-PCR), immunohistochemistry (IHC) and double immunofluorescence. In addition, the capacity of IL-1β to stimulate epithelial IL-8 production in colon cancer Caco-2 cells was examined in vitro and the IL-8 product was measured by enzyme-linked immunosorbent assay (ELISA). RESULTS IHC observation showed increased expression of both CSCs and IL-8 in the adenoma and CRC epithelium, and q-PCR results revealed that increased expression of IL-1β transcript was strongly correlated with increased IL-8 transcript levels in both adenoma and CRC tissues. Double immunofluorescence images demonstrated the coexpression of the IL-8 receptors IL-8RA and IL-8RB with LGR5 labeled CSCs in CRC tissue sections. Consistently, in vitro experiments showed that coculture of Caco-2 cells with IL-1β at concentrations of 1, 5, 10 and 20 ng/ml resulted in a dose-dependent release of IL-8, which could be specifically inhibited by cotreatment with the IL-1β receptor antagonist. CONCLUSIONS These results demonstrate activation of the IL-8 network in the niche of CSCs from the precancerous adenoma stage to the CRC stage, which is potentially stimulated by IL-1β in CRC cells.
Collapse
Affiliation(s)
- Guanglin Cui
- Research Group of Gastrointestinal Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Faculty of Health Science, Nord University, Campus Levanger, Levanger, Norway.
| | - Gui Li
- Research Group of Gastrointestinal Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhigang Pang
- Research Group of Gastrointestinal Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jon Florholmen
- Department of Gastroenterology, University Hospital of North Norway, University of Tromsø, Tromsø, Norway
| | - Rasmus Goll
- Department of Gastroenterology, University Hospital of North Norway, University of Tromsø, Tromsø, Norway
| |
Collapse
|
73
|
Wumei Pill Ameliorates AOM/DSS-Induced Colitis-Associated Colon Cancer through Inhibition of Inflammation and Oxidative Stress by Regulating S-Adenosylhomocysteine Hydrolase- (AHCY-) Mediated Hedgehog Signaling in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4061713. [PMID: 35927991 PMCID: PMC9345734 DOI: 10.1155/2022/4061713] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 07/06/2022] [Indexed: 02/07/2023]
Abstract
Wumei Pill (WMP) is a traditional Chinese herbal formulation and widely used to treat digestive system diseases in clinical. S-Adenosylhomocysteine hydrolase (AHCY) can catalyze the hydrolysis of S-adenosylhomocysteine to adenosine and homocysteine in living organisms, and its abnormal expression is linked to the pathogenesis of many diseases including colorectal cancer (CRC). A previous study reported that WMP could prevent CRC in mice; however, the underlying mechanisms especially the roles of AHCY in WMP-induced anti-CRC remain largely unknown. Here, we investigated the regulatory roles and potential mechanisms of AHCY in WMP-induced anti-CRC. WMP notably alleviated the azoxymethane/dextran sulfate sodium- (AOM/DSS-) induced colitis-associated colon cancer (CAC) in mice. Besides, WMP inhibited the inflammation and oxidative stress in AOM/DSS-induced CAC mice. AHCY was high expression in clinical samples of colon cancer compared to the adjacent tissues. WMP inhibited the AHCY expression in AOM/DSS-induced CAC mice. An in vitro study found that AHCY overexpression induced cell proliferation, colony formation, invasion, and tumor angiogenesis, whereas its knockdown impaired its oncogenic function. AHCY overexpression enhanced, while its knockdown weakened the inflammation and oxidative stress in colon cancer cells. Interestingly, WMP potently suppressed the hedgehog (Hh) signaling in AOM/DSS-induced CAC mice. A further study showed that AHCY overexpression activated the Hh signaling while AHCY knockdown inactivated the Hh signaling. Moreover, activation of the Hh signaling reversed the effect of AHCY silencing on inflammation and oxidative stress in vitro. In conclusion, WMP alleviated the AOM/DSS-induced CAC through inhibition of inflammation and oxidative stress by regulating AHCY-mediated hedgehog signaling in mice. These findings uncovered a potential molecular mechanism underlying the anti-CAC effect of WMP and suggested WMP as a promising therapeutic candidate for CRC.
Collapse
|
74
|
Cao W, Zhang B, Liu Y. Expression of Long Nonencoding Ribonucleic Acid SNHG20 in Colon Cancer Tissue in Its Influences on Chemotherapeutic Sensitivity of Colon Cancer Cells. BIOMED RESEARCH INTERNATIONAL 2022; 2022:4752782. [PMID: 35915794 PMCID: PMC9338858 DOI: 10.1155/2022/4752782] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/17/2022] [Accepted: 05/25/2022] [Indexed: 11/17/2022]
Abstract
Noncoding RNA (ncRNA) is a kind of RNA that plays a key role in a variety of biological processes, illnesses, and tumours despite the fact that it cannot be translated into proteins. The HT29 colon cancer cell line was utilized to create a 5-FU drug-resistant cell strain (control group), a lentivirus SNHG20 carrier (OE-SNHG20 group), and an SNHG20 shRNA carrier (SNHG20 shRNA carrier group) (SE-SNHG20 group). To determine the expression of cell SNHG20, a real-time quantitative reverse transcription-polymerase chain reaction (RT-qPCR) was utilized, and cholecystokinin-octapeptide (CCK-8) was used to detect the difference in 5-FU inhibitory concentration 50. The goal of the study was to see how variations in long nonencoding ribonucleic acid (lncRNA) SNHG20 expression affect colon cancer cell 5-fluorouracil (5-FU) chemotherapeutic sensitivity by collecting colon cancer and normal para cancer tissues and analysing the differences in SNHG20 expression. The ability of cell cladogenesis was tested using platform cladogenesis. Cell apoptosis was detected using flow cytometry. Western blots revealed the presence of protein phosphatidylinositol kinase (PI3K), protein kinase B (AKT), caspase-3, e-cadherin, and matrix metalloproteinase 9 (MMP-9) enzymes. The findings revealed that SNHG20 expression was considerably upregulated (P < 0.05) in colon cancer tissue and 5-FU drug-resistant colon cancer cells. Cell 5-FU IC50, cell cladogenesis, cell survival rate, and MMP-9, P-PI3K, and P-AKT expression were all significantly improved. Cell apoptosis and expressions of E-cadherin and caspase-3, on the other hand, were considerably decreased (P < 0.05). Cell 5-FU IC50, cell cladogenesis, cell survival rate, and the expressions of MMP-9, P-PI3K, and P-AKT were all significantly lower in the SE-SNHG20 group, although cell apoptosis and the expressions of E-cadherin and caspase-3 were significantly higher (P < 0.05). The results revealed that lncRNA SNHG20 could inhibit the chemotherapeutic sensitivity of colon cancer cells to 5-FU by regulating PI3K/AKT pathways. The inhibition of lncRNA SNHG20 expression could promote the apoptosis and proliferation of 5-FU-resistant colon cancer cells.
Collapse
Affiliation(s)
- Wenbin Cao
- North China University of Science and Technology Affiliated Hospital, Tangshan, 063000 Hebei, China
| | - Bo Zhang
- North China University of Science and Technology Affiliated Hospital, Tangshan, 063000 Hebei, China
| | - Yang Liu
- North China University of Science and Technology Affiliated Hospital, Tangshan, 063000 Hebei, China
| |
Collapse
|
75
|
Novoa Díaz MB, Martín MJ, Gentili C. Tumor microenvironment involvement in colorectal cancer progression via Wnt/β-catenin pathway: Providing understanding of the complex mechanisms of chemoresistance. World J Gastroenterol 2022; 28:3027-3046. [PMID: 36051330 PMCID: PMC9331520 DOI: 10.3748/wjg.v28.i26.3027] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 04/29/2022] [Accepted: 06/20/2022] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) continues to be one of the main causes of death from cancer because patients progress unfavorably due to resistance to current therapies. Dysregulation of the Wnt/β-catenin pathway plays a fundamental role in the genesis and progression of several types of cancer, including CRC. In many subtypes of CRC, hyperactivation of the β-catenin pathway is associated with mutations of the adenomatous polyposis coli gene. However, it can also be associated with other causes. In recent years, studies of the tumor microenvironment (TME) have demonstrated its importance in the development and progression of CRC. In this tumor nest, several cell types, structures, and biomolecules interact with neoplastic cells to pave the way for the spread of the disease. Cross-communications between tumor cells and the TME are then established primarily through paracrine factors, which trigger the activation of numerous signaling pathways. Crucial advances in the field of oncology have been made in the last decade. This Minireview aims to actualize what is known about the central role of the Wnt/β-catenin pathway in CRC chemoresistance and aggressiveness, focusing on cross-communication between CRC cells and the TME. Through this analysis, our main objective was to increase the understanding of this complex disease considering a more global context. Since many treatments for advanced CRC fail due to mechanisms involving chemoresistance, the data here exposed and analyzed are of great interest for the development of novel and effective therapies.
