1
|
Xu DQ, Geng JX, Gao ZK, Fan CY, Zhang BW, Han X, He LQ, Dai L, Gao S, Yang Z, Zhang Y, Arshad M, Fu Y, Mu XQ. To explore the potential combined treatment strategy for colorectal cancer: Inhibition of cancer stem cells and enhancement of intestinal immune microenvironment. Eur J Pharmacol 2025; 998:177533. [PMID: 40120791 DOI: 10.1016/j.ejphar.2025.177533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 03/19/2025] [Accepted: 03/19/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND The antibiotic salinomycin, a well-known cancer stem cell inhibitor, may impact the diversity of the intestinal microbiota in colorectal cancer (CRC) mice, which plays a pivotal role in shaping the immune system. This study explores the anti-cancer effects and mechanisms of combining salinomycin and fecal microbiota transplantation (FMT) in treating CRC. METHODS FMT was given via enema, while salinomycin was injected intraperitoneally into the CRC mouse model induced by azoxymethane/dextran sodium sulfate. RESULTS In CRC mice, a large number of LGR5-labeled cancer stem cells and severe disturbances in the intestinal microbiota were observed. Interestingly, salinomycin inhibited the proliferation of cancer stem cells without exacerbating the microbial disorder as expected. In comparison to salinomycin treatment, the combination of salinomycin and FMT significantly improved pathological damage and restored intestinal microbial diversity, which is responsible for shaping the anti-cancer immune microenvironment. The supplementation of FMT significantly increased the levels of propionic acid and butyric acid while also promoting the infiltration of CD8+ T cells and Ly6G+ neutrophils, as well as reducing F4/80+ macrophage recruitment. Notably, cytokines that were not impacted by salinomycin exhibited robust reactions to alterations in the gut microbiota. These included pro-inflammatory factors (IL6, IL12b, IL17, and IL22), chemokine-like protein OPN, and immunosuppressive factor PD-L1. CONCLUSIONS Salinomycin plays the role of "eliminating pathogenic qi," targeting cancer stem cells; FMT plays the role of "strengthening vital qi," reversing the intestinal microbiota disorder and enhancing anti-cancer immunity. They have a synergistic effect on the development of CRC.
Collapse
Affiliation(s)
- Dan-Qi Xu
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Jia-Xin Geng
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Zhan-Kui Gao
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Chao-Yuan Fan
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Bo-Wen Zhang
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Xing Han
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Li-Qian He
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Lin Dai
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Shuo Gao
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Zhou Yang
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Yang Zhang
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Muhammad Arshad
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Yin Fu
- School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, 150006, China.
| | - Xiao-Qin Mu
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China.
| |
Collapse
|
2
|
Kanameda K, Honda A, Hirao-Suzuki M, Sugihara N, Takiguchi M, Takeda S. Interactions between cadmium and 17β-estradiol at physiologically relevant levels evoke unsynchronized events in MCF-7 breast cancer cells: Impaired cell growth and activation of estrogen receptor α-related pathways. Toxicol Appl Pharmacol 2025; 500:117360. [PMID: 40320013 DOI: 10.1016/j.taap.2025.117360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 04/22/2025] [Accepted: 05/01/2025] [Indexed: 05/07/2025]
Abstract
Cadmium (Cd), a heavy metal, is implicated in the development of estrogen receptor α (ERα)-positive breast cancers (BCs). However, controversy surrounds whether Cd is estrogenic or anti-estrogenic for the malignancy of in vivo animal models and human observational/epidemiological studies, a debate also presents in in vitro experiments. The development of ERα-positive BCs is stimulated by circulating 17β-estradiol (E2). Thus, potential biological interactions between E2 and Cd in the progression of ERα-positive BCs exist. Although the interactions between Cd and E2 at physiologically relevant levels (1 nM each) may not have been confirmed in vitro, it is likely to occur. Therefore, this study aims to investigate the interactions of chemicals in human BC MCF-7 cells (ERα-positive) using a sequential exposure system in which chemicals are added to cells every 24-48 h. Pretreatment with Cd, but not secondary treatment, interfered with E2-mediated oncogenic actions by inducing cell cycle arrest at the S phase. This was accompanied by changes in the expression of genes regulating the cell cycle checkpoint and upregulation of the tumor suppressor metallothionein 1F gene, which E2 suppressed. Paradoxically, ERα-mediated estrogenic pathways were upregulated. In conclusion, this study is the first to show that physiologically relevant levels of Cd may dampen E2-induced oncogenic events independent of the E2/ERα-mediated pathway.
Collapse
Affiliation(s)
- Koki Kanameda
- Laboratory of Molecular Life Sciences, Graduate School of Pharmacy and Pharmaceutical Sciences, Fukuyama University, Fukuyama, Japan
| | - Azumi Honda
- Laboratory of Molecular Life Sciences, Faculty of Pharmacy and Pharmaceutical Sciences, Fukuyama University, Fukuyama, Japan
| | - Masayo Hirao-Suzuki
- Laboratory of Xenobiotic Metabolism and Environmental Toxicology, Faculty of Pharmaceutical Sciences, Hiroshima International University, Kure, Japan
| | - Narumi Sugihara
- Laboratory of Molecular Life Sciences, Faculty of Pharmacy and Pharmaceutical Sciences, Fukuyama University, Fukuyama, Japan
| | - Masufumi Takiguchi
- Laboratory of Xenobiotic Metabolism and Environmental Toxicology, Faculty of Pharmaceutical Sciences, Hiroshima International University, Kure, Japan
| | - Shuso Takeda
- Laboratory of Molecular Life Sciences, Graduate School of Pharmacy and Pharmaceutical Sciences, Fukuyama University, Fukuyama, Japan; Laboratory of Molecular Life Sciences, Faculty of Pharmacy and Pharmaceutical Sciences, Fukuyama University, Fukuyama, Japan.
| |
Collapse
|
3
|
Yuan C, Ma Y, Wang W, Wang Y, Shi X, Song Q. Salinomycin inhibits porcine epidemic diarrhea virus infection by targeting Wnt/β-catenin pathway. Int Immunopharmacol 2025; 155:114652. [PMID: 40215770 DOI: 10.1016/j.intimp.2025.114652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 04/08/2025] [Accepted: 04/08/2025] [Indexed: 04/29/2025]
Abstract
Porcine epidemic diarrhea virus (PEDV) is a re-emerging pathogen that causes severe economic losses in the pig industry. Commercial PEDV vaccines provide limited protection against PEDV virulent strains. Therefore, the development of novel vaccines and antiviral drugs is urgently required. In this study, we investigated the inhibitory effects of Salinomycin (SLM) against PEDV infection in vitro. First, the half-maximal cytotoxic concentration (CC50) and half-maximal inhibitory concentration (IC50) of SLM were measured by a cell counting kit 8 (CCK-8) and cytopathic effect (CPE). The results showed that the CC50 of SLM on Vero cells was 7.698 μmol·L-1, and the IC50 for PEDV was 0.998 μmol·L-1. SLM dose-dependently suppressed the PEDV-QY strain infection in vitro. In addition, SLM mainly acted on the internalization and replication stages of the PEDV-QY strain, and had no significant effect on viral inactivation, attachment, and release. Finally, SLM inhibited PEDV infection by suppressing PEDV-induced Wnt/β-catenin activation. Collectively, these results suggest that SLM exerts anti-PEDV effects in vitro and presents a potential as an anti-PEDV drug.
Collapse
Affiliation(s)
- Chen Yuan
- College of Veterinary Medicine, Hebei Agricultural University; Veterinary Biological Technology Innovation Center of Hebei Province, Baoding 071000, China
| | - Yajuan Ma
- College of Veterinary Medicine, Hebei Agricultural University; Veterinary Biological Technology Innovation Center of Hebei Province, Baoding 071000, China
| | - Wei Wang
- College of Veterinary Medicine, Hebei Agricultural University; Veterinary Biological Technology Innovation Center of Hebei Province, Baoding 071000, China
| | - Yawen Wang
- College of Veterinary Medicine, Hebei Agricultural University; Veterinary Biological Technology Innovation Center of Hebei Province, Baoding 071000, China
| | - Xiaojing Shi
- Handan City Rural Revitalization Promotion Center, Handan 056004, Hebei Province, China
| | - Qinye Song
- College of Veterinary Medicine, Hebei Agricultural University; Veterinary Biological Technology Innovation Center of Hebei Province, Baoding 071000, China.
| |
Collapse
|
4
|
Wang Z, Zhang Z, Yue Y, Hou Y, Cao Y, Guo C, Nie X, Hou J. Cross-talk between WNT Signaling and Ferroptosis in Cancer. Mol Cancer Res 2025; 23:175-189. [PMID: 39786453 DOI: 10.1158/1541-7786.mcr-24-0880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/19/2024] [Accepted: 12/23/2024] [Indexed: 01/12/2025]
Abstract
Cancer remains one of the most formidable challenges in the medical field in this century, largely because of its poorly understood pathogenesis. Fortunately, recent advancements in the understanding of cancer pathogenesis have helped identify more therapeutic targets for improved treatment outcomes. The WNT signaling pathways are highly conserved cascades that participate in diverse physiologic processes, such as embryonic development, tissue homeostasis, and tissue regeneration. Ferroptosis, a unique iron-dependent form of cell death that is distinct from apoptosis, is driven by lipid peroxidation and excessive reactive oxygen species production. Emerging evidence shows that the dysregulation of WNT signaling pathways and ferroptosis, as well as their intricate cross-talk, plays crucial roles in cancer progression and therapeutic resistance, indicating their potential as targets for cancer therapies. This review provides a comprehensive overview of the current understanding of the cross-talk between WNT signaling pathways and ferroptosis in the pathogenesis and progression of cancer, with a specific focus on the regulatory role of the canonical WNT cascade in cancer-related ferroptosis. In addition, we discuss the pharmacologic mechanisms of current strategies that inhibit canonical WNT signaling and/or induce ferroptosis in cancer treatment. We propose that combining canonical WNT pathway inhibitors and ferroptosis inducers with current therapies represents a promising therapeutic strategy for personalized cancer treatment.
Collapse
Affiliation(s)
- Zheng Wang
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Zhixiang Zhang
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Yunhui Yue
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Yifan Hou
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Yujia Cao
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Changsheng Guo
- Kaifeng 155 Hospital, China RongTong Medical Healthcare Group Co. Ltd., Kaifeng, China
| | - Xiaobo Nie
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Junqing Hou
- Kaifeng 155 Hospital, China RongTong Medical Healthcare Group Co. Ltd., Kaifeng, China
| |
Collapse
|
5
|
Antoszczak M, Mielczarek-Puta M, Struga M, Huczynski A. Urea and Thiourea Derivatives of Salinomycin as Agents Targeting Malignant Colon Cancer Cells. Anticancer Agents Med Chem 2025; 25:330-338. [PMID: 39390831 DOI: 10.2174/0118715206322603241002064435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/17/2024] [Accepted: 09/23/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND Since it was discovered that a natural polyether ionophore called salinomycin (SAL) selectively inhibits human cancer cells, the scientific world has been paying special attention to this compound. It has been studied for nearly 15 years. OBJECTIVE Thus, a very interesting research direction is the chemical modification of SAL structure, which could give more biologically active agents. METHODS We evaluated the anticancer activity of (thio)urea analogues class of C20-epi-aminosalinomycin (compound 3b). The studies covered the generation of reactive oxygen species (ROS), proapoptotic activity, cytotoxic activity, and lipid peroxidation in vitro. RESULTS Thioureas 5a-5d showed antiproliferative activity against selected human colon cancer cell lines greater than that of chemically unmodified SAL, with a 2~10-fold higher potency towards a metastatic variant of colon cancer cells (SW620). Mechanistically, SAL derivatives showed proapoptotic activity in primary colon cancer cells and induced the production of reactive oxygen species (ROS) in these cells. In SW620 cells, SAL derivatives increased lipid peroxidation with a weak effect on apoptosis and low ROS formation with cytotoxic effects followed by cytostatic ones, suggesting different modes of action of the compounds against primary and metastatic colon cancer cells. CONCLUSION The results of this study suggested that urea and thiourea derivatives of SAL provide promising leads for the rational development of new anticancer active agents.
