1
|
Liu Y, Yan Z, Liu C, Yang R, Zheng Q, Jian J, Wang M, Wang L, Weng X, Chen Z, Liu X. Integrated RNA sequencing analysis and machine learning identifies a metabolism-related prognostic signature in clear cell renal cell carcinoma. Sci Rep 2025; 15:1691. [PMID: 39799252 PMCID: PMC11724983 DOI: 10.1038/s41598-025-85618-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 01/06/2025] [Indexed: 01/15/2025] Open
Abstract
The connection between metabolic reprogramming and tumor progression has been demonstrated in an increasing number of researches. However, further research is required to identify how metabolic reprogramming affects interpatient heterogeneity and prognosis in clear cell renal cell carcinoma (ccRCC). In this work, single-cell RNA sequencing (scRNA-seq) based deconvolution was utilized to create a malignant cell hierarchy with metabolic differences and to investigate the relationship between metabolic biomarkers and prognosis. Simultaneously, we created a machine learning-based approach for creating metabolism-related prognostic signature (MRPS). Gamma-glutamyltransferase 6 (GGT6) was further explored for deep biological insights through in vitro experiments. Compared to 51 published signatures and conventional clinical features, MRPS showed substantially higher accuracy. Meanwhile, high MRPS-risk samples demonstrated an immunosuppressive phenotype with more infiltrations of regulatory T cell (Treg) and tumour-associated macrophage (TAM). Following the administration of immune checkpoint inhibitors (ICIs), MRPS showed consistent and strong performance and was an independent risk factor for overall survival. GGT6, an essential metabolic indicator and component of MRPS, has been proven to support proliferation and invasion in ccRCC. MRPS has the potential to be a highly effective tool in improving the clinical results of patients with ccRCC.
Collapse
Affiliation(s)
- Yunxun Liu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zhiwei Yan
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Cheng Liu
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Rui Yang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Qingyuan Zheng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jun Jian
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Minghui Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Lei Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xiaodong Weng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Zhiyuan Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Xiuheng Liu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
2
|
Dannesboe J, Bastrup JA, Nielsen KH, Munck P, Thomsen MB, Hawkins CL, Jepps TA. Paracetamol metabolism by endothelial cells - Potential mechanism underlying intravenous paracetamol-induced hypotension. Pharmacol Res 2025; 211:107540. [PMID: 39653302 DOI: 10.1016/j.phrs.2024.107540] [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: 07/12/2024] [Revised: 11/24/2024] [Accepted: 12/05/2024] [Indexed: 12/16/2024]
Abstract
It was shown previously that a metabolite of acetaminophen (APAP), N-acetyl-p-benzoquinone imine (NAPQI), is a potent vasodilator, which could underlie the hypotension observed when APAP is administered intravenously. However, it is unknown whether APAP metabolism to NAPQI is possible in the vasculature. In this study, we examine the hypothesis that APAP is metabolized by cytochrome P450 enzymes within the endothelium, which may be accelerated in critically ill patients by the presence of elevated myeloperoxidase (MPO). Exposure of human coronary artery endothelial cells (HCAECs) to APAP resulted in the formation of protein-bound APAP adducts. Proteomic analysis of HCAECs exposed to APAP showed upregulation of CYP20A1, together with proteins involved in the pentose phosphate pathway and maintaining redox homeostasis. Proteomic analyses of mesenteric arteries from rats administered intravenous APAP are consistent with a key role of the vascular wall in APAP metabolism, with similar proteomic pathway changes identified in HCAECs. These changes occurred over a short timeframe and were not seen in the corresponding proteomic analyses of liver tissue. Intracellular thiols were depleted in HCAECs upon APAP treatment, which was partially attenuated by ketoconazole, consistent with the involvement of cytochrome P450 enzymes in the metabolism of APAP to a thiol-reactive metabolite such as NAPQI. Evidence was also obtained for the metabolism of APAP to a thiol-reactive intermediate by MPO in the absence of chloride ions, consistent with NAPQI formation. Taken together, these data provide a putative mechanism to explain the presentation of hypotension in critically ill patients following IV APAP administration.
Collapse
Affiliation(s)
- Johs Dannesboe
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, Copenhagen N 2200, Denmark
| | - Joakim A Bastrup
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, Copenhagen N 2200, Denmark
| | - Kathrine Holm Nielsen
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, Copenhagen N 2200, Denmark
| | - Pelle Munck
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, Copenhagen N 2200, Denmark
| | - Morten B Thomsen
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, Copenhagen N 2200, Denmark
| | - Clare L Hawkins
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, Copenhagen N 2200, Denmark
| | - Thomas A Jepps
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, Copenhagen N 2200, Denmark.
| |
Collapse
|
3
|
Zhai X, Yang R, Chu Q, Guo Z, Hou P, Li X, Bai C, Lu Z, Qiao L, Fu Y, Niu J, Li B. AMPK-regulated glycerol excretion maintains metabolic crosstalk between reductive and energetic stress. Nat Cell Biol 2025; 27:141-153. [PMID: 39747579 DOI: 10.1038/s41556-024-01549-x] [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: 11/20/2023] [Accepted: 09/29/2024] [Indexed: 01/04/2025]
Abstract
Glucose metabolism has been studied extensively, but the role of glucose-derived excretory glycerol remains unclear. Here we show that hypoxia induces NADH accumulation to promote glycerol excretion and this pathway consumes NADH continuously, thus attenuating its accumulation and reductive stress. Aldolase B accounts for glycerol biosynthesis by forming a complex with glycerol 3-phosphate dehydrogenases GPD1 and GPD1L. Blocking GPD1, GPD1L or glycerol 3-phosphate phosphatase exacerbates reductive stress and suppresses cell proliferation under hypoxia and tumour growth in vivo. Overexpression of these enzymes increases glycerol excretion but still reduces cell viability under hypoxia and tumour proliferation due to energy stress. AMPK inactivates aldolase B to mitigate glycerol synthesis that dissipates ATP, alleviating NADH accumulation-induced energy crisis. Therefore, glycerol biosynthesis/excretion regulates the trade-off between reductive stress and energy stress. Moreover, this mode of regulation seems to be prevalent in reductive stress-driven transformations, enhancing our understanding of the metabolic complexity and guiding tumour treatment.