Collapse
Affiliation(s)
- María Belén Novoa Díaz
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS)-INBIOSUR (CONICET-UNS), Bahía Blanca 8000, Argentina
| | - María Julia Martín
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS)-INBIOSUR (CONICET-UNS), Bahía Blanca 8000, Argentina
- Departamento de Química, Universidad Nacional del Sur (UNS)-INQUISUR (CONICET-UNS), Bahía Blanca 8000, Argentina
| | - Claudia Gentili
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS)-INBIOSUR (CONICET-UNS), Bahía Blanca 8000, Argentina
| |
Collapse
|
76
|
Hall DCN, Benndorf RA. Aspirin sensitivity of PIK3CA-mutated Colorectal Cancer: potential mechanisms revisited. Cell Mol Life Sci 2022; 79:393. [PMID: 35780223 PMCID: PMC9250486 DOI: 10.1007/s00018-022-04430-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/01/2022] [Accepted: 06/14/2022] [Indexed: 11/30/2022]
Abstract
PIK3CA mutations are amongst the most prevalent somatic mutations in cancer and are associated with resistance to first-line treatment along with low survival rates in a variety of malignancies. There is evidence that patients carrying PIK3CA mutations may benefit from treatment with acetylsalicylic acid, commonly known as aspirin, particularly in the setting of colorectal cancer. In this regard, it has been clarified that Class IA Phosphatidylinositol 3-kinases (PI3K), whose catalytic subunit p110α is encoded by the PIK3CA gene, are involved in signal transduction that regulates cell cycle, cell growth, and metabolism and, if disturbed, induces carcinogenic effects. Although PI3K is associated with pro-inflammatory cyclooxygenase-2 (COX-2) expression and signaling, and COX-2 is among the best-studied targets of aspirin, the mechanisms behind this clinically relevant phenomenon are still unclear. Indeed, there is further evidence that the protective, anti-carcinogenic effect of aspirin in this setting may be mediated in a COX-independent manner. However, until now the understanding of aspirin's prostaglandin-independent mode of action is poor. This review will provide an overview of the current literature on this topic and aims to analyze possible mechanisms and targets behind the aspirin sensitivity of PIK3CA-mutated cancers.
Collapse
Affiliation(s)
- Daniella C N Hall
- Department of Clinical Pharmacy and Pharmacotherapy, Institute of Pharmacy, Martin-Luther-University Halle-Wittenberg, Kurt-Mothes-Str. 3, 06120, Halle (Saale), Germany
| | - Ralf A Benndorf
- Department of Clinical Pharmacy and Pharmacotherapy, Institute of Pharmacy, Martin-Luther-University Halle-Wittenberg, Kurt-Mothes-Str. 3, 06120, Halle (Saale), Germany.
| |
Collapse
|
77
|
Lazer LM, Kesavan Y, Gor R, Ramachandran I, Pathak S, Narayan S, Anbalagan M, Ramalingam S. Targeting colon cancer stem cells using novel doublecortin like kinase 1 antibody functionalized folic acid conjugated hesperetin encapsulated chitosan nanoparticles. Colloids Surf B Biointerfaces 2022; 217:112612. [PMID: 35738074 DOI: 10.1016/j.colsurfb.2022.112612] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 05/25/2022] [Accepted: 06/02/2022] [Indexed: 01/05/2023]
Abstract
The cancer stem cell (CSC) hypothesis is an evolving oncogenesis concept. CSCs have a distinct ability to self-renew themselves and also give rise to a phenotypically diverse population of cells. Targeting CSCs represents a promising strategy for cancer treatment. Plant-derived compounds are potent in restricting the expansion of CSCs. DCLK1 has been already reported as a colon CSC specific marker. Nanoparticles can effectively inhibit multiple types of CSCs by targeting specific markers. We have synthesized DCLK1 functionalized folic acid conjugated hesperetin encapsulated chitosan nanoparticles (CFH-DCLK1), specifically to target CSCs. In this regard, we have performed proliferation assay, colony formation assay, cell migration assay, apoptosis assay, flow cytometry analysis, real-time RT- PCR and western blot analyses to determine the effect of CFH-DCLK1 and CFH nanoparticles in HCT116-colon cancer cells. In our study, we have determined the median inhibitory concentration (IC50) of CFH (47.8 µM) and CFH-DCLK1 (4.8 µM) nanoparticles in colon cancer cells. CFH-DCLK1 nanoparticles induced apoptosis and inhibited the migration and invasion of colon cancer cells. Real time PCR and western blot results have demonstrated that the treatment with CFH-DCLK1 nanoparticles significantly reduced the expression of CSC markers such as DCLK1, STAT1 and NOTCH1 compared to the CFH alone in HCT116 colon cancer cells. Finally, in the 3D spheroid model, CFH-DCLK1 nanoparticles significantly inhibited the colonosphere growth. Overall, our results highlight the effectiveness of CFH-DCLK1 nanoparticles in targeting the colon cancer cells and CSCs. This study would lead to the development of therapies targeting both cancer cells and CSCs simultaneously using nanoformulated drugs, which could bring changes in the current cancer treatment strategies.
Collapse
Affiliation(s)
- Lizha Mary Lazer
- Department of Genetic Engineering, School of Bio-Engineering, SRM Institute of Science and Technology, Kattankulathur, Kanchipuram 603203, Tamil Nadu, India
| | - Yasodha Kesavan
- Department of Genetic Engineering, School of Bio-Engineering, SRM Institute of Science and Technology, Kattankulathur, Kanchipuram 603203, Tamil Nadu, India
| | - Ravi Gor
- Department of Genetic Engineering, School of Bio-Engineering, SRM Institute of Science and Technology, Kattankulathur, Kanchipuram 603203, Tamil Nadu, India
| | - Ilangovan Ramachandran
- Department of Endocrinology, Dr. ALM PG Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai 600113, Tamil Nadu, India
| | - Surajit Pathak
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Tamilnadu, 603103, India
| | - Shoba Narayan
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Tamilnadu, 603103, India
| | - Muralidharan Anbalagan
- Structural & Cellular Biology, Pre-clinical small animal Imaging Facility, Tulane University School of Medicine, New Orleans, Louisiana, 70112, USA
| | - Satish Ramalingam
- Department of Genetic Engineering, School of Bio-Engineering, SRM Institute of Science and Technology, Kattankulathur, Kanchipuram 603203, Tamil Nadu, India.
| |
Collapse
|
78
|
Wu L, Sun S, Qu F, Liu X, Sun M, Pan Y, Zheng Y, Su G. ASCL2 Affects the Efficacy of Immunotherapy in Colon Adenocarcinoma Based on Single-Cell RNA Sequencing Analysis. Front Immunol 2022; 13:829640. [PMID: 35774798 PMCID: PMC9237783 DOI: 10.3389/fimmu.2022.829640] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 05/06/2022] [Indexed: 11/13/2022] Open
Abstract
Colon adenocarcinoma (COAD) is one of the leading causes of cancer-associated deaths worldwide. Patients with microsatellite instability-high (MSI-H) tumors were shown to highly benefit from immune checkpoint inhibitors (ICIs) than patients with microsatellite stable (MSS) tumors. Furthermore, the infiltration of immune cells and the expression of cancer stem cells (CSCs) in COAD were associated with the anti-tumor immune response. However, the potential mechanisms showing the relationship between microsatellite instability and CSCs or tumor-infiltrating immune cells (TIICs) have not been elucidated. Accumulating evidence reveals that achaete-scute family bHLH transcription factor 2 (ASCL2) plays a crucial role in the initiation and progression of COAD and drug resistance. However, the specific biological functions of ASCL2 in COAD remain unknown. In this study, we performed weighted gene co-expression network analysis (WGCNA) between MSS and MSI-H subsets of COAD. The results revealed that ASCL2 was a potential key candidate in COAD. Subsequently, the single-cell RNA-seq revealed that ASCL2 was positively associated with CSCs. Further, ASCL2 was shown to indirectly affect tumor immune cell infiltration by negatively regulating the expression of DUSP4. Finally, we inferred that the immunotherapy-sensitive role of ASCL2/DUSP4 axis on COAD is partly attributed to the activation of WNT/β-catenin pathway. In conclusion, this study revealed that ASCL2 was positively correlated to CSCs and tumor immune infiltration in COAD. Therefore, ASCL2 is a promising predictor of clinical responsiveness to anti-PD-1/PD-L1 therapy in COAD.
