1
|
Liang H, Zhou B, Li P, Zhang X, Zhang S, Zhang Y, Yao S, Qu S, Chen J. Stemness regulation in prostate cancer: prostate cancer stem cells and targeted therapy. Ann Med 2025; 57:2442067. [PMID: 39711287 DOI: 10.1080/07853890.2024.2442067] [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: 11/27/2023] [Revised: 11/07/2024] [Accepted: 11/22/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND Increasing evidence indicates that cancer stem cells (CSCs) and cancer stem-like cells form a special subpopulation of cells that are ubiquitous in tumors. These cells exhibit similar characteristics to those of normal stem cells in tissues; moreover, they are capable of self-renewal and differentiation, as well as high tumorigenicity and drug resistance. In prostate cancer (PCa), it is difficult to kill these cells using androgen signaling inhibitors and chemotherapy drugs. Consequently, the residual prostate cancer stem cells (PCSCs) mediate tumor recurrence and progression. OBJECTIVE This review aims to provide a comprehensive and up-to-date overview of PCSCs, with a particular emphasis on potential therapeutic strategies targeting these cells. METHODS After searching in PubMed and Embase databases using 'prostate cancer' and 'cancer stem cells' as keywords, studies related were compiled and examined. RESULTS In this review, we detail the origin and characteristics of PCSCs, introduce the regulatory pathways closely related to CSC survival and stemness maintenance, and discuss the link between epithelial-mesenchymal transition, tumor microenvironment and tumor stemness. Furthermore, we introduce the currently available therapeutic strategies targeting CSCs, including signaling pathway inhibitors, anti-apoptotic protein inhibitors, microRNAs, nanomedicine, and immunotherapy. Lastly, we summarize the limitations of current CSC research and mention future research directions. CONCLUSION A deeper understanding of the regulatory network and molecular markers of PCSCs could facilitate the development of novel therapeutic strategies targeting these cells. Previous preclinical studies have demonstrated the potential of this treatment approach. In the future, this may offer alternative treatment options for PCa patients.
Collapse
Affiliation(s)
- Hao Liang
- Department of Urology, Qilu Hospital of Shandong University (Qingdao), Qingdao, China
| | - Bin Zhou
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Peixin Li
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaoyi Zhang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Shijie Zhang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Yaozhong Zhang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Shengwen Yao
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Sifeng Qu
- Department of Urology, Qilu Hospital of Shandong University (Qingdao), Qingdao, China
| | - Jun Chen
- Department of Urology, Qilu Hospital of Shandong University (Qingdao), Qingdao, China
| |
Collapse
|
2
|
Lu T, Wang Q, Xin Y, Wu X, Wang Y, Xia Y, Xun L, Liu H. Knockout of the sulfide: quinone oxidoreductase SQR reduces growth of HCT116 tumor xenograft. Redox Biol 2025; 83:103650. [PMID: 40305883 DOI: 10.1016/j.redox.2025.103650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 04/22/2025] [Accepted: 04/23/2025] [Indexed: 05/02/2025] Open
Abstract
Colorectal cancer (CRC) exhibits significant diversity and heterogeneity, posing a requirement for novel therapeutic targets. Polysulfides are associated with CRC progression and immune evasion, but the underlying mechanisms are not fully understood. Sulfide: quinone oxidoreductase (SQR), a mitochondrial flavoprotein, catalyzes hydrogen sulfide (H2S) oxidation and polysulfides production. Herein, we explored its role in CRC pathogenesis and its potential as a therapeutic target. Our findings revealed that SQR knockout disrupted polysulfides homeostasis, diminished mitochondrial function, impaired cell proliferation, and triggered early apoptosis in HCT116 CRC cells. Moreover, the SQR knockout led to markedly reduced tumor sizes in mice models of colon xenografts. Although the transcription of glycolytic genes remained largely unchanged, metabolomic analysis demonstrated a reprogramming of glycolysis at the fructose-1,6-bisphosphate degradation step, catalyzed by aldolase A (ALDOA). Both Western blot analysis and enzymatic assays confirmed the decrease in ALDOA levels and activity. In conclusion, the study establishes the critical role of SQR in mitochondrial function and metabolic regulation in CRC, with its knockout leading to metabolic reprogramming and diminished tumor growth in HCT116 tumor xenografts. These insights lay a foundation for the development of SQR-targeted therapies for CRC.
Collapse
Affiliation(s)
- Ting Lu
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital), University of Health and Rehabilitation Sciences, Qingdao, 266071, People's Republic of China
| | - Qingda Wang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266200, People's Republic of China
| | - Yuping Xin
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266200, People's Republic of China
| | - Xiaohua Wu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266200, People's Republic of China
| | - Yang Wang
- Origin Biotechnology Private Limited, 2 Venture Drive, 608526, Singapore
| | - Yongzhen Xia
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266200, People's Republic of China
| | - Luying Xun
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266200, People's Republic of China; School of Molecular Biosciences, Washington State University, Pullman, WA, 991647520, USA
| | - Huaiwei Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266200, People's Republic of China.
| |
Collapse
|
3
|
Han J, Yuan Y, Zhang J, Hou Y, Xu H, Nie X, Zhao Z, Hou J. Regulatory effect of Wnt signaling on mitochondria in cancer: from mechanism to therapy. Apoptosis 2025:10.1007/s10495-025-02114-z. [PMID: 40257508 DOI: 10.1007/s10495-025-02114-z] [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] [Accepted: 04/06/2025] [Indexed: 04/22/2025]
Abstract
Cancer is one of the most significant public health challenges in the new millennium, and complex mechanisms are at work to contribute to its pathogenesis and progression. The Wnt signaling pathways, which are crucial conserved cascades involved in embryological development and tissue homeostasis, and mitochondria, the intracellular powerhouses responsible for energy production, calcium and iron homeostasis, as well as mitochondrial apoptosis in eukaryotic cells, have their own mechanisms regulating these pathological processes. In the past decade, accumulating evidence has indicated that Wnt signaling pathways directly regulate mitochondrial biogenesis and function under physiological and pathological conditions. In this review, we systemically summarize the current understanding of how Wnt signaling pathways, particularly the canonical Wnt cascade, regulate mitochondrial fission, respiration, metabolism, and mitochondrial-dependent apoptosis in cancer. In addition, we discuss recent advancements in the research of anticancer agents and related pharmacological mechanisms targeting the signaling transduction of canonical Wnt pathway and/or mitochondrial function. We believe that the combined use of pharmaceuticals targeting Wnt signaling and/or mitochondria with conventional therapies, immunotherapy and targeted therapy based on accurate molecular pathological diagnosis will undoubtedly be the future mainstream direction of personalized cancer treatment, which could benefit more cancer patients.
Collapse
Affiliation(s)
- Jinping Han
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, 475004, Kaifeng, China
| | - Yimeng Yuan
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, 475004, Kaifeng, China
| | - Jianhua Zhang
- Kaifeng 155 Hospital, China RongTong Medical Healthcare Group Co. Ltd, 475003, Kaifeng, China
| | - Yifan Hou
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, 475004, Kaifeng, China
| | - Hongtao Xu
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, 475004, Kaifeng, China
| | - Xiaobo Nie
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, 475004, Kaifeng, China.
| | - Zhenhua Zhao
- Ma'anshan 86 Hospital, China RongTong Medical Healthcare Group Co. Ltd, 243100, Ma'anshan, China
| | - Junqing Hou
- Kaifeng 155 Hospital, China RongTong Medical Healthcare Group Co. Ltd, 475003, Kaifeng, China
| |
Collapse
|
4
|
Xie X, Zhang B, Peng J, Ma N, Pan Q, Wei Y, Jin H, Yu F, Huang X, Zhang P, Wang J, Zheng J, Ying X, Liu RY, Yu H, Lee MH, Meng X. EGF-Upregulated lncRNA ESSENCE Promotes Colorectal Cancer Growth through Stabilizing CAD and Ferroptosis Defense. RESEARCH (WASHINGTON, D.C.) 2025; 8:0649. [PMID: 40190348 PMCID: PMC11969792 DOI: 10.34133/research.0649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 03/03/2025] [Accepted: 03/08/2025] [Indexed: 04/09/2025]
Abstract
Epidermal growth factor receptor/mitogen-activated protein kinase (EGFR/MAPK) signaling is highly activated in various types of cancer. The long noncoding RNAs induced by this pathway and their roles in colorectal cancer (CRC) have not been fully elucidated. In this study, based on the profiling of long noncoding RNAs triggered by EGFR/MAPK signaling, we identified that ESSENCE (EGF [epidermal growth factor] Signal Sensing CAD's Effect; ENST00000415336), which is mediated by the transcription factor early growth response factor 1, functions as a potent oncogenic molecule that predicts poor prognosis in CRC. Mechanistically, ESSENCE directly interacts with carbamoyl-phosphate synthetase 2, aspartate transcarbamylase, and dihydroorotase (CAD) and competitively attenuates CAD degradation mediated by its newly discovered E3 ligase KEAP1, thereby suppressing ferroptosis and promoting CRC progression. Importantly, combinational treatment of the mitogen-activated extracellular signal-regulated kinase inhibitor selumetinib and ferroptosis inducer sulfasalazine synergistically suppresses ESSENCE-high CRC in a patient-derived xenograft mouse model. Taken together, these findings demonstrate the crucial role of ESSENCE in mediating CRC progression by regulating CAD stabilization and suggest a therapeutic strategy of targeting the ESSENCE-CAD axis in CRC.