Collapse
Affiliation(s)
- Xuewei Zhai
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Ronghui Yang
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Qiaoyun Chu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Zihao Guo
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Pengjiao Hou
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Xuexue Li
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Changsen Bai
- Department of Cancer Cell Biology and National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Ziwen Lu
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Luxin Qiao
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yanxia Fu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Jing Niu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Binghui Li
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China.
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.
- Department of Cancer Cell Biology and National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.
| |
Collapse
|
4
|
Liu M, Yang W, Qu S, Zhao T, Jiang S, Peng S, Zhang M, Xuan J, Liu Z, Zen K. Loss of glomerular aldolase B in diabetic nephropathy promotes renal fibrosis via activating Akt/GSK/β-catenin axis. J Adv Res 2024:S2090-1232(24)00605-2. [PMID: 39725005 DOI: 10.1016/j.jare.2024.12.027] [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: 08/17/2024] [Revised: 11/13/2024] [Accepted: 12/16/2024] [Indexed: 12/28/2024] Open
Abstract
OBJECTIVE Diabetic nephropathy (DN), characterized by a complex and multifaceted pathogenesis, stands as the foremost catalyst behind end-stage renal disease (ESRD). This study aims to analyze the level and non-metabolic role of glomerular aldolase B (ALDOB) in DN progression. METHODS Glomerular proteomics and transcriptome are analyzed from 50 DN patients and 25 controls, respectively. Human kidney biopsy, cultured podocytes and mouse models are employed to study ALDOB levels and function. RESULTS ALDOB is strongly downregulated in DN-affected glomeruli, as well as in human and murine podocytes exposed to inflammatory cytokines. ALDOB reduction increases podocyte injury, while adenovirus-mediated ALDOB overexpression leads to substantial alleviation of renal injuries in a diabetic mouse model. Mechanistically, ALDOB reduction triggers the Akt/GSK/β-catenin signaling cascade within podocytes. CONCLUSION Our findings reveal a novel non-metabolic role of glomerular ALDOB in protecting against podocyte injury and renal fibrosis.
Collapse
Affiliation(s)
- Minghui Liu
- School of Pharmaceutical Science, Nanjing Tech University, 30 South Puzhu Road, Nanjing, Jiangsu 211816, China; State Key Laboratory of Pharmaceutical Biotechnology, Department of Gastroenterology, Drum Tower Hospital, Nanjing University, School of Life Sciences, Nanjing, Jiangsu 210093, China
| | - Wenwen Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Gastroenterology, Drum Tower Hospital, Nanjing University, School of Life Sciences, Nanjing, Jiangsu 210093, China
| | - Shuang Qu
- Geriatric Hospital of Nanjing Medical University, Nanjing, Jiangsu 210024, China
| | - Tingting Zhao
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu 210002, China
| | - Song Jiang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu 210002, China
| | - Suming Peng
- School of Life Science and Technology, Chinese Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Mingchao Zhang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu 210002, China
| | - Ji Xuan
- Department of Gastroenterology, Jinling Hospital, Nanjing University, School of Medicine, Nanjing, Jiangsu 210002, China.
| | - Zhihong Liu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu 210002, China.
| | - Ke Zen
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Gastroenterology, Drum Tower Hospital, Nanjing University, School of Life Sciences, Nanjing, Jiangsu 210093, China.
| |
Collapse
|
5
|
Nelson AB, Reese LE, Rono E, Queathem ED, Qiu Y, McCluskey BM, Crampton A, Conniff E, Cummins K, Boytim E, Dansou S, Hwang J, Safo S, Puchalska P, Wood DK, Schwertfeger KL, Crawford PA. Deciphering Colorectal Cancer-Hepatocyte Interactions: A Multiomic Platform for Interrogation of Metabolic Crosstalk in the Liver-Tumor Microenvironment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.06.627264. [PMID: 39713297 PMCID: PMC11661097 DOI: 10.1101/2024.12.06.627264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Metabolic reprogramming is a hallmark of cancer, enabling tumor cells to adapt to and exploit their microenvironment for sustained growth. The liver is a common site of metastasis, but the interactions between tumor cells and hepatocytes remain poorly understood. In the context of liver metastasis, these interactions play a crucial role in promoting tumor survival and progression. This study leverages multiomics coverage of the microenvironment via liquid chromatography and high-resolution, high-mass accuracy mass spectrometry-based untargeted metabolomics, 13C-stable isotope tracing, and RNA sequencing to uncover the metabolic impact of co-localized primary hepatocytes and a colon adenocarcinoma cell line, SW480, using a 2D co-culture model. Metabolic profiling revealed disrupted Warburg metabolism with an 80% decrease in glucose consumption and 94% decrease in lactate production by hepatocyte-SW480 co-cultures relative to SW480 control cultures. Decreased glucose consumption was coupled with alterations in glutamine and ketone body metabolism, suggesting a possible fuel switch upon co-culturing. Further, integrated multiomic analysis indicates that disruptions in metabolic pathways, including nucleoside biosynthesis, amino acids, and TCA cycle, correlate with altered SW480 transcriptional profiles and highlight the importance of redox homeostasis in tumor adaptation. Finally, these findings were replicated in 3-dimensional microtissue organoids. Taken together, these studies support a bioinformatic approach to study metabolic crosstalk and discovery of potential therapeutic targets in preclinical models of the tumor microenvironment.