Collapse
Affiliation(s)
- Lei Wu
- Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
- Research Center of Translational Medicine, Jinan Central Hospital, Shandong University, Jinan, China
- Department of Oncology, Central Hospital Affiliated to Shandong First Medical University, Zhuhai, China
| | - Shengnan Sun
- Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
- Research Center of Translational Medicine, Jinan Central Hospital, Shandong University, Jinan, China
| | - Fei Qu
- Department of Pathology, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xiuxiu Liu
- Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
- Research Center of Translational Medicine, Jinan Central Hospital, Shandong University, Jinan, China
| | - Meili Sun
- Department of Oncology, Central Hospital Affiliated to Shandong First Medical University, Zhuhai, China
| | - Ying Pan
- Department of Oncology, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated With Jinan University), Zhuhai, China
| | - Yan Zheng
- Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
- *Correspondence: Yan Zheng, ; Guohai Su,
| | - Guohai Su
- Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
- Research Center of Translational Medicine, Jinan Central Hospital, Shandong University, Jinan, China
- *Correspondence: Yan Zheng, ; Guohai Su,
| |
Collapse
|
79
|
Shang Y, Jiang T, Ran L, Hu W, Wu Y, Ye J, Peng Z, Chen L, Wang R. TET2-BCLAF1 transcription repression complex epigenetically regulates the expression of colorectal cancer gene Ascl2 via methylation of its promoter. J Biol Chem 2022; 298:102095. [PMID: 35660018 PMCID: PMC9251787 DOI: 10.1016/j.jbc.2022.102095] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 05/17/2022] [Accepted: 05/18/2022] [Indexed: 11/08/2022] Open
Abstract
Ascl2 has been shown to be involved in tumorigenesis in colorectal cancer (CRC), although its epigenetic regulatory mechanism is largely unknown. Here, we found that methylation of the Ascl2 promoter (bp -1670 ∼ -1139) was significantly increased compared to the other regions of the Ascl2 locus in CRC cells and was associated with elevated Ascl2 mRNA expression. Furthermore, we found that promoter methylation was predictive of CRC patient survival after analyzing DNA methylation data, RNA-Seq data, and clinical data of 410 CRC patient samples from the MethHC database, the MEXPRESS database, and the Cbioportal website. Using the established TET methylcytosine dioxygenase 2 (TET2) knockdown and ectopic TET2 catalytic domain–expression cell models, we performed glucosylated hydroxymethyl–sensitive quatitative PCR (qPCR), real-time PCR, and Western blot assays to further confirm that hypermethylation of the Ascl2 promoter, and elevated Ascl2 expression in CRC cells was partly due to the decreased expression of TET2. Furthermore, BCLAF1 was identified as a TET2 interactor in CRC cells by LC-MS/MS, coimmunoprecipitation, immunofluorescence colocalization, and proximity ligation assays. Subsequently, we found the TET2–BCLAF1 complex bound to multiple elements around CCGG sites at the Ascl2 promoter and further restrained its hypermethylation by inducing its hydroxymethylation using chromatin immunoprecipitation-qPCR and glucosylated hydroxymethyl-qPCR assays. Finally, we demonstrate that TET2-modulated Ascl2-targeted stem gene expression in CRC cells was independent of Wnt signaling. Taken together, our data suggest an additional option for inhibiting Ascl2 expression in CRC cells through TET2–BCLAF1–mediated promoter methylation, Ascl2-dependent self-renewal of CRC progenitor cells, and TET2–BCLAF1–related CRC progression.
Collapse
Affiliation(s)
- Yangyang Shang
- Institute of Gastroenterology of PLA, Southwest Hospital, Army Medical University (Third Military Medical University) Chongqing 400038, China
| | - Tao Jiang
- Institute of Gastroenterology of PLA, Southwest Hospital, Army Medical University (Third Military Medical University) Chongqing 400038, China
| | - Lijian Ran
- Institute of Gastroenterology of PLA, Southwest Hospital, Army Medical University (Third Military Medical University) Chongqing 400038, China
| | - Wenjing Hu
- Institute of Gastroenterology of PLA, Southwest Hospital, Army Medical University (Third Military Medical University) Chongqing 400038, China
| | - Yun Wu
- Institute of Gastroenterology of PLA, Southwest Hospital, Army Medical University (Third Military Medical University) Chongqing 400038, China
| | - Jun Ye
- Department of Gastroenterology of 958 Hospital, Army Medical University (Third Military Medical University) Chongqing 400038, China
| | - Zhihong Peng
- Institute of Gastroenterology of PLA, Southwest Hospital, Army Medical University (Third Military Medical University) Chongqing 400038, China
| | - Lei Chen
- Institute of Gastroenterology of PLA, Southwest Hospital, Army Medical University (Third Military Medical University) Chongqing 400038, China
| | - Rongquan Wang
- Institute of Gastroenterology of PLA, Southwest Hospital, Army Medical University (Third Military Medical University) Chongqing 400038, China.
| |
Collapse
|
80
|
Cancer stem cell marker expression and methylation status in patients with colorectal cancer. Oncol Lett 2022; 24:231. [PMID: 35720495 PMCID: PMC9185140 DOI: 10.3892/ol.2022.13352] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 03/29/2022] [Indexed: 11/21/2022] Open
Abstract
The number of individuals diagnosed with colorectal cancer (CRC) has been on an alarming upward trajectory over the past decade. In some countries, this cancer represents one of the most frequently diagnosed types of neoplasia. Therefore, it is an important demand to study the pathology underlying this disease to gain insights into the mechanism of resistance to treatment. Resistance of tumors to chemotherapy and tumor aggressiveness have been associated with a minor population of neoplastic cells, which are considered to be responsible for tumor recurrence. These types of neoplastic cells are known as cancer stem cells, which have been previously reported to serve an important role in pathogenesis of this malignant disease. Slovakia has one of the highest incidence rates of CRC worldwide. In the present study, the aim was to classify the abundance of selected stem cell markers (CD133, CD166 and Lgr5) in CRC tumors using flow cytometry. In addition, the methylation status of selected genomic regions of CRC biomarkers (ADAMTS16, MGMT, PROM1 (CD133), LGR5 and ALCAM) was investigated by pyrosequencing in a cohort of patients from Martin University Hospital, Martin, Slovakia. Samples from both primary tumors and metastatic tumors were tested. Analysis of DNA methylation in the genomic regions of indicated five CRC biomarkers was also performed, which revealed the highest levels of methylation in the A disintegrin and metalloproteinase with thrombospondin motifs 16 and O6-methyguanine-DNA methyl transferase genes, whereas the lowest levels of methylation were found in genes expressing prominin-1, leucine-rich repeat-containing G-protein-coupled receptor 5 and activated leukocyte cell adhesion molecule. Furthermore, tumor tissues from metastases showed significantly higher levels of CD133+ cells compared with that in primary tumors. Higher levels of CD133+ cells correlated with TNM stage and the invasiveness of CRC into the lymphatic system. Although relatively small number of samples was processed, CD133 marker was consider to be important marker in pathology of CRC.