Collapse
Affiliation(s)
- Michal Antoszczak
- Department of Medical Chemistry, Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznańskiego 8, Poznań, 61-614, Poland
| | - Magdalena Mielczarek-Puta
- Chair and Department of Biochemistry, Medical University of Warsaw, Banacha 1, Warszawa, 02-097, Poland
| | - Marta Struga
- Chair and Department of Biochemistry, Medical University of Warsaw, Banacha 1, Warszawa, 02-097, Poland
| | - Adam Huczynski
- Department of Medical Chemistry, Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznańskiego 8, Poznań, 61-614, Poland
| |
Collapse
|
6
|
Cheng Y, Xiao S, Lan L, Liu D, Tang R, Gu J, Ma L, He Z, Chen X, Geng L, Chen P, Li H, Ren L, Zhu Y, Cheng Y, Gong S. WNT2B high‑expressed fibroblasts induce the fibrosis of IBD by promoting NK cells secreting IL-33. J Mol Med (Berl) 2024; 102:1199-1215. [PMID: 39138828 DOI: 10.1007/s00109-024-02477-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 08/02/2024] [Accepted: 08/05/2024] [Indexed: 08/15/2024]
Abstract
Fibrosis is an important pathological change in inflammatory bowel disease (IBD), but the mechanism has yet to be elucidated. WNT2B high‑expressed fibroblasts are enriched in IBD intestinal tissues, although the precise function of this group of fibroblasts remains unclear. This study investigated whether WNT2B high‑expressed fibroblasts aggravated intestinal tissue damage and fibrosis. Our study provides evidence that WNT2B high‑expressed fibroblasts and NK cells were enriched in colitis tissue of patients with IBD. WNT2B high‑expressed fibroblasts secreted wnt2b, which bound to FZD4 on NK cells and activated the NF-κB and STAT3 pathways to enhance IL-33 expression. TCF4, a downstream component of the WNT/β-catenin pathway, bound to p65 and promoted binding to IL-33 promoter. Furthermore, Salinomycin, an inhibitor of the WNT/β-catenin pathway, inhibited IL-33 secretion in colitis, thereby reducing intestinal inflammation.Knocking down WNT2B reduces NK cell infiltration and IL-33 secretion in colitis, and reduce intestinal inflammation and fibrosis. In conclusion, WNT2B high‑expressed fibroblasts activate NK cells by secreting wnt2b, which activates the WNT/β-catenin and NF-κB pathways to promote IL-33 expression and secretion, potentially culminating in the induction of colonic fibrosis in IBD. KEY MESSAGES: WNT2B high-expressed fibroblasts and NK cells are enriched in colitis tissue, promoting NK cells secreting IL-33. Wnt2b activates NF-κB and STAT3 pathways promotes IL-33 expression by activating p65 and not STAT3. syndrome TCF4 binds to p65 and upregulates the NF- κB pathway. Salinomycin reduces NK cell infiltration and IL-33 secretion in colitis. Knocking down WNT2B mitigates inflammation and fibrosis in chronic colitis.
Collapse
Affiliation(s)
- Yanling Cheng
- Department of Digestive Diseases, Guangzhou Women and Children's Medical Center,Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, 510120, China
- Department of Pediatrics, Shantou Central Hospital, Shantou, 515031, China
| | - Shuzhe Xiao
- Department of Digestive Diseases, Guangzhou Women and Children's Medical Center,Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, 510120, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Lin Lan
- Department of Digestive Diseases, Guangzhou Women and Children's Medical Center,Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, 510120, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Danqiong Liu
- Department of Digestive Diseases, Guangzhou Women and Children's Medical Center,Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, 510120, China
| | - Rui Tang
- Department of Digestive Diseases, Guangzhou Women and Children's Medical Center,Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, 510120, China
| | - Jianbiao Gu
- Department of Digestive Diseases, Guangzhou Women and Children's Medical Center,Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, 510120, China
| | - Li Ma
- Department of Digestive Diseases, Guangzhou Women and Children's Medical Center,Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, 510120, China
| | - Zhihua He
- Department of Digestive Diseases, Guangzhou Women and Children's Medical Center,Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, 510120, China
| | - Xirong Chen
- Nanshan School, Guangzhou Medical University, Guangzhou, 511436, China
| | - Lanlan Geng
- Department of Digestive Diseases, Guangzhou Women and Children's Medical Center,Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, 510120, China
| | - Peiyu Chen
- Department of Digestive Diseases, Guangzhou Women and Children's Medical Center,Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, 510120, China
| | - Huiwen Li
- Department of Digestive Diseases, Guangzhou Women and Children's Medical Center,Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, 510120, China
| | - Lu Ren
- Department of Digestive Diseases, Guangzhou Women and Children's Medical Center,Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, 510120, China
| | - Yun Zhu
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yang Cheng
- Department of Digestive Diseases, Guangzhou Women and Children's Medical Center,Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, 510120, China.
| | - Sitang Gong
- Department of Digestive Diseases, Guangzhou Women and Children's Medical Center,Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, 510120, China.
| |
Collapse
|
7
|
Zhao W, Tang H, Liang Z, Wang N, Sun R, Su R, Yang Z, Zhou K, Peng Y, Zheng S, Xie H. Carvacrol ameliorates skin allograft rejection through modulating macrophage polarization by activating the Wnt signalling pathway. Phytother Res 2024; 38:4675-4694. [PMID: 39120138 DOI: 10.1002/ptr.8282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 05/22/2024] [Accepted: 06/13/2024] [Indexed: 08/10/2024]
Abstract
Post-transplantation immune rejection remains an important factor for transplant patients. However, conventional immunosuppressants are associated with substantial adverse effects. Natural immunosuppressants present a promising alternative to conventional counterparts, boasting exceptional biological activity, minimal toxicity and reduced side effects. We identified carvacrol as a prospective immunosuppressive agent following T cell proliferation experiment and validated carvacrol's immunosuppressive efficacy in the murine allogeneic skin graft model. T cell proliferation assay was used to screen natural small molecule compounds and the immunosuppressive effect of compounds was evaluated in MHC-mismatched murine allogeneic skin graft model. H&E and immunohistochemical staining were applied to evaluate the pathological grade. Furthermore, flow cytometry was uitlized to analyse the immunophenotype changes of immune cells. Western blotting and q-PCR were used to detect the expression of key molecules in macrophages. In vitro, carvacrol demonstrates significant inhibition of the proliferation of CD4+ T and CD8+ T cells. It notably reduces inflammatory factor expression within the allografts, suppresses T cell differentiation toward Th1 phenotype and expansion. Furthermore, carvacrol prominently hinders M1-type macrophages polarization by activating Wnt signaling. Notably, the anti-rejection efficacy of carvacrol was significantly weakened upon the removal of macrophages in mice using chlorophosphate liposomes. Carvacrol could significantly inhibit T cell proliferation, alleviate graft rejection and has outstanding toxicological safety. The molecular mechanism of the anti-rejection effect of carvacrol is closely related to its mediating activation of macrophage Wnt pathway, inhibiting M1 polarization and inducing T cell differentiation.
Collapse
Affiliation(s)
- Wentao Zhao
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hong Tang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhi Liang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ning Wang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ruiqi Sun
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Rong Su
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
| | - Zhentao Yang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ke Zhou
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yiyang Peng
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Haiyang Xie
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| |
Collapse
|
8
|
Zhao X, Ma Y, Luo J, Xu K, Tian P, Lu C, Song J. Blocking the WNT/β-catenin pathway in cancer treatment:pharmacological targets and drug therapeutic potential. Heliyon 2024; 10:e35989. [PMID: 39253139 PMCID: PMC11381626 DOI: 10.1016/j.heliyon.2024.e35989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 08/03/2024] [Accepted: 08/07/2024] [Indexed: 09/11/2024] Open
Abstract
The WNT/β-catenin signaling pathway plays crucial roles in tumorigenesis and relapse, metastasis, drug resistance, and tumor stemness maintenance. In most tumors, the WNT/β-catenin signaling pathway is often aberrantly activated. The therapeutic usefulness of inhibition of WNT/β-catenin signaling has been reported to improve the efficiency of different cancer treatments and this inhibition of signaling has been carried out using different methods including pharmacological agents, short interfering RNA (siRNA), and antibodies. Here, we review the WNT-inhibitory effects of some FDA-approved drugs and natural products in cancer treatment and focus on recent progress of the WNT signaling inhibitors in improving the efficiency of chemotherapy, immunotherapy, gene therapy, and physical therapy. We also classified these FDA-approved drugs and natural products according to their structure and physicochemical properties, and introduced briefly their potential mechanisms of inhibiting the WNT signaling pathway. The review provides a comprehensive understanding of inhibitors of WNT/β-catenin pathway in various cancer therapeutics. This will benefit novel WNT inhibitor development and optimal clinical use of WNT signaling-related drugs in synergistic cancer therapy.
Collapse
Affiliation(s)
- Xi Zhao
- Medical Scientific Research Center, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, China
- China Medical College of Guangxi University, Guangxi University, Nanning, 530004, China
| | - Yunong Ma
- Medical Scientific Research Center, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, China
- China Medical College of Guangxi University, Guangxi University, Nanning, 530004, China
| | - Jiayang Luo
- Medical Scientific Research Center, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, China
| | - Kexin Xu
- Medical Scientific Research Center, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, China
| | - Peilin Tian
- Medical Scientific Research Center, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, China
| | - Cuixia Lu
- Medical Scientific Research Center, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, China
| | - Jiaxing Song
- China Medical College of Guangxi University, Guangxi University, Nanning, 530004, China
| |
Collapse
|
9
|
Wang D, Chen M, Tao Z, Du J, Tian K, Chen Z, Yu B, Chen Y, Lv L. Overexpression of Extracellular Superoxide Dismutase 3 Inhibits Cancer Cell Growth and Migration in Colorectal Cancer. THE TURKISH JOURNAL OF GASTROENTEROLOGY : THE OFFICIAL JOURNAL OF TURKISH SOCIETY OF GASTROENTEROLOGY 2024; 35:465-474. [PMID: 39128081 PMCID: PMC11232067 DOI: 10.5152/tjg.2024.23232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
BACKGROUND/AIMS Incidence of colorectal cancer is rapidly increasing worldwide. Extracellular superoxide dismutase (EcSOD; SOD3) is an antioxidant enzyme. However, SOD3 roles in colorectal cancer progression remain uncertain. MATERIALS AND METHODS Superoxide dismutase 3 expression was evaluated, and we analyzed clinical relevance of SOD3 expression in colorectal cancer. Subsequently, SOD3 roles in colorectal cancer progression were detected by gain of function experiments. Changes in subcutaneous tumor and liver nodule size after SOD3 overexpression were examined in nude mice. The expression of proliferation marker Ki67 was assessed by immunohistochemical staining. RESULTS Supperoxide dismutase 3 was downregulated in colorectal cancer (P <.01). Downregulation of SOD3 was correlated with unfavorable outcomes (P < .05). Superoxide dismutase 3 upregulation limited the proliferative (P <.05), migrative (P <.01) and invasive actions of colorectal cancer cells (P <.01) by suppressing epithelial-mesenchymal transition. Moreover, SOD3 overexpression reduced Ki67 expression (P <.01) and blocked tumor growth (P <01) and liver metastasis (P <.001) in mouse tumor model. CONCLUSION Superoxide dismutase 3 upregulation attenuates tumor growth and liver metastasis in colorectal cancer, suggesting that SOD3 has potential diagnostic and prognostic values regarding colorectal cancer treatment.
Collapse
Affiliation(s)
- Donghua Wang
- Department of Coloproctological Surgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Manyu Chen
- Department of Coloproctological Surgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Zhenggui Tao
- Department of Coloproctological Surgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Jinghu Du
- Department of Coloproctological Surgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Kui Tian
- Department of Coloproctological Surgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Zhen Chen
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Yu
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Chen
- Department of Coloproctological Surgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Long Lv
- Department of Coloproctological Surgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| |
Collapse
|
10
|
Wang Y, Li C, Jiang T, Yin Y, Wang Y, Zhao H, Yu L. A comprehensive exploration of twist1 to identify a biomarker for tumor immunity and prognosis in pan-cancer. Medicine (Baltimore) 2024; 103:e37790. [PMID: 38608058 PMCID: PMC11018223 DOI: 10.1097/md.0000000000037790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 03/14/2024] [Indexed: 04/14/2024] Open
Abstract
Twist1 has been identified as a critical gene in tumor, but current study of this gene remains limitative. This study aims to investigate its roles and potential mechanisms across pan-cancer. The study used various databases and computational techniques to analyze twist's RNA expression, clinical data, gene mutations, tumor stemness, tumor microenvironment, immune regulation. Furthermore, the experimental method of fluorescence staining was carried out to identify twist1 expression in various tumor masses. After analyzing the protein-protein interaction of TWIST, enrichment analysis and predictive potential drugs were performed, and molecular docking was conducted to validate. We found that twist1 expression was significantly higher in various types of cancer and associated with tumor stage, grade, and poor prognosis in multiple cancers. Differential expression of twist1 was linked to gene mutation, RNA modifications, and tumor stemness. Additionally, twist1 expression was positively associated with tumor immunoregulation and immune checkpoint. Salinomycin, klugline, isocephaelince, manassantin B, and pimonidazole are predictive potential drugs targeting TWIST1. This study revealed that twist1 plays an important role in tumor, and might be a curial marker in tumor diagnose and prognosis. The study also highlighted twist1 as a promising therapeutic target for cancer treatment and provided a foundation for future research.