Collapse
Affiliation(s)
- Alisa B. Nelson
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN USA
| | - Lyndsay E. Reese
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN USA
| | - Elizabeth Rono
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN USA
| | - Eric D. Queathem
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN USA
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN USA
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Yinjie Qiu
- Minnesota Supercomputing Institute, Minneapolis, MN USA
| | | | - Alexandra Crampton
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN USA
| | - Eric Conniff
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN USA
| | - Katherine Cummins
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN USA
| | - Ella Boytim
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN USA
| | - Senali Dansou
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN USA
| | - Justin Hwang
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN USA
| | - Sandra Safo
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, MN, USA
| | - Patrycja Puchalska
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN USA
| | - David K. Wood
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN USA
| | - Kathryn L. Schwertfeger
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN USA
| | - Peter A. Crawford
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN USA
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN USA
| |
Collapse
|
6
|
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) 2024:e2406562. [PMID: 39665272 DOI: 10.1002/advs.202406562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [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 Surgery, The Second Hospital of Dalian Medical University, Dalian, 116023, China
- Department of Clinical Biochemistry, College of Laoratory Medicine, Dalian Medical University, Dalian, 116044, China
| | - Yina Gao
- Department of General Surgery, The Second Hospital of Dalian Medical University, Dalian, 116023, China
- Department of Clinical Biochemistry, College of Laoratory Medicine, Dalian Medical University, Dalian, 116044, China
| | - Yuanyuan Qian
- Department of Clinical Biochemistry, College of Laoratory Medicine, Dalian Medical University, Dalian, 116044, China
| | - Bowen Wei
- Department of Clinical Biochemistry, College of Laoratory Medicine, Dalian Medical University, Dalian, 116044, China
| | - Kexin Jiang
- Department of Clinical Biochemistry, College of Laoratory Medicine, Dalian Medical University, Dalian, 116044, China
| | - Zhiwei Sun
- Department of General Surgery, The Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Feifan Zhang
- Department of General Surgery, The Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Mingming Yang
- Department of Clinical Biochemistry, College of Laoratory Medicine, Dalian Medical University, Dalian, 116044, China
| | - Salem Baldi
- Department of Clinical Biochemistry, College of Laoratory Medicine, Dalian Medical University, Dalian, 116044, China
| | - Xiaoqi Yu
- Department of Clinical Biochemistry, College of Laoratory Medicine, Dalian Medical University, Dalian, 116044, China
| | - Yunfei Zuo
- Department of Clinical Biochemistry, College of Laoratory Medicine, Dalian Medical University, Dalian, 116044, China
| | - Shuangyi Ren
- Department of General Surgery, The Second Hospital of Dalian Medical University, Dalian, 116023, China
| |
Collapse
|
7
|
Leven AS, Wagner N, Nienaber S, Messiha D, Tasdogan A, Ugurel S. Changes in tumor and cardiac metabolism upon immune checkpoint. Basic Res Cardiol 2024:10.1007/s00395-024-01092-8. [PMID: 39658699 DOI: 10.1007/s00395-024-01092-8] [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: 07/01/2024] [Revised: 11/06/2024] [Accepted: 11/25/2024] [Indexed: 12/12/2024]
Abstract
Cardiovascular disease and cancer are the leading causes of death in the Western world. The associated risk factors are increased by smoking, hypertension, diabetes, sedentary lifestyle, aging, unbalanced diet, and alcohol consumption. Therefore, the study of cellular metabolism has become of increasing importance, with current research focusing on the alterations and adjustments of the metabolism of cancer patients. This may also affect the efficacy and tolerability of anti-cancer therapies such as immune-checkpoint inhibition (ICI). This review will focus on metabolic adaptations and their consequences for various cell types, including cancer cells, cardiac myocytes, and immune cells. Focusing on ICI, we illustrate how anti-cancer therapies interact with metabolism. In addition to the desired tumor response, we highlight that ICI can also lead to a variety of side effects that may impact metabolism or vice versa. With regard to the cardiovascular system, ICI-induced cardiotoxicity is increasingly recognized as one of the most life-threatening adverse events with a mortality of up to 50%. As such, significant efforts are being made to assess the specific interactions and associated metabolic changes associated with ICIs to improve both efficacy and management of side effects.
Collapse
Affiliation(s)
- Anna-Sophia Leven
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, University Duisburg-Essen, Essen, Germany.
| | - Natalie Wagner
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Stephan Nienaber
- Clinic III for Internal Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Daniel Messiha
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Centre, University of Duisburg-Essen, Essen, Germany
| | - Alpaslan Tasdogan
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany
- National Center for Tumor Diseases (NCT)-West, Campus Essen, and Research Alliance Ruhr, Research Center One Health, University Duisburg-Essen, Essen, Germany
| | - Selma Ugurel
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany
- National Center for Tumor Diseases (NCT)-West, Campus Essen, and Research Alliance Ruhr, Research Center One Health, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
8
|
Wang C, Wang L, Zhao Q, Ma J, Li Y, Kuang J, Yang X, Bi H, Lu A, Cheung KCP, Melino G, Jia W. Exploring fructose metabolism as a potential therapeutic approach for pancreatic cancer. Cell Death Differ 2024; 31:1625-1635. [PMID: 39406919 PMCID: PMC11618635 DOI: 10.1038/s41418-024-01394-3] [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: 06/08/2024] [Revised: 09/20/2024] [Accepted: 09/27/2024] [Indexed: 12/06/2024] Open
Abstract
Excessive fructose intake has been associated with the development and progression of pancreatic cancer. This study aimed to elucidate the relationship between fructose utilization and pancreatic cancer progression. Our findings revealed that pancreatic cancer cells have a high capacity to utilize fructose and are capable of converting glucose to fructose via the AKR1B1-mediated polyol pathway, in addition to uptake via the fructose transporter GLUT5. Fructose metabolism exacerbates pancreatic cancer proliferation by enhancing glycolysis and accelerating the production of key metabolites that regulate angiogenesis. However, pharmacological blockade of fructose metabolism has been shown to slow pancreatic cancer progression and synergistically enhance anti-tumor capabilities when combined with anti-angiogenic agents. Overall, targeting fructose metabolism may prove to be a promising therapeutic approach in the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Chengqiang Wang
- Chinese Medicine Phenome Research Centre, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Lu Wang
- Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong, China
| | - Qing Zhao
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Jiao Ma
- Chinese Medicine Phenome Research Centre, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Yitao Li
- Chinese Medicine Phenome Research Centre, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Junliang Kuang
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Xintong Yang
- Chinese Medicine Phenome Research Centre, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Huichang Bi
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Aiping Lu
- Chinese Medicine Phenome Research Centre, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Kenneth C P Cheung
- Chinese Medicine Phenome Research Centre, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
| | - Gerry Melino
- Department of Experimental Medicine, University of Rome "Tor Vergata", 00133, Rome, Italy.