Collapse
|
81
|
Pączek S, Łukaszewicz-Zając M, Mroczko B. Granzymes-Their Role in Colorectal Cancer. Int J Mol Sci 2022; 23:ijms23095277. [PMID: 35563668 PMCID: PMC9104098 DOI: 10.3390/ijms23095277] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/06/2022] [Accepted: 05/07/2022] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer (CRC) is among the most common malignancies worldwide. CRC is considered a heterogeneous disease due to various clinical symptoms, biological behaviours, and a variety of mutations. A number of studies demonstrate that as many as 50% of CRC patients have distant metastases at the time of diagnosis. However, despite the fact that social and medical awareness of CRC has increased in recent years and screening programmes have expanded, there is still an urgent need to find new diagnostic tools for early detection of CRC. The effectiveness of the currently used classical tumour markers in CRC diagnostics is very limited. Therefore, new proteins that play an important role in the formation and progression of CRC are being sought. A number of recent studies show the potential significance of granzymes (GZMs) in carcinogenesis. These proteins are released by cytotoxic lymphocytes, which protect the body against viral infection as well specific signalling pathways that ultimately lead to cell death. Some studies suggest a link between GZMs, particularly the expression of Granzyme A, and inflammation. This paper summarises the role of GZMs in CRC pathogenesis through their involvement in the inflammatory process. Therefore, it seems that GZMs could become the focus of research into new CRC biomarkers.
Collapse
Affiliation(s)
- Sara Pączek
- Department of Biochemical Diagnostics, Medical University in Bialystok, 15-269 Bialystok, Poland; (M.Ł.-Z.); (B.M.)
- Correspondence: ; Tel.: +48-85-831-8587
| | - Marta Łukaszewicz-Zając
- Department of Biochemical Diagnostics, Medical University in Bialystok, 15-269 Bialystok, Poland; (M.Ł.-Z.); (B.M.)
| | - Barbara Mroczko
- Department of Biochemical Diagnostics, Medical University in Bialystok, 15-269 Bialystok, Poland; (M.Ł.-Z.); (B.M.)
- Department of Neurodegeneration Diagnostics, Medical University in Bialystok, 15-269 Bialystok, Poland
| |
Collapse
|
82
|
Wu L, Li S, Shu P, Liu Q. Effect of miR-488 on Colon Cancer Biology and Clinical Applications. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:2138954. [PMID: 35571741 PMCID: PMC9098289 DOI: 10.1155/2022/2138954] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/24/2022] [Accepted: 03/28/2022] [Indexed: 11/22/2022]
Abstract
Objective To explore the expression levels of miR-488, miR-29c-3p, and growth differentiation factor 15 (GDF15) in colon cancer tissue and analyze their relationship with clinicopathological features in patients with colon cancer. Methods The study was conducted from November 2012 to November 2020. A total of 200 patients with colon cancer were treated in our hospital during this period. During the operation, the colon cancer tissues and the adjacent tissues whose distance from the cancer tissues were more than 5 cm were collected, and the expression levels of miR-488, miR-29c-3p, and GDF15 mRNA in colon cancer tissues were detected by qRT-PCR (real-time fluorescence quantitative). The relationship between them and the clinicopathological features and prognosis of patients with colon cancer were analyzed and discussed. Results The level of miR-488 in colon cancer tissues was lower than that in adjacent tissues, but the levels of miR-29c-3p and GDF15 mRNA in colon cancer tissues were higher than those in adjacent tissues (P < 0.05). Compared with paracancerous tissues, the expression rates of miR-29c-3p and GDF15 protein were higher in colon cancer tissues (P < 0.05). There was no difference in age, sex, tumor location, and tumor diameter between high expression of miR-488 group and low expression of miR-488 group (P > 0.05). The degree of differentiation, depth of invasion, TNM stage, lymph node metastasis, and other factors have a direct impact on the level of miR-488 and the expression of miR-29c-3p (P < 0.05). The depth of invasion, TNM stage, and lymph node metastasis could affect the expression of GDF15 in patients with colon cancer (P < 0.05). Conclusion miR-488, miR-29c-3p, and GDF15 in colon cancer tissue are related to the clinicopathological features of patients in varying degrees and may become markers after early warning of colon cancer, which can provide effective guidance for clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Liangqin Wu
- Department of Gastroenterology, Anhui No.2 Provincial People's Hospital, Hefei, Anhui, China
| | - Songguo Li
- Department of Pathology, Anhui No.2 Provincial People's Hospital, Hefei, Anhui, China
| | - Peng Shu
- Department of Gastroenterology, Anhui No.2 Provincial People's Hospital, Hefei, Anhui, China
| | - Qian Liu
- Department of Pathology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Nanjing, Jiangsu, China
| |
Collapse
|
83
|
Anthocyanins in Red Jasmine Rice (Oryza sativa L.) Extracts and Efficacy on Inhibition of Herpes Simplex Virus, Free Radicals and Cancer Cell. Nutrients 2022; 14:nu14091905. [PMID: 35565872 PMCID: PMC9101121 DOI: 10.3390/nu14091905] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 04/26/2022] [Accepted: 04/28/2022] [Indexed: 12/10/2022] Open
Abstract
Rice is one of the most important food crops in many countries, with nutritional value and health benefits. In this study, the ethanolic and aqueous extracts of red jasmine rice from Chiang Mai, Thailand were examined for their anthocyanins and phenolic contents. The antioxidant and antiviral activity against herpes simplex virus type 1 (HSV-1) and type 2 (HSV-2), as well as anticancer activity, were investigated. The total anthocyanins content of 708.03 ± 11.56 mg Cy-3-glc equivalent/g extract, determined from the ethanolic extract, was higher than the aqueous extract. However, the aqueous extract showed the highest total phenolic compound of 81.91 ± 0.51 mg GAE/g extract. In addition, the ethanolic extract demonstrated higher antioxidant activity than aqueous extract using DPPH, ABTS, and FRAP assays by 28.91 ± 3.26 mg GAE/g extract, 189.45 ± 11.58 mg 24 TEAC/g extract, and 3292.46 ± 259.64 g FeSO4/g extract, respectively. In the antiviral assay, it was found that the ethanolic extract of red jasmine rice could inhibit HSV-1 more effectively than HSV-2 when treated before, during, and after the viral attachment on Vero cells, with 50% effective doses of 227.53 ± 2.41, 189.59 ± 7.76, and 192.62 ± 2.40 µg/mL, respectively. The extract also demonstrated the highest reduction of HSV-1 particles at 4 h after treatment and the inhibition of HSV-1 replication. The ethanolic extract exhibited a higher toxicity level than the aqueous extract, as well as the potential to induce DNA fragmentation by intrinsic and extrinsic apoptosis pathways on the Caco-2 cells. These findings suggest that red jasmine rice extract demonstrates nutritional value and biological activity on HSV, free radicals, and cancer cell inhibition.
Collapse
|
84
|
In vivo antitumor activity of Euphorbia lathyris ethanol extract in colon cancer models. Biomed Pharmacother 2022; 149:112883. [PMID: 35692123 DOI: 10.1016/j.biopha.2022.112883] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/09/2022] [Accepted: 03/23/2022] [Indexed: 11/20/2022] Open
Abstract
Euphorbia lathyris seeds have been used to treat various medical conditions. We previously reported that ethanolic extract from the defatted seed of Euphorbia lathyris (EE) (variety S3201) possesses a potent in vitro antitumor activity against colon cancer (CRC) cell lines. However, the effects of EE on CRC in vivo models and its possible preventive activity have not been elucidated. The aim of this study is to develop an in vivo study to corroborate its efficacy. For this purpose, two tumor induction models have been developed. In orthotopic xenograft model, it has been shown that EE reduces tumor size without hematological toxicity. The ethanolic extract induced an intense apoptosis in tumors mediated by caspase 3. Using the Azoxymethane/Dextran Sulfate Sodium model, a reduction of dysplastic polyps has been demonstrated, showing its preventive power. Furthermore, EE promoted the presence of an eubiotic microbiotal environment in the mucosa of the colon and induced an increase in antioxidant enzyme activity. This fact was accompanied by a modulation of cytokine expression that could be related to its protective mechanism. Therefore, although further experiments will be necessary to determine its applicability in the treatment of CRC, ES could be a new prevention strategy as well as treatment for this type of tumor, being a powerful candidate for future clinical trials.