Collapse
Affiliation(s)
- Yue Wang
- Department of Otolaryngology–Head and Neck Surgery, The first affiliated hospital of Ningbo University, Ningbo, China
- Department of Otorhinolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
| | - Chunhao Li
- Department of Otorhinolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
| | - Tianjiao Jiang
- Department of Otorhinolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
| | - Yiqiang Yin
- Department of Otorhinolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
- Department of Pathology, Jinan Fourth People’s Hospital, Jinan, China
| | - Yaowen Wang
- Department of Otolaryngology–Head and Neck Surgery, The first affiliated hospital of Ningbo University, Ningbo, China
| | - Hui Zhao
- Department of Otorhinolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
- Department of Otorhinolaryngology-Head and Neck Surgery, Linyi People’s Hospital, Linyi, China
| | - Liang Yu
- Department of Otorhinolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
| |
Collapse
|
11
|
Yang K, Yi T. Tumor cell stemness in gastrointestinal cancer: regulation and targeted therapy. Front Mol Biosci 2024; 10:1297611. [PMID: 38455361 PMCID: PMC10918437 DOI: 10.3389/fmolb.2023.1297611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 11/14/2023] [Indexed: 03/09/2024] Open
Abstract
The cancer stem cells are a rare group of self-renewable cancer cells capable of the initiation, progression, metastasis and recurrence of tumors, and also a key contributor to the therapeutic resistance. Thus, understanding the molecular mechanism of tumor stemness regulation, especially in the gastrointestinal (GI) cancers, is of great importance for targeting CSC and designing novel therapeutic strategies. This review aims to elucidate current advancements in the understanding of CSC regulation, including CSC biomarkers, signaling pathways, and non-coding RNAs. We will also provide a comprehensive view on how the tumor microenvironment (TME) display an overall tumor-promoting effect, including the recruitment and impact of cancer-associated fibroblasts (CAFs), the establishment of an immunosuppressive milieu, and the induction of angiogenesis and hypoxia. Lastly, this review consolidates mainstream novel therapeutic interventions targeting CSC stemness regulation.
Collapse
Affiliation(s)
- Kangqi Yang
- School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Tuo Yi
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
12
|
Peri SS, Narayanaa Y K, Hubert TD, Rajaraman R, Arfuso F, Sundaram S, Archana B, Warrier S, Dharmarajan A, Perumalsamy LR. Navigating Tumour Microenvironment and Wnt Signalling Crosstalk: Implications for Advanced Cancer Therapeutics. Cancers (Basel) 2023; 15:5847. [PMID: 38136392 PMCID: PMC10741643 DOI: 10.3390/cancers15245847] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/06/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023] Open
Abstract
Cancer therapeutics face significant challenges due to drug resistance and tumour recurrence. The tumour microenvironment (TME) is a crucial contributor and essential hallmark of cancer. It encompasses various components surrounding the tumour, including intercellular elements, immune system cells, the vascular system, stem cells, and extracellular matrices, all of which play critical roles in tumour progression, epithelial-mesenchymal transition, metastasis, drug resistance, and relapse. These components interact with multiple signalling pathways, positively or negatively influencing cell growth. Abnormal regulation of the Wnt signalling pathway has been observed in tumorigenesis and contributes to tumour growth. A comprehensive understanding and characterisation of how different cells within the TME communicate through signalling pathways is vital. This review aims to explore the intricate and dynamic interactions, expressions, and alterations of TME components and the Wnt signalling pathway, offering valuable insights into the development of therapeutic applications.
Collapse
Affiliation(s)
- Shraddha Shravani Peri
- Department of Biomedical Sciences, Faculty of Biomedical Sciences, Technology and Research, Sri Ramachandra Institute of Higher Education and Research, Chennai 600116, India; (S.S.P.); (K.N.Y.); (T.D.H.); (R.R.)
| | - Krithicaa Narayanaa Y
- Department of Biomedical Sciences, Faculty of Biomedical Sciences, Technology and Research, Sri Ramachandra Institute of Higher Education and Research, Chennai 600116, India; (S.S.P.); (K.N.Y.); (T.D.H.); (R.R.)
| | - Therese Deebiga Hubert
- Department of Biomedical Sciences, Faculty of Biomedical Sciences, Technology and Research, Sri Ramachandra Institute of Higher Education and Research, Chennai 600116, India; (S.S.P.); (K.N.Y.); (T.D.H.); (R.R.)
| | - Roshini Rajaraman
- Department of Biomedical Sciences, Faculty of Biomedical Sciences, Technology and Research, Sri Ramachandra Institute of Higher Education and Research, Chennai 600116, India; (S.S.P.); (K.N.Y.); (T.D.H.); (R.R.)
| | - Frank Arfuso
- School of Human Sciences, The University of Western Australia, Nedlands, WA 6009, Australia;
| | - Sandhya Sundaram
- Department of Pathology, Sri Ramachandra Institute of Higher Education and Research, Chennai 600116, India; (S.S.); (B.A.)
| | - B. Archana
- Department of Pathology, Sri Ramachandra Institute of Higher Education and Research, Chennai 600116, India; (S.S.); (B.A.)
| | - Sudha Warrier
- Department of Biotechnology, Sri Ramachandra Institute of Higher Education and Research, Chennai 600116, India;
| | - Arun Dharmarajan
- Department of Biomedical Sciences, Faculty of Biomedical Sciences, Technology and Research, Sri Ramachandra Institute of Higher Education and Research, Chennai 600116, India; (S.S.P.); (K.N.Y.); (T.D.H.); (R.R.)
- School of Human Sciences, The University of Western Australia, Nedlands, WA 6009, Australia;
- Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia
- Curtin Medical School, Curtin University, Perth, WA 6102, Australia
| | - Lakshmi R. Perumalsamy
- Department of Biomedical Sciences, Faculty of Biomedical Sciences, Technology and Research, Sri Ramachandra Institute of Higher Education and Research, Chennai 600116, India; (S.S.P.); (K.N.Y.); (T.D.H.); (R.R.)
| |
Collapse
|
13
|
Zhao H, Han R, Wang Z, Xian J, Bai X. Colorectal Cancer Stem Cells and Targeted Agents. Pharmaceutics 2023; 15:2763. [PMID: 38140103 PMCID: PMC10748092 DOI: 10.3390/pharmaceutics15122763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/30/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
Since their discovery, cancer stem cells have become a hot topic in cancer therapy research. These cells possess stem cell-like self-renewal and differentiation capacities and are important factors that dominate cancer metastasis, therapy-resistance and recurrence. Worse, their inherent characteristics make them difficult to eliminate. Colorectal cancer is the third-most common cancer and the second leading cause of cancer death worldwide. Targeting colorectal cancer stem cells (CR-CSCs) can inhibit colorectal cancer metastasis, enhance therapeutic efficacy and reduce recurrence. Here, we introduced the origin, biomarker proteins, identification, cultivation and research techniques of CR-CSCs, and we summarized the signaling pathways that regulate the stemness of CR-CSCs, such as Wnt, JAK/STAT3, Notch and Hh signaling pathway. In addition to these, we also reviewed recent anti-CR-CSC drugs targeting signaling pathways, biomarkers and other regulators. These will help researchers gain insight into the current agents targeting to CR-CSCs, explore new cancer drugs and propose potential therapies.
Collapse
Affiliation(s)
- Haobin Zhao
- Department of General Practice, People’s Hospital of Longhua, 38 Jinglong Jianshe Road, Shenzhen 518109, China; (H.Z.); (J.X.)
- Endocrinology Department, People’s Hospital of Longhua, 38 Jinglong Jianshe Road, Shenzhen 518109, China
| | - Ruining Han
- Obstetric Department, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518033, China;
| | - Zhankun Wang
- Emergency Department, People’s Hospital of Longhua, 38 Jinglong Jianshe Road, Shenzhen 518109, China;
| | - Junfang Xian
- Department of General Practice, People’s Hospital of Longhua, 38 Jinglong Jianshe Road, Shenzhen 518109, China; (H.Z.); (J.X.)
| | - Xiaosu Bai
- Endocrinology Department, People’s Hospital of Longhua, 38 Jinglong Jianshe Road, Shenzhen 518109, China
| |
Collapse
|
14
|
Guan XY, Guan XL, Zhu JR. Mechanisms and applications of ferroptosis-associated regulators in cancer therapy and drug resistance. J Chemother 2023; 35:671-688. [PMID: 36764828 DOI: 10.1080/1120009x.2023.2177808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 01/08/2023] [Accepted: 02/01/2023] [Indexed: 02/12/2023]
Abstract
Iron is an essential element for almost all living things. Both iron excess and iron deficiency can damage the body's health, but the body has developed complex mechanisms to regulate iron balance. The imbalance of iron homeostasis and lipid peroxidation are important features of ferroptosis. In this review, we summarize the latest regulatory mechanisms of ferroptosis, the roles of relevant regulators that target ferroptosis for cancer therapy, and their relationship to drug resistance. In conclusion, targeting ferroptosis is an important strategy for cancer therapy.
Collapse
Affiliation(s)
- Xiao-Ying Guan
- Pathology Department, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Xiao-Li Guan
- General Medicine Department, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Jia-Rui Zhu
- Cuiying Biomedical Center, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| |
Collapse
|
15
|
Sun Q, Liu B, Lan Q, Su Z, Fu Q, Wang L, Deng Y, Li C, Xue VW, Liu S, Chen X, Yang G, Lu D. Antimicrobial agent chloroxylenol targets β‑catenin‑mediated Wnt signaling and exerts anticancer activity in colorectal cancer. Int J Oncol 2023; 63:121. [PMID: 37681484 PMCID: PMC10546378 DOI: 10.3892/ijo.2023.5569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 07/18/2023] [Indexed: 09/09/2023] Open
Abstract
Chloroxylenol is the active ingredient of the antibacterial agent Dettol. The anticancer effect and underlying mechanisms of this compound and other common antimicrobial agents have not been clearly elucidated. In the present study, the effects of chloroxylenol, benzalkonium chloride, benzethonium chloride, triclosan and triclocarban on β‑catenin‑mediated Wnt signaling in colorectal cancer were evaluated using the SuperTOPFlash reporter assay. It was demonstrated that chloroxylenol, but not the other antimicrobial agents tested, inhibited the Wnt/β‑catenin signaling pathway by decreasing the nuclear translocation of β‑catenin and disrupting β‑catenin/T‑cell factor 4 complex, which resulted in the downregulation of the Wnt target genes Axin2, Survivin and Leucine‑rich G protein‑coupled receptor‑5. Chloroxylenol effectively inhibited the viability, proliferation, migration and invasion, and sphere formation, and induced apoptosis in HCT116 and SW480 cells. Notably, chloroxylenol attenuated the growth of colorectal cancer in the MC38 cell xenograft model and inhibited organoid formation by the patient‑derived cells. Chloroxylenol also demonstrated inhibitory effects on the stemness of colorectal cancer cells. The results of the present study demonstrated that chloroxylenol could exert anti‑tumor activities in colorectal cancer by targeting the Wnt/β‑catenin signaling pathway, which provided an insight into its therapeutic potential as an anticancer agent.
Collapse
Affiliation(s)
- Qi Sun
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Department of Pharmacology, Shenzhen University Medical School, Shenzhen, Guangdong 518060, P.R. China
| | - Boxin Liu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Department of Pharmacology, Shenzhen University Medical School, Shenzhen, Guangdong 518060, P.R. China
| | - Quanxue Lan
- Shenzhen Longgang District Center for Disease Control and Prevention, Shenzhen, Guangdong 518100, P.R. China
| | - Zijie Su
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Department of Pharmacology, Shenzhen University Medical School, Shenzhen, Guangdong 518060, P.R. China
- Department of Research, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Qiuxia Fu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Department of Pharmacology, Shenzhen University Medical School, Shenzhen, Guangdong 518060, P.R. China
| | - Lian Wang
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Department of Pharmacology, Shenzhen University Medical School, Shenzhen, Guangdong 518060, P.R. China
| | - Yingying Deng
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Department of Pharmacology, Shenzhen University Medical School, Shenzhen, Guangdong 518060, P.R. China
| | - Chuanli Li
- Shenzhen Academy of Metrology and Quality Inspection, National Nutrition Food Testing Center, Shenzhen, Guangdong 518102, P.R. China
| | - Vivian Weiwen Xue
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Department of Pharmacology, Shenzhen University Medical School, Shenzhen, Guangdong 518060, P.R. China
| | - Shanshan Liu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Department of Pharmacology, Shenzhen University Medical School, Shenzhen, Guangdong 518060, P.R. China
| | - Xianxiong Chen
- Department of Physiology, Shenzhen University Medical School, Shenzhen, Guangdong 518060, P.R. China
| | - Guowu Yang
- Shenzhen Academy of Metrology and Quality Inspection, National Nutrition Food Testing Center, Shenzhen, Guangdong 518102, P.R. China
| | - Desheng Lu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Department of Pharmacology, Shenzhen University Medical School, Shenzhen, Guangdong 518060, P.R. China
| |
Collapse
|
16
|
Du Y, Chen Y, Li F, Mao Z, Ding Y, Wang W. Genetically Engineered Cellular Nanovesicle as Targeted DNase I Delivery System for the Clearance of Neutrophil Extracellular Traps in Acute Lung Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303053. [PMID: 37759381 PMCID: PMC10646266 DOI: 10.1002/advs.202303053] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 08/18/2023] [Indexed: 09/29/2023]
Abstract
Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) are prevalent critical illnesses with a high mortality rate among patients in intensive care units. Neutrophil extracellular traps (NETs) are implicated in the pathogenesis of ALI/ARDS and represent a promising therapeutic target. However, the clinical application of deoxyribonuclease I (DNase I), the only drug currently available to clear NETs, is limited due to the lack of precise and efficient delivery strategies. Therefore, targeted delivery of DNase I to the inflamed lung remains a critical issue to be addressed. Herein, a novel biomimetic DNase I delivery system is developed (DCNV) that employs genetically and bioorthogonally engineered cellular nanovesicles for pulmonary NETs clearance. The CXC motif chemokine receptor 2 overexpressed cellular nanovesicles can mimic the inflammatory chemotaxis of neutrophils in ALI/ARDS, leading to enhanced lung accumulation. Furthermore, DNase I immobilized through bioorthogonal chemistry exhibits remarkable enzymatic activity in NETs degradation, thus restraining inflammation and safeguarding lung tissue in the lipopolysaccharide-induced ALI murine model. Collectively, the findings present a groundbreaking proof-of-concept in the utilization of biomimetic cellular nanovesicles to deliver DNase I for treating ALI/ARDS. This innovative strategy may usher in a new era in the development of pharmacological interventions for various inflammation-related diseases.