| | - Wei Jia
- Chinese Medicine Phenome Research Centre, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
- Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong, China.
| |
Collapse
|
9
|
Yufa M, Dongmei C, Wei L, Shuangxing L, Li S, Xingchao G. Peripheral serum iTRAQ-based proteomic characteristics of carbon tetrachloride-induced acute liver injury in Macaca fascicularis. Toxicol Rep 2024; 13:101689. [PMID: 39184831 PMCID: PMC11342196 DOI: 10.1016/j.toxrep.2024.101689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 05/25/2024] [Accepted: 07/06/2024] [Indexed: 08/27/2024] Open
Abstract
Carbon tetrachloride (CCl4) is a potent chemical compound that can induce liver cells necrosis. The purpose of this study was to evaluate the hepatic toxicity of CCl4 exposure in Macaca fascicularis to explore the liver toxicity mechanism using a proteomic approach. One animal (no.F6) was intoxicated by oral gavage with 15 % CCl4 solution (10 mL/kg, dissolved in edible peanut oil), and was sacrificed at 48 h after CCl4 administration. Another blank control animal (no.F4) was sacrificed at the same time. The liver cells of the blank control animal showed normal hepatocyte morphology. However, the hepatocytes at 48 h time point after CCl4 administration showed necrosis and vacuolation histopathologically. The animal No.F7∼F12 and no.M7∼M12 were administrated by gavage with 15 % CCl4 solution (10 mL/kg, dissolved in edible peanut oil). Blood samples were collected before gavage administration, and served as the 0 h blank control samples. Then, blood samples were collected at 2 h, 48 h, 72 h and 168 h after CCl4 exposure, and served as the test samples. Routine biochemistry and immunical parameters were performed using biochemistry analyzer for all serum. Then the serum from male and female animals at 0 h, 2 h, 48 h, and 72 h was mixed, respectively. The peripheral serum proteins at 0 h, 2 h, 48 h, and 72 h were extracted, then the proteins were enzymatically hydrolyzed and the peptides were isotopic labeled by isobaric tags for relative and absolute quantification (iTRAQ). Finally, the UniProt Protein Sequence Library of Macaca fascicularis was queried to identify and compare the differential proteins between different time points. The results showed that, as traditional biomarkers of liver injury, alanine aminotransferases (ALT) and aspartate aminotransferases (AST) showed a typical time-effect curve. Compared with 0 h, there were totally 55, 323, and 158 differential proteins (P value <0.05, Ratio fold >1.5, FDR<0.05) at 2 h, 48 h and 72 h, respectively. GO enrichment analysis of differentially expressed proteins only at 48 h involved 3 cellular components (P adjust value <0.05), and differential proteins at other time points had no significant enrichment. Furthermore, KEGG enrichment analysis showed that the toxicity effect of CCl4 at different time points after administration was mediated through 22 pathways such as biosynthesis of antibiotics, carbon metabolism, biosynthesis of amino acids, peroxisome, cysteine and methionine metabolism, arginine biosynthesis, and complement and coagulation cascades (P adjust value <0.05). Among them, the counts of signaling pathway involved biosynthesis of antibiotics, carbon metabolism and biosynthesis of amino acids were more than 10 and the three pathways may play a greater role in toxicity progress after administration of CCl4. PPI network analysis showed that there were 3, 52, and 13 nodes in the interaction of differential proteins at 2 h, 48 h, and 72 h, respectively. In conclusion, many differential proteins in peripheral blood were detected after CCl4 administration, and the GO and KEGG enrichment analysis showed the toxicological mechanisms of CCl4-induced liver injury and potential protection reaction mechanism for CCl4 detoxication may be related with multi biological processes, signaling pathway and targets.
Collapse
Affiliation(s)
- Miao Yufa
- National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, Beijing Key Laboratory for Safety Evaluation of Drugs, Beijing 100176, China
| | - Chen Dongmei
- Beijing Red Cross Blood Center, Beijing 100088, China
| | - Li Wei
- National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, Beijing Key Laboratory for Safety Evaluation of Drugs, Beijing 100176, China
| | - Li Shuangxing
- National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, Beijing Key Laboratory for Safety Evaluation of Drugs, Beijing 100176, China
| | - Sun Li
- National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, Beijing Key Laboratory for Safety Evaluation of Drugs, Beijing 100176, China
| | - Geng Xingchao
- National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, Beijing Key Laboratory for Safety Evaluation of Drugs, Beijing 100176, China
| |
Collapse
|
10
|
Fowle-Grider R, Rowles JL, Shen I, Wang Y, Schwaiger-Haber M, Dunham AJ, Jayachandran K, Inkman M, Zahner M, Naser FJ, Jackstadt MM, Spalding JL, Chiang S, McCommis KS, Dolle RE, Kramer ET, Zimmerman SM, Souroullas GP, Finck BN, Shriver LP, Kaufman CK, Schwarz JK, Zhang J, Patti GJ. Dietary fructose enhances tumour growth indirectly via interorgan lipid transfer. Nature 2024; 636:737-744. [PMID: 39633044 DOI: 10.1038/s41586-024-08258-3] [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: 06/18/2020] [Accepted: 10/21/2024] [Indexed: 12/07/2024]
Abstract
Fructose consumption has increased considerably over the past five decades, largely due to the widespread use of high-fructose corn syrup as a sweetener1. It has been proposed that fructose promotes the growth of some tumours directly by serving as a fuel2,3. Here we show that fructose supplementation enhances tumour growth in animal models of melanoma, breast cancer and cervical cancer without causing weight gain or insulin resistance. The cancer cells themselves were unable to use fructose readily as a nutrient because they did not express ketohexokinase-C (KHK-C). Primary hepatocytes did express KHK-C, resulting in fructolysis and the excretion of a variety of lipid species, including lysophosphatidylcholines (LPCs). In co-culture experiments, hepatocyte-derived LPCs were consumed by cancer cells and used to generate phosphatidylcholines, the major phospholipid of cell membranes. In vivo, supplementation with high-fructose corn syrup increased several LPC species by more than sevenfold in the serum. Administration of LPCs to mice was sufficient to increase tumour growth. Pharmacological inhibition of ketohexokinase had no direct effect on cancer cells, but it decreased circulating LPC levels and prevented fructose-mediated tumour growth in vivo. These findings reveal that fructose supplementation increases circulating nutrients such as LPCs, which can enhance tumour growth through a cell non-autonomous mechanism.