Collapse
|
85
|
Kim J, Lee S, Sun R, Kim J. Juglone and KPT6566 Suppress the Tumorigenic Potential of CD44+CD133+ Tumor-Initiating Caco-2 Cells In Vitro and In Vivo. Front Cell Dev Biol 2022; 10:861045. [PMID: 35433695 PMCID: PMC9006057 DOI: 10.3389/fcell.2022.861045] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/14/2022] [Indexed: 12/11/2022] Open
Abstract
Pin1, a cis/trans isomerase of peptidyl-prolyl peptide bonds, plays a crucial role in the pathogenesis of many human cancers. Although chemical inhibitors of Pin1 show potent antitumor therapeutic properties against various cancers, their effect on colorectal cancer, especially colorectal tumor-initiating cells, remains unknown. Here, we investigated the effect of Juglone and KPT6566 on Caco-2 cells and tumor-initiating Caco-2 cells. Juglone and KPT6566 inhibited cell growth and colony formation, and induced apoptosis of Caco-2 cells. We also found that Juglone and KPT6566 downregulated expression of G1-phase-specific cyclins and cyclin-dependent kinases in a time-dependent manner, consistent with suppression of Caco-2 cell proliferation and colony formation. Although tumor-initiating cells are thought to be responsible for resistance to traditional chemotherapeutic drugs, our experiments demonstrate that Juglone or KPT6566 kill both tumor-initiating and non-tumor-initiating Caco-2 cells with equal or similar efficacy. Finally, when CD44+CD133+ tumor-initiating Caco-2 cells were injected into NSG mice, Juglone or KPT6566 led to a meaningful reduction in tumor volume and mass compared with tumors isolated from mice that received control treatment. Overall, these results indicate that chemical Pin1 inhibitors may be a valuable therapeutic option against colorectal tumor-initiating cancer cells.
Collapse
|
86
|
Supruniuk K, Czarnomysy R, Muszyńska A, Radziejewska I. Anti-cancer effects of pyrazole-platinum(II) complexes combined with anti-MUC1 monoclonal antibody versus monotherapy in DLD-1 and HT-29 colon cancer cells. Transl Oncol 2022; 18:101348. [PMID: 35121220 PMCID: PMC8818584 DOI: 10.1016/j.tranon.2022.101348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 10/26/2022] Open
Abstract
The membrane-bound MUC1 mucin is overexpressed and aberrantly glycosylated in many epithelium origin cancers. One of the promising strategies in cancer therapy is combining monoclonal antibodies against cancer related antigens, like MUC1, with chemotherapeutics. In the study we evaluated the potency of cisplatin (cisPt), two pyrazole-platinum(II) complexes PtPz4, PtPz6, and anti-MUC1 mAb applied as monotherapy, as well as the chemotherapeutics administrated with antibody, towards apoptotic response and cancer-related carbohydrate antigens (TACAs) in DLD-1 and HT-29 colon cancer cells. To assess the impact of the tested compounds on the examined factors flow cytometry, RT-PCR, Western blotting and ELISA were utilized. The combined therapy was more potent than monotherapy towards Bcl-2, Bid, caspases and TACAs of both cell lines. Combined therapy applied in DLD-1 cells induced apoptosis, was more effective than monotherapy in relation to p53, Bcl-xL, Bax, and Bim. In HT-29 cells, anti-MUC1 administrated with the drugs was more potent than monotherapy towards Bad. The proposed anti-MUC1/cisPt and pyrazole-platinum(II) complexes PtPz4, PtPz6 combined therapy may be promising anti-colon cancer therapy.
Collapse
Affiliation(s)
- Katarzyna Supruniuk
- Department of Medical Chemistry, Medical University of Białystok, ul. Mickiewicza 2a, Białystok 15- 222, Poland
| | - Robert Czarnomysy
- Department of Biotechnology, Medical University of Białystok, ul. Kilińskiego 1, Białystok 15-089, Poland
| | - Anna Muszyńska
- Department of Biotechnology, Medical University of Białystok, ul. Kilińskiego 1, Białystok 15-089, Poland
| | - Iwona Radziejewska
- Department of Medical Chemistry, Medical University of Białystok, ul. Mickiewicza 2a, Białystok 15- 222, Poland.
| |
Collapse
|
87
|
Identification of Growth Factors, Cytokines and Mediators Regulated by Artemisia annua L. Polyphenols (pKAL) in HCT116 Colorectal Cancer Cells: TGF-β1 and NGF-β Attenuate pKAL-Induced Anticancer Effects via NF-κB p65 Upregulation. Int J Mol Sci 2022; 23:ijms23031598. [PMID: 35163520 PMCID: PMC8835737 DOI: 10.3390/ijms23031598] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/26/2022] [Accepted: 01/28/2022] [Indexed: 11/30/2022] Open
Abstract
The anticancer effects of natural phytochemicals are relevant to the modulation of cytokine signaling pathways in various cancer cells with stem-like properties as well as immune cells. The aim of this study was to elucidate a novel anticancer mechanism of Artemisia annua L. polyphenols (pKAL) involved in the regulation of growth factors, cytokines and mediators in stem-like HCT116 colorectal cancer cells. Through RayBiotech human L-1000 antibody array and bioinformatics analysis, we show here that pKAL-induced anticancer effects are associated with downregulation of growth factor and cytokine signaling proteins including TGFA, FGF16, PDGFC, CCL28, CXCR3, IRF6 and SMAD1. Notably, we found that TGF-β signaling proteins such as GDF10, ENG and TGFBR2 and well-known survival proteins such as NGF-β, VEGFD and insulin were significantly upregulated by pKAL. Moreover, the results of hematoxylin staining, cell viability assay and Western blot analysis demonstrated that TGF-β1 and NGF-β attenuated pKAL-induced anticancer effects by inhibiting pKAL-induced downregulation of caspase-8, NF-κB p65 and cyclin D1. These results suggest that certain survival mediators may be activated by pKAL through the TGF-β1 and NGF-β signaling pathways during pKAL-induced cell death and thus, strategies to inhibit the survival signaling are inevitably required for more effective anticancer effects of pKAL.
Collapse
|
88
|
Li M, Wang X, Wang Y, Bao S, Chang Q, Liu L, Zhang S, Sun L. Strategies for Remodeling the Tumor Microenvironment Using Active Ingredients of Ginseng-A Promising Approach for Cancer Therapy. Front Pharmacol 2022; 12:797634. [PMID: 35002732 PMCID: PMC8727883 DOI: 10.3389/fphar.2021.797634] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/06/2021] [Indexed: 12/21/2022] Open
Abstract
The tumor microenvironment (TME) plays a key role in promoting the initiation and progression of tumors, leading to chemoradiotherapy resistance and immunotherapy failure. Targeting of the TME is a novel anti-tumor therapeutic approach and is currently a focus of anti-tumor research. Panax ginseng C. A. Meyer (ginseng), an ingredient of well-known traditional Asia medicines, exerts beneficial anti-tumor effects and can regulate the TME. Here, we present a systematic review that describes the current status of research efforts to elucidate the functions and mechanisms of ginseng active components (including ginsenosides and ginseng polysaccharides) for achieving TME regulation. Ginsenosides have variety effects on TME, such as Rg3, Rd and Rk3 can inhibit tumor angiogenesis; Rg3, Rh2 and M4 can regulate the function of immune cells; Rg3, Rd and Rg5 can restrain the stemness of cancer stem cells. Ginseng polysaccharides (such as red ginseng acidic polysaccharides and polysaccharides extracted from ginseng berry and ginseng leaves) can regulate TME mainly by stimulating immune cells. In addition, we propose a potential mechanistic link between ginseng-associated restoration of gut microbiota and the tumor immune microenvironment. Finally, we describe recent advances for improving ginseng efficacy, including the development of a nano-drug delivery system. Taken together, this review provides novel perspectives on potential applications for ginseng active ingredients as anti-cancer adjuvants that achieve anti-cancer effects by reshaping the tumor microenvironment.
