1
|
Qiu C, Tang C, Tang Y, Su K, Chai X, Zhan Z, Niu X, Li J. RGS5 + lymphatic endothelial cells facilitate metastasis and acquired drug resistance of breast cancer through oxidative stress-sensing mechanism. Drug Resist Updat 2024; 77:101149. [PMID: 39306871 DOI: 10.1016/j.drup.2024.101149] [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: 05/24/2024] [Revised: 08/24/2024] [Accepted: 09/03/2024] [Indexed: 11/12/2024]
Abstract
AIMS Oxidative stress reflected by elevated reactive oxygen species (ROS) in the tumor ecosystem, is a hallmark of human cancers. The mechanisms by which oxidative stress regulate the metastatic ecosystem and resistance remain elusive. This study aimed to dissect the oxidative stress-sensing machinery during the evolvement of early dissemination and acquired drug resistance in breast cancer. METHODS Here, we constructed single-cell landscape of primary breast tumors and metastatic lymph nodes, and focused on RGS5+ endothelial cell subpopulation in breast cancer metastasis and resistance. RESULTS We reported on RGS5 as a master in endothelial cells sensing oxidative stress. RGS5+ endothelial cells facilitated tumor-endothelial adhesion and transendothelial migration of breast cancer cells. Antioxidant suppressed oxidative stress-induced RGS5 expression in endothelial cells, and prevented adhesion and transendothelial migration of cancer cells. RGS5-overexpressed HLECs displayed attenuated glycolysis and oxidative phosphorylation. Drug-resistant HLECs with RGS5 overexpression conferred acquired drug resistance of breast cancer cells. Importantly, genetic knockdown of RGS5 prevented tumor growth and lymph node metastasis. CONCLUSIONS Our work demonstrates that RGS5 in lymphatic endothelial cells senses oxidative stress to promote breast cancer lymph node metastasis and resistance, providing a novel insight into a potentially targetable oxidative stress-sensing machinery in breast cancer treatment.
Collapse
Affiliation(s)
- Caixin Qiu
- Department of Gastrointestine and Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Chaoyi Tang
- Department of Gastrointestine and Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Yujun Tang
- Department of Gastrointestine and Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Ka Su
- Department of Gastrointestine and Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Xiao Chai
- Department of Gastrointestine and Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Zexu Zhan
- Department of Gastrointestine and Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Xing Niu
- China Medical University Shenyang 110122, China; Experimental Center of BIOQGene, YuanDong International Academy of Life Sciences, 999077, Hong Kong, China.
| | - Jiehua Li
- Department of Gastrointestine and Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China.
| |
Collapse
|
2
|
Xiong Z, He L, Pi F, Yu Y, Xiao Z, Chen T. Intracellular Redox Environment Determines Cancer-normal Cell Selectivity of Selenium Nanoclusters. Angew Chem Int Ed Engl 2024:e202416006. [PMID: 39400451 DOI: 10.1002/anie.202416006] [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: 08/21/2024] [Revised: 09/24/2024] [Accepted: 10/11/2024] [Indexed: 10/15/2024]
Abstract
Elucidating the chemical structure and intracellular action mechanisms is still the critical limit for the clinical translation of nanomedicines. Intracellular redox environments originating from cell metabolism are key factors affecting internalized drug efficacy. Herein, we engineer Se-Se/Se-S bond to assemble selenium (Se) nanoclusters (SeClus) with intracellular redox environment-driven selective structure. Chemical structure analysis reveals that, the bonding of sulfur atom in intermediates to the two neighboring or interposition Se atoms in Se rings is the key internal driving force for SeClus formation. This nanocluster can be predominantly transformed to selenocysteine to facilitate selenoproteins synthesis in normal cells, while metabolize to cytotoxic SeO3 2- based on the oxidative intracellular redox environment of cancer cells. Resultantly, SeClus exhibit significant cell proliferation inhibition ability to cancer cells and impressive safety to normal cells. Taken together, this study not only clarifies the chemical nature of the atom engineering of SeClus, but also elucidates its intracellular redox environment-oriented anticancer mechanism.
Collapse
Affiliation(s)
- Zushuang Xiong
- Department of Neurology and Stroke Center of The First Affiliated Hospital, College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China
| | - Lizhen He
- Department of Neurology and Stroke Center of The First Affiliated Hospital, College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China
| | - Fen Pi
- Department of Neurology and Stroke Center of The First Affiliated Hospital, College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China
| | - Yanzi Yu
- Department of Neurology and Stroke Center of The First Affiliated Hospital, College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China
| | - Zhirong Xiao
- Department of Neurology and Stroke Center of The First Affiliated Hospital, College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China
| | - Tianfeng Chen
- Department of Neurology and Stroke Center of The First Affiliated Hospital, College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China
| |
Collapse
|
3
|
Biondini M, Lehuédé C, Tabariès S, Annis MG, Pacis A, Ma EH, Tam C, Hsu BE, Audet-Delage Y, Abu-Thuraia A, Girondel C, Sabourin V, Totten SP, de Sá Tavares Russo M, Bridon G, Avizonis D, Guiot MC, St-Pierre J, Ursini-Siegel J, Jones R, Siegel PM. Metastatic breast cancer cells are metabolically reprogrammed to maintain redox homeostasis during metastasis. Redox Biol 2024; 75:103276. [PMID: 39053265 PMCID: PMC11321393 DOI: 10.1016/j.redox.2024.103276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 07/19/2024] [Indexed: 07/27/2024] Open
Abstract
Metabolic rewiring is essential for tumor growth and progression to metastatic disease, yet little is known regarding how cancer cells modify their acquired metabolic programs in response to different metastatic microenvironments. We have previously shown that liver-metastatic breast cancer cells adopt an intrinsic metabolic program characterized by increased HIF-1α activity and dependence on glycolysis. Here, we confirm by in vivo stable isotope tracing analysis (SITA) that liver-metastatic breast cancer cells retain a glycolytic profile when grown as mammary tumors or liver metastases. However, hepatic metastases exhibit unique metabolic adaptations including elevated expression of genes involved in glutathione (GSH) biosynthesis and reactive oxygen species (ROS) detoxification when compared to mammary tumors. Accordingly, breast-cancer-liver-metastases exhibited enhanced de novo GSH synthesis. Confirming their increased capacity to mitigate ROS-mediated damage, liver metastases display reduced levels of 8-Oxo-2'-deoxyguanosine. Depletion of the catalytic subunit of the rate-limiting enzyme in glutathione biosynthesis, glutamate-cysteine ligase (GCLC), strongly reduced the capacity of breast cancer cells to form liver metastases, supporting the importance of these distinct metabolic adaptations. Loss of GCLC also affected the early steps of the metastatic cascade, leading to decreased numbers of circulating tumor cells (CTCs) and impaired metastasis to the liver and the lungs. Altogether, our results indicate that GSH metabolism could be targeted to prevent the dissemination of breast cancer cells.
Collapse
Affiliation(s)
- Marco Biondini
- Goodman Cancer Institute, McGill University, Montréal, Québec, Qc, H3A 1A3, Canada; Department of Medicine, McGill University, Montréal, Québec, Qc, H3A 1A3, Canada
| | - Camille Lehuédé
- Goodman Cancer Institute, McGill University, Montréal, Québec, Qc, H3A 1A3, Canada; Department of Medicine, McGill University, Montréal, Québec, Qc, H3A 1A3, Canada
| | - Sébastien Tabariès
- Goodman Cancer Institute, McGill University, Montréal, Québec, Qc, H3A 1A3, Canada; Department of Medicine, McGill University, Montréal, Québec, Qc, H3A 1A3, Canada
| | - Matthew G Annis
- Goodman Cancer Institute, McGill University, Montréal, Québec, Qc, H3A 1A3, Canada; Department of Medicine, McGill University, Montréal, Québec, Qc, H3A 1A3, Canada
| | - Alain Pacis
- Goodman Cancer Institute, McGill University, Montréal, Québec, Qc, H3A 1A3, Canada; Canadian Center for Computational Genomics, McGill University, Montréal, Québec, Qc, H3A 1A3, Canada
| | - Eric H Ma
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA
| | - Christine Tam
- Goodman Cancer Institute, McGill University, Montréal, Québec, Qc, H3A 1A3, Canada; Department of Medicine, McGill University, Montréal, Québec, Qc, H3A 1A3, Canada
| | - Brian E Hsu
- Goodman Cancer Institute, McGill University, Montréal, Québec, Qc, H3A 1A3, Canada; Department of Medicine, McGill University, Montréal, Québec, Qc, H3A 1A3, Canada
| | - Yannick Audet-Delage
- Department of Biochemistry, Microbiology and Immunology and Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, K1H 8M5, Canada
| | - Afnan Abu-Thuraia
- Goodman Cancer Institute, McGill University, Montréal, Québec, Qc, H3A 1A3, Canada; Department of Medicine, McGill University, Montréal, Québec, Qc, H3A 1A3, Canada
| | - Charlotte Girondel
- Goodman Cancer Institute, McGill University, Montréal, Québec, Qc, H3A 1A3, Canada; Department of Medicine, McGill University, Montréal, Québec, Qc, H3A 1A3, Canada
| | - Valerie Sabourin
- Segal Cancer Center, Lady Davis Institute for Medical Research, Sir Mortimer B. Davis Jewish General Hospital, Montréal, Québec, Qc, H3A 1A3, Canada
| | - Stephanie P Totten
- Segal Cancer Center, Lady Davis Institute for Medical Research, Sir Mortimer B. Davis Jewish General Hospital, Montréal, Québec, Qc, H3A 1A3, Canada
| | - Mariana de Sá Tavares Russo
- Goodman Cancer Institute, McGill University, Montréal, Québec, Qc, H3A 1A3, Canada; Department of Medicine, McGill University, Montréal, Québec, Qc, H3A 1A3, Canada
| | - Gaëlle Bridon
- Goodman Cancer Institute, McGill University, Montréal, Québec, Qc, H3A 1A3, Canada; Department of Medicine, McGill University, Montréal, Québec, Qc, H3A 1A3, Canada
| | - Daina Avizonis
- Goodman Cancer Institute, McGill University, Montréal, Québec, Qc, H3A 1A3, Canada; Department of Medicine, McGill University, Montréal, Québec, Qc, H3A 1A3, Canada
| | - Marie-Christine Guiot
- Goodman Cancer Institute, McGill University, Montréal, Québec, Qc, H3A 1A3, Canada; Department of Pathology, Montreal Neurological Hospital/Institute, McGill University Health Centre, Montréal, Québec, Qc, H3A 1A3, Canada
| | - Julie St-Pierre
- Department of Biochemistry, Microbiology and Immunology and Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, K1H 8M5, Canada
| | - Josie Ursini-Siegel
- Segal Cancer Center, Lady Davis Institute for Medical Research, Sir Mortimer B. Davis Jewish General Hospital, Montréal, Québec, Qc, H3A 1A3, Canada
| | - Russell Jones
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA
| | - Peter M Siegel
- Goodman Cancer Institute, McGill University, Montréal, Québec, Qc, H3A 1A3, Canada; Department of Medicine, McGill University, Montréal, Québec, Qc, H3A 1A3, Canada.
| |
Collapse
|
4
|
Gu R, Fang H, Wang R, Dai W, Cai G. A comprehensive overview of the molecular features and therapeutic targets in BRAF V600E-mutant colorectal cancer. Clin Transl Med 2024; 14:e1764. [PMID: 39073010 DOI: 10.1002/ctm2.1764] [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: 05/07/2024] [Revised: 06/23/2024] [Accepted: 07/03/2024] [Indexed: 07/30/2024] Open
Abstract
As one of the most prevalent digestive system tumours, colorectal cancer (CRC) poses a significant threat to global human health. With the emergence of immunotherapy and target therapy, the prognosis for the majority of CRC patients has notably improved. However, the subset of patients with BRAF exon 15 p.V600E mutation (BRAFV600E) has not experienced remarkable benefits from these therapeutic advancements. Hence, researchers have undertaken foundational investigations into the molecular pathology of this specific subtype and clinical effectiveness of diverse therapeutic drug combinations. This review comprehensively summarised the distinctive molecular features and recent clinical research advancements in BRAF-mutant CRC. To explore potential therapeutic targets, this article conducted a systematic review of ongoing clinical trials involving patients with BRAFV600E-mutant CRC.
Collapse
Affiliation(s)
- Ruiqi Gu
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hongsheng Fang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Renjie Wang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Weixing Dai
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Guoxiang Cai
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
5
|
Zhou Z, Qu C, Zhou P, Zhou Q, Li D, Wu X, Yang L. Extracellular vesicles activated cancer-associated fibroblasts promote lung cancer metastasis through mitophagy and mtDNA transfer. J Exp Clin Cancer Res 2024; 43:158. [PMID: 38825680 PMCID: PMC11145873 DOI: 10.1186/s13046-024-03077-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 05/21/2024] [Indexed: 06/04/2024] Open
Abstract
BACKGROUND Studies have shown that oxidative stress and its resistance plays important roles in the process of tumor metastasis, and mitochondrial dysfunction caused by mitochondrial DNA (mtDNA) damage is an important molecular event in oxidative stress. In lung cancer, the normal fibroblasts (NFs) are activated as cancer-associated fibroblasts (CAFs), and act in the realms of the tumor microenvironment (TME) with consequences for tumor growth and metastasis. However, its activation mechanism and whether it participates in tumor metastasis through antioxidative stress remain unclear. METHODS The role and signaling pathways of tumor cell derived extracellular vesicles (EVs) activating NFs and the characteristic of induced CAFs (iCAFs) were measured by the transmission electron microscopy, nanoparticle tracking analysis, immunofluorescence, collagen contraction assay, quantitative PCR, immunoblotting, luciferase reporter assay and mitochondrial membrane potential detection. Mitochondrial genome and single nucleotide polymorphism sequencing were used to investigate the transport of mtDNA from iCAFs to ρ0 cells, which were tumor cells with mitochondrial dysfunction caused by depletion of mtDNA. Further, the effects of iCAFs on mitochondrial function, growth and metastasis of tumor cells were analysed in co-culture models both in vitro and in vivo, using succinate dehydrogenase, glutathione and oxygen consumption rate measurements, CCK-8 assay, transwell assay, xenotransplantation and metastasis experiments as well as in situ hybridization and immunohistochemistry. RESULTS Our findings revealed that EVs derived from high-metastatic lung cancer cells packaged miR-1290 that directly targets MT1G, leading to activation of AKT signaling in NFs and inducing NFs conversion to CAFs. The iCAFs exhibit higher levels of autophagy and mitophagy and more mtDNA release, and reactive oxygen species (ROS) could further promote this process. After cocultured with the conditioned medium (CM) of iCAFs, the ρ0 cells may restore its mitochondrial function by acquisition of mtDNA from CAFs, and further promotes tumor metastasis. CONCLUSIONS These results elucidate a novel mechanism that CAFs activated by tumor-derived EVs can promote metastasis by transferring mtDNA and restoring mitochondrial function of tumor cells which result in resistance of oxidative stress, and provide a new therapeutic target for lung cancer metastasis.
Collapse
Affiliation(s)
- Zhuan Zhou
- Department of Oncology, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410078, China
- Cancer Research Institute, School of Basic Medicine Science, Central South University, Xiangya Road 110, Changsha, 410078, China
- Department of Laboratory Medicine, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, 410078, China
| | - Chunhui Qu
- Department of Oncology, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410078, China
- Cancer Research Institute, School of Basic Medicine Science, Central South University, Xiangya Road 110, Changsha, 410078, China
| | - Peijun Zhou
- Department of Oncology, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410078, China
- Cancer Research Institute, School of Basic Medicine Science, Central South University, Xiangya Road 110, Changsha, 410078, China
| | - Qin Zhou
- Department of Oncology, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410078, China
| | - Dan Li
- Department of Life Science, College of Biology, Hunan University, Changsha, 410012, China
| | - Xia Wu
- Cancer Research Institute, School of Basic Medicine Science, Central South University, Xiangya Road 110, Changsha, 410078, China.
- Department of Pathology, The Second Xiangya Hospital, Central South University, Renmin Middle Road 139, Changsha, 410011, China.
| | - Lifang Yang
- Department of Oncology, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410078, China.
- Cancer Research Institute, School of Basic Medicine Science, Central South University, Xiangya Road 110, Changsha, 410078, China.
| |
Collapse
|
6
|
Lewis K, La Selva R, Maldonado E, Annis MG, Najyb O, Cepeda Cañedo E, Totten S, Hébert S, Sabourin V, Mirabelli C, Ciccolini E, Lehuédé C, Choinière L, Russo M, Avizonis D, Park M, St-Pierre J, Kleinman CL, Siegel PM, Ursini-Siegel J. p66ShcA promotes malignant breast cancer phenotypes by alleviating energetic and oxidative stress. Redox Biol 2024; 70:103028. [PMID: 38211442 PMCID: PMC10821068 DOI: 10.1016/j.redox.2024.103028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 01/02/2024] [Indexed: 01/13/2024] Open
Abstract
Significant efforts have focused on identifying targetable genetic drivers that support the growth of solid tumors and/or increase metastatic ability. During tumor development and progression to metastatic disease, physiological and pharmacological selective pressures influence parallel adaptive strategies within cancer cell sub-populations. Such adaptations allow cancer cells to withstand these stressful microenvironments. This Darwinian model of stress adaptation often prevents durable clinical responses and influences the emergence of aggressive cancers with increased metastatic fitness. However, the mechanisms contributing to such adaptive stress responses are poorly understood. We now demonstrate that the p66ShcA redox protein, itself a ROS inducer, is essential for survival in response to physiological stressors, including anchorage independence and nutrient deprivation, in the context of poor outcome breast cancers. Mechanistically, we show that p66ShcA promotes both glucose and glutamine metabolic reprogramming in breast cancer cells, to increase their capacity to engage catabolic metabolism and support glutathione synthesis. In doing so, chronic p66ShcA exposure contributes to adaptive stress responses, providing breast cancer cells with sufficient ATP and redox balance needed to withstand such transient stressed states. Our studies demonstrate that p66ShcA functionally contributes to the maintenance of aggressive phenotypes and the emergence of metastatic disease by forcing breast tumors to adapt to chronic and moderately elevated levels of oxidative stress.
Collapse
Affiliation(s)
- Kyle Lewis
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada; Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Rachel La Selva
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada; Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Elias Maldonado
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada; Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Matthew G Annis
- Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Ouafa Najyb
- Department of Biochemistry, McGill University, Montreal, QC, Canada; Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Eduardo Cepeda Cañedo
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada; Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Stephanie Totten
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada; Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Steven Hébert
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada
| | - Valérie Sabourin
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada
| | - Caitlynn Mirabelli
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada; Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Emma Ciccolini
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada; Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Camille Lehuédé
- Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Luc Choinière
- Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Mariana Russo
- Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Daina Avizonis
- Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Morag Park
- Department of Biochemistry, McGill University, Montreal, QC, Canada; Division of Experimental Medicine, McGill University, Montreal, QC, Canada; Goodman Cancer Institute, McGill University, Montreal, QC, Canada; Gerald Bronfman Department of Oncology, McGill University, Montreal, QC, Canada
| | - Julie St-Pierre
- Department of Biochemistry, Microbiology and Immunology and Ottawa Institute of Systems Biology, University of Ottawa, ON, Canada
| | - Claudia L Kleinman
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada; Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Peter M Siegel
- Department of Biochemistry, McGill University, Montreal, QC, Canada; Division of Experimental Medicine, McGill University, Montreal, QC, Canada; Goodman Cancer Institute, McGill University, Montreal, QC, Canada; Gerald Bronfman Department of Oncology, McGill University, Montreal, QC, Canada
| | - Josie Ursini-Siegel
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada; Department of Biochemistry, McGill University, Montreal, QC, Canada; Division of Experimental Medicine, McGill University, Montreal, QC, Canada; Gerald Bronfman Department of Oncology, McGill University, Montreal, QC, Canada.
| |
Collapse
|
7
|
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
|
8
|
Karno B, Edwards DN, Chen J. Metabolic control of cancer metastasis: role of amino acids at secondary organ sites. Oncogene 2023; 42:3447-3456. [PMID: 37848626 PMCID: PMC11323979 DOI: 10.1038/s41388-023-02868-3] [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: 07/13/2023] [Revised: 09/29/2023] [Accepted: 10/10/2023] [Indexed: 10/19/2023]
Abstract
Most cancer-related deaths are caused by the metastases, which commonly develop at multiple organ sites including the brain, bone, and lungs. Despite longstanding observations that the spread of cancer is not random, our understanding of the mechanisms that underlie metastatic spread to specific organs remains limited. However, metabolism has recently emerged as an important contributor to metastasis. Amino acids are a significant nutrient source to cancer cells and their metabolism which can serve to fuel biosynthetic pathways capable of facilitating cell survival and tumor expansion while also defending against oxidative stress. Compared to the primary tumor, each of the common metastatic sites exhibit vastly different nutrient compositions and environmental stressors, necessitating the need of cancer cells to metabolically thrive in their new environment during colonization and outgrowth. This review seeks to summarize the current literature on amino acid metabolism pathways that support metastasis to common secondary sites, including impacts on immune responses. Understanding the role of amino acids in secondary organ sites may offer opportunities for therapeutic inhibition of cancer metastasis.
Collapse
Affiliation(s)
- Breelyn Karno
- Program in Cancer Biology, Vanderbilt University, Nashville, TN, 37232, USA
| | - Deanna N Edwards
- Department of Medicine, Division of Rheumatology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jin Chen
- Program in Cancer Biology, Vanderbilt University, Nashville, TN, 37232, USA.
- Department of Medicine, Division of Rheumatology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, USA.
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|