Collapse
Affiliation(s)
- Ronald Fowle-Grider
- Department of Chemistry, Washington University, St Louis, MO, USA
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St Louis, MO, USA
- Center for Mass Spectrometry and Metabolic Tracing, Washington University, St Louis, MO, USA
| | - Joe L Rowles
- Department of Chemistry, Washington University, St Louis, MO, USA
- Center for Mass Spectrometry and Metabolic Tracing, Washington University, St Louis, MO, USA
| | - Isabel Shen
- Department of Chemistry, Washington University, St Louis, MO, USA
- Center for Mass Spectrometry and Metabolic Tracing, Washington University, St Louis, MO, USA
| | - Yahui Wang
- Department of Chemistry, Washington University, St Louis, MO, USA
- Center for Mass Spectrometry and Metabolic Tracing, Washington University, St Louis, MO, USA
| | - Michaela Schwaiger-Haber
- Department of Chemistry, Washington University, St Louis, MO, USA
- Center for Mass Spectrometry and Metabolic Tracing, Washington University, St Louis, MO, USA
| | - Alden J Dunham
- Department of Chemistry, Washington University, St Louis, MO, USA
- Center for Mass Spectrometry and Metabolic Tracing, Washington University, St Louis, MO, USA
| | - Kay Jayachandran
- Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO, USA
| | - Matthew Inkman
- Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO, USA
| | - Michael Zahner
- Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO, USA
- Division of Medical Oncology, Washington University School of Medicine, St Louis, MO, USA
| | - Fuad J Naser
- Department of Chemistry, Washington University, St Louis, MO, USA
- Center for Mass Spectrometry and Metabolic Tracing, Washington University, St Louis, MO, USA
| | - Madelyn M Jackstadt
- Department of Chemistry, Washington University, St Louis, MO, USA
- Center for Mass Spectrometry and Metabolic Tracing, Washington University, St Louis, MO, USA
| | - Jonathan L Spalding
- Department of Chemistry, Washington University, St Louis, MO, USA
- Center for Mass Spectrometry and Metabolic Tracing, Washington University, St Louis, MO, USA
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO, USA
| | - Sarah Chiang
- Department of Chemistry, Washington University, St Louis, MO, USA
- Center for Mass Spectrometry and Metabolic Tracing, Washington University, St Louis, MO, USA
| | - Kyle S McCommis
- Department of Biochemistry & Molecular Biology, Saint Louis University School of Medicine, St Louis, MO, USA
| | - Roland E Dolle
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St Louis, MO, USA
| | - Eva T Kramer
- Division of Medical Oncology, Washington University School of Medicine, St Louis, MO, USA
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO, USA
| | - Sarah M Zimmerman
- Division of Medical Oncology, Washington University School of Medicine, St Louis, MO, USA
| | - George P Souroullas
- Division of Medical Oncology, Washington University School of Medicine, St Louis, MO, USA
- Siteman Cancer Center, Washington University School of Medicine, St Louis, MO, USA
| | - Brian N Finck
- Division of Geriatrics and Nutritional Sciences, Washington University School of Medicine, St Louis, MO, USA
| | - Leah P Shriver
- Department of Chemistry, Washington University, St Louis, MO, USA
- Center for Mass Spectrometry and Metabolic Tracing, Washington University, St Louis, MO, USA
| | - Charles K Kaufman
- Division of Medical Oncology, Washington University School of Medicine, St Louis, MO, USA
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO, USA
| | - Julie K Schwarz
- Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO, USA
- Siteman Cancer Center, Washington University School of Medicine, St Louis, MO, USA
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO, USA
| | - Jin Zhang
- Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO, USA
- Siteman Cancer Center, Washington University School of Medicine, St Louis, MO, USA
- Institute for Informatics, Data Science & Biostatistics (I2DB), Washington University School of Medicine, St Louis, MO, USA
| | - Gary J Patti
- Department of Chemistry, Washington University, St Louis, MO, USA.
- Center for Mass Spectrometry and Metabolic Tracing, Washington University, St Louis, MO, USA.
- Siteman Cancer Center, Washington University School of Medicine, St Louis, MO, USA.
- Center for Human Nutrition, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA.
| |
Collapse
|
11
|
Wan Y, Ge RL, Cao Y, Luo L, Ji W. Chronic Hypobaric Hypoxia Stimulates Differential Expression of Cognitive Proteins in Hippocampal Tissue. High Alt Med Biol 2024. [PMID: 39602167 DOI: 10.1089/ham.2024.0105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2024] Open
Abstract
Objective: We aimed to determine changes in cognitive function resulting from chronic hypobaric hypoxia through proteomic analysis of hippocampal tissue. We screened cognition-related proteins to provide ideas and directions that could help prevent and treat hypoxia-associated cognitive impairment. Methods: We analyzed hippocampal tissues from mice exposed to high altitudes and control mice using 4 D label-free quantitative proteomics. The data were analyzed by protein quantitative analysis, functional annotation, differential protein screening, clustering analyses, and functional classification and enrichment. Differential protein expression was investigated using targeted quantitative omics based on parallel response monitoring. Results: We identified and quantified 20 target proteins in 12 samples, of which 18 were significant validated proteins that were or might be related to cognitive functions. Signaling pathways that were significantly enriched in differentially expressed proteins were pyrimidine metabolism, 5'-Adenosine Triphosphate-activated protein kinase signaling, phospholipase D signaling, purine metabolism, inflammatory mediator regulation of transient receptor potential channels, hedgehog signaling pathways, dilated cardiomyopathy, platelet activation, insulin resistance, mRNA surveillance pathways, drug metabolism-other enzymes, and drug metabolism-cytochrome P450. Conclusion: Chronic hypoxia alters protein expression in murine hippocampal tissues. Eighteen differentially expressed cognition-related proteins might be related to cognitive impairment in mice exposed to chronic high-altitude hypoxia.