Collapse
Affiliation(s)
- Mo Li
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, China.,Department of Thyroid Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Xin Wang
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Ying Wang
- Department of Breast Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Shunchao Bao
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, China
| | - Qing Chang
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, China
| | - Linlin Liu
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, China
| | - Shuai Zhang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, China
| | - Liwei Sun
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
89
|
Wang N, Pei B, Yuan X, Yi C, Wiredu Ocansey DK, Qian H, Mao F. Emerging roles of mesenchymal stem cell-derived exosomes in gastrointestinal cancers. Front Bioeng Biotechnol 2022; 10:1019459. [PMID: 36338118 PMCID: PMC9631450 DOI: 10.3389/fbioe.2022.1019459] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 10/10/2022] [Indexed: 02/05/2023] Open
Abstract
Gastrointestinal tumours are the most common solid tumours, with a poor prognosis and remain a major challenge in cancer treatment. Mesenchymal stem cells (MSC) are multipotent stromal cells with the potential to differentiate into multiple cell types. Several studies have shown that MSC-derived exosomes have become essential regulators of intercellular communication in a variety of physiological and pathological processes. Notably, MSC-derived exosomes support or inhibit tumour progression in different cancers through the delivery of proteins, RNA, DNA, and bioactive lipids. Herein, we summarise current advances in MSC-derived exosomes in cancer research, with particular reference to their role in gastrointestinal tumour development. MSC-derived exosomes are expected to be a novel potential strategy for the treatment of gastrointestinal cancers.
Collapse
Affiliation(s)
- Naijian Wang
- Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu, China
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Bing Pei
- Department of Clinical Laboratory, The Affiliated Suqian First People’s Hospital of Nanjing Medical University, Suqian, Jiangsu, China
| | - Xinyi Yuan
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Chengxue Yi
- School of Medical Technology, Zhenjiang College, Zhenjiang, Jiangsu, China
| | - Dickson Kofi Wiredu Ocansey
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
- Directorate of University Health Services, University of Cape Coast, Cape Coast, Ghana
| | - Hua Qian
- Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu, China
- *Correspondence: Hua Qian,
| | - Fei Mao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
90
|
Bhanu H, Mittal R, Raman S. Evaluation and Clinicopathological Correlation of CD44 in Colorectal Adenoma with Low/High-Grade Dysplasia and Carcinoma. CLINICAL CANCER INVESTIGATION JOURNAL 2022. [DOI: 10.51847/q4yjbhtgzg] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
91
|
Manoochehri H, Asadi S, Tanzadehpanah H, Sheykhhasan M, Ghorbani M. CDC25A is strongly associated with colorectal cancer stem cells and poor clinical outcome of patients. GENE REPORTS 2021; 25:101415. [DOI: 10.1016/j.genrep.2021.101415] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
92
|
Parihar N, Bhatt LK. Deubiquitylating enzymes: potential target in autoimmune diseases. Inflammopharmacology 2021; 29:1683-1699. [PMID: 34792672 DOI: 10.1007/s10787-021-00890-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 10/28/2021] [Indexed: 12/28/2022]
Abstract
The ubiquitin-proteasome pathway is responsible for the turnover of different cellular proteins, such as transport proteins, presentation of antigens to the immune system, control of the cell cycle, and activities that promote cancer. The enzymes which remove ubiquitin, deubiquitylating enzymes (DUBs), play a critical role in central and peripheral immune tolerance to prevent the development of autoimmune diseases and thus present a potential therapeutic target for the treatment of autoimmune diseases. DUBs function by removing ubiquitin(s) from target protein and block ubiquitin chain elongation. The addition and removal of ubiquitin molecules have a significant impact on immune responses. DUBs and E3 ligases both specifically cleave target protein and modulate protein activity and expression. The balance between ubiquitylation and deubiquitylation modulates protein levels and also protein interactions. Dysregulation of the ubiquitin-proteasome pathway results in the development of various autoimmune diseases such as inflammatory bowel diseases (IBD), psoriasis, multiple sclerosis (MS), systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). This review summarizes the current understanding of ubiquitination in autoimmune diseases and focuses on various DUBs responsible for the progression of autoimmune diseases.
Collapse
Affiliation(s)
- Niraj Parihar
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India
| | - Lokesh Kumar Bhatt
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India.
| |
Collapse
|
93
|
Rainho MDA, Mencalha AL, Thole AA. Hypoxia effects on cancer stem cell phenotype in colorectal cancer: a mini-review. Mol Biol Rep 2021; 48:7527-7535. [PMID: 34637098 DOI: 10.1007/s11033-021-06809-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 10/05/2021] [Indexed: 12/15/2022]
Abstract
Colorectal cancer (CRC) is ranked third most incident and second most deadly around the world, and even though treatments significantly developed over the years, overall survival remains low. This scenario has the contribution of cancer stem cells (CSC), a subpopulation of the heterogeneous tumor bulk, considered to be responsible for the tumor maintenance, conventional therapies resistance, metastasis, and recurrence. In this regard, hypoxia appears as an important component of tumor microenvironment and CSC niche, being associated with a worse prognosis. Therefore, it is vital the study of hypoxia influence on CSC phenotype in CRC. The aim of this mini-review article is to present a brief overview on this field. Recent articles discoursed about CSC molecular regulation, signalling pathways, methods for the study of the topic, as well as molecules and drugs capacity of inhibiting the interplay of hypoxia-CSC. Finally, the studies demonstrated important results, extensively accessing the topics of cellular and molecular regulation and therapeutic intervention, being morphology an area to be more explored.
Collapse
Affiliation(s)
- Mateus de Almeida Rainho
- LPCT - Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | - Andre Luiz Mencalha
- LABICAN - Laboratory of Cancer Biology, Biometry and Biophysics Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Alessandra Alves Thole
- LPCT - Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
94
|
Malla RR, Padmaraju V, Marni R, Kamal MA. Natural products: Potential targets of TME related long non-coding RNAs in lung cancer. PHYTOMEDICINE 2021; 93:153782. [PMID: 34627097 DOI: 10.1016/j.phymed.2021.153782] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 09/08/2021] [Accepted: 09/26/2021] [Indexed: 02/05/2023]
Abstract
BACKGROUND Lung cancer is a significant health concern worldwide due to high mortality and morbidity, despite the advances in diagnosis, treatment, and management. Recent experimental evidence from different models suggested long non-coding RNAs (lncRNAs) as major modulators of cancer stem cells (CSCs) in the tumor microenvironment (TME) to support metastasis and drug resistance in lung cancer. Evidence-based studies demonstrated that natural products interfere with TME functions. PURPOSE OF STUDY To establish lncRNAs of TME as novel targets of natural compounds for lung cancer management. STUDY DESIGN Current study used a combination of TME and lung CSCs, lncRNAs and enrichment and stemness maintenance, natural products and stem cell management, natural products and lncRNAs, natural products and targeted delivery as keywords to retrieve the literature from Scopus, Web of Science, PubMed, and Google Scholar. This study critically reviewed the current literature and presented cancer stem cells' ability in reprogramming lung TME. RESULTS This review found that TME related oncogenic and tumor suppressor lncRNAs and their signaling pathways control the maintenance of stemness in lung TME. This review explored natural phenolic compounds and found that curcumin, genistein, quercetin epigallocatechin gallate and ginsenoside Rh2 are efficient in managing lung CSCs. They modulate lncRNAs and their upstream mediators by targeting signaling and epigenetic pathways. This review also identified relevant nanotechnology-based phytochemical delivery approaches for targeting lung cancer. CONCLUSION By critical literature analysis, TME related lncRNAs were identified as potential therapeutic targets, aiming to develop natural product-based therapeutics to treat metastatic and drug-resistant lung cancers.