Collapse
Affiliation(s)
- Yang Du
- Department of Hepatobiliary and Pancreatic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang310009China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang ProvinceHangzhouZhejiang310009China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang ProvinceHangzhouZhejiang310009China
- National Innovation Center for Fundamental Research on Cancer MedicineHangzhouZhejiang310009China
- Cancer CenterZhejiang UniversityHangzhouZhejiang310058China
- ZJU‐Pujian Research & Development Center of Medical Artificial Intelligence for Hepatobiliary and Pancreatic DiseaseHangzhouZhejiang310058China
| | - Yining Chen
- Department of Hepatobiliary and Pancreatic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang310009China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang ProvinceHangzhouZhejiang310009China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang ProvinceHangzhouZhejiang310009China
- National Innovation Center for Fundamental Research on Cancer MedicineHangzhouZhejiang310009China
- Cancer CenterZhejiang UniversityHangzhouZhejiang310058China
- ZJU‐Pujian Research & Development Center of Medical Artificial Intelligence for Hepatobiliary and Pancreatic DiseaseHangzhouZhejiang310058China
| | - Fangyuan Li
- Department of Hepatobiliary and Pancreatic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang310009China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang ProvinceHangzhouZhejiang310009China
- Institute of PharmaceuticsHangzhou Institute of Innovative MedicineCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Zhengwei Mao
- Department of Hepatobiliary and Pancreatic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang310009China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang ProvinceHangzhouZhejiang310009China
- MOE Key Laboratory of Macromolecular Synthesis and FunctionalizationDepartment of Polymer Science and EngineeringZhejiang UniversityHangzhouZhejiang310027China
| | - Yuan Ding
- Department of Hepatobiliary and Pancreatic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang310009China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang ProvinceHangzhouZhejiang310009China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang ProvinceHangzhouZhejiang310009China
- National Innovation Center for Fundamental Research on Cancer MedicineHangzhouZhejiang310009China
- Cancer CenterZhejiang UniversityHangzhouZhejiang310058China
- ZJU‐Pujian Research & Development Center of Medical Artificial Intelligence for Hepatobiliary and Pancreatic DiseaseHangzhouZhejiang310058China
| | - Weilin Wang
- Department of Hepatobiliary and Pancreatic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang310009China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang ProvinceHangzhouZhejiang310009China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang ProvinceHangzhouZhejiang310009China
- National Innovation Center for Fundamental Research on Cancer MedicineHangzhouZhejiang310009China
- Cancer CenterZhejiang UniversityHangzhouZhejiang310058China
- ZJU‐Pujian Research & Development Center of Medical Artificial Intelligence for Hepatobiliary and Pancreatic DiseaseHangzhouZhejiang310058China
| |
Collapse
|
17
|
Lu J, Yang Y, Liu X, Chen X, Song W, Liu Z. FTO-mediated LINC01134 stabilization to promote chemoresistance through miR-140-3p/WNT5A/WNT pathway in PDAC. Cell Death Dis 2023; 14:713. [PMID: 37914721 PMCID: PMC10620239 DOI: 10.1038/s41419-023-06244-7] [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: 01/13/2023] [Revised: 10/13/2023] [Accepted: 10/23/2023] [Indexed: 11/03/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive cancer most frequently detected at an advanced stage that limits treatment options to systemic chemotherapy, which has provided only marginal positive clinical outcomes. Currently, the first-line chemotherapeutic agent for PDAC is gemcitabine (GEM). However, the chemotherapy resistance to GEM is often overlooked in the clinical treatment of PDAC due to the lack of effective biological markers. Therefore, it is crucial to find new prognostic markers and therapeutic targets for patients with PDAC. In this study, we identified a novel regulatory mechanism in the development of resistance to GEM in PDAC. Here, we report that LINC01134 was significantly upregulated in primary tumors from PDAC patients. In vitro and in vivo functional studies revealed that LINC01134 promotes PDAC resistance to GEM through facilitating stem cell features and modulating the cell cycle. Mechanistically, LINC01134 interactes with tumor suppressor miR-497-5p in PDAC cells. Increased LINC01134 downregulates miR-140-3p to promotes the oncogenic WNT5A expression. Moreover, m6A demethylase FTO participated in the upregulation of LINC01134 by maintaining LINC01134 mRNA stability through YTHDF2. Taken together, the present study suggested FTO-mediated LINC01134 stabilization to promote chemotherapy resistance to GEM through miR-140-3p/WNT5A/WNT pathway in PDAC. Our study identified new prognostic markers and new therapeutic targets for patients with PDAC.
Collapse
Affiliation(s)
- Jin Lu
- Cancer Center, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China
| | - Yongsheng Yang
- Department of Hepatobiliary Pancreatic Surgery, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun, 130041, China
| | - Xiangliang Liu
- Cancer Center, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China
| | - Xiao Chen
- Cancer Center, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China
| | - Wei Song
- Cancer Center, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China
| | - Zefeng Liu
- Department of Hepatobiliary Pancreatic Surgery, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun, 130041, China.
- Jilin Engineering Laboratory for Translational Medicine of Hepatobiliary and Pancreatic Diseases, Changchun, 130041, China.
| |
Collapse
|
18
|
Zaarour RF, Ribeiro M, Azzarone B, Kapoor S, Chouaib S. Tumor microenvironment-induced tumor cell plasticity: relationship with hypoxic stress and impact on tumor resistance. Front Oncol 2023; 13:1222575. [PMID: 37886168 PMCID: PMC10598765 DOI: 10.3389/fonc.2023.1222575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 09/27/2023] [Indexed: 10/28/2023] Open
Abstract
The role of tumor interaction with stromal components during carcinogenesis is crucial for the design of efficient cancer treatment approaches. It is widely admitted that tumor hypoxic stress is associated with tumor aggressiveness and thus impacts susceptibility and resistance to different types of treatments. Notable biological processes that hypoxia functions in include its regulation of tumor heterogeneity and plasticity. While hypoxia has been reported as a major player in tumor survival and dissemination regulation, the significance of hypoxia inducible factors in cancer stem cell development remains poorly understood. Several reports indicate that the emergence of cancer stem cells in addition to their phenotype and function within a hypoxic tumor microenvironment impacts cancer progression. In this respect, evidence showed that cancer stem cells are key elements of intratumoral heterogeneity and more importantly are responsible for tumor relapse and escape to treatments. This paper briefly reviews our current knowledge of the interaction between tumor hypoxic stress and its role in stemness acquisition and maintenance. Our review extensively covers the influence of hypoxia on the formation and maintenance of cancer stem cells and discusses the potential of targeting hypoxia-induced alterations in the expression and function of the so far known stem cell markers in cancer therapy approaches. We believe that a better and integrated understanding of the effect of hypoxia on stemness during carcinogenesis might lead to new strategies for exploiting hypoxia-associated pathways and their targeting in the clinical setting in order to overcome resistance mechanisms. More importantly, at the present time, efforts are oriented towards the design of innovative therapeutical approaches that specifically target cancer stem cells.
Collapse
Affiliation(s)
- RF. Zaarour
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - M. Ribeiro
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - B. Azzarone
- Tumor Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - S. Kapoor
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - S. Chouaib
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Faculty of Medicine, University Paris-Saclay, Villejuif, France
| |
Collapse
|
19
|
Yang G, He Y, Chen Y, Huang Z, Huang J, Ren X, Xu S, Li P. Antitumor activity of galaxamide involved in cell apoptosis and stemness by inhibiting Wnt/β-catenin pathway in cervical cancer. Drug Dev Res 2023; 84:1114-1126. [PMID: 37154105 DOI: 10.1002/ddr.22073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/21/2023] [Accepted: 04/24/2023] [Indexed: 05/10/2023]
Abstract
Our previous work reported that galaxamide, a cyclopeptide extracted from the seaweed Galaxaura filamentosa, showed antiproliferative activity against HeLa cells by MTT assay. In this study, the growth-inhibitory effects of galaxamide in HeLa cells and xenograft mouse models were investigated. It was found galaxamide significantly inhibited cell growth, colony formation, migration, and invasion and induced cell apoptosis by inhibiting the Wnt signaling pathway in HeLa cells. RNA sequencing revealed that galaxamide regulated stemness by Wnt6 signaling pathway in HeLa cells. By analyzing The Cancer Genome Atlas database, Wnt6 was found to be negatively/positively correlated with stemness- and apoptosis-related genes in human cervical cancer. Cancer stem-like cells (CSCs) isolated and enriched from HeLa cells demonstrated elevated Wnt6 and β-catenin genes compared with nonstem HeLa cells. After galaxamide treatment, CSCs showed abrogation of sphere-forming ability, along with inhibition of stemness-related and Wnt pathway genes. Galaxamide treatment was also accompanied by the induction of apoptosis in HeLa cells, which was consistent with the results in BALB/c nude mice. Our results provide evidence that suppression of stemness by downregulating the Wnt signaling pathway is the molecular mechanism by which galaxamide effectively inhibits cell growth and induces apoptosis in cervical cancer cells.
Collapse
Affiliation(s)
- Guang Yang
- Department of Pathology, Jinan University School of Medicine, Guangzhou, China
- Department of Chemistry, College of Chemistry and Material Science, Jinan University, Guangzhou, China
| | - Yunbiao He
- Department of Medical Statistics, Jinan University School of Medicine, Guangzhou, China
| | - Yingxing Chen
- Department of Gynecology & Obstetrics, The First Affiliated Hospital of Jinan University, Jian University, Guangzhou, China
| | - Zhihan Huang
- Department of Chemistry, College of Chemistry and Material Science, Jinan University, Guangzhou, China
| | - Jieqiong Huang
- Department of Pathology, Jinan University School of Medicine, Guangzhou, China
| | - Xinyi Ren
- Department of Pathology, Jinan University School of Medicine, Guangzhou, China
| | - Shihai Xu
- Department of Chemistry, College of Chemistry and Material Science, Jinan University, Guangzhou, China
| | - Ping Li
- Department of Pathology, Jinan University School of Medicine, Guangzhou, China
| |
Collapse
|
20
|
Tang L, Duan W, Zhang C, Shi Y, Tu W, Lei K, Zhang W, Wu S, Zhang J. Potent salinomycin C20-O-alkyl oxime derivative SAL-98 efficiently inhibits tumor growth and metastasis by affecting Wnt/β-catenin signal pathway. Biochem Pharmacol 2023:115666. [PMID: 37391086 DOI: 10.1016/j.bcp.2023.115666] [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: 03/14/2023] [Revised: 06/16/2023] [Accepted: 06/19/2023] [Indexed: 07/02/2023]
Abstract
The dysregulation of Wnt/β-catenin signaling pathway is closely related to tumorigenesis, metastasis and cancer stem cell maintenance. Salinomycin is a polyether ionophore antibiotic that selectively eliminates cancer stem cells by inhibiting the Wnt/β-catenin signal pathway. Salinomycin selectively target cancer stem cells, but the toxicity limits its further use. In this study, we explore the anti-tumor mechanism of one most active salinomycin C20-O-alkyl oximederivative SAL-98 and found that SAL-98 exerts 10 times higher anti-tumor and anti-CSCs activities compared with salinomycin, which induces cell cycle arrest, ER stress and mitochondria dysfunction and inhibits Wnt/β-catenin signal pathway in vitro with high efficacy. Moreover, SAL-98 shows good anti-metastasis effect in vivo. In addition, SAL-98 demonstrates same anti-tumor activities as salinomycin with less 5 times concentration in vivo, the ER stress, autophagy and anti-CSCs effects were also confirmed in vivo. Mechanistically, SAL-98 inhibits the Wnt/β-catenin signaling pathway associated with CHOP expression induced by ER stress, the induced CHOP disrupts the β-catenin/TCF4 complex and represses the Wnt targeted genes. This study provides an alternative strategy for rational drug development to target Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Lei Tang
- Faculty of Life Science, Kunming University of Science and Technology, Kunming 650500, China; Medical School, Kunming University of Science and Technology, Kunming, 650500, China
| | - Wenfang Duan
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China
| | - Chi Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, China
| | - Yulu Shi
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China
| | - Wenlian Tu
- The First Hospital of Yunnan Province, the affiliated Hospital of Kunming University of Science and Technology, 650032, China
| | - Kangfan Lei
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, China
| | - Wenxuan Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, China.
| | - Song Wu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, China.
| | - Jihong Zhang
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China; Yunnan Province Clinical Research Center for Hematologic Disease, Kunming, 650032, China.
| |
Collapse
|
21
|
Mak PHW, Rehman MA, Kiarie EG, Topp E, Diarra MS. Production systems and important antimicrobial resistant-pathogenic bacteria in poultry: a review. J Anim Sci Biotechnol 2022; 13:148. [PMID: 36514172 DOI: 10.1186/s40104-022-00786-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 09/18/2022] [Indexed: 12/15/2022] Open
Abstract
Economic losses and market constraints caused by bacterial diseases such as colibacillosis due to avian pathogenic Escherichia coli and necrotic enteritis due to Clostridium perfringens remain major problems for poultry producers, despite substantial efforts in prevention and control. Antibiotics have been used not only for the treatment and prevention of such diseases, but also for growth promotion. Consequently, these practices have been linked to the selection and spread of antimicrobial resistant bacteria which constitute a significant global threat to humans, animals, and the environment. To break down the antimicrobial resistance (AMR), poultry producers are restricting the antimicrobial use (AMU) while adopting the antibiotic-free (ABF) and organic production practices to satisfy consumers' demands. However, it is not well understood how ABF and organic poultry production practices influence AMR profiles in the poultry gut microbiome. Various Gram-negative (Salmonella enterica serovars, Campylobacter jejuni/coli, E. coli) and Gram-positive (Enterococcus spp., Staphylococcus spp. and C. perfringens) bacteria harboring multiple AMR determinants have been reported in poultry including organically- and ABF-raised chickens. In this review, we discussed major poultry production systems (conventional, ABF and organic) and their impacts on AMR in some potential pathogenic Gram-negative and Gram-positive bacteria which could allow identifying issues and opportunities to develop efficient and safe production practices in controlling pathogens.
Collapse
Affiliation(s)
- Philip H W Mak
- Guelph Research and Development Centre, Agriculture and Agri-Food Canada (AAFC), Guelph, ON, Canada.,Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada
| | - Muhammad Attiq Rehman
- Guelph Research and Development Centre, Agriculture and Agri-Food Canada (AAFC), Guelph, ON, Canada
| | - Elijah G Kiarie
- Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada
| | - Edward Topp
- London Research and Development Center, AAFC, London, ON, Canada
| | - Moussa S Diarra
- Guelph Research and Development Centre, Agriculture and Agri-Food Canada (AAFC), Guelph, ON, Canada.
| |
Collapse
|
22
|
Yang P, Zhu Y, Zheng Q, Meng S, Wu Y, Shuai W, Sun Q, Wang G. Recent advances of β-catenin small molecule inhibitors for cancer therapy: Current development and future perspectives. Eur J Med Chem 2022; 243:114789. [DOI: 10.1016/j.ejmech.2022.114789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/17/2022] [Accepted: 09/18/2022] [Indexed: 11/28/2022]
|
23
|
Liao M, Du H, Wang B, Huang J, Huang D, Tong G. Anticancer Effect of Polyphyllin I in Suppressing Stem Cell-Like Properties of Hepatocellular Carcinoma via the AKT/GSK-3 β/ β-Catenin Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4031008. [PMID: 36317061 PMCID: PMC9617736 DOI: 10.1155/2022/4031008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 08/29/2022] [Accepted: 09/28/2022] [Indexed: 12/24/2022]
Abstract
Polyphyllin I (PPI), also called Chong Lou saponin I, is a steroidal saponin isolated from the rhizome of Paris polyphylla. PPI has been demonstrated to have strong anticancer activity. However, its effect on the stemness of liver cancer stem cells (LCSCs) is not completely understood. Herein, we aimed to investigate the effect of PPI on the stem cell-like features of LCSCs and hepatocellular carcinoma (HCC). LCSCs were enriched in a serum-free medium and treated with PPI, sorafenib (Sora), or PPI and Sora. Several endpoints, including spheroid formation and differentiation, cell proliferation, surface markers of LCSCs, PPI binding targets, and stemness-associated protein expression, were evaluated. Immunofluorescence staining, quantitative real-time polymerase chain reaction, siRNA transfection, and coimmunoprecipitation ubiquitination assays were conducted for in-depth mechanistic studies. Evaluation of in vivo antitumor efficacy demonstrated that PPI effectively inhibited the proliferation of liver cancer cells and the self-renewal and differentiation of LCSCs. Flow cytometry indicated that PPI suppressed the expression of the stem cell surface markers EpCAM and CD13. Molecular docking showed a high affinity between PPI and proteins of the Wnt/β-catenin signaling pathway, including AKT, GSK-3β, and β-catenin, with the binding energies of -5.51, -5.32, and -5.40 kcal/mol, respectively, which suggested that PPI might regulate the Wnt/β-catenin signaling pathway to affect the stem cell-like properties of HCC. Further ex vivo experiments implied that PPI activated the AKT/GSK-3β-mediated ubiquitin proteasomal degradation of β-catenin and subsequently attenuated the prooncogenic effect of LCSCs. Finally, the anticancer property of PPI was confirmed in vivo. It was found that PPI inhibited the tumor growth in an HCC cell line xenograft model. Taken together, molecular docking analysis and experimental data highlighted the novel function of PPI in suppressing the stem cell-like characteristics of LCSCs via the AKT/GSK-3β/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Mianmian Liao
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Haiyan Du
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Bing Wang
- Nanjing University of Traditional Chinese Medicine, Nanjing, China
| | - Jinzhen Huang
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Danping Huang
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
- Department of Integrated Traditional Chinese and Western Medicine, School of Clinical Medicine of Guangdong Pharmaceuticcal University, Guangzhou, China
| | - Guangdong Tong
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| |
Collapse
|
24
|
Chen J, Liu J, Chen S, Lai R, Zheng C, Lu J, Jiang X, He F, Yang C, Li K, Xie K, Tang Y, Wang L. Salinomycin alleviates osteoarthritis progression via inhibiting Wnt/β-catenin signaling. Int Immunopharmacol 2022; 112:109225. [PMID: 36095950 DOI: 10.1016/j.intimp.2022.109225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/23/2022] [Accepted: 08/31/2022] [Indexed: 11/20/2022]
Abstract
Osteoarthritis (OA) is the most prevalent degenerative whole-joint disease characterized by cartilage degeneration, synovial hyperplasia, osteophyte formation, and subchondral bone sclerosis. Currently there are no disease-modifying treatments available for OA because its etiology and pathogenesis are largely unknown. Here we report that a natural carboxylic polyether ionophore that is used as an anti-tumor drug, salinomycin (SAL), may be a promising therapeutic drug for OA in the future. We found that SAL showed no cytotoxicity on mouse chondrocytes and displayed a protective effect against interleukin-1β (IL-1β), in cultured mouse chondrocytes and cartilage explants. Treatment with low SAL concentrations directly upregulated the anabolism factors collagen II and aggrecan, while it inhibited the catabolic factors matrix metalloproteinase-13 (MMP13) and metalloproteinase with thrombospondin motifs-5 (ADAMTS5) to protect against extracellular matrix (ECM) degradation, and also suppressed inflammatory responses in mouse chondrocytes. Furthermore, SAL reduced the severity of OA-associated changes and delayed cartilage destruction, subchondral bone sclerosis, and osteophyte formation in a destabilized medial meniscus (DMM) surgery-induced mouse OA model. Mechanistically, a low SAL concentration induced anabolism and inhibited catabolism in chondrocytes via inhibiting Lrp6 phosphorylation and Wnt/β-catenin signaling. Our results suggested that SAL may serve as a potential disease-modifying therapeutic against OA pathogenesis.
Collapse
Affiliation(s)
- Jian Chen
- Guangxi Key Laboratory of basic and translational research of Bone and Joint Degenerative Diseases, Guangxi Biomedical Materials Engineering Research Center for Bone and Joint Degenerative Diseases, Department of Orthopedics, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China; The First People's Hospital of Zhaoqing, Zhaoqing 526020, China
| | - Jia Liu
- Guangxi Key Laboratory of basic and translational research of Bone and Joint Degenerative Diseases, Guangxi Biomedical Materials Engineering Research Center for Bone and Joint Degenerative Diseases, Department of Orthopedics, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Shimin Chen
- Guangxi Botanical Garden of Medicinal Plants, Nanning, China; Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Ruijun Lai
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Chuanchuan Zheng
- Guangxi Key Laboratory of basic and translational research of Bone and Joint Degenerative Diseases, Guangxi Biomedical Materials Engineering Research Center for Bone and Joint Degenerative Diseases, Department of Orthopedics, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Jialiang Lu
- Guangxi Key Laboratory of basic and translational research of Bone and Joint Degenerative Diseases, Guangxi Biomedical Materials Engineering Research Center for Bone and Joint Degenerative Diseases, Department of Orthopedics, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Xinshao Jiang
- Guangxi Key Laboratory of basic and translational research of Bone and Joint Degenerative Diseases, Guangxi Biomedical Materials Engineering Research Center for Bone and Joint Degenerative Diseases, Department of Orthopedics, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Feng He
- Guangxi Key Laboratory of basic and translational research of Bone and Joint Degenerative Diseases, Guangxi Biomedical Materials Engineering Research Center for Bone and Joint Degenerative Diseases, Department of Orthopedics, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Chengliang Yang
- Guangxi Key Laboratory of basic and translational research of Bone and Joint Degenerative Diseases, Guangxi Biomedical Materials Engineering Research Center for Bone and Joint Degenerative Diseases, Department of Orthopedics, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Kai Li
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Kegong Xie
- Guangxi Key Laboratory of basic and translational research of Bone and Joint Degenerative Diseases, Guangxi Biomedical Materials Engineering Research Center for Bone and Joint Degenerative Diseases, Department of Orthopedics, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Yujin Tang
- Guangxi Key Laboratory of basic and translational research of Bone and Joint Degenerative Diseases, Guangxi Biomedical Materials Engineering Research Center for Bone and Joint Degenerative Diseases, Department of Orthopedics, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China.
| | - Liqiang Wang
- State Key Laboratory of Metal Matrix Composites, School of Material Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
25
|
He D, Wu B, Du J, Li L, Zhao J. Synergistic inhibition of the growth of MDA-MB-231 cells in triple-negative breast cancer by salinomycin combined with 17-AAG and its mechanism. Oncol Lett 2022; 23:138. [PMID: 35317027 PMCID: PMC8907932 DOI: 10.3892/ol.2022.13258] [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: 04/12/2021] [Accepted: 12/16/2021] [Indexed: 11/06/2022] Open
Abstract
Salinomycin (SAL), a typical ion carrier antibiotic, inhibits tumor growth and metastasis by inducing apoptosis or autophagy in cancer or cancer stem cells and thus overcomes drug resistance. 17-allylamino-17-demethoxygeldanamycin (17-AAG), a heat shock protein Hsp90 competitive inhibitor, also has a role in inhibiting tumor development. However, their combination on the growth of breast cancer cells and its specific mechanism remains to be elucidated. The present study tested the influence of SAL and 17-AAG on cell growth, apoptosis and autophagy by MTT assays, Annexin V-FITC and propidium iodide double staining assay and immunoelectron microscopy. The influence of SAL and 17-AAG on proteomics was investigated by isobaric tag for relative and absolute quantitation. It was found that SAL combined with 17-AAG synergistically inhibited the cell growth and induced the apoptosis in a concentration-dependent manner, with the expression of caspase 3 and Bcl-2 were decreased while the expression of Bax was increased. In addition, SAL combined with 17-AAG inhibited autophagy, with the expression of microtubule-associated protein 1 light chain 3, Beclin1, p62 being decreased. Mechanistically, SAL combined with 17-AAG synergistically inhibited the reactive oxygen species/JNK signaling pathway. In conclusion, SAL combined with 17-AAG had a synergistic inhibitory effect on cell growth of breast cancer via inducing apoptosis and inhibiting autophagy. The present study might provide a new strategy for potential clinical application of SAL as a new anti-tumor drug especially as a drug combination with other molecular targeting therapeutics.
Collapse
Affiliation(s)
- Duo He
- Key Laboratory for Cancer Prevention and Treatment, Medical College of Yan'an University, Yan'an, Shaanxi 716000, P.R. China
- National Translational Science Center for Molecular Medicine, Department of Cell Biology, School of Basic Medicine, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
- Department of Pathology, Yan'an People's Hospital, Yan'an, Shaanxi 716000, P.R. China
| | - Bo Wu
- Key Laboratory for Cancer Prevention and Treatment, Medical College of Yan'an University, Yan'an, Shaanxi 716000, P.R. China
| | - Juan Du
- Key Laboratory for Cancer Prevention and Treatment, Medical College of Yan'an University, Yan'an, Shaanxi 716000, P.R. China
| | - Ling Li
- National Translational Science Center for Molecular Medicine, Department of Cell Biology, School of Basic Medicine, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Jumei Zhao
- Key Laboratory for Cancer Prevention and Treatment, Medical College of Yan'an University, Yan'an, Shaanxi 716000, P.R. China
| |
Collapse
|
26
|
Niwa AM, Semprebon SC, D'Epiro GFR, Marques LA, Zanetti TA, Mantovani MS. Salinomycin induces cell cycle arrest and apoptosis and modulates hepatic cytochrome P450 mRNA expression in HepG2/C3a cells. Toxicol Mech Methods 2021; 32:341-351. [PMID: 34806536 DOI: 10.1080/15376516.2021.2008570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Salinomycin (SAL) is a monocarboxylic polyether ionophore antibiotic isolated from Streptomyces albus. It exhibits an effective antitumor potential against numerous human cancer cells. This study aimed to assess the antiproliferative effects of SAL in human hepatocellular carcinoma HepG2/C3a cell line. We investigated the effects of SAL on cell growth, DNA damage induction, cell cycle changes and apoptosis; and relative changes in expression of cell cycle-related, apoptosis-related, and CYP450 genes. SAL induced cell cycle arrest in the G2/M phase, upregulation of CDKN1A and GADD45A and downregulation of cyclin genes including CCNB1 and CCNA2. SAL effectively suppressed mRNA levels of CTNNB1 gene, an important oncogene that promotes tumorigenesis. The decrease of HepG2/C3A cells' survival can also be due to downregulation of antiapoptotic BCL-2 expression, thus promoting the induction of apoptosis by SAL. This study also demonstrated the ability of SAL in modulating hepatic cytochrome P450 (CYP) mRNA expression, such that SAL caused the upregulation of CYP1A members and CYP3A5; and downregulation of CYP3A4. Taken together, these data contribute to the understanding of the mechanism of action of SAL, highlighting that metabolizing enzymes modulated by SAL can interfere with chemotherapy treatment and it must be considered in associated treatments.
Collapse
Affiliation(s)
- Andressa Megumi Niwa
- Department of General Biology, Center of Biological Sciences, Londrina State University - UEL, Londrina, Brazil
| | - Simone Cristine Semprebon
- Department of General Biology, Center of Biological Sciences, Londrina State University - UEL, Londrina, Brazil
| | | | - Lilian Areal Marques
- Department of General Biology, Center of Biological Sciences, Londrina State University - UEL, Londrina, Brazil
| | - Thalita Alves Zanetti
- Department of General Biology, Center of Biological Sciences, Londrina State University - UEL, Londrina, Brazil
| | - Mário Sérgio Mantovani
- Department of General Biology, Center of Biological Sciences, Londrina State University - UEL, Londrina, Brazil
| |
Collapse
|
27
|
Silva VR, Santos LDS, Dias RB, Quadros CA, Bezerra DP. Emerging agents that target signaling pathways to eradicate colorectal cancer stem cells. Cancer Commun (Lond) 2021; 41:1275-1313. [PMID: 34791817 PMCID: PMC8696218 DOI: 10.1002/cac2.12235] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/28/2021] [Accepted: 10/25/2021] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) represents the third most commonly diagnosed cancer and the second leading cause of cancer death worldwide. The modern concept of cancer biology indicates that cancer is formed of a small population of cells called cancer stem cells (CSCs), which present both pluripotency and self-renewal properties. These cells are considered responsible for the progression of the disease, recurrence and tumor resistance. Interestingly, some cell signaling pathways participate in CRC survival, proliferation, and self-renewal properties, and most of them are dysregulated in CSCs, including the Wingless (Wnt)/β-catenin, Notch, Hedgehog, nuclear factor kappa B (NF-κB), Janus kinase/signal transducer and activator of transcription (JAK/STAT), peroxisome proliferator-activated receptor (PPAR), phosphatidyl-inositol-3-kinase/Akt/mechanistic target of rapamycin (PI3K/Akt/mTOR), and transforming growth factor-β (TGF-β)/Smad pathways. In this review, we summarize the strategies for eradicating CRC stem cells by modulating these dysregulated pathways, which will contribute to the study of potential therapeutic schemes, combining conventional drugs with CSC-targeting drugs, and allowing better cure rates in anti-CRC therapy.
Collapse
Affiliation(s)
- Valdenizia R Silva
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Luciano de S Santos
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Rosane B Dias
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Claudio A Quadros
- São Rafael Hospital, Rede D'Or/São Luiz, Salvador, Bahia, 41253-190, Brazil.,Bahia State University, Salvador, Bahia, 41150-000, Brazil
| | - Daniel P Bezerra
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| |
Collapse
|
28
|
Qi D, Liu Y, Li J, Huang JH, Hu X, Wu E. Salinomycin as a potent anticancer stem cell agent: State of the art and future directions. Med Res Rev 2021; 42:1037-1063. [PMID: 34786735 PMCID: PMC9298915 DOI: 10.1002/med.21870] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 10/22/2021] [Accepted: 10/26/2021] [Indexed: 12/11/2022]
Abstract
Cancer stem cells (CSCs) are a small subpopulation of cells within a tumor that can both self‐renew and differentiate into other cell types forming the heterogeneous tumor bulk. Since CSCs are involved in all aspects of cancer development, including tumor initiation, cell proliferation, metastatic dissemination, therapy resistance, and recurrence, they have emerged as attractive targets for cancer treatment and management. Salinomycin, a widely used antibiotic in poultry farming, was identified by the Weinberg group as a potent anti‐CSC agent in 2009. As a polyether ionophore, salinomycin exerts broad‐spectrum activities, including the important anti‐CSC function. Studies on the mechanism of action of salinomycin against cancer have been continuously and rapidly published since then. Thus, it is imperative for us to update its literature of recent research findings in this area. We here summarize the notable work reported on salinomycin's anticancer activities, intracellular binding target(s), effects on tumor microenvironment, safety, derivatives, and tumor‐specific drug delivery; after that we also discuss the translational potential of salinomycin toward clinical application based on current multifaceted understandings.
Collapse
Affiliation(s)
- Dan Qi
- Department of Neurosurgery, Baylor Scott & White Health, Temple, Texas, USA.,Neuroscience Institute, Baylor Scott & White Health, Temple, Texas, USA
| | - Yunyi Liu
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Molecular Engineering for Theranostics, Hunan University, Changsha, China
| | - Juan Li
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Molecular Engineering for Theranostics, Hunan University, Changsha, China
| | - Jason H Huang
- Department of Neurosurgery, Baylor Scott & White Health, Temple, Texas, USA.,Neuroscience Institute, Baylor Scott & White Health, Temple, Texas, USA.,Department of Surgery, Texas A&M University College of Medicine, Temple, Texas, USA
| | - Xiaoxiao Hu
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Molecular Engineering for Theranostics, Hunan University, Changsha, China.,Shenzhen Research Institute, Hunan University, Shenzhen, Guangdong, China
| | - Erxi Wu
- Department of Neurosurgery, Baylor Scott & White Health, Temple, Texas, USA.,Neuroscience Institute, Baylor Scott & White Health, Temple, Texas, USA.,Department of Surgery, Texas A&M University College of Medicine, Temple, Texas, USA.,LIVESTRONG Cancer Institutes and Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, Texas, USA.,Department of Pharmaceutical Sciences, Texas A&M University College of Pharmacy, College Station, Texas, USA
| |
Collapse
|
29
|
Huang B, Yan X, Li Y. Cancer Stem Cell for Tumor Therapy. Cancers (Basel) 2021; 13:cancers13194814. [PMID: 34638298 PMCID: PMC8508418 DOI: 10.3390/cancers13194814] [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: 08/24/2021] [Revised: 09/13/2021] [Accepted: 09/23/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Although many methods have been applied in clinical treatment for tumors, they still always show a poor prognosis. Molecule targeted therapy has revolutionized tumor therapy, and a proper target must be found urgently. With a crucial role in tumor development, metastasis and recurrence, cancer stem cells have been found to be a feasible and potential target for tumor therapy. We list the unique biological characteristics of cancer stem cells and summarize the recent strategies to target cancer stem cells for tumor therapy, through which we hope to provide a comprehensive understanding of cancer stem cells and find a better combinational strategy to target cancer stem cells for tumor therapy. Abstract Tumors pose a significant threat to human health. Although many methods, such as operations, chemotherapy and radiotherapy, have been proposed to eliminate tumor cells, the results are unsatisfactory. Targeting therapy has shown potential due to its specificity and efficiency. Meanwhile, it has been revealed that cancer stem cells (CSCs) play a crucial role in the genesis, development, metastasis and recurrence of tumors. Thus, it is feasible to inhibit tumors and improve prognosis via targeting CSCs. In this review, we provide a comprehensive understanding of the biological characteristics of CSCs, including mitotic pattern, metabolic phenotype, therapeutic resistance and related mechanisms. Finally, we summarize CSCs targeted strategies, including targeting CSCs surface markers, targeting CSCs related signal pathways, targeting CSC niches, targeting CSC metabolic pathways, inducing differentiation therapy and immunotherapy (tumor vaccine, CAR-T, oncolytic virus, targeting CSCs–immune cell crosstalk and immunity checkpoint inhibitor). We highlight the potential of immunity therapy and its combinational anti-CSC therapies, which are composed of different drugs working in different mechanisms.
Collapse
Affiliation(s)
- Binjie Huang
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou 730030, China; (B.H.); (X.Y.)
- Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou 730030, China
| | - Xin Yan
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou 730030, China; (B.H.); (X.Y.)
- Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou 730030, China
| | - Yumin Li
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou 730030, China; (B.H.); (X.Y.)
- Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou 730030, China
- Correspondence: ; Tel.: +86-138-9361-5421
| |
Collapse
|
30
|
Saydé T, Manczak R, Saada S, Bégaud G, Bessette B, Lespes G, Le Coustumer P, Gaudin K, Dalmay C, Pothier A, Lalloué F, Battu S. Characterization of Glioblastoma Cancer Stem Cells Sorted by Sedimentation Field-Flow Fractionation Using an Ultrahigh-Frequency Range Dielectrophoresis Biosensor. Anal Chem 2021; 93:12664-12671. [PMID: 34491042 DOI: 10.1021/acs.analchem.1c02466] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cancer stem cells (CSCs) appear to be an essential target for cancer therapies, in particular, in brain tumors such as Glioblastoma. Nevertheless, their isolation is made difficult by their low content in culture or tumors (<5% of the tumor mass) and is essentially based on the use of fluorescent or magnetic labeling techniques, increasing the risk of differentiation induction. The use of label-free separation methods such as sedimentation field-flow fractionation (SdFFF) is promising, but it becomes necessary to consider a coupling with a detection and characterization method for future identification and purification of CSCs from patient-derived tumors. In this study, we demonstrate for the first time the capability of using an ultrahigh-frequency range dielectrophoresis fluidic biosensor as a detector. This implies an important methodological adaptation of SdFFF cell sorting by the use of a new compatible carrier liquid DEP buffer (DEP-B). After SdFFF sorting, subpopulations derived from U87-MG and LN18 cell lines undergo biological characterization, demonstrating that using DEP-B as a carrier liquid, we sorted by SdFFF subpopulations with specific differentiation characteristics: F1 = differentiated cells/F2 = CSCs. These subpopulations presented high-frequency crossover (HFC) values similar to those measured for standard differentiated (around 110 MHz) and CSC (around 80 MHz) populations. This coupling appeared as a promising solution for the development of an online integration of these two complementary label-free separation/detection technologies.
Collapse
Affiliation(s)
- Tarek Saydé
- EA3842-CAPTuR, GEIST, Faculté de Médecine, Université de Limoges, 2 rue du Dr Marcland, Limoges 87025, France.,ARNA, INSERM U1212, UMR CNRS 5320, Université de Bordeaux, 146 rue Léo Saignat, Bordeaux 33076, France
| | - Rémi Manczak
- XLIM-UMR CNRS 7252, Université de Limoges, 123, avenue Albert Thomas, Limoges 87060 LIMOGES CEDEX, France
| | - Sofiane Saada
- EA3842-CAPTuR, GEIST, Faculté de Médecine, Université de Limoges, 2 rue du Dr Marcland, Limoges 87025, France
| | - Gaelle Bégaud
- EA3842-CAPTuR, GEIST, Faculté de Médecine, Université de Limoges, 2 rue du Dr Marcland, Limoges 87025, France
| | - Barbara Bessette
- EA3842-CAPTuR, GEIST, Faculté de Médecine, Université de Limoges, 2 rue du Dr Marcland, Limoges 87025, France
| | - Gaëtane Lespes
- CNRS, Institut des Sciences Analytiques et de Physico-Chimie pour l'Environnement et les Matériaux (IPREM), UMR 5254, Université de Pau et des Pays de l'Adour (E2S/UPPA), 2 Avenue Pierre Angot, Pau 64053, France
| | - Philippe Le Coustumer
- Bordeaux Imaging Center, UMS 3420 CNRS-INSERM, Université de Bordeaux, 146 rue Léo Saignat, Bordeaux 33076, France
| | - Karen Gaudin
- ARNA, INSERM U1212, UMR CNRS 5320, Université de Bordeaux, 146 rue Léo Saignat, Bordeaux 33076, France
| | - Claire Dalmay
- XLIM-UMR CNRS 7252, Université de Limoges, 123, avenue Albert Thomas, Limoges 87060 LIMOGES CEDEX, France
| | - Arnaud Pothier
- XLIM-UMR CNRS 7252, Université de Limoges, 123, avenue Albert Thomas, Limoges 87060 LIMOGES CEDEX, France
| | - Fabrice Lalloué
- EA3842-CAPTuR, GEIST, Faculté de Médecine, Université de Limoges, 2 rue du Dr Marcland, Limoges 87025, France
| | - Serge Battu
- EA3842-CAPTuR, GEIST, Faculté de Médecine, Université de Limoges, 2 rue du Dr Marcland, Limoges 87025, France
| |
Collapse
|
31
|
Wang Z, Zhou L, Wang Y, Peng Q, Li H, Zhang X, Su Z, Song J, Sun Q, Sayed S, Liu S, Lu D. The CK1δ/ε-AES axis regulates tumorigenesis and metastasis in colorectal cancer. Am J Cancer Res 2021; 11:4421-4435. [PMID: 33754069 PMCID: PMC7977458 DOI: 10.7150/thno.53901] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 01/22/2021] [Indexed: 12/13/2022] Open
Abstract
Background: Amino-terminal enhancer of split (AES) has been identified as a tumor and metastasis suppressor in some cancers including colorectal cancer (CRC), but very little is known about the regulation of AES expression. Methods: Bioinformatics analysis was used to investigate the expression patterns of AES, CK1δ and CK1ε. The co-immunoprecipitation, GST pull-down, Western Blot, real-time PCR and immunohistochemistry were performed to study the mechanism underlying the regulation of AES expression by CK1δ/ε. The biological function was assessed by in vitro colony formation, transwell, sphere formation, tumor organoids, in vivo tumor metastasis model and patient-derived colorectal tumor xenografts (PDTX) model. Results: A strong inverse relationship was observed between the expression of AES and the expression of CK1δ/ε. Mechanically, AES could interact with CK1δ/ε and SKP2 using its Q domain. SKP2 mediated the ubiquitination and degradation of AES in a CK1δ/ε-dependent manner. CK1δ/ε phosphorylated AES at Ser121 and accelerated the SKP2-mediated ubiquitination and degradation of AES. In colon cancer cells, CK1δ/ε antagonized the effect of wild-type AES but not that of its mutant (S121A) on Wnt and Notch signaling, leading to an increase in the expression of Wnt target genes and Notch target genes. By downregulating the expression of AES, CK1δ/ε enhanced anchorage-independent growth, migration, invasion and sphere formation in colon cancer cells. CK1δ/ε also promoted the growth of APCmin/+ colorectal tumor organoids and liver metastasis in colon cancer mouse models through the regulation of AES degradation. Furthermore, CK1 inhibitor SR3029 treatment suppressed tumor growth via stabilizing AES in APCmin/+ colorectal tumor organoids and patient-derived colorectal tumor xenografts (PDTX). Conclusions: Our results revealed that the CK1δ/ε-AES axis is important for CRC tumorigenesis and metastasis, and targeted inhibition of this axis may be a potential therapeutic strategy for CRC.
Collapse
|
32
|
MicroRNA-194: a novel regulator of glucagon-like peptide-1 synthesis in intestinal L cells. Cell Death Dis 2021; 12:113. [PMID: 33479193 PMCID: PMC7820456 DOI: 10.1038/s41419-020-03366-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 09/18/2020] [Accepted: 10/09/2020] [Indexed: 12/24/2022]
Abstract
In the status of obesity, the glucagon-like peptide-1 (GLP-1) level usually declines and results in metabolic syndrome. This study aimed to investigate the intracellular mechanism of GLP-1 synthesis in L cells from the perspective of microRNA (miRNA). In the present study, we found that GLP-1 level was down-regulated in the plasma and ileum tissues of obese mice, while the ileac miR-194 expression was up-regulated. In vitro experiments indicated that miR-194 overexpression down-regulated GLP-1 level, mRNA levels of proglucagon gene (gcg) and prohormone convertase 1/3 gene (pcsk1), and the nuclear protein level of beta-catenin (β-catenin). Further investigation confirmed that β-catenin could promote gcg transcription through binding to transcription factor 7-like 2 (TCF7L2). miR-194 suppressed gcg mRNA level via negatively regulating TCF7L2 expression. What’s more, forkhead box a1 (Foxa1) could bind to the promoter of pcsk1 and enhanced its transcription. miR-194 suppressed pcsk1 transcription through targeting Foxa1. Besides, the interference of miR-194 reduced palmitate (PA)-induced cell apoptosis and the anti-apoptosis effect of miR-194 inhibitor was abolished by TCF7L2 knockdown. Finally, in HFD-induced obese mice, the silence of miR-194 significantly elevated GLP-1 level and improved the metabolic symptoms caused by GLP-1 deficiency. To sum up, our study found that miR-194 suppressed GLP-1 synthesis in L cells via inhibiting TCF7L2-mediated gcg transcription and Foxa1-mediated pcsk1 transcription. Meanwhile, miR-194 took part in the PA-induced apoptosis of L cells.
Collapse
|
33
|
Heffer AM, Wang V, Libby RT, Feldon SE, Woeller CF, Kuriyan AE. Salinomycin inhibits proliferative vitreoretinopathy formation in a mouse model. PLoS One 2020; 15:e0243626. [PMID: 33347461 PMCID: PMC7751870 DOI: 10.1371/journal.pone.0243626] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 11/24/2020] [Indexed: 01/22/2023] Open
Abstract
Proliferative vitreoretinopathy (PVR) is a progressive disease that develops in a subset of patients who undergo surgery for retinal detachment repair, and results in significant vision loss. PVR is characterized by the migration of retinal pigment epithelial (RPE) cells into the vitreous cavity, where they undergo epithelial-to-mesenchymal transition and form contractile membranes within the vitreous and along the retina, resulting in recurrent retinal detachments. Currently, surgical intervention is the only treatment for PVR and there are no pharmacological agents that effectively inhibit or prevent PVR formation. Here, we show that a single intravitreal injection of the polyether ionophore salinomycin (SNC) effectively inhibits the formation of PVR in a mouse model with no evidence of retinal toxicity. After 4 weeks, fundus photography and optical coherence tomography (OCT) demonstrated development of mean PVR grade of 3.5 (SD: 1.3) in mouse eyes injected with RPE cells/DMSO (vehicle), compared to mean PVR grade of 1.6 (SD: 1.3) in eyes injected with RPE cells/SNC (p = 0.001). Additionally, immunohistochemistry analysis showed RPE cells/SNC treatment reduced both fibrotic (αSMA, FN1, Vim) and inflammatory (GFAP, CD3, CD20) markers compared to control RPE cells/DMSO treatment. Finally, qPCR analysis confirmed that Tgfβ, Tnfα, Mcp1 (inflammatory/cytokine markers), and Fn1, Col1a1 and Acta2 (fibrotic markers) were significantly attenuated in the RPE cells/SNC group compared to RPE/DMSO control. These results suggest that SNC is a potential pharmacologic agent for the prevention of PVR in humans and warrants further investigation.
Collapse
Affiliation(s)
- Alison M. Heffer
- Flaum Eye Institute, University of Rochester, Rochester, NY, United States of America
| | - Victor Wang
- Flaum Eye Institute, University of Rochester, Rochester, NY, United States of America
| | - Richard T. Libby
- Flaum Eye Institute, University of Rochester, Rochester, NY, United States of America
- Center for Visual Sciences, University of Rochester, Rochester, NY, United States of America
| | - Steven E. Feldon
- Flaum Eye Institute, University of Rochester, Rochester, NY, United States of America
- Center for Visual Sciences, University of Rochester, Rochester, NY, United States of America
| | - Collynn F. Woeller
- Flaum Eye Institute, University of Rochester, Rochester, NY, United States of America
| | - Ajay E. Kuriyan
- Flaum Eye Institute, University of Rochester, Rochester, NY, United States of America
- Center for Visual Sciences, University of Rochester, Rochester, NY, United States of America
- Retina Service, Wills Eye Hospital, Thomas Jefferson University, Philadelphia, PA, United States of America
| |
Collapse
|
34
|
Singla A, Wang J, Yang R, Geller DS, Loeb DM, Hoang BH. Wnt Signaling in Osteosarcoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1258:125-139. [PMID: 32767238 DOI: 10.1007/978-3-030-43085-6_8] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Wnt molecules are a class of cysteine-rich secreted glycoproteins that participate in various developmental events during embryogenesis and adult tissue homeostasis. Since its discovery in 1982, the roles of Wnt signaling have been established in various key regulatory systems in biology. Wnt signals exert pleiotropic effects, including mitogenic stimulation, cell fate specification, and differentiation. The Wnt signaling pathway in humans has been shown to be involved in a wide variety of disorders including colon cancer, sarcoma, coronary artery disease, tetra-amelia, Mullerian duct regression, eye vascular defects, and abnormal bone mass. The canonical Wnt pathway functions by regulating the function of the transcriptional coactivator β-catenin, whereas noncanonical pathways function independent of β-catenin. Although the role of Wnt signaling is well established in epithelial malignancies, its role in mesenchymal tumors is more controversial. Some studies have suggested that Wnt signaling plays a pro-oncogenic role in various sarcomas by driving cell proliferation and motility; however, others have reported that Wnt signaling acts as a tumor suppressor by committing tumor cells to differentiate into a mature lineage. Wnt signaling pathway also plays an important role in regulating cancer stem cell function. In this review, we will discuss Wnt signaling pathway and its role in osteosarcoma.
Collapse
Affiliation(s)
- Amit Singla
- Department of Orthopedic Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jichuan Wang
- Department of Orthopedic Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA.,Musculoskeletal Tumor Center, Beijing Key Laboratory for Musculoskeletal Tumors, Peking University People's Hospital, Beijing, China
| | - Rui Yang
- Department of Orthopedic Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - David S Geller
- Department of Orthopedic Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - David M Loeb
- Departments of Pediatrics and Developmental and Molecular Biology, Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Bang H Hoang
- Department of Orthopedic Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
35
|
Wang Z, Feng T, Zhou L, Jiang D, Zhang Y, He G, Lin J, Huang P, Lu D. Salinomycin nanocrystals for colorectal cancer treatment through inhibition of Wnt/β-catenin signaling. NANOSCALE 2020; 12:19931-19938. [PMID: 32990713 DOI: 10.1039/d0nr04552g] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Salinomycin (SAL) is one of the first discovered inhibitors of human cancer stem cells (CSCs), which acts via blocking the Wnt/β-catenin pathway. However, SAL has not been clinically used to treat human diseases due to its poor aqueous solubility and considerable toxicity. In this study, we developed salinomycin nanocrystals (SAL NCs) to treat colorectal cancer through the inhibitory enhancement of Wnt/β-catenin signaling. The as-prepared SAL NCs exhibited excellent size distribution, stability, and improved water solubility. In vitro cellular uptake and in vivo fluorescence imaging studies showed that SAL NCs increased cellular uptake efficiency compared with free SAL. As a result, SAL NCs exhibited significant higher cytotoxicity, 1.5-3 times better Wnt inhibitory effect, and 10 times better cancer stem cell inhibitory effect than free SAL. Furthermore, compared with free SAL, SAL NCs exhibited 2 times better anti-colon tumor effect in APCmin/+ transgenic mice through oral administration. Our results indicated that SAL NCs with enhanced cellular internalization and tumor tissue accumulation may be a promising agent for colorectal cancer management.
Collapse
Affiliation(s)
- Zhongyuan Wang
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, International Cancer Center, Department of Pharmacology, Shenzhen University Health Science Center, Shenzhen 518060, China
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Wang L, Deng K, Gong L, Zhou L, Sayed S, Li H, Sun Q, Su Z, Wang Z, Liu S, Zhu H, Song J, Lu D. Chlorquinaldol targets the β-catenin and T-cell factor 4 complex and exerts anti-colorectal cancer activity. Pharmacol Res 2020; 159:104955. [PMID: 32485279 DOI: 10.1016/j.phrs.2020.104955] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/06/2020] [Accepted: 05/21/2020] [Indexed: 02/07/2023]
Abstract
Aberrant activation of Wnt signaling plays a critical role in the initiation and progression of colorectal cancer (CRC). Chlorquinaldol (CQD) is a topical antimicrobial agent used to treat skin infections. Little is known about the anticancer activity of CQD and its underlying mechanisms. In this study, CQD was demonstrated to inhibit Wnt/β-catenin signaling through targeting the downstream part of this pathway. The results showed that CQD could inhibit the acetylation of β-catenin and disrupt the interaction of β-catenin with T-cell factor 4 (TCF4), leading to reduced binding of β-catenin to the promoters of Wnt target genes and downregulation of the expression of these target genes. Moreover, treatment with CQD suppressed the proliferation, migration, invasion and stemness of CRC cells. In APCmin/+ mice and CRC cell xenografts, administration of CQD suppressed tumor growth and the expression of Wnt target genes c-Myc and Leucine-rich G protein-coupled receptor-5 (LGR5). These results strongly suggest that CQD may be a promising therapeutic agent in the treatment of CRC.
Collapse
Affiliation(s)
- Ling Wang
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Cancer Research Center, Department of Pharmacology, Shenzhen University Health Science Center, Shenzhen, Guangdong, 518060, China
| | - Ke Deng
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Cancer Research Center, Department of Pharmacology, Shenzhen University Health Science Center, Shenzhen, Guangdong, 518060, China
| | - Liang Gong
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Cancer Research Center, Department of Pharmacology, Shenzhen University Health Science Center, Shenzhen, Guangdong, 518060, China
| | - Liang Zhou
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Cancer Research Center, Department of Pharmacology, Shenzhen University Health Science Center, Shenzhen, Guangdong, 518060, China
| | - Sapna Sayed
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Cancer Research Center, Department of Pharmacology, Shenzhen University Health Science Center, Shenzhen, Guangdong, 518060, China
| | - Huan Li
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Cancer Research Center, Department of Pharmacology, Shenzhen University Health Science Center, Shenzhen, Guangdong, 518060, China
| | - Qi Sun
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Cancer Research Center, Department of Pharmacology, Shenzhen University Health Science Center, Shenzhen, Guangdong, 518060, China
| | - Zijie Su
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Cancer Research Center, Department of Pharmacology, Shenzhen University Health Science Center, Shenzhen, Guangdong, 518060, China
| | - Zhongyuan Wang
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Cancer Research Center, Department of Pharmacology, Shenzhen University Health Science Center, Shenzhen, Guangdong, 518060, China
| | - Shanshan Liu
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Cancer Research Center, Department of Pharmacology, Shenzhen University Health Science Center, Shenzhen, Guangdong, 518060, China
| | - Huifang Zhu
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Cancer Research Center, Department of Pharmacology, Shenzhen University Health Science Center, Shenzhen, Guangdong, 518060, China
| | - Jiaxing Song
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Cancer Research Center, Department of Pharmacology, Shenzhen University Health Science Center, Shenzhen, Guangdong, 518060, China.
| | - Desheng Lu
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Cancer Research Center, Department of Pharmacology, Shenzhen University Health Science Center, Shenzhen, Guangdong, 518060, China.
| |
Collapse
|
37
|
Tang C, Gong L, Lvzi Xu, Qiu K, Zhang Z, Wan L. Echinacoside inhibits breast cancer cells by suppressing the Wnt/β-catenin signaling pathway. Biochem Biophys Res Commun 2020; 526:170-175. [PMID: 32201078 DOI: 10.1016/j.bbrc.2020.03.050] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 03/05/2020] [Indexed: 12/11/2022]
Abstract
Echinacoside, a small molecule derived from the natural herbs Cistanche and Echinacea, shows effective anticancer abilities, but the mechanism remains unclear. By using colony formation, scratch, and transwell assays in MDA-MB-231 breast cancer cells, we confirmed the anti-breast cancer ability of Echinacoside in vitro. In addition, we found that Echinacoside can dose-dependently reduce phosho-LRP6, total LRP6, phosho-Dvl2, active β-catenin, and total β-catenin protein expression level in MDA-MB-231 and MDA-MB-468 cells by western blot. We also detected well-known Wnt targets genes, including LEF1, CD44, and cyclin D1 by real-time PCR and western blot, and Echinacoside significantly shows inhibition effect in these two breast cancer cell lines. Furthermore, we investigated its anti-breast cancer ability in an MDA-MB-231 xenograft model in vivo. Echinacoside treatment significantly reduced tumor growth, which was accompanied by a reduction in Wnt/β-catenin signaling. In summary, our results demonstrate that Echinacoside can effectively inhibit Wnt/β-catenin signaling, and therefore, it may be a promising therapeutic target to treat breast cancer.
Collapse
Affiliation(s)
- Chenghuai Tang
- Department of Forensic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Liuping Gong
- Department of Forensic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Lvzi Xu
- Forensic Identification Center, College of Criminal Investigation, Southwest University of Political Science and Law, Chongqing, 401120, China
| | - Kaijin Qiu
- Department of Forensic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Zhong Zhang
- Department of Forensic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Lihua Wan
- Department of Forensic Medicine, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
38
|
Huang H, Cai H, Zhang L, Hua Z, Shi J, Wei Y. Oroxylin A inhibits carcinogen-induced skin tumorigenesis through inhibition of inflammation by regulating SHCBP1 in mice. Int Immunopharmacol 2020; 80:106123. [PMID: 31927505 DOI: 10.1016/j.intimp.2019.106123] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 12/29/2022]
Abstract
Accumulating evidence has shown that SHC SH2 domain-binding protein 1 (SHCBP1) functions as an oncogene and participated in the progression of various cancers. Oroxylin A, an active ingredient extracted from Chinese Medicine Scutellaria baicalensis, shows strong anticancer effects on multiple cancers, however, the pharmacological effect of oroxylin A on skin cancer and the regulatory effect of SHCBP1 on this process have never been evaluated. The present study was aimed at elucidating the effect of oroxylin A on carcinogen (DMBA/TPA)-induced skin tumorigenesis, and to further clarify the role of SHCBP1 in oroxylin A induced antitumor effect. Pretreatment with oroxylin A remarkably inhibited DMBA/TPA-induced tumor formation and growth, and significantly reduced tumor incidence and the average number of tumors per mouse. Oroxylin A suppressed DMBA/TPA-induced skin hyperplasia and tumor proliferation. Oroxylin A significantly inhibited the expression of several inflammatory factors in vivo. In vitro experiments found that oroxylin A inhibited TPA-induced cell malignant transformation of skin epidermal JB6 P + cells. Besides, oroxylin A significantly suppressed the levels of TPA-induced inflammatory factors in vitro. Mechanistic studies showed that oroxylin A remarkably inhibited TPA-induced increased expression of SHCBP1. Overexpression of SHCBP1 attenuated the oroxylin A-induced anti-inflammatory effect. In addition, TPA increased the expression of nuclear NF-κB p65, and SHCBP1 siRNA notably decreased the nuclear NF-κB p65 expression in JB6 P + cells. Collectively, the anti-skin cancer effect of oroxylin A may possibly by inhibiting inflammation via suppression of SHCBP1. Oroxylin A might be a potential candidate compound for the treatment of skin cancer.
Collapse
Affiliation(s)
- Hongjuan Huang
- First Clinical Medical College, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210046, Jiangsu, China
| | - Hengji Cai
- Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong 226001, Jiangsu, China
| | - Li Zhang
- Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong 226001, Jiangsu, China
| | - Zhixiang Hua
- The First People's Hospital of Nantong, Nantong, Jiangsu 226001, China
| | - Jian Shi
- The First People's Hospital of Nantong, Nantong, Jiangsu 226001, China
| | - Yuegang Wei
- First Clinical Medical College, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210046, Jiangsu, China.
| |
Collapse
|
39
|
Wang Z, Zhou L, Xiong Y, Yu S, Li H, Fan J, Li F, Su Z, Song J, Sun Q, Liu S, Xia Y, Zhao L, Li S, Guo F, Huang P, Carson DA, Lu D. Salinomycin exerts anti-colorectal cancer activity by targeting the β-catenin/T-cell factor complex. Br J Pharmacol 2019; 176:3390-3406. [PMID: 31236922 PMCID: PMC6692576 DOI: 10.1111/bph.14770] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 05/24/2019] [Accepted: 06/04/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND PURPOSE Salinomycin is a well-known inhibitor of human cancer stem cells (CSCs). However, the molecular mechanism(s) by which salinomycin targets colorectal CSCs is poorly understood. Here, we have investigated underlying antitumour mechanisms of salinomycin in colorectal cancer cells and three tumour models. EXPERIMENTAL APPROACH The inhibitory effect of salinomycin on the Wnt/β-catenin pathway was analysed with the SuperTopFlash reporter system. The mRNA expression of Wnt target genes was evaluated with real-time PCR. Effects of salinomycin on β-catenin/TCF4E interaction were examined using co-immunoprecipitation and an in vitro GST pull-down assay. Cell proliferation was determined by BrdU incorporation and soft agar colony formation assay. The stemness of the cells was assessed by sphere formation assay. Antitumour effects of salinomycin on colorectal cancers was evaluated with colorectal CSC xenografts, APCmin/+ transgenic mice, and patient-derived colorectal tumour xenografts. KEY RESULTS Salinomycin blocked β-catenin/TCF4E complex formation in colorectal cancer cells and in an in vitro GST pull-down assay, thus decreasing expression of Wnt target genes. Salinomycin also suppressed the transcriptional activity mediated by β-catenin/LEF1 or β-catenin/TCF4E complex and exhibited an inhibitory effect on the sphere formation, proliferation, and anchorage-independent growth of colorectal cancer cells. In colorectal tumour xenografts and APCmin/+ transgenic mice, administration of salinomycin significantly reduced tumour growth and the expression of CSC-related Wnt target genes including LGR5. CONCLUSIONS AND IMPLICATIONS Our study suggested that salinomycin could suppress the growth of colorectal cancer by disrupting the β-catenin/TCF complex and thus may be a promising agent for colorectal cancer treatment.
Collapse
Affiliation(s)
- Zhongyuan Wang
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Carson International Cancer Center, Department of PharmacologyShenzhen University Health Science CenterShenzhenChina
| | - Liang Zhou
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Carson International Cancer Center, Department of PharmacologyShenzhen University Health Science CenterShenzhenChina
| | - Yanpeng Xiong
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Carson International Cancer Center, Department of PharmacologyShenzhen University Health Science CenterShenzhenChina
| | - Shubin Yu
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Carson International Cancer Center, Department of PharmacologyShenzhen University Health Science CenterShenzhenChina
| | - Huan Li
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Carson International Cancer Center, Department of PharmacologyShenzhen University Health Science CenterShenzhenChina
| | - Jiaoyang Fan
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Carson International Cancer Center, Department of PharmacologyShenzhen University Health Science CenterShenzhenChina
| | - Fan Li
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Laboratory of Evolutionary Theranostics, School of Biomedical EngineeringShenzhen University Health Science CenterShenzhenChina
| | - Zijie Su
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Carson International Cancer Center, Department of PharmacologyShenzhen University Health Science CenterShenzhenChina
| | - Jiaxing Song
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Carson International Cancer Center, Department of PharmacologyShenzhen University Health Science CenterShenzhenChina
| | - Qi Sun
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Carson International Cancer Center, Department of PharmacologyShenzhen University Health Science CenterShenzhenChina
| | - Shan‐Shan Liu
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Carson International Cancer Center, Department of PharmacologyShenzhen University Health Science CenterShenzhenChina
| | - Yuqing Xia
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Carson International Cancer Center, Department of PharmacologyShenzhen University Health Science CenterShenzhenChina
| | - Liang Zhao
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Carson International Cancer Center, Department of PharmacologyShenzhen University Health Science CenterShenzhenChina
| | - Shiyue Li
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Carson International Cancer Center, Department of PharmacologyShenzhen University Health Science CenterShenzhenChina
| | - Fang Guo
- Center for Systems BiomedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Peng Huang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Laboratory of Evolutionary Theranostics, School of Biomedical EngineeringShenzhen University Health Science CenterShenzhenChina
| | - Dennis A. Carson
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Carson International Cancer Center, Department of PharmacologyShenzhen University Health Science CenterShenzhenChina
- Moores Cancer CenterUniversity of California San Diego (UCSD)La JollaCalifornia
| | - Desheng Lu
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Carson International Cancer Center, Department of PharmacologyShenzhen University Health Science CenterShenzhenChina
| |
Collapse
|