Collapse
Affiliation(s)
- Xiaoshan Xie
- Department of General Surgery, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
| | - Boyu Zhang
- Department of General Surgery, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
| | - Jingxuan Peng
- Department of General Surgery, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
| | - Ning Ma
- Department of General Surgery, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
| | - Qihao Pan
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
- Department of Obstetrics and Gynecology, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
| | - Yue Wei
- Department of General Surgery, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
| | - Huilin Jin
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
- Department of Hepatobiliary, Pancreatic and Splenic Surgery, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
| | - Fenghai Yu
- Department of General Surgery, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
| | - Xiaoling Huang
- Department of General Surgery, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
| | - Peng Zhang
- Department of General Surgery, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
| | - Jiarui Wang
- Department of General Surgery, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
| | - Jiaying Zheng
- Department of General Surgery, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
| | - Xiaofang Ying
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430079, China
| | - Ran-yi Liu
- State Key Laboratory of Oncology in South China & Collaborative Innovation Center for Cancer Medicine,
Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Hongyan Yu
- Department of Clinical Biological Resource Bank, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center,
Guangzhou Medical University, Guangzhou 510623, China
| | - Mong-Hong Lee
- Department of General Surgery, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
| | - Xiangqi Meng
- Department of General Surgery, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital,
Sun Yat-sen University, Guangzhou 510655, China
| |
Collapse
|
5
|
Zhang S, Chen Y, Lv Y, Feng Y, Gao C. Mitochondrial PGAM5 modulates methionine metabolism and feather follicle development by targeting Wnt/β-catenin signaling pathway in broiler chickens. J Anim Sci Biotechnol 2025; 16:35. [PMID: 40038789 DOI: 10.1186/s40104-025-01176-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 02/08/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Poor feather growth not only affects the appearance of the organism but also decreases the feed efficiency. Methionine (Met) is an essential amino acid required for feather follicle development; yet the exact mechanism involved remains insufficiently understood. METHODS A total of 180 1-day-old broilers were selected and randomly divided into 3 treatments: control group (0.45% Met), Met-deficiency group (0.25% Met), and Met-rescue group (0.45% Met in the pre-trial period and 0.25% Met in the post-trial period). The experimental period lasted for 56 d, with a pre-trial period of 1-28 d and a post-trial period of 29-56 d. In addition, Met-deficiency and Met-rescue models were constructed in feather follicle epidermal stem cell by controlling the supply of Met in the culture medium. RESULTS Dietary Met-deficiency significantly (P < 0.05) reduced the ADG, ADFI and F/G, and inhibited feather follicle development. Met supplementation significantly (P < 0.05) improved growth performance and the feather growth in broilers. Met-rescue may promote feather growth in broilers by activating the Wnt/β-catenin signaling pathway (GSK-3β, CK1, Axin1, β-catenin, Active β-catenin, TCF4, and Cyclin D1). Compared with Met-deficiency group, Met-rescue significantly (P < 0.05) increased the activity of feather follicle epidermal stem cell and mitochondrial membrane potential, activated Wnt/β-catenin signaling pathway, and decreased the content of reactive oxygen species (P < 0.05). CO-IP confirmed that mitochondrial protein PGAM5 interacted with Axin1, the scaffold protein of the disruption complex of the Wnt/β-catenin signaling pathway, and directly mediated Met regulation of Wnt/β-catenin signaling pathway and feather follicle development. CONCLUSIONS PGAM5 binding to Axin1 mediates the regulation of Wnt/β-catenin signaling pathway, and promotes feather follicle development and feather growth of broiler chickens through Met supplementation. These results provide theoretical support for the improvement of economic value and production efficiency of broiler chickens.
Collapse
Affiliation(s)
- Sheng Zhang
- College of Animal Science, Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangdong Laboratory for Lingnan Modern Agriculture, State Key Laboratory of Swine and Poultry Breeding Industry, South China Agricultural University, Guangzhou, 510642, China
| | - Yijun Chen
- College of Animal Science, Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangdong Laboratory for Lingnan Modern Agriculture, State Key Laboratory of Swine and Poultry Breeding Industry, South China Agricultural University, Guangzhou, 510642, China
| | - Yaxue Lv
- College of Animal Science, Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangdong Laboratory for Lingnan Modern Agriculture, State Key Laboratory of Swine and Poultry Breeding Industry, South China Agricultural University, Guangzhou, 510642, China
| | - Yuqing Feng
- College of Animal Science, Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangdong Laboratory for Lingnan Modern Agriculture, State Key Laboratory of Swine and Poultry Breeding Industry, South China Agricultural University, Guangzhou, 510642, China
| | - Chunqi Gao
- College of Animal Science, Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangdong Laboratory for Lingnan Modern Agriculture, State Key Laboratory of Swine and Poultry Breeding Industry, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
6
|
Jin J, Luo J, Jin X, Lim KS, He Y, Ding J, Shen Y, Hou Y, Liu H, Zhu X, Zhao J, Zhou W, Huang H, Gao Y, Xiao J, He H, Li Q, Liu L, Chen L, He Q, Zhang C. Chromatin Helicase CHD6 Establishes Proinflammatory Enhancers and Is a Synthetic Lethal Target in FH-Deficient Renal Cell Carcinoma. Cancer Res 2025; 85:675-691. [PMID: 39589780 DOI: 10.1158/0008-5472.can-24-0787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 07/01/2024] [Accepted: 11/20/2024] [Indexed: 11/27/2024]
Abstract
Fumarate hydratase (FH) deficiency causes hereditary leiomyomatosis and renal cell carcinoma (RCC). FH-deficient tumors lack effective therapeutic options. Here, we utilized an epigenetic-focused single-guide RNA library to elucidate potential drug targets in FH-deficient tumors. The screen identified chromodomain helicase DNA-binding protein 6 (CHD6) as an essential regulator of the growth of FH-mutated RCC. Mechanically, FH loss induced fumarate-mediated succinylation and inactivation of KEAP1, blocking subsequent ubiquitin-proteasome degradation of CHD6. Stabilized CHD6 formed a complex with p65 to establish proinflammatory enhancers and thereby regulate NF-κB-mediated transcription. Moreover, CHD6 recruited mSWI/SNF ATPases to maintain chromatin accessibility at CHD6-bound enhancers. The PROTAC degrader of SMARCA2/4 AU-15330 effectively abolished structures of cis-regulatory elements bound by CHD6 and suppressed the growth of FH-mutated, but not FH-intact, RCC in vivo. Collectively, these data indicate that CHD6 is a molecular bridge between FH deficiency and proinflammatory enhancer assembly that endows FH-deficient tumors with epigenetic vulnerabilities. Significance: CHD6 links FH deficiency to aberrant NF-κB activity in renal cell carcinoma, highlighting an epigenetic vulnerability for this rare tumor subtype.
Collapse
Affiliation(s)
- Juan Jin
- Department of Nephrology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Jun Luo
- Department of Urology, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiaodong Jin
- Department of Urology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Kiat Shenq Lim
- Department of Urology, Shanghai Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yang He
- The 2nd Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiawei Ding
- Department of Urology, Shanghai Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yan Shen
- Department of Urology, Shanghai Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yuchen Hou
- Division of Life Sciences and Medicine, Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Hanqing Liu
- Division of Life Sciences and Medicine, Department of Urology, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Xiaoyu Zhu
- Hunter Biotechnology, Inc., Hangzhou, China
| | - Jing Zhao
- Hunter Biotechnology, Inc., Hangzhou, China
- National Clinical Research Center for Child Health of the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China
| | - Wenjie Zhou
- Department of Urology, Shanghai Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hai Huang
- Department of Urology, Shanghai Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yi Gao
- Department of Urology, Shanghai Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jun Xiao
- Division of Life Sciences and Medicine, Department of Urology, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Hongchao He
- Department of Urology, Shanghai Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Genitourinary Medical Oncology and Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Qunyi Li
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Lianxin Liu
- Division of Life Sciences and Medicine, Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Li Chen
- Department of Pharmacy, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Shanghai, China
| | - Qiang He
- Department of Nephrology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Chuanjie Zhang
- Department of Nephrology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
- Department of Urology, Shanghai Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
- Division of Life Sciences and Medicine, Department of Urology, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| |
Collapse
|
7
|
Zhang Z, Gao Y, Qian Y, Wei B, Jiang K, Sun Z, Zhang F, Yang M, Baldi S, Yu X, Zuo Y, Ren S. The Lyn/RUVBL1 Complex Promotes Colorectal Cancer Liver Metastasis by Regulating Arachidonic Acid Metabolism Through Chromatin Remodeling. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2406562. [PMID: 39665272 PMCID: PMC11792055 DOI: 10.1002/advs.202406562] [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: 06/13/2024] [Revised: 11/26/2024] [Indexed: 12/13/2024]
Abstract
Liver metastasis is a common cause of death in colorectal cancer (CRC) patients, but epigenetic remodeling and metabolic reprogramming for CRC liver metastasis remain unclear. The study revealed that the Lyn/RUVBL1 complex is highly expressed in CRC and is closely correlated with liver metastasis. On the one hand, ATAC-seq and HiCut suggested that Lyn/RUVBL1 regulates the expression of TRIB3 through the POL II-mediated chromatin conformation of TRIB3 and thus the expression of β-catenin. This promotes the proliferation and migration of CRC through β-catenin-mediated upregulation of MMP9 and VEGF. On the other hand, metabolomics revealed that Lyn/RUVBL1 regulates the expression of PGE2 through the enzyme COX2, thereby promoting arachidonic acid (AA) metabolism. CUT-Tag showed that Lyn/RUVBL1 silencing reduces the H3K27ac level in the COX2 promoter. Then, it is found that COX2 is regulated by the transcription factor FOXA1. Lyn/RUVBL1 modulates AA metabolism by regulating the chromatin accessibility of FOXA1. AA metabolism promotes the metastasis of CRC by affecting β-catenin nuclear translocation and upregulating MMP9 and VEGF. These findings suggest that the Lyn/RUVBL1 complex mediates epigenetic remodeling to regulate the metabolic reprogramming of AA, highlighting its role in promoting the metastasis of CRC.