Collapse
Affiliation(s)
- Yaqi Wan
- Department of Neurology, Qinghai Provincial People's Hospital, Xining, China
| | - Ri-Li Ge
- Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province, Research Center for High Altitude Medicine, Qinghai University, Xining, China
| | - Yaxin Cao
- Tang Du hospital of Air Force Military Medical University, Xi'an, China
| | - Lan Luo
- Department of Neurology, Qinghai Provincial People's Hospital, Xining, China
| | - Weizhong Ji
- Department of Neurology, Qinghai Provincial People's Hospital, Xining, China
| |
Collapse
|
12
|
Miller PG, Huang E, Fisher R, Shuler ML. Development of a Microphysiological System to Model Human Cancer Metastasis From the Colon to the Liver. Biotechnol Bioeng 2024. [PMID: 39587032 DOI: 10.1002/bit.28890] [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: 07/28/2024] [Revised: 11/05/2024] [Accepted: 11/10/2024] [Indexed: 11/27/2024]
Abstract
We describe a novel device to mimic the metastasis of cancer cells from the colon into the liver in a human model. The colon mimic is connected to the liver model by a gravity-driven recirculating unidirectional flow of a blood surrogate and can mimic the five steps of the metastatic cascade: invasion in the colon, intravasation into the bloodstream, systemic transportation, extravasation into the liver, and colonization in the liver. The colon mimic uses established normal colon epithelial organoid cells (NL) and human umbilical vein endothelial cells (HUVEC) plated on opposite sides of a membrane. To better mimic the colon structure the NL side of the membrane is exposed to air to establish an air-liquid interface. The liver mimic consists of human liver sinusoidal endothelial cells (HHSEC) and epithelial hepatic cells (HepG2 C3A) plated in Matrigel on opposite sides of a membrane. Labeled colorectal cancer cells/clusters (CA) from organoids are introduced into an established normal colon epithelial cell (NL) layer from the same patient before assembly of the system or alternatively NL organoids and fluorescently labeled CA organoids from the same patient were prepared as a ratio of 10:1 NL:CA and established together before assembly of the system. Cell viability is greater than 85% in this system. We demonstrate that over 5 days of operation that the five steps of the metastatic cascade are replicated. This novel device allows an in vitro estimate of metastatic capability (as measured by using percentages of the labeled areas per device through ImageJ) in response to selected variables. In this study, the metastatic capability depends on the source of cancer cells (e.g., the patient), the clumping of cancer cells, glucose concentration, and oxygen levels (hypoxia). For the first time, this new in vitro system mimics all five steps of the metastatic cascade in a single device and provides a new device to probe and observe the process of metastasis in a human-based model in only 5 days. The rapid observation is due to the use of a high concentration of cancer cells in the colon (e.g. 10%) and the absence of the immune system. Our device makes it possible to probe aspects of each step of metastasis and interactions between steps.
Collapse
Affiliation(s)
- Paula G Miller
- Department of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Emina Huang
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Robert Fisher
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Michael L Shuler
- Department of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| |
Collapse
|
13
|
Yan H, Wang Z, Teng D, Chen X, Zhu Z, Chen H, Wang W, Wei Z, Wu Z, Chai Q, Zhang F, Wang Y, Shu K, Li S, Shi G, Zhu M, Piao HL, Shen X, Bu P. Hexokinase 2 senses fructose in tumor-associated macrophages to promote colorectal cancer growth. Cell Metab 2024; 36:2449-2467.e6. [PMID: 39471815 DOI: 10.1016/j.cmet.2024.10.002] [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: 11/16/2023] [Revised: 05/14/2024] [Accepted: 10/01/2024] [Indexed: 11/01/2024]
Abstract
Fructose is associated with colorectal cancer tumorigenesis and metastasis through ketohexokinase-mediated metabolism in the colorectal epithelium, yet its role in the tumor immune microenvironment remains largely unknown. Here, we show that a modest amount of fructose, without affecting obesity and associated complications, promotes colorectal cancer tumorigenesis and growth by suppressing the polarization of M1-like macrophages. Fructose inhibits M1-like macrophage polarization independently of fructose-mediated metabolism. Instead, it serves as a signal molecule to promote the interaction between hexokinase 2 and inositol 1,4,5-trisphophate receptor type 3, the predominant Ca2+ channel on the endoplasmic reticulum. The interaction reduces Ca2+ levels in cytosol and mitochondria, thereby suppressing the activation of mitogen-activated protein kinase (MAPK) and signal transducer and activator of transcription 1 (STAT1) as well as NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome activation. Consequently, this impedes M1-like macrophage polarization. Our study highlights the critical role of fructose as a signaling molecule that impairs the polarization of M1-like macrophages for tumor growth.
Collapse
Affiliation(s)
- Huiwen Yan
- Key Laboratory of Epigenetic Regulation and Intervention, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhi Wang
- Key Laboratory of Epigenetic Regulation and Intervention, Chinese Academy of Sciences, Beijing 100101, China; Department of Ophthalmology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Da Teng
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, China
| | - Xiaodong Chen
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zijing Zhu
- Key Laboratory of Epigenetic Regulation and Intervention, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Huan Chen
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Wen Wang
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Ziyuan Wei
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhenzhen Wu
- Key Laboratory of Epigenetic Regulation and Intervention, Chinese Academy of Sciences, Beijing 100101, China
| | - Qian Chai
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Fei Zhang
- Key Laboratory of Epigenetic Regulation and Intervention, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Youwang Wang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Kaile Shu
- Key Laboratory of Epigenetic Regulation and Intervention, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shaotang Li
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Guizhi Shi
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mingzhao Zhu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Hai-Long Piao
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.
| | - Xian Shen
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Pengcheng Bu
- Key Laboratory of Epigenetic Regulation and Intervention, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
14
|
Schwärzler J, Mayr L, Grabherr F, Tilg H, Adolph TE. Epithelial metabolism as a rheostat for intestinal inflammation and malignancy. Trends Cell Biol 2024; 34:913-927. [PMID: 38341347 DOI: 10.1016/j.tcb.2024.01.004] [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: 11/20/2023] [Revised: 01/06/2024] [Accepted: 01/12/2024] [Indexed: 02/12/2024]
Abstract
The gut epithelium protects the host from a potentially hostile environment while allowing nutrient uptake that is vital for the organism. To maintain this delicate task, the gut epithelium has evolved multilayered cellular functions ranging from mucus production to hormone release and orchestration of mucosal immunity. Here, we review the execution of intestinal epithelial metabolism in health and illustrate how perturbation of epithelial metabolism affects experimental gut inflammation and tumorigenesis. We also discuss the impact of environmental factors and host-microbe interactions on epithelial metabolism in the context of inflammatory bowel disease and colorectal cancer. Insights into epithelial metabolism hold promise to unravel mechanisms of organismal health that may be therapeutically exploited in humans in the future.