Collapse
Affiliation(s)
- Rama Rao Malla
- Cancer Biology Lab, Department of Biochemistry and Bioinformatics, GIS, GITAM (Deemed to be University), Visakhapatnam, Andhra Pradesh 530045, India; Department of Biochemistry and Bioinformatics, GIS, GITAM (Deemed to be) University, Visakhapatnam, Andhra Pradesh 530045, India.
| | - Vasudevaraju Padmaraju
- Department of Biochemistry and Bioinformatics, GIS, GITAM (Deemed to be) University, Visakhapatnam, Andhra Pradesh 530045, India
| | - Rakshmitha Marni
- Cancer Biology Lab, Department of Biochemistry and Bioinformatics, GIS, GITAM (Deemed to be University), Visakhapatnam, Andhra Pradesh 530045, India; Department of Biochemistry and Bioinformatics, GIS, GITAM (Deemed to be) University, Visakhapatnam, Andhra Pradesh 530045, India
| | - Mohammad Amjad Kamal
- West China School of Nursing / Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China; King Fahd Medical Research Center, King Abdulaziz University, P. O. Box 80216, Jeddah 21589, Saudi Arabia; Enzymoics, Novel Global Community Educational Foundation, Australia
| |
Collapse
|
95
|
Oliveira BSAD, de Assis ACC, Souza NM, Ferreira LFR, Soriano RN, Bilal M, Iqbal HMN. Nanotherapeutic approach to tackle chemotherapeutic resistance of cancer stem cells. Life Sci 2021; 279:119667. [PMID: 34087280 DOI: 10.1016/j.lfs.2021.119667] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 05/16/2021] [Accepted: 05/22/2021] [Indexed: 02/07/2023]
Abstract
Estimates indicate that cancer will become the leading cause of mortality worldwide in the future. Tumorigenesis is a complex process that involves self-sufficiency in signs of growth, insensitivity to anti-growth signals, prevention of apoptosis, unlimited replication, sustained angiogenesis, tissue invasion, and metastasis. Cancer stem cells (CSCs) have an important role in tumor development and resistance. Here we will approach phenotypic plasticity capacity, highly efficient DNA repair systems, anti-apoptotic machinery, sustained stemness features, interaction with the tumor microenvironment, and Notch, Wnt, and Hedgehog signaling pathways. The researches about CSCs as a target in cancer treatment has been growing. Many different options have pointed beneficial results, such as pathways and CSC-surface markers targeting. Besides its limitations, nanotherapeutics have emerged as a potential strategy in this context since they aim to improve pharmacokinetics, biodistribution, and reduce the side effects observed in traditional treatments. Nanoparticles have been studied in this field, mostly for drug delivery and a multitherapy approach. Another widely researched approaches in this area are related to heat therapy, such as photothermal therapy, photodynamic therapy and magnetic hyperthermia, besides molecular targeting. This review will contemplate the most relevant studies that have shown the effects of nanotherapeutics. In conclusion, although the studies analyzed are mostly preclinical, we believe that there is strong evidence that nanoparticles can increase the chances of a better prognosis to cancer in the future. It is also essential to transpose these findings to the clinic to confirm and better understand the role of nanotherapeutics in this context.
Collapse
Affiliation(s)
- Bruna Stefane Alves de Oliveira
- Undergradute student, Department of Medicine, Federal University of Juiz de Fora, Governador Valadares, MG 35032-620, Brazil
| | - Ana Carolina Correa de Assis
- Undergradute student, Department of Medicine, Federal University of Juiz de Fora, Governador Valadares, MG 35032-620, Brazil
| | - Natália Melo Souza
- Undergradute student, Department of Medicine, Federal University of Juiz de Fora, Governador Valadares, MG 35032-620, Brazil
| | - Luiz Fernando Romanholo Ferreira
- Graduate Program in Process Engineering, Tiradentes University (UNIT), Av. Murilo Dantas, 300, Farolândia, 49032-490 Aracaju, Sergipe, Brazil; Institute of Technology and Research (ITP), Tiradentes University (UNIT), Av. Murilo Dantas, 300, Farolândia, 49032-490 Aracaju, Sergipe, Brazil
| | - Renato Nery Soriano
- Division of Physiology and Biophysics, Department of Basic Life Sciences, Federal University of Juiz de Fora, Governador Valadares, MG 35010-177, Brazil
| | - Muhammad Bilal
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huaian 223003, China.
| | - Hafiz M N Iqbal
- Tecnologico de Monterrey, School of Engineering and Sciences, Monterrey 64849, Mexico.
| |
Collapse
|
96
|
Chen ZQ, Yuan T, Jiang H, Yang YY, Wang L, Fu RM, Luo SQ, Zhang T, Wu ZY, Wen KM. MicroRNA‑8063 targets heterogeneous nuclear ribonucleoprotein AB to inhibit the self‑renewal of colorectal cancer stem cells via the Wnt/β‑catenin pathway. Oncol Rep 2021; 46:219. [PMID: 34396427 PMCID: PMC8377466 DOI: 10.3892/or.2021.8170] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 07/27/2021] [Indexed: 12/13/2022] Open
Abstract
The presence of cancer stem cells (CSCs) is a major cause of therapeutic failure in a variety of cancer types, including colorectal cancer (CRC). However, the underlying mechanisms that regulate the self-renewal of colorectal cancer stem cells (CRCSCs) remain unclear. Our previous study utilized CRCSCs and their parent cells; through gene microarray screening and bioinformatics analysis, we hypothesized that microRNA (miR)-8063 may bind to, and regulate the expression of, heterogeneous nuclear ribonucleoprotein AB (hnRNPAB) to facilitate the regulation of CRCSC self-renewal. The aim of the present study was to confirm this conjecture through relevant experiments. The results indicated that compared with that in parent cells, miR-8063 expression was significantly downregulated in CRCSCs, while hnRNPAB expression was increased. Furthermore, hnRNPAB was identified as a direct target of miR-8063 using a dual-Luciferase assay. Overexpression of hnRNPAB promoted the acquisition of CSC characteristics in CRC cells (increased colony formation ability, enhanced tumorigenicity, and upregulated expression of CSC markers), as well as the upregulation of key proteins (Wnt3a, Wnt5a and β-catenin) in the Wnt/β-catenin signaling pathway. Similarly, after silencing miR-8063 in CRC cells, the characteristics of CSC were altered, and the expression of hnRNPAB protein was promoted. However, post overexpression of miR-8063 in CRCSCs, the self-renewal ability of CSCs was weakened with the downregulation of hnRNPAB protein, Wnt3a, Wnt5a and β-catenin. These results suggest that as a tumor suppressor, miR-8063 is involved in regulating the self-renewal of CRCSCs, where loss of miR-8063 expression weakens its inhibition on hnRNPAB, which leads to the activation of Wnt/β-catenin signaling to promote the self-renewal of CRCSCs.
Collapse
Affiliation(s)
- Zheng-Quan Chen
- Department of Gastrointestinal Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Tao Yuan
- Department of Gastrointestinal Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Hang Jiang
- Department of Gastrointestinal Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Yuan-Yuan Yang
- Department of Gastrointestinal Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Lin Wang
- Department of Gastrointestinal Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Rui-Min Fu
- Department of Gastrointestinal Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Sheng-Qiang Luo
- Department of Gastrointestinal Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Tao Zhang
- Department of Gastrointestinal Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Zhen-Yu Wu
- Department of Gastrointestinal Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Kun-Ming Wen
- Department of Gastrointestinal Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| |
Collapse
|
97
|
Dutta R, Khalil R, Mayilsamy K, Green R, Howell M, Bharadwaj S, Mohapatra SS, Mohapatra S. Combination Therapy of Mithramycin A and Immune Checkpoint Inhibitor for the Treatment of Colorectal Cancer in an Orthotopic Murine Model. Front Immunol 2021; 12:706133. [PMID: 34381456 PMCID: PMC8350740 DOI: 10.3389/fimmu.2021.706133] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 06/25/2021] [Indexed: 01/10/2023] Open
Abstract
The axis of Programmed cell death-1 receptor (PD-1) with its ligand (PD-L1) plays a critical role in colorectal cancer (CRC) in escaping immune surveillance, and blocking this axis has been found to be effective in a subset of patients. Although blocking PD-L1 has been shown to be effective in 5-10% of patients, the majority of the cohorts show resistance to this checkpoint blockade (CB) therapy. Multiple factors assist in the growth of resistance to CB, among which T cell exhaustion and immunosuppressive effects of immune cells in the tumor microenvironment (TME) play a critical role along with other tumor intrinsic factors. We have previously shown the polyketide antibiotic, Mithramycin-A (Mit-A), an effective agent in killing cancer stem cells (CSCs) in vitro and in vivo in a subcutaneous murine model. Since TME plays a pivotal role in CB therapy, we tested the immunomodulatory efficacy of Mit-A with anti-PD-L1 mAb (αPD-L1) combination therapy in an immunocompetent MC38 syngeneic orthotopic CRC mouse model. Tumors and spleens were analyzed by flow cytometry for the distinct immune cell populations affected by the treatment, in addition to RT-PCR for tumor samples. We demonstrated the combination treatment decreases tumor growth, thus increasing the effectiveness of the CB. Mit-A in the presence of αPD-L1 significantly increased CD8+ T cell infiltration and decreased immunosuppressive granulocytic myeloid-derived suppressor cells and anti-inflammatory macrophages in the TME. Our results revealed Mit-A in combination with αPD-L1 has the potential for augmented CB therapy by turning an immunologically "cold" into "hot" TME in CRC.
Collapse
Affiliation(s)
- Rinku Dutta
- James A. Haley Veterans’ Hospital, Tampa, FL, United States
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
- Center for Research and Education in Nano-Bioengineering, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Roukiah Khalil
- James A. Haley Veterans’ Hospital, Tampa, FL, United States
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
- Center for Research and Education in Nano-Bioengineering, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Karthick Mayilsamy
- James A. Haley Veterans’ Hospital, Tampa, FL, United States
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
- Center for Research and Education in Nano-Bioengineering, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Ryan Green
- James A. Haley Veterans’ Hospital, Tampa, FL, United States
- Center for Research and Education in Nano-Bioengineering, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Mark Howell
- James A. Haley Veterans’ Hospital, Tampa, FL, United States
- Center for Research and Education in Nano-Bioengineering, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Srinivas Bharadwaj
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Shyam S. Mohapatra
- James A. Haley Veterans’ Hospital, Tampa, FL, United States
- Center for Research and Education in Nano-Bioengineering, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Subhra Mohapatra
- James A. Haley Veterans’ Hospital, Tampa, FL, United States
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
- Center for Research and Education in Nano-Bioengineering, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| |
Collapse
|
98
|
Research for Expression and Prognostic Value of GABRD in Colon Cancer and Coexpressed Gene Network Construction Based on Data Mining. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2021; 2021:5544182. [PMID: 34194536 PMCID: PMC8203377 DOI: 10.1155/2021/5544182] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/21/2021] [Accepted: 05/10/2021] [Indexed: 02/07/2023]
Abstract
Colon cancer is one of the top five cancers with the highest incidence rate in the world. In order to better understand the pathogenesis and progression of colon cancer, it is still necessary to investigate the abnormally expressed genes in cancer tissue. In this study, the Oncomine database was used for expression analysis, and it was found that the expression level of gamma-aminobutyric acid type A receptor subunit delta (GABRD) gene was upregulated in colon cancer tissue compared with that in normal tissue. UALCAN was used to analyze the expression of GABRD in different groups of age, gender, cancer stage, N stage, and histological subtype. Then, it was also found that the expression of GABRD in each subgroup of colon cancer tissue was all high compared with that in normal tissue. LinkedOmics was used to screen out the differentially expressed genes related to GABRD expression in colon cancer. GO annotation and KEGG pathway enrichment analyses found that the correlated genes may be related to breast cancer, human papillomavirus infection, Notch signaling pathway, and other pathways. Thereafter, GSEA was performed to obtain GABRD-related kinases, miRNAs, and transcription factors, and gene interaction networks were constructed. It was found that GABRD may be involved in cell cycle regulation. Finally, websites like GEPIA were used to detect the predictive ability of GABRD on the prognosis of patients with colon cancer. Kaplan-Meier analysis suggested that the upregulation of GABRD expression was related to the poor prognosis of patients with colon cancer. Overall, in this study, the potential role and prognostic ability of GABRD in colon cancer were explored through data mining, which can be a clue for further research on GABRD.
Collapse
|
99
|
Yi Q, Liu Z, Zhang K, Liu X, Wang L, Geng B, Xia Y. The role of long non-coding RNA BCAR4 in human cancers. Hum Cell 2021; 34:1301-1309. [PMID: 34041673 DOI: 10.1007/s13577-021-00556-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 05/20/2021] [Indexed: 01/08/2023]
Abstract
Long non-coding RNA (lncRNA) is a type of non-coding RNA with length of nucleotides > 200 bp. Increasing evidences show that lncRNA breast cancer antiestrogen resistance 4 (BCAR4) plays an important role in the occurrence and development of various human cancers. It is found that BCAR4 is highly expressed in diverse tumor tissues and cells, and the high expression of BCAR4 is usually associated with poor prognosis. BCAR4 is considered as an oncogene in human cancers. By competing endogenous RNA (ceRNA) mechanism and regulating different signaling pathways, BCAR4 participates in the proliferation, apoptosis, migration, invasion, epithelial-mesenchymal transition (EMT), metastasis and tumorigenesis of different cancers. In addition, overexpression of BCAR4 promotes drug resistance of tumor cells. Therefore, BCAR4 is a promising biomarker for cancer diagnosis and prognosis, and it is a potential target for cancer therapy. This paper reviews studies focusing on the relationship between BCAR4 and cancers in recent years and aims to summarize the effect and mechanism of BCAR4 in human cancers.
Collapse
Affiliation(s)
- Qiong Yi
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, 730000, Gansu, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, 730000, Gansu, China.,Orthopaedic Clinical Medical Research Center of Gansu Province, Lanzhou, 730000, Gansu, China
| | - Zhongcheng Liu
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, 730000, Gansu, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, 730000, Gansu, China.,Orthopaedic Clinical Medical Research Center of Gansu Province, Lanzhou, 730000, Gansu, China
| | - Kun Zhang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, 730000, Gansu, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, 730000, Gansu, China.,Orthopaedic Clinical Medical Research Center of Gansu Province, Lanzhou, 730000, Gansu, China
| | - Xuening Liu
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, 730000, Gansu, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, 730000, Gansu, China.,Orthopaedic Clinical Medical Research Center of Gansu Province, Lanzhou, 730000, Gansu, China
| | - Lifu Wang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, 730000, Gansu, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, 730000, Gansu, China.,Orthopaedic Clinical Medical Research Center of Gansu Province, Lanzhou, 730000, Gansu, China
| | - Bin Geng
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, 730000, Gansu, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, 730000, Gansu, China.,Orthopaedic Clinical Medical Research Center of Gansu Province, Lanzhou, 730000, Gansu, China
| | - Yayi Xia
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, 730000, Gansu, China. .,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, 730000, Gansu, China. .,Orthopaedic Clinical Medical Research Center of Gansu Province, Lanzhou, 730000, Gansu, China.
| |
Collapse
|
100
|
Zalewski A, Snook AE, Waldman SA. Stem cells as therapeutic targets in colorectal cancer. Per Med 2021; 18:171-183. [PMID: 33565332 PMCID: PMC8190705 DOI: 10.2217/pme-2020-0099] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 12/02/2020] [Indexed: 12/31/2022]
Abstract
Colorectal cancer continues to represent a significant burden on public health as the second highest cause of cancer mortality, when men and women are combined, in the US. About 50% of patients either present with late-stage metastatic disease, or develop metastatic recurrences, and ultimately die. In turn, this mortality largely reflects cancer stem cells, tumor-initiating cells that are responsible for cancer progression, drug resistance, recurrence and metastasis. This review summarizes the unique properties of colorectal cancer stem cells, and the emerging strategies by which they can be selectively targeted as a therapeutic approach to eradicating this disease.
Collapse
Affiliation(s)
- Alicja Zalewski
- Department of Surgery, Thomas Jefferson University Hospital, 1020 Locust St, JAH368, Philadelphia, PA 19107, USA
| | - Adam E Snook
- Department of Pharmacology & Experimental Therapeutics, Thomas Jefferson University, 1020 Locust St, JAH368, Philadelphia, PA 19107, USA
| | - Scott A Waldman
- Department of Surgery, Thomas Jefferson University Hospital, 1020 Locust St, JAH368, Philadelphia, PA 19107, USA
| |
Collapse
|