Collapse
Affiliation(s)
- Zhenyu Zhang
- Department of General SurgeryThe Second Hospital of Dalian Medical UniversityDalian116023China
- Department of Clinical BiochemistryCollege of Laoratory Medicine, Dalian Medical UniversityDalian116044China
| | - Yina Gao
- Department of General SurgeryThe Second Hospital of Dalian Medical UniversityDalian116023China
- Department of Clinical BiochemistryCollege of Laoratory Medicine, Dalian Medical UniversityDalian116044China
| | - Yuanyuan Qian
- Department of Clinical BiochemistryCollege of Laoratory Medicine, Dalian Medical UniversityDalian116044China
| | - Bowen Wei
- Department of Clinical BiochemistryCollege of Laoratory Medicine, Dalian Medical UniversityDalian116044China
| | - Kexin Jiang
- Department of Clinical BiochemistryCollege of Laoratory Medicine, Dalian Medical UniversityDalian116044China
| | - Zhiwei Sun
- Department of General SurgeryThe Second Hospital of Dalian Medical UniversityDalian116023China
| | - Feifan Zhang
- Department of General SurgeryThe Second Hospital of Dalian Medical UniversityDalian116023China
| | - Mingming Yang
- Department of Clinical BiochemistryCollege of Laoratory Medicine, Dalian Medical UniversityDalian116044China
| | - Salem Baldi
- Department of Clinical BiochemistryCollege of Laoratory Medicine, Dalian Medical UniversityDalian116044China
| | - Xiaoqi Yu
- Department of Clinical BiochemistryCollege of Laoratory Medicine, Dalian Medical UniversityDalian116044China
| | - Yunfei Zuo
- Department of Clinical BiochemistryCollege of Laoratory Medicine, Dalian Medical UniversityDalian116044China
| | - Shuangyi Ren
- Department of General SurgeryThe Second Hospital of Dalian Medical UniversityDalian116023China
| |
Collapse
|
8
|
Gao Y, Siyu zhang, Zhang X, Du Y, Ni T, Hao S. Crosstalk between metabolic and epigenetic modifications during cell carcinogenesis. iScience 2024; 27:111359. [PMID: 39660050 PMCID: PMC11629229 DOI: 10.1016/j.isci.2024.111359] [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] [Indexed: 12/12/2024] Open
Abstract
Genetic mutations arising from various internal and external factors drive cells to become cancerous. Cancerous cells undergo numerous changes, including metabolic reprogramming and epigenetic modifications, to support their abnormal proliferation. This metabolic reprogramming leads to the altered expression of many metabolic enzymes and the accumulation of metabolites. Recent studies have shown that these enzymes and metabolites can serve as substrates or cofactors for chromatin-modifying enzymes, thereby participating in epigenetic modifications and promoting carcinogenesis. Additionally, epigenetic modifications play a role in the metabolic reprogramming and immune evasion of cancer cells, influencing cancer progression. This review focuses on the origins of cancer, particularly the metabolic reprogramming of cancer cells and changes in epigenetic modifications. We discuss how metabolites in cancer cells contribute to epigenetic remodeling, including lactylation, acetylation, succinylation, and crotonylation. Finally, we review the impact of epigenetic modifications on tumor immunity and the latest advancements in cancer therapies targeting these modifications.
Collapse
Affiliation(s)
- Yue Gao
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China
| | - Siyu zhang
- Key Lab of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, School of Life Sciences, Ningxia University, Yinchuan 750021, China
| | - Xianhong Zhang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China
| | - Yitian Du
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China
| | - Ting Ni
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China
| | - Shuailin Hao
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China
| |
Collapse
|
9
|
Bu L, Huang S, Rao Z, Wu C, Sun BY, Liu Y, He L, Zhao D. CHD6 eviction of promoter nucleosomes maintains housekeeping transcriptional program in prostate cancer. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102397. [PMID: 39717618 PMCID: PMC11665337 DOI: 10.1016/j.omtn.2024.102397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 11/13/2024] [Indexed: 12/25/2024]
Abstract
CHD6, a member of the chromodomain helicase DNA-binding protein family, has been implicated in various diseases and tumors. However, its precise binding model of CHD6 on regulatory functional genes remains poorly understood. In this study, we discovered sharp peaks of CHD6, as the first member of CHD family for housekeeping process, binding only to the promoter region of genes in the C4-2 cell line. These genes, with conserved sharp CHD6 peaks across tumor cells, likely represent housekeeping genes ADNP and GOLGA5. Genes with sharp CHD6 peaks exhibit stable and low expression levels, sharing epigenetic features similar to housekeeping genes. Furthermore, this regulatory model also exists in both HEK293 cells and cardiomyocytes. Overall, the results of this study demonstrate that CHD6 binds to the promoter regions of housekeeping genes, regulating their histone modifications, chromatin structure, and gene expression.
Collapse
Affiliation(s)
- Lina Bu
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Shaodong Huang
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Ziyan Rao
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Chenyang Wu
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Bryan-Yu Sun
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Yanhua Liu
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Lin He
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Dongyu Zhao
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| |
Collapse
|
10
|
Cao P, Chen H, Zhang Y, Zhang Q, Shi M, Han H, Wang X, Jin L, Guo B, Hao R, Zhao X, Li Y, Gao C, Liu X, Wang Y, Yang A, Yang C, Si A, Li H, Song Q, He F, Zhou G. Genomic Amplification of TBC1D31 Promotes Hepatocellular Carcinoma Through Reducing the Rab22A-Mediated Endolysosomal Trafficking and Degradation of EGFR. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405459. [PMID: 39206796 PMCID: PMC11516053 DOI: 10.1002/advs.202405459] [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: 05/19/2024] [Revised: 07/29/2024] [Indexed: 09/04/2024]
Abstract
Hepatocellular carcinomas (HCCs) are characterized by a vast spectrum of somatic copy number alterations (CNAs); however, their functional relevance is largely unknown. By performing a genome-wide survey on prognosis-associated focal CNAs in 814 HCC patients by an integrative computational framework based on transcriptomic data, genomic amplification is identified at 8q24.13 as a promising candidate. Further evidence is provided that the 8q24.13 amplification-driven overexpression of Rab GTPase activating protein TBC1D31 exacerbates HCC growth and metastasis both in vitro and in vivo through activating Epidermal growth factor receptor (EGFR) signaling. Mechanistically, TBC1D31 acts as a Rab GTPase activating protein to catalyze GTP hydrolysis for Rab22A and then reduces the Rab22A-mediated endolysosomal trafficking and degradation of EGFR. Notably, overexpression of TBC1D31 markedly increases the resistance of HCC cells to lenvatinib, whereas inhibition of the TBC1D31-EGFR axis can reverse this resistance phenotype. This study highlights that TBC1D31 at 8q24.13 is a new critical oncogene, uncovers a novel mechanism of EGFR activation in HCC, and proposes the potential strategies for treating HCC patients with TBC1D31 amplification or overexpression.
Collapse
Affiliation(s)
- Pengbo Cao
- State Key Laboratory of Medical ProteomicsNational Center for Protein Sciences at BeijingBeijing Institute of Radiation MedicineBeijing100850China
| | - Hongxia Chen
- State Key Laboratory of Medical ProteomicsNational Center for Protein Sciences at BeijingBeijing Institute of Radiation MedicineBeijing100850China
| | - Ying Zhang
- School of Life SciencesTsinghua UniversityBeijing100084China
| | - Qi Zhang
- State Key Laboratory of Medical ProteomicsNational Center for Protein Sciences at BeijingBeijing Institute of Radiation MedicineBeijing100850China
- University of South ChinaHengyang421001China
| | | | - Huihui Han
- State Key Laboratory of Medical ProteomicsNational Center for Protein Sciences at BeijingBeijing Institute of Radiation MedicineBeijing100850China
| | - Xiaowen Wang
- State Key Laboratory of Medical ProteomicsNational Center for Protein Sciences at BeijingBeijing Institute of LifeomicsBeijing102206China
| | - Liang Jin
- State Key Laboratory of Medical ProteomicsNational Center for Protein Sciences at BeijingBeijing Institute of Radiation MedicineBeijing100850China
| | - Bingqian Guo
- State Key Laboratory of Medical ProteomicsNational Center for Protein Sciences at BeijingBeijing Institute of Radiation MedicineBeijing100850China
| | | | - Xi Zhao
- State Key Laboratory of Medical ProteomicsNational Center for Protein Sciences at BeijingBeijing Institute of Radiation MedicineBeijing100850China
| | - Yuanfeng Li
- State Key Laboratory of Medical ProteomicsNational Center for Protein Sciences at BeijingBeijing Institute of Radiation MedicineBeijing100850China
| | - Chengming Gao
- State Key Laboratory of Medical ProteomicsNational Center for Protein Sciences at BeijingBeijing Institute of Radiation MedicineBeijing100850China
| | - Xinyi Liu
- State Key Laboratory of Medical ProteomicsNational Center for Protein Sciences at BeijingBeijing Institute of Radiation MedicineBeijing100850China
| | - Yahui Wang
- State Key Laboratory of Medical ProteomicsNational Center for Protein Sciences at BeijingBeijing Institute of Radiation MedicineBeijing100850China
| | - Aiqing Yang
- State Key Laboratory of Medical ProteomicsNational Center for Protein Sciences at BeijingBeijing Institute of Radiation MedicineBeijing100850China
| | - Chenning Yang
- State Key Laboratory of Medical ProteomicsNational Center for Protein Sciences at BeijingBeijing Institute of Radiation MedicineBeijing100850China
| | - Anfeng Si
- Jinling HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjing210002China
| | - Hua Li
- Department of OncologyChengdu Military General HospitalChengdu610083China
| | - Qingfeng Song
- Affiliated Cancer Hospital of Guangxi Medical UniversityNanning530021China
| | - Fuchu He
- School of Life SciencesTsinghua UniversityBeijing100084China
- State Key Laboratory of Medical ProteomicsNational Center for Protein Sciences at BeijingBeijing Institute of LifeomicsBeijing102206China
| | - Gangqiao Zhou
- State Key Laboratory of Medical ProteomicsNational Center for Protein Sciences at BeijingBeijing Institute of Radiation MedicineBeijing100850China
- School of Life SciencesTsinghua UniversityBeijing100084China
- University of South ChinaHengyang421001China
- Guangxi Medical UniversityNanning530021China
- Hebei UniversityBaoding071000China
| |
Collapse
|
11
|
Amor M, Diaz M, Bianco V, Svecla M, Schwarz B, Rainer S, Pirchheim A, Schooltink L, Mukherjee S, Grabner GF, Beretta G, Lamina C, Norata GD, Hackl H, Kratky D. Identification of regulatory networks and crosstalk factors in brown adipose tissue and liver of a cold-exposed cardiometabolic mouse model. Cardiovasc Diabetol 2024; 23:298. [PMID: 39143620 PMCID: PMC11325583 DOI: 10.1186/s12933-024-02397-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 08/07/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND Activation of brown adipose tissue (BAT) has gained attention due to its ability to dissipate energy and counteract cardiometabolic diseases (CMDs). METHODS This study investigated the consequences of cold exposure on the BAT and liver proteomes of an established CMD mouse model based on LDL receptor-deficient (LdlrKO) mice fed a high-fat, high-sucrose, high-cholesterol diet for 16 weeks. We analyzed energy metabolism in vivo and performed untargeted proteomics on BAT and liver of LdlrKO mice maintained at 22 °C or 5 °C for 7 days. RESULTS We identified several dysregulated pathways, miRNAs, and transcription factors in BAT and liver of cold-exposed Ldlrko mice that have not been previously described in this context. Networks of regulatory interactions based on shared downstream targets and analysis of ligand-receptor pairs identified fibrinogen alpha chain (FGA) and fibronectin 1 (FN1) as potential crosstalk factors between BAT and liver in response to cold exposure. Importantly, genetic variations in the genes encoding FGA and FN1 have been associated with cardiometabolic-related phenotypes and traits in humans. DISCUSSION This study describes the key factors, pathways, and regulatory networks involved in the crosstalk between BAT and the liver in a cold-exposed CMD mouse model. These findings may provide a basis for future studies aimed at testing whether molecular mediators, as well as regulatory and signaling mechanisms involved in tissue adaption upon cold exposure, could represent a target in cardiometabolic disorders.
Collapse
Affiliation(s)
- Melina Amor
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, Graz, 8010, Austria
| | - Malena Diaz
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, Graz, 8010, Austria
| | - Valentina Bianco
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, Graz, 8010, Austria
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Monika Svecla
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, Graz, 8010, Austria
- Department of Neurosurgery, Charité- Universitätsmedizin Berlin, Berlin, Germany
| | - Birgit Schwarz
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, Graz, 8010, Austria
| | - Silvia Rainer
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, Graz, 8010, Austria
| | - Anita Pirchheim
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, Graz, 8010, Austria
| | - Laszlo Schooltink
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, Graz, 8010, Austria
| | - Suravi Mukherjee
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, Graz, 8010, Austria
| | - Gernot F Grabner
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, Graz, 8010, Austria
| | - Giangiacomo Beretta
- Department of Environmental Science and Policy, Università degli Studi di Milano, Milan, Italy
| | - Claudia Lamina
- Department of Genetics and Pharmacology, Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Hubert Hackl
- Institute of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Dagmar Kratky
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, Graz, 8010, Austria.
- BioTechMed-Graz, Graz, Austria.
| |
Collapse
|
12
|
Guo M, Yang F, Zhu L, Wang L, Li Z, Qi Z, Fotopoulos V, Yu J, Zhou J. Loss of cold tolerance is conferred by absence of the WRKY34 promoter fragment during tomato evolution. Nat Commun 2024; 15:6667. [PMID: 39107290 PMCID: PMC11303406 DOI: 10.1038/s41467-024-51036-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 07/28/2024] [Indexed: 08/10/2024] Open
Abstract
Natural evolution has resulted in reduced cold tolerance in cultivated tomato (Solanum lycopersicum). Herein, we perform a combined analysis of ATAC-Seq and RNA-Seq in cold-sensitive cultivated tomato and cold-tolerant wild tomato (S. habrochaites). We identify that WRKY34 has the most significant association with differential chromatin accessibility and expression patterns under cold stress. We find that a 60 bp InDel in the WRKY34 promoter causes differences in its transcription and cold tolerance among 376 tomato accessions. This 60 bp fragment contains a GATA cis-regulatory element that binds to SWIBs and GATA29, which synergistically suppress WRKY34 expression under cold stress. Moreover, WRKY34 interferes with the CBF cold response pathway through regulating transcription and protein levels. Our findings emphasize the importance of polymorphisms in cis-regulatory regions and their effects on chromatin structure and gene expression during crop evolution.
Collapse
Affiliation(s)
- Mingyue Guo
- Department of Horticulture, Zhejiang Provincial Key Laboratory of Horticultural Crop Quality Regulation, Zhejiang University, Yuhangtang Road 866, Hangzhou, 310058, China
| | - Fengjun Yang
- Department of Horticulture, Zhejiang Provincial Key Laboratory of Horticultural Crop Quality Regulation, Zhejiang University, Yuhangtang Road 866, Hangzhou, 310058, China
| | - Lijuan Zhu
- Department of Horticulture, Zhejiang Provincial Key Laboratory of Horticultural Crop Quality Regulation, Zhejiang University, Yuhangtang Road 866, Hangzhou, 310058, China
| | - Leilei Wang
- Department of Horticulture, Zhejiang Provincial Key Laboratory of Horticultural Crop Quality Regulation, Zhejiang University, Yuhangtang Road 866, Hangzhou, 310058, China
| | - Zhichao Li
- Department of Horticulture, Zhejiang Provincial Key Laboratory of Horticultural Crop Quality Regulation, Zhejiang University, Yuhangtang Road 866, Hangzhou, 310058, China
| | - Zhenyu Qi
- Hainan Institute, Zhejiang University, Sanya, 572000, China
- Agricultural Experiment Station, Zhejiang University, Hangzhou, 310058, China
| | - Vasileios Fotopoulos
- Cyprus University of Technology, Department of Agricultural Sciences, Biotechnology and Food Science, Lemesos, 3036, Cyprus
| | - Jingquan Yu
- Department of Horticulture, Zhejiang Provincial Key Laboratory of Horticultural Crop Quality Regulation, Zhejiang University, Yuhangtang Road 866, Hangzhou, 310058, China
- Hainan Institute, Zhejiang University, Sanya, 572000, China
- Key Laboratory of Horticultural Plants Growth, Development and Quality Improvement, Ministry of Agriculture and Rural Affairs of China, Yuhangtang Road 866, Hangzhou, 310058, China
| | - Jie Zhou
- Department of Horticulture, Zhejiang Provincial Key Laboratory of Horticultural Crop Quality Regulation, Zhejiang University, Yuhangtang Road 866, Hangzhou, 310058, China.
- Hainan Institute, Zhejiang University, Sanya, 572000, China.
- Key Laboratory of Horticultural Plants Growth, Development and Quality Improvement, Ministry of Agriculture and Rural Affairs of China, Yuhangtang Road 866, Hangzhou, 310058, China.
| |
Collapse
|
13
|
Qian Z, Liang J, Huang R, Song W, Ying J, Bi X, Zhao J, Shi Z, Liu W, Liu J, Li Z, Zhou J, Huang Z, Zhang Y, Zhao D, Wu J, Wang L, Chen X, Mao R, Zhou Y, Guo L, Hu H, Ge D, Li X, Luo Z, Yao J, Li T, Chen Q, Wang B, Wei Z, Chen K, Qu C, Cai J, Jiao Y, Bao L, Zhao H. HBV integrations reshaping genomic structures promote hepatocellular carcinoma. Gut 2024; 73:1169-1182. [PMID: 38395437 PMCID: PMC11187386 DOI: 10.1136/gutjnl-2023-330414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 02/01/2024] [Indexed: 02/25/2024]
Abstract
OBJECTIVE Hepatitis B virus (HBV)-related hepatocellular carcinoma (HCC), mostly characterised by HBV integrations, is prevalent worldwide. Previous HBV studies mainly focused on a few hotspot integrations. However, the oncogenic role of the other HBV integrations remains unclear. This study aimed to elucidate HBV integration-induced tumourigenesis further. DESIGN Here, we illuminated the genomic structures encompassing HBV integrations in 124 HCCs across ages using whole genome sequencing and Nanopore long reads. We classified a repertoire of integration patterns featured by complex genomic rearrangement. We also conducted a clustered regularly interspaced short palindromic repeat (CRISPR)-based gain-of-function genetic screen in mouse hepatocytes. We individually activated each candidate gene in the mouse model to uncover HBV integration-mediated oncogenic aberration that elicits tumourigenesis in mice. RESULTS These HBV-mediated rearrangements are significantly enriched in a bridge-fusion-bridge pattern and interchromosomal translocations, and frequently led to a wide range of aberrations including driver copy number variations in chr 4q, 5p (TERT), 6q, 8p, 16q, 9p (CDKN2A/B), 17p (TP53) and 13q (RB1), and particularly, ultra-early amplifications in chr8q. Integrated HBV frequently contains complex structures correlated with the translocation distance. Paired breakpoints within each integration event usually exhibit different microhomology, likely mediated by different DNA repair mechanisms. HBV-mediated rearrangements significantly correlated with young age, higher HBV DNA level and TP53 mutations but were less prevalent in the patients subjected to prior antiviral therapies. Finally, we recapitulated the TONSL and TMEM65 amplification in chr8q led by HBV integration using CRISPR/Cas9 editing and demonstrated their tumourigenic potentials. CONCLUSION HBV integrations extensively reshape genomic structures and promote hepatocarcinogenesis (graphical abstract), which may occur early in a patient's life.
Collapse
Affiliation(s)
- Zhaoyang Qian
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Junbo Liang
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Rong Huang
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Wei Song
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Jianming Ying
- Department of Pathology, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xinyu Bi
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianjun Zhao
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhenyu Shi
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Liver Cancer Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Wenjie Liu
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianmei Liu
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhiyu Li
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianguo Zhou
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhen Huang
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yefan Zhang
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dongbing Zhao
- Department of Pancreatic and Gastric Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianxiong Wu
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liming Wang
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiao Chen
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rui Mao
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yanchi Zhou
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lei Guo
- Department of Pathology, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hanjie Hu
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dazhuang Ge
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xingchen Li
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhiwen Luo
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jinjie Yao
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tengyan Li
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qichen Chen
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bingzhi Wang
- Department of Pathology, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhewen Wei
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kun Chen
- Department of Immunology, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chunfeng Qu
- Department of Immunology, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianqiang Cai
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of Gene Editing Screening and R&D of Digestive System Tumor Drugs, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuchen Jiao
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li Bao
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Liver Cancer Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Hong Zhao
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of Gene Editing Screening and R&D of Digestive System Tumor Drugs, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
14
|
Li Z, Wang D, Li J, Liu H, Nie L, Li C. Bone Regeneration Facilitated by Autologous Bioscaffold Material: Liquid Phase of Concentrated Growth Factor with Dental Follicle Stem Cell Loading. ACS Biomater Sci Eng 2024; 10:3173-3187. [PMID: 38605468 DOI: 10.1021/acsbiomaterials.3c01981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2024]
Abstract
The application of bioengineering techniques for achieving bone regeneration in the oral environment is an increasingly prominent field. However, the clinical use of synthetic materials carries certain risks. The liquid phase of concentrated growth factor (LPCGF), as a biologically derived material, exhibits superior biocompatibility. In this study, LPCGF was employed as a tissue engineering scaffold, hosting dental follicle cells (DFCs) to facilitate bone regeneration. Both in vivo and in vitro experimental results demonstrate that this platform significantly enhances the expression of osteogenic markers in DFCs, such as alkaline phosphatase (ALP), runt-related transcription factor 2 (Runx2), and type I collagen (Col1a1). Simultaneously, it reduces the expression of inflammation-related genes, particularly interleukin-6 (IL-6) and interleukin-8 (IL-8), thereby alleviating the negative impact of the inflammatory microenvironment on DFCs. Further investigation into potential mechanisms reveals that this process is regulated over time by the WNT pathway. Our research results demonstrate that LPCGF, with its favorable physical characteristics, holds great potential as a scaffold. It can effectively carry DFCs, thereby providing an optimal initial environment for bone regeneration. Furthermore, LPCGF endeavors to closely mimic the mechanisms of bone healing post-trauma to facilitate bone formation. This offers new perspectives and insights into bone regeneration engineering.
Collapse
Affiliation(s)
- Zhentao Li
- Stomatological Hospital of Chongqing Medical University, No. 426 Songshi North Road, Yubei District, Chongqing 401147, China
| | - Di Wang
- Stomatological Hospital of Chongqing Medical University, No. 426 Songshi North Road, Yubei District, Chongqing 401147, China
| | - Jie Li
- College of Stomatology, Chongqing Medical University, No. 426 Songshi North Road, Yubei District, Chongqing 401147, China
| | - Hao Liu
- Stomatological Hospital of Chongqing Medical University, No. 426 Songshi North Road, Yubei District, Chongqing 401147, China
| | - Li Nie
- Stomatological Hospital of Chongqing Medical University, No. 426 Songshi North Road, Yubei District, Chongqing 401147, China
| | - Conghua Li
- Stomatological Hospital of Chongqing Medical University, No. 426 Songshi North Road, Yubei District, Chongqing 401147, China
| |
Collapse
|
15
|
Tzeng YDT, Chu PY, Yong SB, Hsu TS, Tseng LM, Hou MF, Sheu JJC, Hsiao JH, Li CJ. Spatial and Single-Cell Investigations Illuminate Theragnostic value and Immune Landscape of Mitochondrial Dynamin-Like GTPase in Breast Cancer. J Cancer 2024; 15:3633-3644. [PMID: 38911373 PMCID: PMC11190754 DOI: 10.7150/jca.96100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 04/30/2024] [Indexed: 06/25/2024] Open
Abstract
Background: As we delve into the intricate world of mitochondrial inner membrane proteins, particularly Optic Atrophy types 1 and 3 (OPA1/3), we uncover their pivotal role in maintaining mitochondrial dynamic equilibrium and fusion, crucial for cellular energy production and synthesis. Despite extensive scrutiny, the significance of OPA1/3 in breast cancer (BRCA) and its interplay with the immune microenvironment remain elusive. Materials and Methods: We meticulously sourced BRCA data from renowned repositories such as The Cancer Genome Atlas (TCGA), Genotype-Tissue Expression (GTEx), Gene Expression Omnibus (GEO), and the Human Protein Atlas (HPA), leveraging cutting-edge techniques including single-cell RNA-sequencing (scRNA-seq), spatial transcriptomics, and pharmacogenomics. Through multifaceted data analysis, we endeavored to unravel the intricate role and potential value of OPA1/3 in BRCA tumorigenesis and progression. Results: Our investigation reveals a conspicuous upregulation of OPA1/3 expression in BRCA, correlating with dismal prognoses. Kaplan-Meier plot analysis underscores that heightened OPA1/3 levels are associated with poor survival rates. Both clinical specimens and biobank biopsies corroborate the elevated expression of OPA1/3 in breast cancer patients. Moreover, scRNA-seq unveils a strong correlation between OPA1/3 and macrophage infiltration in the BRCA immune milieu, alongside its association with the cellular communication network involving CXCL, TGFb, VEGF, and IL16. Conclusion: In light of these findings, OPA1/3 emerges as a promising contender for therapeutic targeting and as a potential diagnostic, prognostic, and survival biomarker in BRCA. The implications of our study underscore the pressing need to explore these novel biomarkers to enhance patient outcomes.
Collapse
Affiliation(s)
- Yen-Dun Tony Tzeng
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan
- Department of Surgery, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan
| | - Pei-Yi Chu
- Department of Pathology, Show Chwan Memorial Hospital, Changhua 500, Taiwan
| | - Su-Boon Yong
- Department of Allergy and Immunology, China Medical University Children's Hospital, Taichung 404, Taiwan
- Research Center for Allergy, Immunology, and Microbiome, China Medical University Hospital, Taichung 404, Taiwan
| | - Tzu-Sheng Hsu
- Institute of Molecular & Cellular Biology, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Ling-Ming Tseng
- Comprehensive Breast Health Center, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Ming-Feng Hou
- Division of Breast Surgery, Department of Surgery, Center for Cancer Research, Kaohsiung Medical University Chung-Ho Memorial Hospital, Kaohsiung 807, Taiwan
| | - Jim Jinn-Chyuan Sheu
- Institute of Clinical Medicine, National Cheng Kung University, Tainan 704, Taiwan
| | - Jui-Hu Hsiao
- Department of Surgery, Kaohsiung Municipal Minsheng Hospital, Kaohsiung 802, Taiwan
| | - Chia-Jung Li
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan
- Institute of BioPharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan
| |
Collapse
|
16
|
Zhang M, Wu K, Zhang W, Lin X, Cao Q, Zhang L, Chen K. The therapeutic potential of targeting the CHD protein family in cancer. Pharmacol Ther 2024; 256:108610. [PMID: 38367868 PMCID: PMC10942663 DOI: 10.1016/j.pharmthera.2024.108610] [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: 09/28/2023] [Revised: 01/06/2024] [Accepted: 02/02/2024] [Indexed: 02/19/2024]
Abstract
Accumulating evidence indicates that epigenetic events undergo deregulation in various cancer types, playing crucial roles in tumor development. Among the epigenetic factors involved in the epigenetic remodeling of chromatin, the chromodomain helicase DNA-binding protein (CHD) family frequently exhibits gain- or loss-of-function mutations in distinct cancer types. Therefore, targeting CHD remodelers holds the potential for antitumor treatment. In this review, we discuss epigenetic regulations of cancer development. We emphasize proteins in the CHD family, delving deeply into the intricate mechanisms governing their functions. Additionally, we provide an overview of current therapeutic strategies targeting CHD family members in preclinical trials. We further discuss the promising approaches that have demonstrated early signs of success in cancer treatment.
Collapse
Affiliation(s)
- Min Zhang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Kaiyuan Wu
- Basic and Translational Research Division, Department of Cardiology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Department of Bioengineering, Rice University, Houston, TX 77005, USA
| | - Weijie Zhang
- Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China; Department of Orthopaedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xia Lin
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Qi Cao
- Department of Urology, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Lili Zhang
- Basic and Translational Research Division, Department of Cardiology, Boston Children's Hospital, Boston, MA 02115, USA; Prostate Cancer Program, Dana-Farber and Harvard Cancer Center, Harvard University, Boston, MA 02115, USA
| | - Kaifu Chen
- Basic and Translational Research Division, Department of Cardiology, Boston Children's Hospital, Boston, MA 02115, USA; Prostate Cancer Program, Dana-Farber and Harvard Cancer Center, Harvard University, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
17
|
Li K, Zhang J, Lyu H, Yang J, Wei W, Wang Y, Luo H, Zhang Y, Jiang X, Yi H, Wang M, Zhang C, Wu K, Xiao L, Wen W, Xu H, Li G, Wan Y, Yang F, Yang R, Fu X, Qin B, Zhou Z, Zhang H, Lee M. CSN6-SPOP-HMGCS1 Axis Promotes Hepatocellular Carcinoma Progression via YAP1 Activation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306827. [PMID: 38308184 PMCID: PMC11005689 DOI: 10.1002/advs.202306827] [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: 09/18/2023] [Revised: 01/15/2024] [Indexed: 02/04/2024]
Abstract
Cholesterol metabolism has important roles in maintaining membrane integrity and countering the development of diseases such as obesity and cancers. Cancer cells sustain cholesterol biogenesis for their proliferation and microenvironment reprograming even when sterols are abundant. However, efficacy of targeting cholesterol metabolism for cancer treatment is always compromised. Here it is shown that CSN6 is elevated in HCC and is a positive regulator of hydroxymethylglutaryl-CoA synthase 1 (HMGCS1) of mevalonate (MVA) pathway to promote tumorigenesis. Mechanistically, CSN6 antagonizes speckle-type POZ protein (SPOP) ubiquitin ligase to stabilize HMGCS1, which in turn activates YAP1 to promote tumor growth. In orthotopic liver cancer models, targeting CSN6 and HMGCS1 hinders tumor growth in both normal and high fat diet. Significantly, HMGCS1 depletion improves YAP inhibitor efficacy in patient derived xenograft models. The results identify a CSN6-HMGCS1-YAP1 axis mediating tumor outgrowth in HCC and propose a therapeutic strategy of targeting non-alcoholic fatty liver diseases- associated HCC.
Collapse
|
18
|
Jin H, Huang X, Pan Q, Ma N, Xie X, Wei Y, Yu F, Wen W, Zhang B, Zhang P, Chen X, Wang J, Liu RY, Lin J, Meng X, Lee MH. The EIF3H-HAX1 axis increases RAF-MEK-ERK signaling activity to promote colorectal cancer progression. Nat Commun 2024; 15:2551. [PMID: 38514606 PMCID: PMC10957977 DOI: 10.1038/s41467-024-46521-3] [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: 05/03/2023] [Accepted: 02/29/2024] [Indexed: 03/23/2024] Open
Abstract
Eukaryotic initiation translation factor 3 subunit h (EIF3H) plays critical roles in regulating translational initiation and predicts poor cancer prognosis, but the mechanism underlying EIF3H tumorigenesis remains to be further elucidated. Here, we report that EIF3H is overexpressed in colorectal cancer (CRC) and correlates with poor prognosis. Conditional Eif3h deletion suppresses colorectal tumorigenesis in AOM/DSS model. Mechanistically, EIF3H functions as a deubiquitinase for HAX1 and stabilizes HAX1 via antagonizing βTrCP-mediated ubiquitination, which enhances the interaction between RAF1, MEK1 and ERK1, thereby potentiating phosphorylation of ERK1/2. In addition, activation of Wnt/β-catenin signaling induces EIF3H expression. EIF3H/HAX1 axis promotes CRC tumorigenesis and metastasis in mouse orthotopic cancer model. Significantly, combined targeting Wnt and RAF1-ERK1/2 signaling synergistically inhibits tumor growth in EIF3H-high patient-derived xenografts. These results uncover the important roles of EIF3H in mediating CRC progression through regulating HAX1 and RAF1-ERK1/2 signaling. EIF3H represents a promising therapeutic target and prognostic marker in CRC.
Collapse
Affiliation(s)
- Huilin Jin
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Hepatobiliary, Pancreatic and Splenic surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoling Huang
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qihao Pan
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Obstetrics and Gynecology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ning Ma
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoshan Xie
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yue Wei
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Fenghai Yu
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Weijie Wen
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Boyu Zhang
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Peng Zhang
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xijie Chen
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jie Wang
- Department of Radiation Oncology, Dalian Municipal Central Hospital, Dalian, China
| | - Ran-Yi Liu
- State Key Laboratory of Oncology in South China & Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Junzhong Lin
- State Key Laboratory of Oncology in South China & Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiangqi Meng
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Mong-Hong Lee
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
19
|
Guo P, Jiang J, Chu R, He F, Ge M, Fang R, Guan Q, Cheng H, Jiang C, Su T, Zhu Z, Liu H, Wei W, Zhang S, Wang Q. GRK2 mediated degradation of SAV1 initiates hyperplasia of fibroblast-like synoviocytes in rheumatoid arthritis. Acta Pharm Sin B 2024; 14:1222-1240. [PMID: 38486990 PMCID: PMC10935169 DOI: 10.1016/j.apsb.2023.12.007] [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: 07/19/2023] [Revised: 10/27/2023] [Accepted: 11/29/2023] [Indexed: 03/17/2024] Open
Abstract
Hyperplasia and migration of fibroblast-like synoviocytes (FLSs) are the key drivers in the pathogenesis of rheumatoid arthritis (RA) and joint destruction. Abundant Yes-associated protein (YAP), which is a powerful transcription co-activator for proliferative genes, was observed in the nucleus of inflammatory FLSs with unknown upstream mechanisms. Using Gene Expression Omnibus database analysis, it was found that Salvador homolog-1 (SAV1), the pivotal negative regulator of the Hippo-YAP pathway, was slightly downregulated in RA synovium. However, SAV1 protein expression is extremely reduced. Subsequently, it was revealed that SAV1 is phosphorylated, ubiquitinated, and degraded by interacting with an important serine-threonine kinase, G protein-coupled receptor (GPCR) kinase 2 (GRK2), which was predominately upregulated by GPCR activation induced by ligands such as prostaglandin E2 (PGE2) in RA. This process further contributes to the decreased phosphorylation, nuclear translocation, and transcriptional potency of YAP, and leads to aberrant FLSs proliferation. Genetic depletion of GRK2 or inhibition of GRK2 by paroxetine rescued SAV1 expression and restored YAP phosphorylation and finally inhibited RA FLSs proliferation and migration. Similarly, paroxetine treatment effectively reduced the abnormal proliferation of FLSs in a rat model of collagen-induced arthritis which was accompanied by a significant improvement in clinical manifestations. Collectively, these results elucidate the significance of GRK2 regulation of Hippo-YAP signaling in FLSs proliferation and migration and the potential application of GRK2 inhibition in the treatment of FLSs-driven joint destruction in RA.
Collapse
Affiliation(s)
- Paipai Guo
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicines, Hefei 230032, China
| | - Ji Jiang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicines, Hefei 230032, China
| | - Rui Chu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicines, Hefei 230032, China
| | - Feng He
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicines, Hefei 230032, China
| | - Mingli Ge
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicines, Hefei 230032, China
| | - Ruhong Fang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicines, Hefei 230032, China
| | - Qiuyun Guan
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicines, Hefei 230032, China
| | - Huijuan Cheng
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicines, Hefei 230032, China
| | - Chunru Jiang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicines, Hefei 230032, China
| | - Tiantian Su
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicines, Hefei 230032, China
| | - Zhenduo Zhu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicines, Hefei 230032, China
| | - Hao Liu
- School of Pharmacy, Bengbu Medical College, Bengbu 233030, China
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicines, Hefei 230032, China
| | - Shihao Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicines, Hefei 230032, China
| | - Qingtong Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicines, Hefei 230032, China
- The Third Affiliated Hospital of Anhui Medical University (the First People's Hospital of Hefei), Hefei 230061, China
| |
Collapse
|
20
|
Li J, Zhang Y, You Y, Huang Z, Wu L, Liang C, Weng B, Pan L, Huang Y, Huang Y, Yang M, Lu M, Li R, Yan X, Liu Q, Deng S. Unraveling the mechanisms of NK cell dysfunction in aging and Alzheimer's disease: insights from GWAS and single-cell transcriptomics. Front Immunol 2024; 15:1360687. [PMID: 38464521 PMCID: PMC10920339 DOI: 10.3389/fimmu.2024.1360687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 02/06/2024] [Indexed: 03/12/2024] Open
Abstract
BACKGROUND Aging is an important factor in the development of Alzheimer's disease (AD). The senescent cells can be recognized and removed by NK cells. However, NK cell function is gradually inactivated with age. Therefore, this study used senescence as an entry point to investigate how NK cells affect AD. METHODS The study validated the correlation between cognition and aging through a prospective cohort of the National Health and Nutrition Examination Survey database. A cellular trajectory analysis of the aging population was performed using single-cell nuclear transcriptome sequencing data from patients with AD and different ages. The genome-wide association study (GWAS) cohort of AD patients was used as the outcome event, and the expression quantitative trait locus was used as an instrumental variable. Causal associations between genes and AD were analyzed by bidirectional Mendelian randomization (MR) and co-localization. Finally, clinical cohorts were constructed to validate the expression of key genes. RESULTS A correlation between cognition and aging was demonstrated using 2,171 older adults over 60 years of age. Gene regulation analysis revealed that most of the highly active transcription factors were concentrated in the NK cell subpopulation of AD. NK cell trajectories were constructed for different age populations. MR and co-localization analyses revealed that CHD6 may be one of the factors influencing AD. CONCLUSION We explored different levels of AD and aging from population cohorts, single-cell data, and GWAS cohorts and found that there may be some correlations of NK cells between aging and AD. It also provides some basis for potential causation.