Collapse
Affiliation(s)
- Julian Schwärzler
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, and Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| | - Lisa Mayr
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Felix Grabherr
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Timon E Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, and Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
15
|
Ma Z, Wang Y, Shen J, Yan W, Cao C, Feng M, Zhu J, Pang W. Overexpression of ALDOC Promotes Porcine Intramuscular and Intermuscular Fat Deposition by Activating the AKT-mTORC1 Signaling Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:23893-23907. [PMID: 39428631 DOI: 10.1021/acs.jafc.4c07781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
Skeletal muscle fat accumulation, a common complication of obesity, has garnered increasing attention. For livestock animals, intramuscular fat content is a key determinant of meat quality and directly influences consumer preferences for different meat products. However, genes effectively regulating intramuscular and intermuscular fat (IIMF) deposition remain largely unexplored. To address this gap, we employed transcriptome sequencing of muscle from pigs with high and low IIMF contents, uncovering Aldolase C (ALDOC) as a novel key gene controlling IIMF. ALDOC overexpression significantly enhanced the IIMF content both in vivo and in vitro. Mechanistic investigations revealed that ALDOC activates the AKT-mTORC1 axis to promote lipid accumulation in myotubes and intramuscular adipocytes. Notably, the mTORC1 inhibitor rapamycin abrogated ALDOC's proadipogenic effect. Our findings establish ALDOC as a novel key gene regulating IIMF deposition and identify the ALDOC-AKT-mTORC1 signaling pathway as a promising therapeutic target for treating skeletal muscle fat infiltration and improving pork quality.
Collapse
Affiliation(s)
- Zeqiang Ma
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Yingqian Wang
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Junnan Shen
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Wenyong Yan
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Chaoyue Cao
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Ming Feng
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Jiahua Zhu
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Weijun Pang
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China
| |
Collapse
|
16
|
Ni X, Wei Y, Li X, Pan J, Fang B, Zhang T, Lu Y, Ye D, Zhu Y. From biology to the clinic - exploring liver metastasis in prostate cancer. Nat Rev Urol 2024; 21:593-614. [PMID: 38671281 DOI: 10.1038/s41585-024-00875-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2024] [Indexed: 04/28/2024]
Abstract
Liver metastases from prostate cancer are associated with an aggressive disease course and poor prognosis. Results from autopsy studies indicate a liver metastasis prevalence of up to 25% in patients with advanced prostate cancer. Population data estimate that ~3-10% of patients with metastatic castration-resistant prostate cancer harbour liver metastases at the baseline, rising to 20-30% in post-treatment cohorts, suggesting that selective pressure imposed by novel therapies might promote metastatic spread to the liver. Liver metastases are associated with more aggressive tumour biology than lung metastases. Molecular profiling of liver lesions showed an enrichment of low androgen receptor, neuroendocrine phenotypes and high genomic instability. Despite advancements in molecular imaging modalities such as prostate-specific membrane antigen PET-CT, and liquid biopsy markers such as circulating tumour DNA, early detection of liver metastases from prostate cancer remains challenging, as both approaches are hampered by false positive and false negative results, impeding the accurate identification of early liver lesions. Current therapeutic strategies showed limited efficacy in this patient population. Emerging targeted radionuclide therapies, metastasis-directed therapy, and novel systemic agents have shown preliminary activity against liver metastases, but require further validation. Treatment with various novel prostate cancer therapies might lead to an increase in the prevalence of liver metastasis, underscoring the urgent need for coordinated efforts across preclinical and clinical researchers to improve characterization, monitoring, and management of liver metastases from prostate cancer. Elucidating molecular drivers of liver tropism and interactions with the liver microenvironment might ultimately help to identify actionable targets to enhance survival in this high-risk patient group.
Collapse
Affiliation(s)
- Xudong Ni
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Yu Wei
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Xiaomeng Li
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Jian Pan
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Bangwei Fang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Tingwei Zhang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Ying Lu
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Yao Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
- Shanghai Genitourinary Cancer Institute, Shanghai, China.
| |
Collapse
|
17
|
Huang XH, Huang CY. Fructose shields human colorectal cancer cells from hypoxia-induced necroptosis. NPJ Sci Food 2024; 8:71. [PMID: 39353947 PMCID: PMC11445490 DOI: 10.1038/s41538-024-00318-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 09/24/2024] [Indexed: 10/03/2024] Open
Abstract
Recent studies have shown that high dietary fructose intake enhances intestinal tumor growth in mice. Our previous work indicated that glucose enables hypoxic colorectal cancer (CRC) cells to resist receptor-interacting protein (RIP)-dependent necroptosis. Despite having the same chemical formula, glucose and fructose are absorbed through different transporters yet both can enter the glycolytic metabolic pathway. The excessive intake of dietary fructose, leading to its overflow into the colon, allows colonic cells to absorb fructose apically. This study explores the mechanisms behind apical fructose-mediated death resistance in CRC cells under hypoxic stress. Utilizing three CRC cell lines (Caco-2, HT29, and T84) under normoxic and hypoxic conditions with varying fructose concentrations, we assessed lactate dehydrogenase (LDH) activity, RIP1/3 complex formation (a necroptosis marker), and cell integrity. We investigated the role of fructose in glycolytic-mediated death resistance using glycolytic inhibitors iodoacetate (IA, a glycolytic inhibitor to glyceraldehyde 3-phosphate dehydrogenase), and UK5099 (UK, an inhibitor to mitochondrial pyruvate carrier). Our findings reveal that apical fructose prevents the hypoxia-induced RIP-dependent necroptosis in Caco-2 and HT29 cells. Fructose exposure under hypoxia also preserved epithelial integrity. IA, but not UK, blocked fructose-mediated glycolytic metabolite production and necrosis, indicating that anaerobic glycolytic metabolites facilitate death resistance. Notably, fructose treatment upregulated pyruvate kinase (PK)-M1 mRNA in hypoxic Caco-2 and HT29 cells, while PKM2 upregulation was exclusive to HT29 cells. In conclusion, apical fructose utilization through glycolysis effectively inhibits hypoxia-induced RIP-dependent necroptosis in CRC cells, shedding light on potential metabolic adaptation mechanisms in the tumor microenvironment and suggesting novel targets for therapeutic intervention.
Collapse
Affiliation(s)
- Xiang-Han Huang
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung, Taiwan
| | - Ching-Ying Huang
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung, Taiwan.
| |
Collapse
|
18
|
Jiang J, Meng X, Wang Y, Zhuang Z, Du T, Yan J. Effect of aberrant fructose metabolism following SARS-CoV-2 infection on colorectal cancer patients' poor prognosis. PLoS Comput Biol 2024; 20:e1012412. [PMID: 39331675 PMCID: PMC11463760 DOI: 10.1371/journal.pcbi.1012412] [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: 06/06/2023] [Revised: 10/09/2024] [Accepted: 08/13/2024] [Indexed: 09/29/2024] Open
Abstract
Most COVID-19 patients have a positive prognosis, but patients with additional underlying diseases are more likely to have severe illness and increased fatality rates. Numerous studies indicate that cancer patients are more prone to contract SARS-CoV-2 and develop severe COVID-19 or even dying. In the recent transcriptome investigations, it is demonstrated that the fructose metabolism is altered in patients with SARS-CoV-2 infection. However, cancer cells can use fructose as an extra source of energy for growth and metastasis. Furthermore, enhanced living conditions have resulted in a notable rise in fructose consumption in individuals' daily dietary habits. We therefore hypothesize that the poor prognosis of cancer patients caused by SARS-CoV-2 may therefore be mediated through fructose metabolism. Using CRC cases from four distinct cohorts, we built and validated a predictive model based on SARS-CoV-2 producing fructose metabolic anomalies by coupling Cox univariate regression and lasso regression feature selection algorithms to identify hallmark genes in colorectal cancer. We also developed a composite prognostic nomogram to improve clinical practice by integrating the characteristics of aberrant fructose metabolism produced by this novel coronavirus with age and tumor stage. To obtain the genes with the greatest potential prognostic values, LASSO regression analysis was performed, In the TCGA training cohort, patients were randomly separated into training and validation sets in the ratio of 4: 1, and the best risk score value for each sample was acquired by lasso regression analysis for further analysis, and the fifteen genes CLEC4A, FDFT1, CTNNB1, GPI, PMM2, PTPRD, IL7, ALDH3B1, AASS, AOC3, SEPINE1, PFKFB1, FTCD, TIMP1 and GATM were finally selected. In order to validate the model's accuracy, ROC curve analysis was performed on an external dataset, and the results indicated that the model had a high predictive power for the prognosis prediction of patients. Our study provides a theoretical foundation for the future targeted regulation of fructose metabolism in colorectal cancer patients, while simultaneously optimizing dietary guidance and therapeutic care for colorectal cancer patients in the context of the COVID-19 pandemic.
Collapse
Affiliation(s)
- Jiaxin Jiang
- Department of Biochemistry & Molecular Biology, China Medical University, Shenyang, China
| | - Xiaona Meng
- Teaching Center for Basic Medical Experiment, China Medical University, Shenyang, China
| | - Yibo Wang
- Department of Bioinformatics, China Medical University, Shenyang, China
| | - Ziqian Zhuang
- Department of Bioinformatics, China Medical University, Shenyang, China
| | - Ting Du
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Jing Yan
- Department of Biochemistry & Molecular Biology, China Medical University, Shenyang, China
| |
Collapse
|
19
|
Wu L, Dong J, Fei D, Le T, Xiao L, Liu J, Yu Z. Fructose-1, 6-Bisphosphate Aldolase B Suppresses Glycolysis and Tumor Progression of Gastric Cancer. Dig Dis Sci 2024; 69:3290-3304. [PMID: 39068380 DOI: 10.1007/s10620-024-08568-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 07/15/2024] [Indexed: 07/30/2024]
Abstract
OBJECTIVE Gastric cancer (GC) is believed to be one of the most common digestive tract malignant tumors. However, mounting evidence indicates a link between the glycolysis and tumorigenesis, including gastric cancer. METHODS Our research identified 5508 differently expressed mRNAs in gastric cancer. Then, the genes highly associated with tumorigenesis were identified through weighted correlation network analysis (WGCNA). Bioinformatics analysis observed that these hub genes were significantly linked to the regulation of cell cycle, drug metabolism, and glycolysis. Among these hub genes, there is a critical gene involved in glycolysis regulation, namely fructose-bisphosphate B (ALDOB). RESULTS Analysis based on The Cancer Genome Atlas (TCGA) and three Gene Expression Omnibus (GEO) datasets revealed that ALDOB was significantly downregulated in GC compared with normal tissues. In addition, cell viability assay confirmed that ALDOB acted as a tumor suppressor. Finally, drug sensitivity analysis revealed that ALDOB increased the sensitivity of gastric cancer cells to most antitumor drugs, especially talazoparib, XAV939, and FTI-277. Our results showed that the expression of ALDOB was significantly lower in GC tissues than in normal tissues. And ALDOB significantly inhibited proliferation and migration, delayed glycolysis in GC cells. Consequently, our study suggests that ALDOB may be a potential target for the clinical treatment of gastric cancer.
Collapse
Affiliation(s)
- Liping Wu
- The Department of Science and Education, Zhoushan Hospital, Wenzhou Medical University, Zhoushan, Zhejiang, China
| | - Jinliang Dong
- Department of General Surgery, Zhoushan Hospital, Wenzhou Medical University, No. 739 Dingshen Road, Lincheng New District, Zhoushan, