1
|
Li S, Yang K, Ye J, Xu C, Qin Z, Chen Y, Yu L, Zhou T, Sun B, Xu J. LGALS4 inhibits glycolysis and promotes apoptosis of colorectal cancer cells via β‑catenin signaling. Oncol Lett 2025; 29:126. [PMID: 39807100 PMCID: PMC11726281 DOI: 10.3892/ol.2025.14873] [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: 06/26/2024] [Accepted: 10/31/2024] [Indexed: 01/16/2025] Open
Abstract
Colorectal cancer (CRC) is one of the leading causes of cancer-related deaths worldwide. Glycolysis serves a crucial role in the development of CRC. The aim of the present study was to investigate the function of lectin galactoside-binding soluble 4 (LGALS4) in the regulation of glycolysis and its therapeutic potential in CRC. In the present study, 175 overlapping differentially expressed genes were identified by comprehensive analysis of The Cancer Genome Atlas database and the GSE26571 CRC dataset from the Gene Expression Omnibus database. LGALS4 was identified as the central gene by prognostic analysis using the mimetic map construction method and least absolute shrinkage and selection operator Cox regression. In vitro experiments were performed to evaluate the effects of LGALS4 overexpression on CRC cell phenotype and aerobic glycolysis, as well as its relationship with β-catenin signaling. LGALS4 was significantly downregulated in CRC, with an average 3-fold decrease compared with LGALS4 expression levels in normal tissues. LGALS4 was also significantly associated with patient survival. LGALS4 overexpression inhibited CRC cell growth, induced cell cycle arrest and enhanced 5-fluorouracil (5-FU)-induced apoptosis. Specifically, LGALS4 overexpression resulted in a ~50% decrease in cell proliferation and a ~2-fold increase in apoptosis. In addition, LGALS4 overexpression inhibited aerobic glycolysis and reduced glucose-dependent and glycolytic activity in CRC cells. The downregulatory effect of LGALS4 on glycolysis-related genes was further enhanced by the addition of the β-catenin inhibitor XAV-939. LGALS4 expression decreased CRC progression by inhibiting glycolysis and affecting β-catenin signaling. Overexpression of LGALS4 reduced the proliferation and glycolytic capacity of CRC cells and also enhanced their sensitivity to 5-FU. These results may potentially provide new perspectives for CRC treatment and targets for future clinical intervention strategies.
Collapse
Affiliation(s)
- Shengjie Li
- Department of Gastroenterology Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei 443000, P.R. China
| | - Kaifeng Yang
- Department of Gastroenterology Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei 443000, P.R. China
| | - Jiayou Ye
- Department of Gastroenterology Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei 443000, P.R. China
| | - Chengfan Xu
- Department of Gastroenterology Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei 443000, P.R. China
| | - Zhixiang Qin
- Department of Gastroenterology Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei 443000, P.R. China
| | - Ying Chen
- Center for Clinical Research and Translational Medicine, Yangpu Hospital, School of Medicine, Tongji University, Shanghai 200438, P.R. China
| | - Lanjian Yu
- Department of Gastroenterology Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei 443000, P.R. China
| | - Tianyu Zhou
- Department of Gastroenterology Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei 443000, P.R. China
| | - Bin Sun
- Center for Clinical Research and Translational Medicine, Yangpu Hospital, School of Medicine, Tongji University, Shanghai 200438, P.R. China
| | - Jun Xu
- Department of Gastroenterology Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei 443000, P.R. China
| |
Collapse
|
2
|
Wei G, Fan L, Song X, Zhou W, Sun M, Sun Y, Yao Y, Zhao H, Chen F, Zhu C. Modulation of almonertinib resistance in non-small cell lung cancer by cancer-associated fibroblasts through HK2-mediated glycolysis and SKP2 signaling. Discov Oncol 2025; 16:187. [PMID: 39954036 PMCID: PMC11829888 DOI: 10.1007/s12672-025-01974-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 02/11/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Almonertinib is a third-generation EGFR-TKI, and studies on its resistance mechanisms are lacking. Cancer-associated fibroblasts (CAFs) can influence resistance to targeted therapeutics, but their role and mechanism of action in relation to almonertinib resistance are unclear. The study explored relationships among glycolysis, cancer-associated fibroblasts (CAFs), and almonertinib resistance. METHODS A dose-escalation method was used to develop the almonertinib-resistant cell line H1975AR. Hexokinase 2 (HK2) effects on almonertinib resistance were evaluated using Cell Counting Kit-8 assays, transcriptome sequencing, western blotting, real-time PCR, siRNA assays, glucose consumption, and lactate production assays. Differential gene expression analysis and siRNA assays in H1975 cells cultured with CAF-conditioned medium (H1975/CAF-CM) revealed S-phase kinase-associated protein 2 (SKP2) as a target driving the effects of HK2. The impact of almonertinib and HK2 inhibitors on H1975/CAF-CM cells was assessed using colony formation, wound healing, transwell, and flow cytometry apoptosis assays. RESULTS H1975AR cells displayed elevated glycolysis and HK2. Subsequently, we showed that knockdown reduced almonertinib resistance in cells. H1975/CAF-CM induced almonertinib resistance and upregulation of HK2, which was reversed by knockdown of SKP2. CAFs regulate HK2-mediated glycolysis through SKP2, promoting almonertinib resistance in NSCLC. CONCLUSIONS CAFs regulate HK2-mediated glycolysis through SKP2, which promotes almonertinib resistance in NSCLC.
Collapse
Affiliation(s)
- Guohao Wei
- Department of Oncology, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210003, China
| | - Lingyao Fan
- Department of Oncology, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210003, China
| | - Xiaoyan Song
- Department of Oncology, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210003, China
| | - Wenzheng Zhou
- Department of Oncology, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210003, China
| | - Min Sun
- Department of Oncology, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210003, China
| | - Yan Sun
- Department of Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, 210009, China
| | - Yuhui Yao
- Department of Oncology, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210003, China
| | - Huihui Zhao
- Department of Oncology, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210003, China.
| | - Fangfang Chen
- Department of Clinical Laboratory, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210093, China.
| | - Chuandong Zhu
- Department of Oncology, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210003, China.
| |
Collapse
|
3
|
Praseatsook K, Vachiraarunwong A, Taya S, Setthaya P, Sato K, Wanibuchi H, Wongpoomchai R, Dejkriengkraikul P, Gi M, Yodkeree S. Anticancer and Antioxidant Effects of Bioactive Peptides from Black Soldier Fly Larvae ( Hermetia illucens). Nutrients 2025; 17:645. [PMID: 40004973 PMCID: PMC11858422 DOI: 10.3390/nu17040645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/03/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Protein hydrolysates from insects are recognized for their biological activities. Black soldier fly larvae (BSFL) have drawn attention due to their antioxidant protein hydrolysates. However, research on bioactive peptides derived from these hydrolysates, particularly their cancer chemopreventive potential, remains limited. This study aims to evaluate the antioxidant, anti-inflammatory, antimutagenic, and anticancer activities of BSFL-derived bioactive peptides and explore the molecular mechanisms. METHODS Alkali-soluble BSFL protein (ASBP) was extracted and hydrolyzed using Alcalase and bromelain under optimized conditions. Antioxidant activity was assessed via FRAP, ABTS, and DPPH assays. The hydrolysate with the highest antioxidant activity was fractionated into molecular weight (MW) groups (>30, 10, and <3 kDa). The bioactivity of fractionated peptides was evaluated through antioxidant, anti-inflammatory (nitric oxide production in RAW 264.7 cells), antimutagenic (Ames test), and anticancer (CCK-8 assay on HCT 116, COLO205, Cw-2, and Caco-2 cells) assays. Mechanistic insights were obtained via microarray and Western blot analyses. Peptides were identified by LC-MS/MS. RESULTS The ASBP-Alcalase hydrolysate (ASBP-AH) showed optimal antioxidant activity at 3% (w/w) for 4 h. The ASBP-AH 30 (MW > 30 kDa) fraction exhibited the highest antioxidant capacity. In contrast, the ASBP-AH3 (MW < 3 kDa) fraction exhibited significant antimutagenic effects, reduced nitric oxide production, and decreased COLO205 cell viability. Treatment with ASBP-AH3 at its LC50 dose modulated the SKP2/p21/cyclin D1 pathways. Mostly peptides from ASBP-AH3 were composed of hydrophobic and charged amino acids. CONCLUSIONS BSFL-derived bioactive peptides exhibit potential as multifunctional agents for cancer chemoprevention. In vivo studies are required to explore their clinical applications.
Collapse
Affiliation(s)
- Kwanchanok Praseatsook
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.P.); (R.W.); (P.D.)
| | - Arpamas Vachiraarunwong
- Department of Environmental Risk Assessment, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-8585, Japan; (A.V.); (H.W.)
| | - Sirinya Taya
- Functional Food Research Unit, Multidisciplinary Research Institute, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Phatthawin Setthaya
- Science and Technology Research Institute, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Kenji Sato
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan;
| | - Hideki Wanibuchi
- Department of Environmental Risk Assessment, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-8585, Japan; (A.V.); (H.W.)
| | - Rawiwan Wongpoomchai
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.P.); (R.W.); (P.D.)
| | - Pornngarm Dejkriengkraikul
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.P.); (R.W.); (P.D.)
- Anticarcinogenesis and Apoptosis Research Cluster, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Min Gi
- Department of Environmental Risk Assessment, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-8585, Japan; (A.V.); (H.W.)
| | - Supachai Yodkeree
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.P.); (R.W.); (P.D.)
- Anticarcinogenesis and Apoptosis Research Cluster, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
4
|
Wang M, Zhang Y, Ni S, Sun M, Wu Q, Wu X, Chen Q, Wang S. The anti-cancer activity of Dioscin: an update and future perspective. Med Oncol 2025; 42:63. [PMID: 39899128 PMCID: PMC11790812 DOI: 10.1007/s12032-024-02572-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 11/18/2024] [Indexed: 02/04/2025]
Abstract
Natural drugs have the advantages of multi-pathway, multi-target, low toxicity, and high efficiency, which make them widely used and effective in anti-tumor therapy. Dioscin is a steroidal saponin compound that can be extracted from Dioscaceae plants. In recent years, it has been found that Dioscin has potent anti-tumor effects, can inhibit tumor cell proliferation, induce apoptosis and autophagy, inhibits tumor cell metastasis, reverses multidrug resistance, and increases sensitivity to anticancer drugs, and thus inhibit tumor progression. Meanwhile, the construction of Dioscin nanocarriers can improve the efficiency of drug use, reduce drug toxicity, realize the precise delivery of drugs, and improve the bioavailability of Dioscin. In this paper, the anticancer mechanism and targets of Dioscin in recent years were reviewed, thereby providing new ideas and a theoretical basis for further research and promotion of Dioscin.
Collapse
Affiliation(s)
- MengYue Wang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
| | - YaNan Zhang
- Shandong Co-Innovation Center of Classic TCM Formula, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
- Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
| | - SongLin Ni
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
| | - Mo Sun
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
| | - QiaoLan Wu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
| | - XiaoLin Wu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
| | - Qian Chen
- Shandong Co-Innovation Center of Classic TCM Formula, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China.
- Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China.
| | - ShiJun Wang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China.
- Shandong Co-Innovation Center of Classic TCM Formula, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China.
| |
Collapse
|
5
|
He K, Tao F, Lu Y, Fang M, Huang H, Zhou Y. The Role of HK2 in Tumorigenesis and Development: Potential for Targeted Therapy with Natural Products. Int J Med Sci 2025; 22:790-805. [PMID: 39991762 PMCID: PMC11843137 DOI: 10.7150/ijms.105553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 01/11/2025] [Indexed: 02/25/2025] Open
Abstract
Hexokinase 2 (HK2) is widely distributed in various tissues, particularly showing significantly elevated expression levels in tumor tissues. As the initial rate-limiting enzyme in the glycolysis process, HK2 is believed to directly participate in the metabolic reprogramming of tumor cells. This phenomenon, known as the "Warburg effect," provides the energy and substances necessary for the rapid proliferation, growth, and division of tumor cells. Furthermore, by enhancing glycolysis, HK2 exerts its influence on various metabolic pathways in tumor cells, such as pentose phosphate metabolism, glutamine metabolism, serine metabolism, and glycine metabolism, thereby playing a role in the occurrence and development of cancer. Therefore, HK2 represents a promising target for cancer therapy. Simultaneously, natural products with effects on inhibiting the expression or activity of HK2, have already been discovered to exhibit significant anticancer potential. Flavonoids, pentacyclic triterpenoids, phenolic compounds, and lignans constitute the majority of these natural products, directly inhibiting HK2 or indirectly downregulating it through protein kinase B (AKT), hypoxia-inducible factor 1 alpha (HIF-1α), and c-Myc signaling pathways. However, several challenges remain, such as further screening for natural products that directly target and inhibit HK2, optimizing the selection of natural product inhibitors for HK2, and elucidating the molecular mechanisms by which natural products indirectly inhibit HK2. In conclusion, the potential of targeting HK2 for cancer therapy is promising, and with these challenges addressed, natural products inhibiting HK2 will play an even greater role in the fight against cancer.