Collapse
Affiliation(s)
- Jinwei Li
- Department of Neurosurgery, Liuzhou Workers Hospital, Liuzhou, China
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yang Zhang
- Department of Vascular Surgery, Fuwai Yunnan Cardiovascular Hospital, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yanwei You
- Division of Sports Science and Physical Education, Tsinghua University, Beijing, China
| | - Zhiwei Huang
- Department of Neurosurgery, Liuzhou Workers Hospital, Liuzhou, China
| | - Liya Wu
- Department of Neurology, Liuzhou Workers Hospital, Liuzhou, China
| | - Cong Liang
- Department of Pharmacy, Liuzhou Workers Hospital, Liuzhou, China
| | - Baohui Weng
- Department of Neurology, Liuzhou Workers Hospital, Liuzhou, China
| | - Liya Pan
- Department of Neurology, Liuzhou Workers Hospital, Liuzhou, China
| | - Yan Huang
- Department of Neurology, Liuzhou Workers Hospital, Liuzhou, China
| | - Yushen Huang
- Department of Pharmacy, Liuzhou Workers Hospital, Liuzhou, China
| | - Mengqi Yang
- Department of Neurology, Liuzhou Workers Hospital, Liuzhou, China
| | - Mengting Lu
- Department of Dermatology, Liuzhou Workers Hospital, Liuzhou, China
| | - Rui Li
- Department of Medical Imaging, Liuzhou Workers Hospital, Liuzhou, China
| | - Xianlei Yan
- Department of Neurosurgery, Liuzhou Workers Hospital, Liuzhou, China
| | - Quan Liu
- Department of Neurosurgery, Liuzhou Workers Hospital, Liuzhou, China
| | - Shan Deng
- Department of Neurology, Liuzhou Workers Hospital, Liuzhou, China
| |
Collapse
|
21
|
Wu Z, Xiao C, Long J, Huang W, You F, Li X. Mitochondrial dynamics and colorectal cancer biology: mechanisms and potential targets. Cell Commun Signal 2024; 22:91. [PMID: 38302953 PMCID: PMC10835948 DOI: 10.1186/s12964-024-01490-4] [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: 08/22/2023] [Accepted: 01/11/2024] [Indexed: 02/03/2024] Open
Abstract
Colorectal cancer (CRC) is a significant public health concern, and its development is associated with mitochondrial dysfunction. Mitochondria can adapt to the high metabolic demands of cancer cells owing to their plasticity and dynamic nature. The fusion-fission dynamics of mitochondria play a crucial role in signal transduction and metabolic functions of CRC cells. Enhanced mitochondrial fission promotes the metabolic reprogramming of CRC cells, leading to cell proliferation, metastasis, and chemoresistance. Excessive fission can also trigger mitochondria-mediated apoptosis. In contrast, excessive mitochondrial fusion leads to adenosine triphosphate (ATP) overproduction and abnormal tumor proliferation, whereas moderate fusion protects intestinal epithelial cells from oxidative stress-induced mitochondrial damage, thus preventing colitis-associated cancer (CAC). Therefore, an imbalance in mitochondrial dynamics can either promote or inhibit CRC progression. This review provides an overview of the mechanism underlying mitochondrial fusion-fission dynamics and their impact on CRC biology. This revealed the dual role of mitochondrial fusion-fission dynamics in CRC development and identified potential drug targets. Additionally, this study partially explored mitochondrial dynamics in immune and vascular endothelial cells in the tumor microenvironment, suggesting promising prospects for targeting key fusion/fission effector proteins against CRC.
Collapse
Affiliation(s)
- Zihong Wu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Chong Xiao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
- Oncology Teaching and Research Department of Chengdu, University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Jing Long
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Wenbo Huang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Fengming You
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
- Institute of Oncology, Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| | - Xueke Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
- Oncology Teaching and Research Department of Chengdu, University of Traditional Chinese Medicine, Chengdu, 610072, China.
| |
Collapse
|
22
|
Chen J, Feng H, Wang Y, Bai X, Sheng S, Li H, Huang M, Chu X, Lei Z. The involvement of E3 ubiquitin ligases in the development and progression of colorectal cancer. Cell Death Discov 2023; 9:458. [PMID: 38104139 PMCID: PMC10725464 DOI: 10.1038/s41420-023-01760-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 11/24/2023] [Accepted: 11/30/2023] [Indexed: 12/19/2023] Open
Abstract
To date, colorectal cancer (CRC) still has limited therapeutic efficacy and poor prognosis and there is an urgent need for novel targets to improve the outcome of CRC patients. The highly conserved ubiquitination modification mediated by E3 ubiquitin ligases is an important mechanism to regulate the expression and function of tumor promoters or suppressors in CRC. In this review, we provide an overview of E3 ligases in modulating various biological processes in CRC, including proliferation, migration, stemness, metabolism, cell death, differentiation and immune response of CRC cells, emphasizing the pluripotency of E3 ubiquitin ligases. We further focus on the role of E3 ligases in regulating vital cellular signal pathways in CRC, such as Wnt/β-catenin pathway and NF-κB pathway. Additionally, considering the potential of E3 ligases as novel targets in the treatment of CRC, we discuss what aspects of E3 ligases can be utilized and exploited for efficient therapeutic strategies.
Collapse
Affiliation(s)
- Jie Chen
- Department of Medical Oncology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Haimei Feng
- Department of Medical Oncology, Jinling Hospital, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Yiting Wang
- Department of Medical Oncology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Xiaoming Bai
- Department of Medical Oncology, Jinling Hospital, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Siqi Sheng
- Department of Medical Oncology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Huiyu Li
- Department of Medical Oncology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Mengxi Huang
- Department of Medical Oncology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China.
| | - Xiaoyuan Chu
- Department of Medical Oncology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China.
- Department of Medical Oncology, Jinling Hospital, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China.
- Department of Medical Oncology, Jinling Hospital, Nanjing Medical university, Nanjing, Jiangsu Province, China.
- Department of Medical Oncology, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing, Jiangsu Province, China.
| | - Zengjie Lei
- Department of Medical Oncology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China.
- Department of Medical Oncology, Jinling Hospital, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China.
- Department of Medical Oncology, Jinling Hospital, Nanjing Medical university, Nanjing, Jiangsu Province, China.
- Department of Medical Oncology, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
23
|
Yang JR, Lu YB, Su HX, Xiao Y, Pan Q, Su F, Zhang XB, Zhu KL, Guan QL, Ling XL. USP10 promotes the progression of triple-negative breast cancer by enhancing the stability of TCF4 protein. Biochem Pharmacol 2023; 218:115864. [PMID: 37863330 DOI: 10.1016/j.bcp.2023.115864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 10/15/2023] [Accepted: 10/17/2023] [Indexed: 10/22/2023]
Abstract
Investigating the role of ubiquitin-specific peptidase 10 (USP10) in triple-negative breast cancer (TNBC). Analyzed USP10 expression levels in tumors using public databases. Detected USP10 mRNA and protein levels in cell lines. Examined USP10 expression in tumor tissues from breast cancer patients. Conducted USP10 knockdown experiments and analyzed changes in cell proliferation and metastasis. Confirmed protein-protein interactions with USP10 through mass spectrometry, Co-IP, and fluorescence experiments. Assessed impact of USP10 on transcription factor 4 (TCF4) ubiquitination and validated TCF4's influence on TNBC cells. We initially identified a pronounced overexpression of USP10 across multiple tumor types, including TNBC. Subsequently, we observed a conspicuous upregulation of USP10 expression levels in breast cancer cell lines compared to normal breast epithelial cells. However, upon subsequent depletion of USP10 within cellular contexts, we noted a substantial attenuation of malignant proliferation and metastatic potential in TNBC cells. In subsequent experimental analyses, we elucidated the physical interaction between USP10 and the transcription factor TCF4, whereby USP10 facilitated the deubiquitination modification of TCF4, consequently promoting its protein stability and contributing to the initiation and progression of TNBC. Collectively, this study demonstrates that USP10 facilitated the deubiquitination modification of TCF4, consequently promoting its protein stability and contributing to the initiation and progression of TNBC.
Collapse
Affiliation(s)
- Jing-Ru Yang
- Department of Oncology, The First Hospital of Lanzhou University, Lanzhou 730013, Gansu, PR China; The First Clinical Medical College of Lanzhou University, Lanzhou 730099, Gansu, PR China
| | - Yong-Bin Lu
- The First Clinical Medical College of Lanzhou University, Lanzhou 730099, Gansu, PR China; Scientific Development and Planing Department, The First Hospital of Lanzhou University, Lanzhou 730013, Gansu, PR China
| | - Hong-Xin Su
- Department of Radiotherapy, The First Hospital of Lanzhou University, Lanzhou 730013, Gansu, PR China
| | - Yi Xiao
- The First Clinical Medical College of Lanzhou University, Lanzhou 730099, Gansu, PR China; Breast Surgery, The First Hospital of Lanzhou University, Lanzhou 730013, Gansu, PR China
| | - Qing Pan
- Breast Surgery, The First Hospital of Lanzhou University, Lanzhou 730013, Gansu, PR China
| | - Fei Su
- Department of Oncology, The First Hospital of Lanzhou University, Lanzhou 730013, Gansu, PR China; The First Clinical Medical College of Lanzhou University, Lanzhou 730099, Gansu, PR China
| | - Xiao-Bin Zhang
- Breast Surgery, The First Hospital of Lanzhou University, Lanzhou 730013, Gansu, PR China
| | - Kai-Li Zhu
- The First Clinical Medical College of Lanzhou University, Lanzhou 730099, Gansu, PR China
| | - Quan-Lin Guan
- The First Clinical Medical College of Lanzhou University, Lanzhou 730099, Gansu, PR China; Department of Oncology Surgery, The First Hospital of Lanzhou University, Lanzhou 730013, Gansu, PR China.
| | - Xiao-Ling Ling
- Department of Oncology, The First Hospital of Lanzhou University, Lanzhou 730013, Gansu, PR China; The First Clinical Medical College of Lanzhou University, Lanzhou 730099, Gansu, PR China.
| |
Collapse
|
24
|
Dzhalilova D, Zolotova N, Fokichev N, Makarova O. Murine models of colorectal cancer: the azoxymethane (AOM)/dextran sulfate sodium (DSS) model of colitis-associated cancer. PeerJ 2023; 11:e16159. [PMID: 37927787 PMCID: PMC10624171 DOI: 10.7717/peerj.16159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 08/31/2023] [Indexed: 11/07/2023] Open
Abstract
Background Colorectal cancer (CRC) is the third most common cancer. It is a heterogeneous disease, including both hereditary and sporadic types of tumors. CRC results from complex interactions between various genetic and environmental factors. Inflammatory bowel disease is an important risk factor for developing CRC. Despite growing understanding of the CRC biology, preclinical models are still needed to investigate the etiology and pathogenesis of the disease, as well as to find new methods of treatment and prevention. Objectives The purpose of this review is to describe existing murine models of CRC with a focus on the models of colitis-associated CRC. This manuscript could be relevant for experimental biologists and oncologists. Methodology We checked PubMed and Google from 01/2018 to 05/2023 for reviews of CRC models. In addition, we searched PubMed from 01/2022 to 01/2023 for articles using the azoxymethane (AOM)/dextran sulfate sodium (DSS) CRC model. Results Existing murine models of CRC include spontaneous, genetically engineered, transplantation, and chemically induced models. For the study of colitis-associated cancer (CAC), the AOM/DSS model is predominantly used. This model is very similar in histological and molecular characteristics to the human CAC, and is highly reproducible, inexpensive, and easy to use. Despite its popularity, the AOM/DSS model is not standardized, which makes it difficult to analyze and compare data from different studies. Conclusions Each model demonstrates particular advantages and disadvantages, and allows to reproduce different subtypes or aspects of the pathogenesis of CRC.
Collapse
Affiliation(s)
- Dzhuliia Dzhalilova
- Avtsyn Research Institute of Human Morphology, Petrovsky National Research Centre of Surgery, Moscow, Russia
| | - Natalia Zolotova
- Avtsyn Research Institute of Human Morphology, Petrovsky National Research Centre of Surgery, Moscow, Russia
| | - Nikolai Fokichev
- Biological Department, Lomonosov Moscow State University, Moscow, Russian Federation
| | - Olga Makarova
- Avtsyn Research Institute of Human Morphology, Petrovsky National Research Centre of Surgery, Moscow, Russia
| |
Collapse
|
25
|
Moreta-Moraleda C, Queralt C, Vendrell-Ayats C, Forcales S, Martínez-Balibrea E. Chromatin factors: Ready to roll as biomarkers in metastatic colorectal cancer? Pharmacol Res 2023; 196:106924. [PMID: 37709185 DOI: 10.1016/j.phrs.2023.106924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 08/29/2023] [Accepted: 09/12/2023] [Indexed: 09/16/2023]
Abstract
Colorectal cancer (CRC) ranks as the third most prevalent cancer globally and stands as the fourth leading cause of cancer-related fatalities in 2020. Survival rates for metastatic disease have slightly improved in recent decades, with clinical trials showing median overall survival of approximately 24-30 months. This progress can be attributed to the integration of chemotherapeutic treatments alongside targeted therapies and immunotherapy. Despite these modest improvements, the primary obstacle to successful treatment for advanced CRC lies in the development of chemoresistance, whether inherent or acquired, which remains the major cause of treatment failure. Epigenetics has emerged as a hallmark of cancer, contributing to master transcription regulation and genome stability maintenance. As a result, epigenetic factors are starting to appear as potential clinical biomarkers for diagnosis, prognosis, and prediction of treatment response in CRC.In recent years, numerous studies have investigated the influence of DNA methylation, histone modifications, and chromatin remodelers on responses to chemotherapeutic treatments. While there is accumulating evidence indicating their significant involvement in various types of cancers, the exact relationship between chromatin landscapes and treatment modulation in CRC remains elusive. This review aims to provide a comprehensive summary of the most pertinent and extensively researched epigenetic-associated mechanisms described between 2015 and 2022 and their potential usefulness as predictive biomarkers in the metastatic disease.
Collapse
Affiliation(s)
- Cristina Moreta-Moraleda
- Immunology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, University of Barcelona, c/Feixa Llarga s/n, 08917 L'Hospitalet de Llobregat, Barcelona, Spain; Group of Inflammation, Immunity and Cancer, Molecular Mechanisms and Experimental Therapy in Oncology Program (Oncobell), The Bellvitge Biomedical Research Institute ( IDIBELL), Hospital Duran i Reynals 3a Planta, Av. Gran Via de l'Hospitalet 199, 08908 L'Hospitalet de Llobregat, Spain
| | - Cristina Queralt
- ProCURE Program, Catalan Instiute of Oncology, Carretera de Can Ruti, camí de les escoles s/n, 08916 Badalona, Spain
| | - Carla Vendrell-Ayats
- ProCURE Program, Catalan Instiute of Oncology, Carretera de Can Ruti, camí de les escoles s/n, 08916 Badalona, Spain; CARE Program, Germans Trias I Pujol Research Institute (IGTP), Carretera de Can Ruti, camí de les escoles s/n, 08916 Badalona, Spain
| | - Sonia Forcales
- Serra Húnter Programme, Immunology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, University of Barcelona, c/Feixa Llarga s/n, 08917 L'Hospitalet de Llobregat, Barcelona, Spain; Group of Inflammation, Immunity and Cancer, Molecular Mechanisms and Experimental Therapy in Oncology Program (Oncobell), The Bellvitge Biomedical Research Institute ( IDIBELL), Hospital Duran i Reynals 3a Planta, Av. Gran Via de l'Hospitalet 199, 08908 L'Hospitalet de Llobregat, Spain.
| | - Eva Martínez-Balibrea
- ProCURE Program, Catalan Instiute of Oncology, Carretera de Can Ruti, camí de les escoles s/n, 08916 Badalona, Spain; CARE Program, Germans Trias I Pujol Research Institute (IGTP), Carretera de Can Ruti, camí de les escoles s/n, 08916 Badalona, Spain.
| |
Collapse
|
26
|
Wei W, Qin B, Wen W, Zhang B, Luo H, Wang Y, Xu H, Xie X, Liu S, Jiang X, Wang M, Tang Q, Zhang J, Yang R, Fan Z, Lyu H, Lin J, Li K, Lee MH. FBXW7β loss-of-function enhances FASN-mediated lipogenesis and promotes colorectal cancer growth. Signal Transduct Target Ther 2023; 8:187. [PMID: 37202390 PMCID: PMC10195794 DOI: 10.1038/s41392-023-01405-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 02/11/2023] [Accepted: 02/28/2023] [Indexed: 05/20/2023] Open
Abstract
Continuous de novo fatty acid synthesis is required for the biosynthetic demands of tumor. FBXW7 is a highly mutated gene in CRC, but its biological functions in cancer are not fully characterized. Here, we report that FBXW7β, a FBXW7 isoform located in the cytoplasm and frequently mutated in CRC, is an E3 ligase of fatty acid synthase (FASN). Cancer-specific FBXW7β mutations that could not degrade FASN can lead to sustained lipogenesis in CRC. COP9 signalosome subunit 6 (CSN6), an oncogenic marker of CRC, increases lipogenesis via interacting with and stabilizing FASN. Mechanistic studies show that CSN6 associates with both FBXW7β and FASN, and antagonizes FBXW7β's activity by enhancing FBXW7β autoubiquitination and degradation, which in turn prevents FBXW7β-mediated FASN ubiquitination and degradation, thereby regulating lipogenesis positively. Both CSN6 and FASN are positively correlated in CRC, and CSN6-FASN axis, regulated by EGF, is responsible for poor prognosis of CRC. The EGF-CSN6-FASN axis promotes tumor growth and implies a treatment strategy of combination of orlistat and cetuximab. Patient-derived xenograft experiments prove the effectiveness of employing orlistat and cetuximab combination in suppressing tumor growth for CSN6/FASN-high CRC. Thus, CSN6-FASN axis reprograms lipogenesis to promote tumor growth and is a target for cancer intervening strategy in CRC.
Collapse
Affiliation(s)
- Wenxia Wei
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, China
| | - Baifu Qin
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, China
| | - Weijie Wen
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, China
| | - Boyu Zhang
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, China
| | - Haidan Luo
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, China
| | - Yuzhi Wang
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, China
| | - Hui Xu
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, China
| | - Xiaoshan Xie
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, China
| | - Sicheng Liu
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, China
- Second Department of Medical Oncology, The Third Affiliated Hospital of Kunming Medical University, Kunming, 650118, China
| | - Xin Jiang
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, China
| | - Mengan Wang
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, China
| | - Qin Tang
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Jiayu Zhang
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, China
| | - Runxiang Yang
- Second Department of Medical Oncology, The Third Affiliated Hospital of Kunming Medical University, Kunming, 650118, China
| | - Zongmin Fan
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, China
| | - Haiwen Lyu
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, China
| | - Junzhong Lin
- Department of Colorectal Surgery, Cancer Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Kai Li
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China.
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, China.
| | - Mong-Hong Lee
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China.
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, China.
- Department of Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China.
| |
Collapse
|
27
|
Neto Í, Rocha J, Gaspar MM, Reis CP. Experimental Murine Models for Colorectal Cancer Research. Cancers (Basel) 2023; 15:2570. [PMID: 37174036 PMCID: PMC10177088 DOI: 10.3390/cancers15092570] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
Colorectal cancer (CRC) is the third most prevalent malignancy worldwide and in both sexes. Numerous animal models for CRC have been established to study its biology, namely carcinogen-induced models (CIMs) and genetically engineered mouse models (GEMMs). CIMs are valuable for assessing colitis-related carcinogenesis and studying chemoprevention. On the other hand, CRC GEMMs have proven to be useful for evaluating the tumor microenvironment and systemic immune responses, which have contributed to the discovery of novel therapeutic approaches. Although metastatic disease can be induced by orthotopic injection of CRC cell lines, the resulting models are not representative of the full genetic diversity of the disease due to the limited number of cell lines suitable for this purpose. On the other hand, patient-derived xenografts (PDX) are the most reliable for preclinical drug development due to their ability to retain pathological and molecular characteristics. In this review, the authors discuss the various murine CRC models with a focus on their clinical relevance, benefits, and drawbacks. From all models discussed, murine CRC models will continue to be an important tool in advancing our understanding and treatment of this disease, but additional research is required to find a model that can correctly reflect the pathophysiology of CRC.
Collapse
Affiliation(s)
- Íris Neto
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (Í.N.); (J.R.)
| | - João Rocha
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (Í.N.); (J.R.)
| | - Maria Manuela Gaspar
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (Í.N.); (J.R.)
| | - Catarina P. Reis
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (Í.N.); (J.R.)
- Instituto de Biofísica e Engenharia Biomédica (IBEB), Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| |
Collapse
|