Collapse
Affiliation(s)
- Keren He
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Fangfang Tao
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Key Laboratory of Blood-Stasis-Toxin Syndrome, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yangyuxiao Lu
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Mengqi Fang
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Hong Huang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Key Laboratory of Blood-Stasis-Toxin Syndrome, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuan Zhou
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Key Laboratory of Blood-Stasis-Toxin Syndrome, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
6
|
Liu S, Yang Y, Hou X, Zhou N, Zhang B, Li W. Role for the F-box proteins in heart diseases. Pharmacol Res 2024; 210:107514. [PMID: 39577754 DOI: 10.1016/j.phrs.2024.107514] [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: 08/26/2024] [Revised: 10/27/2024] [Accepted: 11/19/2024] [Indexed: 11/24/2024]
Abstract
The maintenance of cardiac homeostasis necessitates proper protein turnover, which is regulated by the ubiquitin-proteasome system. F-box proteins are one type of E3 ubiquitin ligases, and accumulating evidence suggests that dysregulation of FBPs exacerbates heart diseases. Therefore, in this review, we summarized the F-box proteins present in the heart, which can be divided into three types based on their repeated sequences, namely FBXO (Fbxo32, Fbxo25, Fbxo44, Fbxo27 and Fbxo28), FBXW (Fbxw7 and Fbxw5), and FBXL (Fbxl1, Fbxl10, Fbxl16 and Fbxl2). Moreover, the physiological and pathological roles and the functional mechanisms of these F-box proteins were elucidated within the cardiac context, providing new theories and strategies for the prevention and treatment of heart diseases.
Collapse
Affiliation(s)
- Sa Liu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Institute of Clinical Pharmacy, Central South University, Changsha, Hunan 410011, China.
| | - Yuanying Yang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Institute of Clinical Pharmacy, Central South University, Changsha, Hunan 410011, China.
| | - Xingyuan Hou
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Institute of Clinical Pharmacy, Central South University, Changsha, Hunan 410011, China.
| | - Ni Zhou
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Institute of Clinical Pharmacy, Central South University, Changsha, Hunan 410011, China.
| | - Bikui Zhang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Institute of Clinical Pharmacy, Central South University, Changsha, Hunan 410011, China.
| | - Wenqun Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Institute of Clinical Pharmacy, Central South University, Changsha, Hunan 410011, China.
| |
Collapse
|
7
|
Cai XX, Huang ZF, Tu FY, Yu J. Impact and mechanism study of dioscin on biological characteristics of colorectal cancer cells. World J Gastrointest Oncol 2024; 16:4456-4467. [PMID: 39554744 PMCID: PMC11551639 DOI: 10.4251/wjgo.v16.i11.4456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/11/2024] [Accepted: 09/29/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is a considerable global health issue. Dioscin, a compound found in several medicinal plants, has shown potential anticancer effects. AIM To find the relationship between CRC cells (HCT116) and diosgenin and clarified their mechanisms of action. METHODS CRC cell line HCT116 was cultured by dividing cells into control and dioscin groups (dioscin + Jagged 1 group; Jagged 1 group, 5 μg/mL; and dioscin group, 2.5 μg/mL). The dioscin groups were given different concentrations of dioscin. Cell Counting Kit-8 was chosen for testing cell viability in different groups. Flow cytometry was established to undiscover the apoptosis rate of human liver cancer cell line 11. Real-time PCR as well as Western blot analyses were applied to reveal the expression levels of caspase-3, Notch, and other proteins. Transwell and scratch experiments were conducted to assess cell migration and invasion abilities. RESULTS This study indicated that dioscin restricted the growth of HCT116 cells, boosted cell apoptosis, and rose the Bax/Bcl-2 ratio as well as the expression of Caspase-3. Dioscin also inhibited physiological activities, for instance cell migration, and significantly reduced the expression levels of proteins for instance Notch1 (P < 0.05). Dioscin partially reversed the effects of Jagged 1. CONCLUSION Dioscin exerts a certain inhibitory effect on HCT116, and its mechanism of action may be linked, with the inhibition of the Notch1 signaling pathway.
Collapse
Affiliation(s)
- Xie-Xiao Cai
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, Zhejiang Province, China
| | - Zhen-Feng Huang
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, Zhejiang Province, China
| | - Fu-Yang Tu
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, Zhejiang Province, China
| | - Jie Yu
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, Zhejiang Province, China
| |
Collapse
|
8
|
Wang M, Qu L, Du X, Song P, Ng JPL, Wong VKW, Law BYK, Fu X. Natural Products and Derivatives Targeting Metabolic Reprogramming in Colorectal Cancer: A Comprehensive Review. Metabolites 2024; 14:490. [PMID: 39330497 PMCID: PMC11433951 DOI: 10.3390/metabo14090490] [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: 07/17/2024] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 09/28/2024] Open
Abstract
Metabolic reprogramming is a critical pathogenesis of colorectal cancer (CRC), referring to metabolic disorders that cancer cells make in response to the stimulating pressure. Metabolic reprogramming induces changes in genetic material and promotes CRC progression and has been proven to be an efficient target of CRC. As natural products have garnered interest due to notable pharmacological effects and potential in counteracting chemoresistance, an increasing body of research is delving into the impact of these natural products on the metabolic reprogramming associated with CRC. In this review, we collected published data from the Web of Science and PubMed, covering the period from January 1980 to October 2023. This article focuses on five central facets of metabolic alterations in cancer cells, glucose metabolism, mitochondrial oxidative phosphorylation (OXPHOS), amino acid metabolism, fatty acid synthesis, and nucleotide metabolism, to provide an overview of recent advancements in natural product interventions targeting metabolic reprogramming in CRC. Our analysis underscores the potential of natural products in disrupting the metabolic pathways of CRC, suggesting promising therapeutic targets for CRC and expanding treatment options for metabolic-associated ailments.
Collapse
Affiliation(s)
- Mengyu Wang
- Nehr’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China; (M.W.)
- Research Institute for Marine Traditional Chinese Medicine, Key Laboratory of Marine Traditional Chinese Medicine in Shandong Universities, Shandong Engineering and Technology Research Center on Omics of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Liqun Qu
- Research Institute for Marine Traditional Chinese Medicine, Key Laboratory of Marine Traditional Chinese Medicine in Shandong Universities, Shandong Engineering and Technology Research Center on Omics of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
- Qingdao Academy of Chinese Medical Sciences Shandong University of Traditional Chinese Medicine, Qingdao Key Laboratory of Research in Marine Traditional Chinese Medicine, Qingdao Key Technology Innovation Center of Marine Traditional Chinese Medicine’s Deep Development and Industrialization, Qingdao 266114, China
| | - Xinying Du
- Research Institute for Marine Traditional Chinese Medicine, Key Laboratory of Marine Traditional Chinese Medicine in Shandong Universities, Shandong Engineering and Technology Research Center on Omics of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
- Qingdao Academy of Chinese Medical Sciences Shandong University of Traditional Chinese Medicine, Qingdao Key Laboratory of Research in Marine Traditional Chinese Medicine, Qingdao Key Technology Innovation Center of Marine Traditional Chinese Medicine’s Deep Development and Industrialization, Qingdao 266114, China
| | - Peng Song
- Nehr’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China; (M.W.)
- Research Institute for Marine Traditional Chinese Medicine, Key Laboratory of Marine Traditional Chinese Medicine in Shandong Universities, Shandong Engineering and Technology Research Center on Omics of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Jerome P. L. Ng
- Nehr’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China; (M.W.)
| | - Vincent Kam Wai Wong
- Nehr’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China; (M.W.)
| | - Betty Yuen Kwan Law
- Nehr’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China; (M.W.)
| | - Xianjun Fu
- Research Institute for Marine Traditional Chinese Medicine, Key Laboratory of Marine Traditional Chinese Medicine in Shandong Universities, Shandong Engineering and Technology Research Center on Omics of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
- Qingdao Academy of Chinese Medical Sciences Shandong University of Traditional Chinese Medicine, Qingdao Key Laboratory of Research in Marine Traditional Chinese Medicine, Qingdao Key Technology Innovation Center of Marine Traditional Chinese Medicine’s Deep Development and Industrialization, Qingdao 266114, China
| |
Collapse
|
9
|
Khorshid Sokhangouy S, Alizadeh F, Lotfi M, Sharif S, Ashouri A, Yoosefi Y, Bozorg Qomi S, Abbaszadegan MR. Recent advances in CRISPR-Cas systems for colorectal cancer research and therapeutics. Expert Rev Mol Diagn 2024; 24:677-702. [PMID: 39132997 DOI: 10.1080/14737159.2024.2388777] [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: 12/20/2023] [Accepted: 07/28/2024] [Indexed: 08/13/2024]
Abstract
INTRODUCTION Colon cancer, ranked as the fourth leading global cause of cancer death, exhibits a complex progression marked by genetic variations. Over the past decade, the utilization of diverse CRISPR systems has propelled accelerated research into colorectal cancer (CRC) treatment. AREAS COVERED CRISPR/Cas9, a key player in this research, identifies new oncogenes, tumor suppressor genes (TSGs), and drug-resistance genes. Additionally, it facilitates the construction of experimental models, conducts genome-wide library screening, and develops new therapeutic targets, especially for targeted knockout in vivo or molecular targeted drug delivery, contributing to personalized treatments and significantly enhancing the care of colon cancer patients. In this review, we provide insights into the mechanism of the CRISPR/Cas9 system, offering a comprehensive exploration of its applications in CRC, spanning screening, modeling, gene functions, diagnosis, and gene therapy. While acknowledging its transformative potential, the article highlights the challenges and limitations of CRISPR systems. EXPERT OPINION The application of CRISPR/Cas9 in CRC research provides a promising avenue for personalized treatments. Its potential for identifying key genes and enabling experimental models and genome-wide screening enhances patient care. This review underscores the significance of CRISPR-Cas9 gene editing technology across basic research, diagnosis, and the treatment landscape of colon cancer.
Collapse
Affiliation(s)
| | - Farzaneh Alizadeh
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Malihe Lotfi
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Samaneh Sharif
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Atefeh Ashouri
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Yasamin Yoosefi
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saeed Bozorg Qomi
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Reza Abbaszadegan
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
10
|
Chen Z, Zhang J, Gao S, Jiang Y, Qu M, Gu J, Wu H, Nan K, Zhang H, Wang J, Chen W, Miao C. Suppression of Skp2 contributes to sepsis-induced acute lung injury by enhancing ferroptosis through the ubiquitination of SLC3A2. Cell Mol Life Sci 2024; 81:325. [PMID: 39079969 PMCID: PMC11335248 DOI: 10.1007/s00018-024-05348-3] [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: 03/24/2024] [Revised: 06/19/2024] [Accepted: 07/03/2024] [Indexed: 08/22/2024]
Abstract
Sepsis is a life-threatening organ dysfunction caused by a dysregulated host response to infection. The inflammatory cytokine storm causes systemic organ damage, especially acute lung injury in sepsis. In this study, we found that the expression of S-phase kinase-associated protein 2 (Skp2) was significantly decreased in sepsis-induced acute lung injury (ALI). Sepsis activated the MEK/ERK pathway and inhibited Skp2 expression in the pulmonary epithelium, resulting in a reduction of K48 ubiquitination of solute carrier family 3 member 2 (SLC3A2), thereby impairing its membrane localization and cystine/glutamate exchange function. Consequently, the dysregulated intracellular redox reactions induced ferroptosis in pulmonary epithelial cells, leading to lung injury. Finally, we demonstrated that intravenous administration of Skp2 mRNA-encapsulating lipid nanoparticles (LNPs) inhibited ferroptosis in the pulmonary epithelium and alleviated lung injury in septic mice. Taken together, these data provide an innovative understanding of the underlying mechanisms of sepsis-induced ALI and a promising therapeutic strategy for sepsis.
Collapse
Affiliation(s)
- Zhaoyuan Chen
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Jie Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Shenjia Gao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Yi Jiang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Mengdi Qu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Jiahui Gu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Han Wu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Ke Nan
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Hao Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Jun Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Wankun Chen
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China.
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China.
- Department of Anesthesiology, QingPu Branch of Zhongshan Hospital Affiliated to Fudan University, 1158# Gongyuan Dong Road, Shanghai, 201700, China.
| | - Changhong Miao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China.
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China.
| |
Collapse
|
11
|
Alhasan BA, Morozov AV, Guzhova IV, Margulis BA. The ubiquitin-proteasome system in the regulation of tumor dormancy and recurrence. Biochim Biophys Acta Rev Cancer 2024; 1879:189119. [PMID: 38761982 DOI: 10.1016/j.bbcan.2024.189119] [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: 01/01/2024] [Revised: 05/12/2024] [Accepted: 05/15/2024] [Indexed: 05/20/2024]
Abstract
Tumor recurrence is a mechanism triggered in sparse populations of cancer cells that usually remain in a quiescent state after strict stress and/or therapeutic factors, which is affected by a variety of autocrine and microenvironmental cues. Despite thorough investigations, the biology of dormant and/or cancer stem cells is still not fully elucidated, as for the mechanisms of their reawakening, while only the major molecular patterns driving the relapse process have been identified to date. These molecular patterns profoundly interfere with the elements of cellular proteostasis systems that support the efficiency of the recurrence process. As a major proteostasis machinery, we review the role of the ubiquitin-proteasome system (UPS) in tumor cell dormancy and reawakening, devoting particular attention to the functions of its components, E3 ligases, deubiquitinating enzymes and proteasomes in cancer recurrence. We demonstrate how UPS components functionally or mechanistically interact with the pivotal proteins implicated in the recurrence program and reveal that modulators of the UPS hold promise to become an efficient adjuvant therapy for eradicating refractory tumor cells to impede tumor relapse.
Collapse
Affiliation(s)
- Bashar A Alhasan
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia.
| | - Alexey V Morozov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Street 32, 119991 Moscow, Russia.
| | - Irina V Guzhova
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia.
| | - Boris A Margulis
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia.
| |
Collapse
|
12
|
He YF, Liu YP, Liao JZ, Gan Y, Li X, Wang RR, Wang F, Zhou J, Zhou L. Xanthohumol Promotes Skp2 Ubiquitination Leading to the Inhibition of Glycolysis and Tumorigenesis in Ovarian Cancer. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:865-884. [PMID: 38790085 DOI: 10.1142/s0192415x24500356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2024]
Abstract
Ovarian cancer is a common, highly lethal tumor. Herein, we reported that S-phase kinase-associated protein 2 (Skp2) is essential for the growth and aerobic glycolysis of ovarian cancer cells. Skp2 was upregulated in ovarian cancer tissues and associated with poor clinical outcomes. Using a customized natural product library screening, we found that xanthohumol inhibited aerobic glycolysis and cell viability of ovarian cancer cells. Xanthohumol facilitated the interaction between E3 ligase Cdh1 and Skp2 and promoted the Ub-K48-linked polyubiquitination of Skp2 and degradation. Cdh1 depletion reversed xanthohumol-induced Skp2 downregulation, enhancing HK2 expression and glycolysis in ovarian cancer cells. Finally, a xenograft tumor model was employed to examine the antitumor efficacy of xanthohumol in vivo. Collectively, we discovered that xanthohumol promotes the binding between Skp2 and Cdh1 to suppress the Skp2/AKT/HK2 signal pathway and exhibits potential antitumor activity for ovarian cancer cells.
Collapse
Affiliation(s)
- Yi-Fu He
- Department of Obstetrics and Gynecology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, P. R. China
| | - Yi-Ping Liu
- Department of Oncology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, P. R. China
| | - Jin-Zhuang Liao
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha 410013, P. R. China
| | - Yu Gan
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha 410013, P. R. China
| | - Xiaoying Li
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha 410013, P. R. China
| | - Rui-Rui Wang
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha 410013, P. R. China
| | - Fang Wang
- Tengzhou Central People's Hospital, Tengzhou 277500, P. R. China
| | - Jun Zhou
- Department of Medical Science Research Center, Xiangya Hospital, Central South University, Changsha 410008, P. R. China
| | - Li Zhou
- Department of Pathology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, P. R. China
| |
Collapse
|
13
|
Han S, Chen Y, Huang Y, Jin L, Ma Y. Arecoline promotes Akt-c-Myc-driven aerobic glycolysis in esophageal epithelial cells. ENVIRONMENTAL TOXICOLOGY 2024; 39:2794-2802. [PMID: 38282581 DOI: 10.1002/tox.24159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 01/02/2024] [Accepted: 01/18/2024] [Indexed: 01/30/2024]
Abstract
Aerobic glycolysis is a typical metabolic rearrangement for tumorigenesis. Arecoline is of explicit carcinogenicity, numerous works demonstrate its mutagenicity, genotoxicity, and cytotoxicity. However, the effects of arecoline on aerobic glycolysis of esophageal epithelial cells remain unclear. In the present study, 5 μM arecoline efficiently increased HK2 expression to induce aerobic glycolysis in Het-1A-Are and NE2-Are cells. The mechanistic analysis showed that arecoline activated the Akt-c-Myc signaling pathway and reduced the GSK3β-mediated phosphorylation of c-Myc on Thr58 to prevent its ubiquitination and destruction, subsequently promoting HK2 transcription and expression. Taken together, these results suggest that arecoline can induce aerobic glycolysis of esophageal epithelial cells and further confirm that arecoline is a carcinogen harmful to human health.
Collapse
Affiliation(s)
- Shuangze Han
- Department of Cardiothoracic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Ultrasound, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingji Chen
- Department of Cardiothoracic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yu Huang
- Department of Cardiothoracic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Longyu Jin
- Department of Cardiothoracic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yuchao Ma
- Department of Cardiothoracic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
14
|
Rong Z, Zheng K, Chen J, Jin X. The cross talk of ubiquitination and chemotherapy tolerance in colorectal cancer. J Cancer Res Clin Oncol 2024; 150:154. [PMID: 38521878 PMCID: PMC10960765 DOI: 10.1007/s00432-024-05659-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 02/20/2024] [Indexed: 03/25/2024]
Abstract
Ubiquitination, a highly adaptable post-translational modification, plays a pivotal role in maintaining cellular protein homeostasis, encompassing cancer chemoresistance-associated proteins. Recent findings have indicated a potential correlation between perturbations in the ubiquitination process and the emergence of drug resistance in CRC cancer. Consequently, numerous studies have spurred the advancement of compounds specifically designed to target ubiquitinates, offering promising prospects for cancer therapy. In this review, we highlight the role of ubiquitination enzymes associated with chemoresistance to chemotherapy via the Wnt/β-catenin signaling pathway, epithelial-mesenchymal transition (EMT), and cell cycle perturbation. In addition, we summarize the application and role of small compounds that target ubiquitination enzymes for CRC treatment, along with the significance of targeting ubiquitination enzymes as potential cancer therapies.
Collapse
Affiliation(s)
- Ze Rong
- Department of Chemoradiotherapy, the Affiliated People's Hospital of Ningbo University, Ningbo, 315040, China.
| | - Kaifeng Zheng
- Department of Chemoradiotherapy, the Affiliated People's Hospital of Ningbo University, Ningbo, 315040, China
| | - Jun Chen
- Department of Chemoradiotherapy, the Affiliated People's Hospital of Ningbo University, Ningbo, 315040, China.
| | - Xiaofeng Jin
- Department of Chemoradiotherapy, the Affiliated People's Hospital of Ningbo University, Ningbo, 315040, China.
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo, 315211, China.
| |
Collapse
|
15
|
William JNG, Dhar R, Gundamaraju R, Sahoo OS, Pethusamy K, Raj AFPAM, Ramasamy S, Alqahtani MS, Abbas M, Karmakar S. SKping cell cycle regulation: role of ubiquitin ligase SKP2 in hematological malignancies. Front Oncol 2024; 14:1288501. [PMID: 38559562 PMCID: PMC10978726 DOI: 10.3389/fonc.2024.1288501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 02/15/2024] [Indexed: 04/04/2024] Open
Abstract
SKP2 (S-phase kinase-associated protein 2) is a member of the F-box family of substrate-recognition subunits in the SCF ubiquitin-protein ligase complexes. It is associated with ubiquitin-mediated degradation in the mammalian cell cycle components and other target proteins involved in cell cycle progression, signal transduction, and transcription. Being an oncogene in solid tumors and hematological malignancies, it is frequently associated with drug resistance and poor disease outcomes. In the current review, we discussed the novel role of SKP2 in different hematological malignancies. Further, we performed a limited in-silico analysis to establish the involvement of SKP2 in a few publicly available cancer datasets. Interestingly, our study identified Skp2 expression to be altered in a cancer-specific manner. While it was found to be overexpressed in several cancer types, few cancer showed a down-regulation in SKP2. Our review provides evidence for developing novel SKP2 inhibitors in hematological malignancies. We also investigated the effect of SKP2 status on survival and disease progression. In addition, the role of miRNA and its associated families in regulating Skp2 expression was explored. Subsequently, we predicted common miRNAs against Skp2 genes by using miRNA-predication tools. Finally, we discussed current approaches and future prospective approaches to target the Skp2 gene by using different drugs and miRNA-based therapeutics applications in translational research.
Collapse
Affiliation(s)
- Jonahunnatha Nesson George William
- Department of Medical, Oral and Biotechnological Sciences (DSMOB), Ageing Research Center and Translational Medicine-CeSI-MeT, “G. d’Annunzio” University Chieti-Pescara, Chieti, Italy
| | - Ruby Dhar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Rohit Gundamaraju
- ER Stress and Intestinal Mucosal Biology Lab, School of Health Sciences, University of Tasmania, Launceston, TAS, Australia
| | - Om Saswat Sahoo
- Department of Biotechnology, National Institute of Technology, Durgapur, India
| | - Karthikeyan Pethusamy
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | | | - Subbiah Ramasamy
- Cardiac Metabolic Disease Laboratory, Department Of Biochemistry, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | - Mohammed S. Alqahtani
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
- BioImaging Unit, Space Research Centre, University of Leicester, Leicester, United Kingdom
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, Abha, Saudi Arabia
| | - Subhradip Karmakar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
16
|
Runa F, Ortiz-Soto G, de Barros NR, Kelber JA. Targeting SMAD-Dependent Signaling: Considerations in Epithelial and Mesenchymal Solid Tumors. Pharmaceuticals (Basel) 2024; 17:326. [PMID: 38543112 PMCID: PMC10975212 DOI: 10.3390/ph17030326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/19/2024] [Accepted: 02/23/2024] [Indexed: 04/01/2024] Open
Abstract
SMADs are the canonical intracellular effector proteins of the TGF-β (transforming growth factor-β). SMADs translocate from plasma membrane receptors to the nucleus regulated by many SMAD-interacting proteins through phosphorylation and other post-translational modifications that govern their nucleocytoplasmic shuttling and subsequent transcriptional activity. The signaling pathway of TGF-β/SMAD exhibits both tumor-suppressing and tumor-promoting phenotypes in epithelial-derived solid tumors. Collectively, the pleiotropic nature of TGF-β/SMAD signaling presents significant challenges for the development of effective cancer therapies. Here, we review preclinical studies that evaluate the efficacy of inhibitors targeting major SMAD-regulating and/or -interacting proteins, particularly enzymes that may play important roles in epithelial or mesenchymal compartments within solid tumors.
Collapse
Affiliation(s)
- Farhana Runa
- Department of Biology, California State University Northridge, Northridge, CA 91330, USA
| | | | | | - Jonathan A Kelber
- Department of Biology, California State University Northridge, Northridge, CA 91330, USA
- Department of Biology, Baylor University, Waco, TX 76706, USA
| |
Collapse
|
17
|
Feng T, Wang P, Zhang X. Skp2: A critical molecule for ubiquitination and its role in cancer. Life Sci 2024; 338:122409. [PMID: 38184273 DOI: 10.1016/j.lfs.2023.122409] [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: 10/09/2023] [Revised: 12/24/2023] [Accepted: 12/29/2023] [Indexed: 01/08/2024]
Abstract
The ubiquitin-proteasome system (UPS) is a multi-step process that serves as the primary pathway for protein degradation within cells. UPS activity also plays a crucial role in regulating various life processes, including the cell cycle, signal transduction, DNA repair, and others. The F-box protein Skp2, a crucial member of the UPS, plays a central role in the development of various diseases. Skp2 controls cancer cell growth and drug resistance by ubiquitinating modifications to a variety of proteins. This review emphasizes the multifaceted role of Skp2 in a wide range of cancers and the mechanisms involved, highlighting the potential of Skp2 as a therapeutic target in cancer. Additionally, we describe the impactful influence exerted by Skp2 in various other diseases beyond cancer.
Collapse
Affiliation(s)
- Tianyang Feng
- The Fourth Affiliated Hospital of China Medical University, Department of Urology, Shenyang 110032, China; Liaoning Provincial Key Laboratory of Basic Research for Bladder Diseases, Shenyang 110000, China
| | - Ping Wang
- The Fourth Affiliated Hospital of China Medical University, Department of Urology, Shenyang 110032, China; Liaoning Provincial Key Laboratory of Basic Research for Bladder Diseases, Shenyang 110000, China
| | - Xiling Zhang
- The Fourth Affiliated Hospital of China Medical University, Department of Urology, Shenyang 110032, China; Liaoning Provincial Key Laboratory of Basic Research for Bladder Diseases, Shenyang 110000, China.
| |
Collapse
|
18
|
Wang R, Li X, Gan Y, Liao J, Han S, Li W, Deng G. Dioscin inhibits non-small cell lung cancer cells and activates apoptosis by downregulation of Survivin. J Cancer 2024; 15:1366-1377. [PMID: 38356707 PMCID: PMC10861826 DOI: 10.7150/jca.89831] [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: 09/04/2023] [Accepted: 12/22/2023] [Indexed: 02/16/2024] Open
Abstract
Human malignancies exhibit elevated levels of survivin, and have been linked to poor prognosis. Targeting survivin expression is a promising therapeutic strategy against cancer cells. Natural compounds have become a hot topic in research due to their non-toxic, non-invasive, and efficient treatment of multiple diseases. In this current investigation, it was discovered that Dioscin, as a natural compound, exerted profound antitumor activity against NSCLC cell lines, inhibiting NSCLC cell viability and promoting apoptosis. Further mechanistic studies showed that Dioscin promoted ubiquitination-mediated survivin degradation via strengthening the interaction between survivin and the E3 ubiquitin ligase Fbxl7. Furthermore, Dioscin exhibited a strong tumor suppressive effect in xenograft tumor models, and Dioscin treatment led to a notable decrease in tumor volume and weight. Based on our findings, Dioscin is expected to be a potential antitumor agent for non-small cell lung cancer treatment.
Collapse
Affiliation(s)
- Ruirui Wang
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
| | - Xiaoying Li
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
| | - Yu Gan
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
| | - Jinzhuang Liao
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
| | - Shuangze Han
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
- Department of Ultrasound, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Wei Li
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
| | - Gaoyan Deng
- Department of Thoracic Surgery, Hunan Chest Hospital, Changsha 410013, Hunan, China
| |
Collapse
|
19
|
Yang ZY, Zhao YW, Xue JR, Guo R, Zhao Z, Liu HD, Ren ZG, Shi M. Thioridazine reverses trastuzumab resistance in gastric cancer by inhibiting S-phase kinase associated protein 2-mediated aerobic glycolysis. World J Gastroenterol 2023; 29:5974-5987. [PMID: 38130998 PMCID: PMC10731152 DOI: 10.3748/wjg.v29.i45.5974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 10/19/2023] [Accepted: 11/17/2023] [Indexed: 12/01/2023] Open
Abstract
BACKGROUND Trastuzumab constitutes the fundamental component of initial therapy for patients with advanced human epidermal growth factor receptor 2 (HER-2)-positive gastric cancer (GC). However, the efficacy of this treatment is hindered by substantial challenges associated with both primary and acquired drug resistance. While S-phase kinase associated protein 2 (Skp2) overexpression has been implicated in the malignant progression of GC, its role in regulating trastuzumab resistance in this context remains uncertain. Despite the numerous studies investigating Skp2 inhibitors among small molecule compounds and natural products, there has been a lack of successful commercialization of drugs specifically targeting Skp2. AIM To discover a Skp2 blocker among currently available medications and develop a therapeutic strategy for HER2-positive GC patients who have experienced progression following trastuzumab-based treatment. METHODS Skp2 exogenous overexpression plasmids and small interfering RNA vectors were utilized to investigate the correlation between Skp2 expression and trastuzumab resistance in GC cells. Q-PCR, western blot, and immunohistochemical analyses were conducted to evaluate the regulatory effect of thioridazine on Skp2 expression. A cell counting kit-8 assay, flow cytometry, a amplex red glucose/glucose oxidase assay kit, and a lactate assay kit were utilized to measure the proliferation, apoptosis, and glycolytic activity of GC cells in vitro. A xenograft model established with human GC in nude mice was used to assess thioridazine's effectiveness in vivo. RESULTS The expression of Skp2 exhibited a negative correlation with the sensitivity of HER2-positive GC cells to trastuzumab. Thioridazine demonstrated the ability to directly bind to Skp2, resulting in a reduction in Skp2 expression at both the transcriptional and translational levels. Moreover, thioridazine effectively inhibited cell proliferation, exhibited antiapoptotic properties, and decreased the glucose uptake rate and lactate production by suppressing Skp2/protein kinase B/mammalian target of rapamycin/glucose transporter type 1 signaling pathways. The combination of thioridazine with either trastuzumab or lapatinib exhibited a more pronounced anticancer effect in vivo, surpassing the efficacy of either monotherapy. CONCLUSION Thioridazine demonstrates promising outcomes in preclinical GC models and offers a novel therapeutic approach for addressing trastuzumab resistance, particularly when used in conjunction with lapatinib. This compound has potential benefits for patients with Skp2-proficient tumors.
Collapse
Affiliation(s)
- Zheng-Yan Yang
- Department of Pathology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, Henan Province, China
| | - Yi-Wei Zhao
- Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng 475004, Henan Province, China
| | - Jing-Rui Xue
- Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng 475004, Henan Province, China
| | - Ran Guo
- Department of Pathology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, Henan Province, China
| | - Zhi Zhao
- Department of Pathology, Henan University-affiliated Zhengzhou Yihe Hospital, Zhengzhou 450000, Henan Province, China
| | - Han-Di Liu
- Department of Pathology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, Henan Province, China
| | - Zhi-Guang Ren
- Department of Pathology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, Henan Province, China
- Key Laboratory of Clinical Resources Translation, The First Affiliated Hospital, Henan University, Kaifeng 475004, Henan Province, China
| | - Ming Shi
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, Jiangsu Province, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, China
| |
Collapse
|
20
|
Zhan L, Su F, Li Q, Wen Y, Wei F, He Z, Chen X, Yin X, Wang J, Cai Y, Gong Y, Chen Y, Ma X, Zeng J. Phytochemicals targeting glycolysis in colorectal cancer therapy: effects and mechanisms of action. Front Pharmacol 2023; 14:1257450. [PMID: 37693915 PMCID: PMC10484417 DOI: 10.3389/fphar.2023.1257450] [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: 07/12/2023] [Accepted: 08/10/2023] [Indexed: 09/12/2023] Open
Abstract
Colorectal cancer (CRC) is the third most common malignant tumor in the world, and it is prone to recurrence and metastasis during treatment. Aerobic glycolysis is one of the main characteristics of tumor cell metabolism in CRC. Tumor cells rely on glycolysis to rapidly consume glucose and to obtain more lactate and intermediate macromolecular products so as to maintain growth and proliferation. The regulation of the CRC glycolysis pathway is closely associated with several signal transduction pathways and transcription factors including phosphatidylinositol 3-kinases/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR), adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK), hypoxia-inducible factor-1 (HIF-1), myc, and p53. Targeting the glycolytic pathway has become one of the key research aspects in CRC therapy. Many phytochemicals were shown to exert anti-CRC activity by targeting the glycolytic pathway. Here, we review the effects and mechanisms of phytochemicals on CRC glycolytic pathways, providing a new method of drug development.
Collapse
Affiliation(s)
- Lu Zhan
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fangting Su
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiang Li
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yueqiang Wen
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Feng Wei
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhelin He
- Guang’an Hospital of Traditional Chinese Medicine, Guang’an, China
| | - Xiaoyan Chen
- Guang’an Hospital of Traditional Chinese Medicine, Guang’an, China
| | - Xiang Yin
- Guang’an Hospital of Traditional Chinese Medicine, Guang’an, China
| | - Jian Wang
- Guang’an Hospital of Traditional Chinese Medicine, Guang’an, China
| | - Yilin Cai
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuxia Gong
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu Chen
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinhao Zeng
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
21
|
Bouabdallah S, Al-Maktoum A, Amin A. Steroidal Saponins: Naturally Occurring Compounds as Inhibitors of the Hallmarks of Cancer. Cancers (Basel) 2023; 15:3900. [PMID: 37568716 PMCID: PMC10417465 DOI: 10.3390/cancers15153900] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/21/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
Cancer is a global health burden responsible for an exponentially growing number of incidences and mortalities, regardless of the significant advances in its treatment. The identification of the hallmarks of cancer is a major milestone in understanding the mechanisms that drive cancer initiation, development, and progression. In the past, the hallmarks of cancer have been targeted to effectively treat various types of cancers. These conventional cancer drugs have shown significant therapeutic efficacy but continue to impose unfavorable side effects on patients. Naturally derived compounds are being tested in the search for alternative anti-cancer drugs. Steroidal saponins are a group of naturally occurring compounds that primarily exist as secondary metabolites in plant species. Recent studies have suggested that steroidal saponins possess significant anti-cancer capabilities. This review aims to summarize the recent findings on steroidal saponins as inhibitors of the hallmarks of cancer and covers key studies published between the years 2014 and 2024. It is reported that steroidal saponins effectively inhibit the hallmarks of cancer, but poor bioavailability and insufficient preclinical studies limit their utilization.
Collapse
Affiliation(s)
- Salwa Bouabdallah
- Theranostic Biomarkers, LR23ES02, Faculty of Medicine of Tunis, Université Tunis El Manar, Tunis 1006, Tunisia
| | - Amna Al-Maktoum
- Biology Department, College of Science, United Arab Emirates University, Al Ain 15551, United Arab Emirates;
| | - Amr Amin
- Biology Department, College of Science, United Arab Emirates University, Al Ain 15551, United Arab Emirates;
| |
Collapse
|
22
|
Koo SY, Park EJ, Noh HJ, Jo SM, Ko BK, Shin HJ, Lee CW. Ubiquitination Links DNA Damage and Repair Signaling to Cancer Metabolism. Int J Mol Sci 2023; 24:ijms24098441. [PMID: 37176148 PMCID: PMC10179089 DOI: 10.3390/ijms24098441] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023] Open
Abstract
Changes in the DNA damage response (DDR) and cellular metabolism are two important factors that allow cancer cells to proliferate. DDR is a set of events in which DNA damage is recognized, DNA repair factors are recruited to the site of damage, the lesion is repaired, and cellular responses associated with the damage are processed. In cancer, DDR is commonly dysregulated, and the enzymes associated with DDR are prone to changes in ubiquitination. Additionally, cellular metabolism, especially glycolysis, is upregulated in cancer cells, and enzymes in this metabolic pathway are modulated by ubiquitination. The ubiquitin-proteasome system (UPS), particularly E3 ligases, act as a bridge between cellular metabolism and DDR since they regulate the enzymes associated with the two processes. Hence, the E3 ligases with high substrate specificity are considered potential therapeutic targets for treating cancer. A number of small molecule inhibitors designed to target different components of the UPS have been developed, and several have been tested in clinical trials for human use. In this review, we discuss the role of ubiquitination on overall cellular metabolism and DDR and confirm the link between them through the E3 ligases NEDD4, APC/CCDH1, FBXW7, and Pellino1. In addition, we present an overview of the clinically important small molecule inhibitors and implications for their practical use.
Collapse
Affiliation(s)
- Seo-Young Koo
- Department of Molecular Cell Biology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea
| | - Eun-Ji Park
- Department of Molecular Cell Biology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea
| | - Hyun-Ji Noh
- Department of Molecular Cell Biology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea
| | - Su-Mi Jo
- Department of Molecular Cell Biology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea
| | - Bo-Kyoung Ko
- Department of Molecular Cell Biology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea
| | - Hyun-Jin Shin
- Team of Radiation Convergence Research, Korea Institute of Radiological & Medical Sciences, Seoul 01812, Republic of Korea
| | - Chang-Woo Lee
- Department of Molecular Cell Biology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea
- SKKU Institute for Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
23
|
Johnson MS, Cook JG. Cell cycle exits and U-turns: Quiescence as multiple reversible forms of arrest. Fac Rev 2023; 12:5. [PMID: 36923701 PMCID: PMC10009890 DOI: 10.12703/r/12-5] [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] [Indexed: 03/10/2023] Open
Abstract
Cell proliferation control is essential during development and for maintaining adult tissues. Loss of that control promotes not only oncogenesis when cells proliferate inappropriately but also developmental abnormalities or degeneration when cells fail to proliferate when and where needed. To ensure that cells are produced at the right place and time, an intricate balance of pro-proliferative and anti-proliferative signals impacts the probability that cells undergo cell cycle exit to quiescence, or G0 phase. This brief review describes recent advances in our understanding of how and when quiescence is initiated and maintained in mammalian cells. We highlight the growing appreciation for quiescence as a collection of context-dependent distinct states.
Collapse
Affiliation(s)
- Martha Sharisha Johnson
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, NC, USA
| | - Jeanette Gowen Cook
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, NC, USA
| |
Collapse
|
24
|
Sampson C, Wang Q, Otkur W, Zhao H, Lu Y, Liu X, Piao H. The roles of E3 ubiquitin ligases in cancer progression and targeted therapy. Clin Transl Med 2023; 13:e1204. [PMID: 36881608 PMCID: PMC9991012 DOI: 10.1002/ctm2.1204] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 02/07/2023] [Accepted: 02/13/2023] [Indexed: 03/08/2023] Open
Abstract
Ubiquitination is one of the most important post-translational modifications which plays a significant role in conserving the homeostasis of cellular proteins. In the ubiquitination process, ubiquitin is conjugated to target protein substrates for degradation, translocation or activation, dysregulation of which is linked to several diseases including various types of cancers. E3 ubiquitin ligases are regarded as the most influential ubiquitin enzyme owing to their ability to select, bind and recruit target substrates for ubiquitination. In particular, E3 ligases are pivotal in the cancer hallmarks pathways where they serve as tumour promoters or suppressors. The specificity of E3 ligases coupled with their implication in cancer hallmarks engendered the development of compounds that specifically target E3 ligases for cancer therapy. In this review, we highlight the role of E3 ligases in cancer hallmarks such as sustained proliferation via cell cycle progression, immune evasion and tumour promoting inflammation, and in the evasion of apoptosis. In addition, we summarise the application and the role of small compounds that target E3 ligases for cancer treatment along with the significance of targeting E3 ligases as potential cancer therapy.
Collapse
Affiliation(s)
- Chibuzo Sampson
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina
- University of Chinese Academy of SciencesBeijingChina
| | - Qiuping Wang
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina
| | - Wuxiyar Otkur
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina
| | - Haifeng Zhao
- Department of OrthopedicsDalian Second People's HospitalDalianChina
| | - Yun Lu
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina
- Department of StomatologyDalian Medical UniversityDalianChina
| | - Xiaolong Liu
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina
| | - Hai‐long Piao
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina
- University of Chinese Academy of SciencesBeijingChina
| |
Collapse
|
25
|
Zhang J, Zou S, Fang L. Metabolic reprogramming in colorectal cancer: regulatory networks and therapy. Cell Biosci 2023; 13:25. [PMID: 36755301 PMCID: PMC9906896 DOI: 10.1186/s13578-023-00977-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 02/01/2023] [Indexed: 02/10/2023] Open
Abstract
With high prevalence and mortality, together with metabolic reprogramming, colorectal cancer is a leading cause of cancer-related death. Metabolic reprogramming gives tumors the capacity for long-term cell proliferation, making it a distinguishing feature of cancer. Energy and intermediate metabolites produced by metabolic reprogramming fuel the rapid growth of cancer cells. Aberrant metabolic enzyme-mediated tumor metabolism is regulated at multiple levels. Notably, tumor metabolism is affected by nutrient levels, cell interactions, and transcriptional and posttranscriptional regulation. Understanding the crosstalk between metabolic enzymes and colorectal carcinogenesis factors is particularly important to advance research for targeted cancer therapy strategies via the investigation into the aberrant regulation of metabolic pathways. Hence, the abnormal roles and regulation of metabolic enzymes in recent years are reviewed in this paper, which provides an overview of targeted inhibitors for targeting metabolic enzymes in colorectal cancer that have been identified through tumor research or clinical trials.
Collapse
Affiliation(s)
- Jieping Zhang
- grid.12981.330000 0001 2360 039XDepartment of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-Sen University, 26 Yuanchun Er Heng Road, Guangzhou, 510655 Guangdong China ,Guangdong Institute of Gastroenterology, Guangzhou, 510655 China
| | - Shaomin Zou
- grid.12981.330000 0001 2360 039XDepartment of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-Sen University, 26 Yuanchun Er Heng Road, Guangzhou, 510655 Guangdong China ,Guangdong Institute of Gastroenterology, Guangzhou, 510655 China
| | - Lekun Fang
- Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-Sen University, 26 Yuanchun Er Heng Road, Guangzhou, 510655, Guangdong, China. .,Guangdong Institute of Gastroenterology, Guangzhou, 510655, China.
| |
Collapse
|
26
|
Kou Y, Sun Q, Zhu R, Lin Z, Li Z, Xu H, Feng X, Liu Y. Dioscin induces M1 macrophage polarization through Connexin-43 Channels in Tumor-associated-macrophages-mediated melanoma metastasis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 109:154559. [PMID: 36610151 DOI: 10.1016/j.phymed.2022.154559] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 11/01/2022] [Accepted: 11/19/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND Tumor-associated macrophages (TAMs) are important constituent parts of tumor microenvironment that connected with tumor metastasis in melanoma. Connexin 43 (Cx43) was expressed in all the immune cells which modulated different aspects of immune response. However, the concrete molecular mechanism maintains unclear. PURPOSE The study aimed to find a natural drug monomer effectively reversed the polarity of tumor-associated macrophages inhibiting melanoma metastasis and improving survival time. METHODS Flow cytometry was used to determine the effects of dioscin on the macrophage phenotype. Western bolt and ELISA were performed to explore the underlying mechanism of dioscin and a co-culture experiment in vitro was applied to assess the role of dioscin on TAMs-mediated melanoma proliferation, invasion and migration. Moreover, in vivo melanoma metastasis models were established for examining effects of dioscin on TAMs-mediated melanoma metastasis. RESULTS Dioscin repolarized macrophages from M2 towards M1-like phenotype. Dioscin suppressed M2-like phenotype macrophages through enhanced the expression and transport function of Cx43. Furthermore, the stimulation IFN-γ/STAT1 pathway and suppression IL-4/JAK2/STAT3 pathway were major mechanism of dioscin. Importantly, dioscin suppressed Cx43G21R mutation TAMs induced proliferation, invasion, migration and metastasis of melanoma cells. It worthily noting that dioscin ameliorated tumor-associated-macrophages-mediated melanoma metastasis in vitro and vivo. CONCLUSION Dioscin re-polarized macrophages from M2 to M1 phenotype through activation of Cx43-gap-junction-intercellular-communications (Cx43-GJs)/IFN-γ/STAT1 pathway and inhibition of Cx43-GJs/IL-4/JAK2/STAT3 suppressing migration, invasion and metastasis of melanoma, which provided a theoretical and experimental basis for treating melanoma metastasis.
Collapse
Affiliation(s)
- Yu Kou
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225000, Jiangsu, China; The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225000, Jiangsu, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medcine for Prevention and Treatment of Senile Diseases, Medical College of Yangzhou University, Yangzhou 225000, Jiangsu, China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225000, Jiangsu, China.
| | - Qidi Sun
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225000, Jiangsu, China; The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225000, Jiangsu, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medcine for Prevention and Treatment of Senile Diseases, Medical College of Yangzhou University, Yangzhou 225000, Jiangsu, China
| | - Rentao Zhu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225000, Jiangsu, China; The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225000, Jiangsu, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medcine for Prevention and Treatment of Senile Diseases, Medical College of Yangzhou University, Yangzhou 225000, Jiangsu, China
| | - Zhijie Lin
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225000, Jiangsu, China
| | - Zhuoqun Li
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225000, Jiangsu, China; The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225000, Jiangsu, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medcine for Prevention and Treatment of Senile Diseases, Medical College of Yangzhou University, Yangzhou 225000, Jiangsu, China
| | - Hairong Xu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225000, Jiangsu, China; The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225000, Jiangsu, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medcine for Prevention and Treatment of Senile Diseases, Medical College of Yangzhou University, Yangzhou 225000, Jiangsu, China
| | - Xuan Feng
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225000, Jiangsu, China; The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225000, Jiangsu, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medcine for Prevention and Treatment of Senile Diseases, Medical College of Yangzhou University, Yangzhou 225000, Jiangsu, China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225000, Jiangsu, China
| | - Yanqing Liu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225000, Jiangsu, China; The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225000, Jiangsu, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medcine for Prevention and Treatment of Senile Diseases, Medical College of Yangzhou University, Yangzhou 225000, Jiangsu, China.
| |
Collapse
|
27
|
Jing J, Rui L, Junyuan S, Jinfeng Y, Zhihao H, Weiguo L, Zhenyu J. Small-molecule compounds inhibiting S-phase kinase-associated protein 2: A review. Front Pharmacol 2023; 14:1122008. [PMID: 37089937 PMCID: PMC10113621 DOI: 10.3389/fphar.2023.1122008] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 03/20/2023] [Indexed: 04/25/2023] Open
Abstract
S-phase kinase-associated protein 2 (Skp2) is a substrate-specific adaptor in Skp1-CUL1-ROC1-F-box E3 ubiquitin ligases and widely regarded as an oncogene. Therefore, Skp2 has remained as an active anticancer research topic since its discovery. Accordingly, the structure of Skp2 has been solved and numerous Skp2 inhibiting compounds have been identified. In this review, we would describe the structural features of Skp2, introduce the ubiquitination function of SCFSkp2, and summarize the diverse natural and synthetic Skp2 inhibiting compounds reported to date. The IC50 data of the Skp2 inhibitors or inhibiting compounds in various kinds of tumors at cellular levels implied that the cancer type, stage and pathological mechanisms should be taken into consideration when selecting Skp2-inhibiting compound for cancer treatment.
Collapse
Affiliation(s)
- Jia Jing
- Schools of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang Province, China
| | - Li Rui
- Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Sun Junyuan
- Schools of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang Province, China
| | - Yang Jinfeng
- School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang Province, China
| | - Hong Zhihao
- School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang Province, China
| | - Lu Weiguo
- Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Women′s Reproductive Health Research of Zhejiang Province, Hangzhou, Zhejiang Province, China
- *Correspondence: Lu Weiguo, ; Jia Zhenyu,
| | - Jia Zhenyu
- Institute of Occupation Diseases, Hangzhou Medical College, Hangzhou, Zhejiang Province, China
- *Correspondence: Lu Weiguo, ; Jia Zhenyu,
| |
Collapse
|
28
|
Li M, Gao F, Li X, Gan Y, Han S, Yu X, Liu H, Li W. Stabilization of MCL-1 by E3 ligase TRAF4 confers radioresistance. Cell Death Dis 2022; 13:1053. [PMID: 36535926 PMCID: PMC9763423 DOI: 10.1038/s41419-022-05500-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 12/01/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022]
Abstract
The E3 ligase TNF receptor-associated factor 4 (TRAF4) is frequently overexpressed and closely related to poor prognosis in human malignancies. However, its effect on carcinogenesis and radiosensitivity in oral squamous cell carcinoma (OSCC) remains unclear. The present study found that TRAF4 was significantly upregulated in primary and relapsed OSCC tumor tissues. Depletion of TRAF4 markedly improved the sensitivity of OSCC cells to irradiation (IR) treatment, showing that tumor cell proliferation, colony formation and xenograft tumor growth were reduced. Mechanistically, IR promoted the interaction between TRAF4 and Akt to induce Akt K63-mediated ubiquitination and activation. TRAF4 knockout inhibited the phosphorylation of Akt and upregulated GSK3β activity, resulting in increased myeloid cell leukemia-1 (MCL-1) S159 phosphorylation, which disrupted the interaction of MCL-1 with Josephin domain containing 1 (JOSD1), and ultimately induced MCL-1 ubiquitination and degradation. Moreover, TRAF4 was positively correlated with MCL-1 in primary and in radiotherapy-treated, relapsed tumor tissues. An MCL-1 inhibitor overcame radioresistance in vitro and in vivo. Altogether, the present findings suggest that TRAF4 confers radioresistance in OSCC by stabilizing MCL-1 through Akt signaling, and that targeting TRAF4 may be a promising therapeutic strategy to overcome radioresistance in OSCC.
Collapse
Affiliation(s)
- Ming Li
- grid.431010.7Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013 People’s Republic of China ,Changsha Stomatological Hospital, Changsha, Hunan 410004 People’s Republic of China ,grid.488482.a0000 0004 1765 5169School of Stomatology, Hunan University of Chinese Medicine, Changsha, Hunan 410208 People’s Republic of China ,grid.431010.7Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013 People’s Republic of China
| | - Feng Gao
- grid.431010.7Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013 People’s Republic of China ,grid.431010.7Department of Ultrasonography, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013 People’s Republic of China
| | - Xiaoying Li
- grid.431010.7Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013 People’s Republic of China
| | - Yu Gan
- grid.431010.7Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013 People’s Republic of China
| | - Shuangze Han
- grid.431010.7Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013 People’s Republic of China ,grid.33199.310000 0004 0368 7223Department of Ultrasound, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022 People’s Republic of China
| | - Xinfang Yu
- grid.431010.7Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013 People’s Republic of China ,grid.39382.330000 0001 2160 926XDepartment of Medicine, Baylor College of Medicine, Houston, TX 77030 USA
| | - Haidan Liu
- grid.452708.c0000 0004 1803 0208Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, People’s Republic of China ,grid.452708.c0000 0004 1803 0208Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, People’s Republic of China
| | - Wei Li
- grid.431010.7Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013 People’s Republic of China ,grid.431010.7Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013 People’s Republic of China
| |
Collapse
|
29
|
Skp2-mediated Zeb1 expression facilitates cancer migration by a ubiquitination-independent pathway. Life Sci 2022; 311:121135. [DOI: 10.1016/j.lfs.2022.121135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/23/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022]
|
30
|
Jiang S, Zhang C, Lu Y, Yuan F. Mechanical stress-caused chondrocyte dysfunction and cartilage injury can be attenuated by dioscin via activating sirtuin1/forkhead box O1. J Biochem Mol Toxicol 2022; 36:e23212. [PMID: 36106352 DOI: 10.1002/jbt.23212] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 08/06/2022] [Accepted: 08/30/2022] [Indexed: 12/24/2022]
Abstract
Sirtuin1 (Sirt1)/forkhead box O1 (FoxO1) axis has been reported as a crucial regulator involved in chondral homeostasis of healthy or osteoarthritis (OA) cartilage. In our study, the aim is to investigate whether dioscin functions as an activator of Sirt1/FoxO1 to protect against mechanical stress-induced chondrocyte dysfunction in vitro and in vivo models. HERB and PubChem databases were implemented to predict dioscin-related gene targets. Cell and mouse models of OA were established to determine the pharmacological value of dioscin, a steroidal saponin. Cartilage loss in the knee joint was detected by Safranin O staining. Phosphorylation and nucleocytoplasmic shuttling of FoxO1 was observed in mechanical stress-stimulated chondrocyte and anterior cruciate ligament transection-induced cartilage injury. However, dioscin treatment repressed FoxO1 phosphorylation and cytoplasmic transfer and elevated Sirt1 protein expression. Dioscin treatment reversed mechanical stress-induced growth inhibition and apoptosis of chondrocytes and improved cartilage degradation and bone loss in the epiphysis of the distal femur. Moreover, dioscin could maintain the normal phenotype of chondrocytes via mediating multiple gene expressions. Dioscin inhibited apoptosis and metabolic disorders in OA-like chondrocytes via maintaining the transcriptional activity of FoxO1 and enhancing Sirt1 expression. Dioscin might be a potential Sirt1 activator providing a novel therapeutic schedule for the treatment of OA.
Collapse
Affiliation(s)
- Shilin Jiang
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Chengyuan Zhang
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Ye Lu
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Feng Yuan
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
31
|
Zhao Y, Chard Dunmall LS, Cheng Z, Wang Y, Si L. Natural products targeting glycolysis in cancer. Front Pharmacol 2022; 13:1036502. [PMID: 36386122 PMCID: PMC9663463 DOI: 10.3389/fphar.2022.1036502] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 10/17/2022] [Indexed: 09/05/2023] Open
Abstract
Many energy metabolism pathways exist in cancer, including glycolysis, amino acid metabolism, fatty acid oxidation, and mitochondrial respiration. Tumor cells mainly generate energy through glycolysis to maintain growth and biosynthesis of tumor cells under aerobic conditions. Natural products regulate many steps in glycolysis and targeting glycolysis using natural products is a promising approach to cancer treatment. In this review, we exemplify the relationship between glycolysis and tumors, demonstrate the natural products that have been discovered to target glycolysis for cancer treatment and clarify the mechanisms involved in their actions. Natural products, such as resveratrol mostly found in red grape skin, licochalcone A derived from root of Glycyrrhiza inflate, and brusatol found in Brucea javanica and Brucea mollis, largely derived from plant or animal material, can affect glycolysis pathways in cancer by targeting glycolytic enzymes and related proteins, oncogenes, and numerous glycolytic signal proteins. Knowledge of how natural products regulate aerobic glycolysis will help illuminate the mechanisms by which these products can be used as therapeutics to inhibit cancer cell growth and regulate cellular metabolism. Systematic Review Registration: https://pubmed.ncbi.nlm.nih.gov/, https://clinicaltrials.gov/, http://lib.zzu.edu.cn/.
Collapse
Affiliation(s)
- Yuanyuan Zhao
- National Centre for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Louisa S Chard Dunmall
- Centre for Cancer Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Zhenguo Cheng
- National Centre for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yaohe Wang
- National Centre for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
- Centre for Cancer Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Lingling Si
- National Centre for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
32
|
Ke G, Zhang J, Gao W, Chen J, Liu L, Wang S, Zhang H, Yan G. Application of advanced technology in traditional Chinese medicine for cancer therapy. Front Pharmacol 2022; 13:1038063. [PMID: 36313284 PMCID: PMC9606699 DOI: 10.3389/fphar.2022.1038063] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 09/28/2022] [Indexed: 11/19/2022] Open
Abstract
Although cancer has seriously threatened people’s health, it is also identified by the World Health Organization as a controllable, treatable and even curable chronic disease. Traditional Chinese medicine (TCM) has been extensively used to treat cancer due to its multiple targets, minimum side effects and potent therapeutic effects, and thus plays an important role in all stages of tumor therapy. With the continuous progress in cancer treatment, the overall efficacy of cancer therapy has been significantly improved, and the survival time of patients has been dramatically prolonged. In recent years, a series of advanced technologies, including nanotechnology, gene editing technology, real-time cell-based assay (RTCA) technology, and flow cytometry analysis technology, have been developed and applied to study TCM for cancer therapy, which efficiently improve the medicinal value of TCM and accelerate the research progress of TCM in cancer therapy. Therefore, the applications of these advanced technologies in TCM for cancer therapy are summarized in this review. We hope this review will provide a good guidance for TCM in cancer therapy.
Collapse
Affiliation(s)
- Gaofeng Ke
- Department of Rehabilitation Medicine, The Affiliated Wenling Hospital of Wenzhou Medical University, Wenling, China
| | - Jia Zhang
- School of Pharmacy, Jiangsu Provincial Engineering Research Center of Traditional Chinese Medicine External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wufeng Gao
- School of Pharmacy, Jiangsu Provincial Engineering Research Center of Traditional Chinese Medicine External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiayi Chen
- School of Life Sciences, Jilin University, Changchun, China
| | - Luotong Liu
- School of Life Sciences, Jilin University, Changchun, China
| | - Simiao Wang
- School of Life Sciences, Jilin University, Changchun, China
| | - Huan Zhang
- School of Life Sciences, Jilin University, Changchun, China
- *Correspondence: Huan Zhang, ; Guojun Yan,
| | - Guojun Yan
- School of Pharmacy, Jiangsu Provincial Engineering Research Center of Traditional Chinese Medicine External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing, China
- *Correspondence: Huan Zhang, ; Guojun Yan,
| |
Collapse
|
33
|
Zhao Y, Song J, Dong W, Liu X, Yang C, Wang D, Xue Y, Ruan X, Liu L, Wang P, Zhang M, Liu Y. The MBNL1/circNTRK2/PAX5 pathway regulates aerobic glycolysis in glioblastoma cells by encoding a novel protein NTRK2-243aa. Cell Death Dis 2022; 13:767. [PMID: 36064939 PMCID: PMC9445070 DOI: 10.1038/s41419-022-05219-4] [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: 05/18/2022] [Revised: 08/25/2022] [Accepted: 08/26/2022] [Indexed: 01/21/2023]
Abstract
Glioblastoma multiforme (GBM) is the most common tumor of the human central nervous system. Aerobic glycolysis has been strongly related to tumor development and malignant behavior. In this study, we found that MBNL1, circNTRK2, and NTRK2-243aa were markedly downregulated and inhibited glycolysis in GBM, whereas PAX5 was upregulated and promoted glycolysis. Functionally, MBNL1 promoted the expression of circNTRK2 by binding to NTRK2 pre-mRNA, as validated using RNA pull-down and nascent RNA immunoprecipitation assays. Mass spectrometry, western blotting, and immunofluorescence staining methods were used to detect the expression of NTRK2-243aa. NTRK2-243aa-encoded by circNTRK2-phosphorylated PAX5 at Y102, leading to the attenuation of the half-life of PAX5, as validated by in vitro kinase and MG132 rescue assays. Besides, PAX5 transcriptionally facilitated the expression of PKM2 and HK2 by binding to their promoter regions, as verified by luciferase reporter and chromatin immunoprecipitation assays. Finally, overexpression of MBNL1 and circNTRK2 combined with PAX5 knockdown effectively inhibited the formation of GBM xenograft tumors and significantly prolonged the survival of orthotopic nude mice. We have delineated that the MBNL1/circNTRK2/PAX5 pathway plays a crucial role in regulating GBM glycolysis and could provide potential targets and alternative strategies for the treatment of GBM.
Collapse
Affiliation(s)
- Yubo Zhao
- grid.412467.20000 0004 1806 3501Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004 China ,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004 China ,Liaoning Medical Surgery and Rehabilitation Robot Technology Engineering Research Center, Shenyang, 110004 China
| | - Jian Song
- grid.412467.20000 0004 1806 3501Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004 China ,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004 China ,Liaoning Medical Surgery and Rehabilitation Robot Technology Engineering Research Center, Shenyang, 110004 China
| | - Weiwei Dong
- grid.412467.20000 0004 1806 3501Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004 China ,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004 China ,Liaoning Medical Surgery and Rehabilitation Robot Technology Engineering Research Center, Shenyang, 110004 China
| | - Xiaobai Liu
- grid.412467.20000 0004 1806 3501Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004 China ,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004 China ,Liaoning Medical Surgery and Rehabilitation Robot Technology Engineering Research Center, Shenyang, 110004 China
| | - Chunqing Yang
- grid.412467.20000 0004 1806 3501Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004 China ,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004 China ,Liaoning Medical Surgery and Rehabilitation Robot Technology Engineering Research Center, Shenyang, 110004 China
| | - Di Wang
- grid.412467.20000 0004 1806 3501Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004 China ,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004 China ,Liaoning Medical Surgery and Rehabilitation Robot Technology Engineering Research Center, Shenyang, 110004 China
| | - Yixue Xue
- grid.412449.e0000 0000 9678 1884Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122 China
| | - Xuelei Ruan
- grid.412449.e0000 0000 9678 1884Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122 China
| | - Libo Liu
- grid.412449.e0000 0000 9678 1884Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122 China
| | - Ping Wang
- grid.412449.e0000 0000 9678 1884Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122 China
| | - Mengyang Zhang
- grid.412449.e0000 0000 9678 1884Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122 China
| | - Yunhui Liu
- grid.412467.20000 0004 1806 3501Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004 China ,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004 China ,Liaoning Medical Surgery and Rehabilitation Robot Technology Engineering Research Center, Shenyang, 110004 China
| |
Collapse
|
34
|
Jin S, Guan T, Wang S, Hu M, Liu X, Huang S, Liu Y. Dioscin Alleviates Cisplatin-Induced Mucositis in Rats by Modulating Gut Microbiota, Enhancing Intestinal Barrier Function and Attenuating TLR4/NF-κB Signaling Cascade. Int J Mol Sci 2022; 23:ijms23084431. [PMID: 35457248 PMCID: PMC9025408 DOI: 10.3390/ijms23084431] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/15/2022] [Accepted: 04/16/2022] [Indexed: 12/12/2022] Open
Abstract
Cisplatin-based chemotherapy causes intestinal mucositis, which causes patients immense suffering and hinders the process of cancer treatment. Dioscin is a natural steroid saponin that exhibits strong anti-inflammatory and immunomodulatory properties. Herein, we investigate the protective effect of dioscin on cisplatin induced mucositis in rats from the perspective of gut microbiota and intestinal barrier. We established a rat model of intestinal mucositis by tail vein injection of cisplatin, and concurrently treated with dioscin oral administration. Parameters, such as body weight, diarrheal incidence, and D-Lactate levels, were assessed in order to evaluate the effects of dioscin on intestinal mucositis in rats. Furthermore, biological samples were collected for microscopic gut microbiota, intestinal integrity, and immune inflammation analyses to elucidate the protective mechanisms of dioscin on intestinal mucositis. The results revealed that administration of dioscin significantly attenuated clinical manifestations, histological injury and inflammation in mucositis rats. Besides this, dioscin markedly inhibited the gut microbiota dysbiosis induced by cisplatin. Meanwhile, dioscin partially alleviated junctions between ileum epithelial cells and increased mucus secretion. Moreover, dioscin effectively inhibited the TLR4-MyD88-NF-κB signal transduction pathway and reduced the secretion of subsequent inflammatory mediators. These results suggested that dioscin effectively attenuated cisplatin-induced mucositis in part by modulating the gut microflora profile, maintaining ileum integrity and inhibiting the inflammatory response through the TLR4-MyD88-NF-κB pathway.
Collapse
Affiliation(s)
- Shengzi Jin
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (S.J.); (T.G.); (S.W.); (M.H.); (X.L.); (S.H.)
| | - Tongxu Guan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (S.J.); (T.G.); (S.W.); (M.H.); (X.L.); (S.H.)
| | - Shuang Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (S.J.); (T.G.); (S.W.); (M.H.); (X.L.); (S.H.)
| | - Mengxin Hu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (S.J.); (T.G.); (S.W.); (M.H.); (X.L.); (S.H.)
| | - Xingyao Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (S.J.); (T.G.); (S.W.); (M.H.); (X.L.); (S.H.)
| | - Siqi Huang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (S.J.); (T.G.); (S.W.); (M.H.); (X.L.); (S.H.)
| | - Yun Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (S.J.); (T.G.); (S.W.); (M.H.); (X.L.); (S.H.)
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin 150030, China
- Correspondence:
| |
Collapse
|
35
|
Licochalcone A Promotes the Ubiquitination of c-Met to Abrogate Gefitinib Resistance. BIOMED RESEARCH INTERNATIONAL 2022; 2022:5687832. [PMID: 35309168 PMCID: PMC8930240 DOI: 10.1155/2022/5687832] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 02/15/2022] [Indexed: 11/18/2022]
Abstract
Met proto-oncogene (MET) amplification and tyrosine-protein kinase Met (c-Met) overexpression confer gefitinib resistance in non-small cell lung cancer (NSCLC). The natural product Licochalcone A (Lico A) exhibits a broad range of inhibitory effects against various tumors. However, the effects of Lico A on c-Met signaling and gefitinib resistance in NSCLC remain unclear. In the present study, Lico A efficiently overcame gefitinib-acquired resistance in NSCLC cells by suppressing c-Met signaling. Lico A decreased cell viability and colony formation dose-dependently and impaired in vivo tumorigenesis of gefitinib-resistant HCC827 and PC-9 cells. Furthermore, Lico A induced intrinsic apoptosis and upregulated the protein expression levels of cleaved poly (ADP-ribose) polymerase and cleaved caspase 3. Lico A promoted the interaction between c-Met and E3 ligase c-Casitas B-lineage lymphoma (Cbl), which enhanced c-Cbl-mediated c-Met ubiquitination and degradation. Depletion of c-Cbl compromised Lico A-induced c-Met ubiquitination and its inhibitory efficacy in gefitinib-resistant NSCLC cells. Taken together, the results suggest that Lico A is a promising antitumor agent that might be used to overcome c-Met overexpression-mediated gefitinib resistance in NSCLC cells.
Collapse
|
36
|
NKX6-1 Is a Less Sensitive But Specific Biomarker of Chromophobe Renal Cell Carcinoma. Am J Surg Pathol 2022; 46:809-815. [PMID: 35256556 DOI: 10.1097/pas.0000000000001872] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
NKX6-1 is a transcription factor that plays a key role in the development, differentiation, and identity maintenance of beta cells of pancreatic islets. Although NKX6-1 expression has also been discovered in pancreatic well-differentiated neuroendocrine tumors (WDNETs) and duodenal WDNETs, its expression in chromophobe renal cell carcinoma (chRCC) is unexplored. Analysis of mRNA expression and immunohistochemistry of NKX6-1 was performed using the kidney cancer cohort from The Cancer Genome Atlas (TCGA) and paraffin-embedded whole-tissue slides from our 196 collected cases, including 48 chRCCs (43 classic and 5 eosinophilic subtypes), 24 renal oncocytomas (ROs), 46 clear cell renal cell carcinomas, 41 papillary renal cell carcinomas, 14 renal urothelial carcinomas, 7 low-grade oncocytic renal tumors (LOTs), 8 eosinophilic solid and cystic renal cell carcinomas, 3 succinate dehydrogenase-deficient renal cell carcinomas, and 5 renal oncocytic tumors, not otherwise specified. NKX6-1 expression was almost exclusively upregulated in chRCC at both the mRNA and protein levels compared with other renal tumors. NKX6-1 was immunohistochemically positive in 39 of 48 (81.3%) chRCCs, but negative in 46 clear cell renal cell carcinomas, 24 ROs, 7 low-grade oncocytic renal tumors, 8 eosinophilic solid and cystic renal cell carcinomas, 3 succinate dehydrogenase-deficient renal cell carcinomas, and 5 renal oncocytic tumors, not otherwise specified. Diffuse, moderate, and focal NKX6-1 staining were seen in 21, 4, and 14 of the 39 chRCCs, respectively. In contrast, NKX6-1 was focally positive in only 1 of 41 (2.4%) papillary renal cell carcinomas and 2 of 14 (14.3%) renal urothelial carcinomas. Therefore, the sensitivity and specificity of NKX6-1 staining were 81.3% and 98% for chRCC, respectively. In conclusion, NKX6-1 may be a novel potential marker for differentiating chRCC from other renal neoplasms, especially from RO.
Collapse
|
37
|
Kooshki L, Mahdavi P, Fakhri S, Akkol EK, Khan H. Targeting lactate metabolism and glycolytic pathways in the tumor microenvironment by natural products: A promising strategy in combating cancer. Biofactors 2022; 48:359-383. [PMID: 34724274 DOI: 10.1002/biof.1799] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 10/06/2021] [Indexed: 12/12/2022]
Abstract
Anticancer drugs are not purely effective because of their toxicity, side effects, high cost, inaccessibility, and associated resistance. On the other hand, cancer is a complex public health problem that could intelligently adopt different signaling pathways and alter the body's metabolism to escape from the immune system. One of the cancer strategies to metastasize is modifying pH in the tumor microenvironment, ranging between 6.5 and 6.9. As a powerful determiner, lactate is responsible for this acidosis. It is involved in immune stimulation, including innate and adaptive immunity, apoptotic-related factors (Bax/Bcl-2, caspase), and glycolysis pathways (e.g., GLUT-1, PKM2, PFK, HK2, MCT-1, and LDH). Lactate metabolism, in turn, is interconnected with several dysregulated signaling mediators, including PI3K/Akt/mTOR, AMPK, NF-κB, Nrf2, JAK/STAT, and HIF-1α. Because of lactate's emerging and critical role, targeting lactate production and its transporters is important for preventing and managing tumorigenesis. Hence, exploring and developing novel promising anticancer agents to minimize human cancers is urgent. Based on numerous studies, natural secondary metabolites as multi-target alternative compounds with health-promoting properties possess more high effectiveness and low side effects than conventional agents. Besides, the mechanism of multi-targeted natural sources is related to lactate production and cancer-associated cross-talked factors. This review focuses on targeting the lactate metabolism/transporters, and lactate-associated mediators, including glycolytic pathways. Besides, interconnected mediators to lactate metabolism are also targeted by natural products. Accordingly, plant-derived secondary metabolites are introduced as alternative therapies in combating cancer through modulating lactate metabolism and glycolytic pathways.
Collapse
Affiliation(s)
- Leila Kooshki
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
- USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Parisa Mahdavi
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Esra Küpeli Akkol
- Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, Ankara, Turkey
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan
| |
Collapse
|
38
|
Skp2 stabilizes Mcl-1 and confers radioresistance in colorectal cancer. Cell Death Dis 2022; 13:249. [PMID: 35301297 PMCID: PMC8930992 DOI: 10.1038/s41419-022-04685-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 01/04/2022] [Accepted: 02/24/2022] [Indexed: 11/08/2022]
Abstract
AbstractOverexpression of Skp2 plays a critical role in tumorigenesis and correlates with poor prognosis in human malignancies. Thus, Skp2 has been proposed as an attractive target for anti-tumor interventions. The expression of Skp2 in human colorectal cancer (CRC) and the role of Skp2 in tumorigenic properties and irradiation sensitivities of CRC cells were examined by anchorage-dependent and -independent growth assays, immunoblot, flow cytometry, immunohistochemical staining, ubiquitination analysis, co-immunoprecipitation assay, CRISPR-Cas9-based gene knockout, and xenograft experiments. Skp2 is highly expressed in CRC patient tissues. Blocking Skp2 expression reduces the tumorigenic properties of CRC cells in vitro and in vivo. Depletion of Skp2 confers sensitivity to irradiation of CRC cells. Skp2 deficiency enhances irradiation-induced intrinsic apoptosis by facilitating E3 ligase FBW7-mediated Mcl-1 ubiquitination and degradation. Knockout of Skp2 sensitizes CRC cells to irradiation treatments in vivo. Our findings indicate that Skp2 stabilizes Mcl-1, and targeting Skp2 in combination with traditional radiotherapy might be efficacious in treating CRC.
Collapse
|
39
|
Xie Y, Wang M, Xia M, Guo Y, Zu X, Zhong J. Ubiquitination regulation of aerobic glycolysis in cancer. Life Sci 2022; 292:120322. [PMID: 35031261 DOI: 10.1016/j.lfs.2022.120322] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 01/06/2022] [Accepted: 01/06/2022] [Indexed: 12/18/2022]
Abstract
Aerobic glycolysis, or the Warburg effect, is regarded as a critical part of metabolic reprogramming and plays a crucial role in the occurrence and development of tumours. Ubiquitination and deubiquitination, essential post-translational modifications, have attracted increasing attention with regards to the regulation of metabolic reprogramming in cancer. However, the mechanism of ubiquitination in glycolysis remains unclear. In this review, we discuss the roles of ubiquitination and deubiquitination in regulating glycolysis, and their involvement in regulating important signalling pathways, enzymes, and transcription factors. Focusing on potential mechanisms may provide novel strategies for cancer treatment.
Collapse
Affiliation(s)
- Yao Xie
- Institute of Clinical Medicine, the First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China; Department of Clinical Laboratory, the First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Mu Wang
- Clinical Research Institute, the NanHua Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Min Xia
- Institute of Clinical Medicine, the First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Yinping Guo
- Institute of Clinical Medicine, the First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Xuyu Zu
- Institute of Clinical Medicine, the First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China; Cancer Research Institute, the First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China.
| | - Jing Zhong
- Institute of Clinical Medicine, the First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China; Cancer Research Institute, the First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China.
| |
Collapse
|
40
|
Bandopadhyay S, Anand U, Gadekar VS, Jha NK, Gupta PK, Behl T, Kumar M, Shekhawat MS, Dey A. Dioscin: A review on pharmacological properties and therapeutic values. Biofactors 2022; 48:22-55. [PMID: 34919768 DOI: 10.1002/biof.1815] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 11/27/2021] [Indexed: 02/06/2023]
Abstract
Dioscin has gained immense popularity as a natural, bioactive steroid saponin, which offers numerous medical benefits. The growing global incidence of disease-associated morbidity and mortality continues to compromise human health, facilitating an increasingly urgent need for nontoxic, noninvasive, and efficient treatment alternatives. Natural compounds can contribute vastly to this field. Over recent years, studies have demonstrated the remarkable protective actions of dioscin against a variety of human malignancies, metabolic disorders, organ injuries, and viral/fungal infections. The successful usage of this phytocompound has been widely seen in medical treatment procedures under traditional Chinese medicine, and it is becoming progressively prevalent worldwide. This review provides an insight into the wide spectrum of pharmacological activities of dioscin, as reported and compiled in recent literature. The various novel approaches and applications of dioscin also verify the advantages exhibited by plant extracts against commercially available drugs, highlighting the potential of phytochemical agents like dioscin to be further incorporated into clinical practice.
Collapse
Affiliation(s)
| | - Uttpal Anand
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Vijaykumar Shivaji Gadekar
- Zoology Department, Sangola College (affiliated to Punyashlok Ahilyadevi Holkar Solapur University), Solapur, Maharashtra, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Piyush Kumar Gupta
- Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Tapan Behl
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Chandigarh, Punjab, India
| | - Manoj Kumar
- Chemical and Biochemical Processing Division, ICAR - Central Institute for Research on Cotton Technology, Mumbai, Maharashtra, India
| | - Mahipal S Shekhawat
- Plant Biotechnology Unit, Kanchi Mamunivar Government Institute for Postgraduate Studies and Research, Puducherry, India
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata, West Bengal, India
| |
Collapse
|
41
|
Llombart V, Mansour MR. Therapeutic targeting of "undruggable" MYC. EBioMedicine 2022; 75:103756. [PMID: 34942444 PMCID: PMC8713111 DOI: 10.1016/j.ebiom.2021.103756] [Citation(s) in RCA: 232] [Impact Index Per Article: 77.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 11/23/2021] [Accepted: 12/01/2021] [Indexed: 12/13/2022] Open
Abstract
c-MYC controls global gene expression and regulates cell proliferation, cell differentiation, cell cycle, metabolism and apoptosis. According to some estimates, MYC is dysregulated in ≈70% of human cancers and strong evidence implicates aberrantly expressed MYC in both tumor initiation and maintenance. In vivo studies show that MYC inhibition elicits a prominent anti-proliferative effect and sustained tumor regression while any alteration on healthy tissue remains reversible. This opens an exploitable window for treatment that makes MYC one of the most appealing therapeutic targets for cancer drug development. This review describes the main functional and structural features of the protein structure of MYC and provides a general overview of the most relevant or recently identified interactors that modulate MYC oncogenic activity. This review also summarizes the different approaches aiming to abrogate MYC oncogenic function, with a particular focus on the prototype inhibitors designed for the direct and indirect targeting of MYC.
Collapse
Affiliation(s)
- Victor Llombart
- UCL Cancer Institute, University College London, Department of Haematology, London WC1E 6DD, UK
| | - Marc R Mansour
- UCL Cancer Institute, University College London, Department of Haematology, London WC1E 6DD, UK; UCL Great Ormond Street Institute of Child Health, Developmental Biology and Cancer, London, UK.
| |
Collapse
|
42
|
Advances in the role of natural products in human gene expression. Chin J Nat Med 2022; 20:1-8. [DOI: 10.1016/s1875-5364(22)60147-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Indexed: 11/17/2022]
|
43
|
Ye P, Chi X, Cha JH, Luo S, Yang G, Yan X, Yang WH. Potential of E3 Ubiquitin Ligases in Cancer Immunity: Opportunities and Challenges. Cells 2021; 10:cells10123309. [PMID: 34943817 PMCID: PMC8699390 DOI: 10.3390/cells10123309] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/20/2021] [Accepted: 11/22/2021] [Indexed: 12/13/2022] Open
Abstract
Cancer immunotherapies, including immune checkpoint inhibitors and immune pathway–targeted therapies, are promising clinical strategies for treating cancer. However, drug resistance and adverse reactions remain the main challenges for immunotherapy management. The future direction of immunotherapy is mainly to reduce side effects and improve the treatment response rate by finding new targets and new methods of combination therapy. Ubiquitination plays a crucial role in regulating the degradation of immune checkpoints and the activation of immune-related pathways. Some drugs that target E3 ubiquitin ligases have exhibited beneficial effects in preclinical and clinical antitumor treatments. In this review, we discuss mechanisms through which E3 ligases regulate tumor immune checkpoints and immune-related pathways as well as the opportunities and challenges for integrating E3 ligases targeting drugs into cancer immunotherapy.
Collapse
Affiliation(s)
- Peng Ye
- Key Laboratory of Cell Homeostasis and Cancer Research of Guangdong Higher Education Institutes and Affiliated Cancer Hospital & Institute, Guangzhou Medical University, Guangzhou 910095, China; (P.Y.); (X.C.); (S.L.); (G.Y.)
| | - Xiaoxia Chi
- Key Laboratory of Cell Homeostasis and Cancer Research of Guangdong Higher Education Institutes and Affiliated Cancer Hospital & Institute, Guangzhou Medical University, Guangzhou 910095, China; (P.Y.); (X.C.); (S.L.); (G.Y.)
| | - Jong-Ho Cha
- Department of Biomedical Science and Engineering, Graduate School, Inha University, Incheon 22212, Korea;
- Department of Biomedical Sciences, College of Medicine, Inha University, Incheon 22212, Korea
| | - Shahang Luo
- Key Laboratory of Cell Homeostasis and Cancer Research of Guangdong Higher Education Institutes and Affiliated Cancer Hospital & Institute, Guangzhou Medical University, Guangzhou 910095, China; (P.Y.); (X.C.); (S.L.); (G.Y.)
| | - Guanghui Yang
- Key Laboratory of Cell Homeostasis and Cancer Research of Guangdong Higher Education Institutes and Affiliated Cancer Hospital & Institute, Guangzhou Medical University, Guangzhou 910095, China; (P.Y.); (X.C.); (S.L.); (G.Y.)
| | - Xiuwen Yan
- Key Laboratory of Cell Homeostasis and Cancer Research of Guangdong Higher Education Institutes and Affiliated Cancer Hospital & Institute, Guangzhou Medical University, Guangzhou 910095, China; (P.Y.); (X.C.); (S.L.); (G.Y.)
- Correspondence: (X.Y.); (W.-H.Y.)
| | - Wen-Hao Yang
- Key Laboratory of Cell Homeostasis and Cancer Research of Guangdong Higher Education Institutes and Affiliated Cancer Hospital & Institute, Guangzhou Medical University, Guangzhou 910095, China; (P.Y.); (X.C.); (S.L.); (G.Y.)
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 406040, Taiwan
- Correspondence: (X.Y.); (W.-H.Y.)
| |
Collapse
|
44
|
Harrington CT, Sotillo E, Dang CV, Thomas-Tikhonenko A. Tilting MYC toward cancer cell death. Trends Cancer 2021; 7:982-994. [PMID: 34481764 PMCID: PMC8541926 DOI: 10.1016/j.trecan.2021.08.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 07/30/2021] [Accepted: 08/02/2021] [Indexed: 12/12/2022]
Abstract
MYC oncoprotein promotes cell proliferation and serves as the key driver in many human cancers; therefore, considerable effort has been expended to develop reliable pharmacological methods to suppress its expression or function. Despite impressive progress, MYC-targeting drugs have not reached the clinic. Recent advances suggest that within a limited expression range unique to each tumor, MYC oncoprotein can have a paradoxical, proapoptotic function. Here we introduce a counterintuitive idea that modestly and transiently elevating MYC levels could aid chemotherapy-induced apoptosis and thus benefit the patients as much, if not more than MYC inhibition.
Collapse
Affiliation(s)
- Colleen T Harrington
- Division of Cancer Pathobiology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Cell and Molecular Biology Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Elena Sotillo
- Division of Cancer Pathobiology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Chi V Dang
- Cell and Molecular Biology Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA 19104, USA; Ludwig Institute for Cancer Research, New York, NY 10017, USA
| | - Andrei Thomas-Tikhonenko
- Division of Cancer Pathobiology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Cell and Molecular Biology Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
45
|
Wu MM, Wang QM, Huang BY, Mai CT, Wang CL, Wang TT, Zhang XJ. Dioscin ameliorates murine ulcerative colitis by regulating macrophage polarization. Pharmacol Res 2021; 172:105796. [PMID: 34343656 DOI: 10.1016/j.phrs.2021.105796] [Citation(s) in RCA: 123] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 12/20/2022]
Abstract
Restoring immune balance by targeting macrophage polarization is a potentially valuable therapeutic strategy for ulcerative colitis (UC). Dioscin is a steroidal saponin with potent anti-inflammatory, immunoregulatory, and hypolipidemic effects. This study examined the protective effect of Dioscin on UC in mice and explored the underlying mechanisms. Mice were induced colitis by dextran sulfate sodium (DSS) and concurrently treated with Dioscin oral administration. RAW264.7 cells were skewed to M1 macrophage polarization by lipopolysaccharide (LPS) and interferon-γ (INF-γ) in vitro, and received Dioscin treatment. The results showed that Dioscin ameliorated colitis in mice, reduced macrophage M1 polarization, but markedly promoted M2 polarization in mice colon. Dioscin inhibited mammalian target rapamycin complex 1 (mTORC1)/hypoxia-inducible factor-1α (HIF-1α) signaling and restrained glycolysis in RAW264.7; however, it activated mammalian target rapamycin complex 2 (mTORC2)/peroxisome proliferator-activated receptor-γ (PPAR-γ) signal and facilitated fatty acid oxidation (FAO). The modulation of mTORs signaling may inhibit M1, but promote M2 polarization. Furthermore, the effect of Dioscin on M2 polarization was neutralized by the FAO inhibitor Etomoxir and the mTORC2 inhibitor JR-AB2-011. In parallel, the inhibitory effect of Dioscin on M1 polarization was mitigated by the mTORC1 agonist L-leucine. Both JR-AB2-011 and L-leucine blocked the therapeutic effect of Dioscin in mice with UC. Therefore, Dioscin ameliorated UC in mice, possibly by restraining M1, while skewing M2 polarization of macrophages. Regulation of mTORC1/HIF-1α and mTORC2/PPAR-γ signals is a possible mechanism by which Dioscin inhibited aerobic glycolysis and promoted FAO of macrophages. In summary, Dioscin protected mice against DSS-induced UC by regulating mTOR signaling, thereby adjusting macrophage metabolism and polarization.
Collapse
Affiliation(s)
- Mei-Mei Wu
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China; Clinical Experimental Center, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen 529030, PR China.
| | - Qiu-Mei Wang
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China.
| | - Bao-Yuan Huang
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China.
| | - Chu-Tian Mai
- State Key Laboratory for Quality Research in Chinese Medicines, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau.
| | - Chun-Li Wang
- Guangxi Botanical Garden of Medicinal Plants, Xiangzhu Ave, Nanning 510663, PR China.
| | - Tian-Tian Wang
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China.
| | - Xiao-Jun Zhang
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China.
| |
Collapse
|
46
|
Li L, Liu P, Xie Y, Liu Y, Chen Z, Geng Y, Zhang L. Xanthatin inhibits human colon cancer cells progression via mTOR signaling mediated energy metabolism alteration. Drug Dev Res 2021; 83:119-130. [PMID: 34180556 DOI: 10.1002/ddr.21850] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 06/08/2021] [Accepted: 06/17/2021] [Indexed: 11/05/2022]
Abstract
Tumor cells exhibit higher glycolysis and rely on abnormal energy metabolism to produce ATP, which is essential for cell proliferation and migration. Abnormal energy metabolism inhibition is considered a promising tumor treatment strategy. Xanthatin is an active sesquiterpene lactone isolated from Xanthium strumarium L. This study evaluated the effect of xanthatin on the energy metabolism of human colon cancer cells. The results showed that xanthatin significantly inhibited the migration and invasion of human HT-29 and HCT-116 colon cancer cells. We found that xanthatin effectively reduced the production of ATP and promoted the accumulation of lactate. Xanthatin inhibited glycolysis which may be related to the reduction of glucose transporter 1 (Glut1) and monocarboxylate transporter 4 (MCT4) mRNA and protein levels. Concomitantly, xanthatin promoted complex II activity and oxidative phosphorylation (OXPHOS), resulting in mitochondrial damage and cell death in HT-29 cells. Furthermore, xanthatin inhibited the phosphorylation of mTOR, the phosphorylation of 4E-binding protein 1 (4E-BP1) and c-myc in HT-29 cells. Moreover, rapamycin, a mTOR inhibitor, could enhance the cytotoxicity effect in xanthatin treated HT-29 cells. Additionally, HT-29 cells transfected with si-mTOR aggravated xanthatin induced cell viability inhibition. Based on these results, we observed that the effect of xanthatin on energy metabolism may be related to its inhibition of the mTOR signaling pathway. Collectively, this study provides important insights into xanthatin's anticancer effect, which occurs by regulation of the energy metabolism of human colon cancer cells, and suggest that xanthatin has potential as a botanical drug against abnormal tumor energy metabolism.
Collapse
Affiliation(s)
- Lingli Li
- Department of Pharmacy, Anhui Provincial Hospital, Anhui Medical University, Hefei, Anhui, 230001, China
| | - Ping Liu
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui, China
| | - Yanbo Xie
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Yunxiao Liu
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui, China
| | - Zhaolin Chen
- Department of Pharmacy, Anhui Provincial Hospital, Anhui Medical University, Hefei, Anhui, 230001, China.,Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Yadi Geng
- Department of Pharmacy, Anhui Provincial Hospital, Anhui Medical University, Hefei, Anhui, 230001, China.,Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Lei Zhang
- Department of Pharmacy, Anhui Provincial Hospital, Anhui Medical University, Hefei, Anhui, 230001, China.,Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.,Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
47
|
LaPlante G, Zhang W. Targeting the Ubiquitin-Proteasome System for Cancer Therapeutics by Small-Molecule Inhibitors. Cancers (Basel) 2021; 13:3079. [PMID: 34203106 PMCID: PMC8235664 DOI: 10.3390/cancers13123079] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/16/2021] [Accepted: 06/17/2021] [Indexed: 12/14/2022] Open
Abstract
The ubiquitin-proteasome system (UPS) is a critical regulator of cellular protein levels and activity. It is, therefore, not surprising that its dysregulation is implicated in numerous human diseases, including many types of cancer. Moreover, since cancer cells exhibit increased rates of protein turnover, their heightened dependence on the UPS makes it an attractive target for inhibition via targeted therapeutics. Indeed, the clinical application of proteasome inhibitors in treatment of multiple myeloma has been very successful, stimulating the development of small-molecule inhibitors targeting other UPS components. On the other hand, while the discovery of potent and selective chemical compounds can be both challenging and time consuming, the area of targeted protein degradation through utilization of the UPS machinery has seen promising developments in recent years. The repertoire of proteolysis-targeting chimeras (PROTACs), which employ E3 ligases for the degradation of cancer-related proteins via the proteasome, continues to grow. In this review, we will provide a thorough overview of small-molecule UPS inhibitors and highlight advancements in the development of targeted protein degradation strategies for cancer therapeutics.
Collapse
Affiliation(s)
- Gabriel LaPlante
- Department of Molecular and Cellular Biology, College of Biological Science, University of Guelph, 50 Stone Rd E, Guelph, ON N1G2W1, Canada;
| | - Wei Zhang
- Department of Molecular and Cellular Biology, College of Biological Science, University of Guelph, 50 Stone Rd E, Guelph, ON N1G2W1, Canada;
- CIFAR Azrieli Global Scholars Program, Canadian Institute for Advanced Research, MaRS Centre West Tower, 661 University Avenue, Toronto, ON M5G1M1, Canada
| |
Collapse
|
48
|
Albayrak E, Uslu M, Akgol S, Tuysuz EC, Kocabas F. Small molecule-mediated modulation of ubiquitination and neddylation improves HSC function ex vivo. J Cell Physiol 2021; 236:8122-8136. [PMID: 34101829 DOI: 10.1002/jcp.30466] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 05/04/2021] [Accepted: 05/27/2021] [Indexed: 11/07/2022]
Abstract
Hematopoietic stem cells (HSCs) are particularly characterized by their quiescence and self-renewal. Cell cycle regulators tightly control quiescence and self-renewal capacity. Studies suggest that modulation of ubiquitination and neddylation could contribute to HSC function via cyclin-dependent kinase inhibitors (CDKIs). S-phase kinase-associated protein 2 (SKP2) is responsible for ubiquitin-mediated proteolysis of CDKIs. Here, we modulated overall neddylation and SKP2-associated ubiquitination in HSCs by using SKP2-C25, an SKP2 inhibitor, and MLN4924 (Pevonedistat) as an inhibitor of the NEDD8 system. Treatments of SKP2-C25 and MLN4924 increased both murine and human stem and progenitor cell (HSPC) compartments. This is associated with the improved quiescence of murine HSC by upregulation of p27 and p57 CDKIs. A colony-forming unit assay showed an enhanced in vitro self-renewal potential post inhibition of ubiquitination and neddylation. In addition, MLN4924 triggered the mobilization of bone marrow HSPCs to peripheral blood. Intriguingly, MLN4924 treatment could decrease the proliferation of murine bone marrow mesenchymal stem cells or endothelial cells. These findings shed light on the contribution of SKP2, and associated ubiquitination and neddylation in HSC maintenance, self-renewal, and expansion.
Collapse
Affiliation(s)
- Esra Albayrak
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Merve Uslu
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Sezer Akgol
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Emre Can Tuysuz
- Department of Medical Genetics, Faculty of Medicine, Yeditepe University, Istanbul, Turkey
| | - Fatih Kocabas
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| |
Collapse
|
49
|
Wu T, Gu X, Cui H. Emerging Roles of SKP2 in Cancer Drug Resistance. Cells 2021; 10:cells10051147. [PMID: 34068643 PMCID: PMC8150781 DOI: 10.3390/cells10051147] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 12/14/2022] Open
Abstract
More than half of all cancer patients receive chemotherapy, however, some of them easily acquire drug resistance. Resistance to chemotherapy has become a massive obstacle to achieve high rates of pathological complete response during cancer therapy. S-phase kinase-associated protein 2 (Skp2), as an E3 ligase, was found to be highly correlated with drug resistance and poor prognosis. In this review, we summarize the mechanisms that Skp2 confers to drug resistance, including the Akt-Skp2 feedback loop, Skp2-p27 pathway, cell cycle and mitosis regulation, EMT (epithelial-mesenchymal transition) property, enhanced DNA damage response and repair, etc. We also addressed novel molecules that either inhibit Skp2 expression or target Skp2-centered interactions, which might have vast potential for application in clinics and benefit cancer patients in the future.
Collapse
Affiliation(s)
- Ting Wu
- Institute of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China;
| | - Xinsheng Gu
- Department of Pharmacology, College of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, China;
| | - Hongmei Cui
- Institute of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China;
- Correspondence:
| |
Collapse
|
50
|
Chen RZ, Yang F, Zhang M, Sun ZG, Zhang N. Cellular and Molecular Mechanisms of Pristimerin in Cancer Therapy: Recent Advances. Front Oncol 2021; 11:671548. [PMID: 34026649 PMCID: PMC8138054 DOI: 10.3389/fonc.2021.671548] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 04/13/2021] [Indexed: 12/11/2022] Open
Abstract
Seeking an efficient and safe approach to eliminate tumors is a common goal of medical fields. Over these years, traditional Chinese medicine has attracted growing attention in cancer treatment due to its long history. Pristimerin is a naturally occurring quinone methide triterpenoid used in traditional Chinese medicine to treat various cancers. Recent studies have identified alterations in cellular events and molecular signaling targets of cancer cells under pristimerin treatment. Pristimerin induces cell cycle arrest, apoptosis, and autophagy to exhibit anti-proliferation effects against tumors. Pristimerin also inhibits the invasion, migration, and metastasis of tumor cells via affecting cell adhesion, cytoskeleton, epithelial-mesenchymal transition, cancer stem cells, and angiogenesis. Molecular factors and pathways are associated with the anti-cancer activities of pristimerin. Furthermore, pristimerin reverses multidrug resistance of cancer cells and exerts synergizing effects with other chemotherapeutic drugs. This review aims to discuss the anti-cancer potentials of pristimerin, emphasizing multi-targeted biological and molecular regulations in cancers. Further investigations and clinical trials are warranted to understand the advantages and disadvantages of pristimerin treatment much better.
Collapse
Affiliation(s)
- Run-Ze Chen
- Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Fei Yang
- Department of Pathology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Min Zhang
- Department of Dermatology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhi-Gang Sun
- Department of Thoracic Surgery, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Nan Zhang
- Department of Oncology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|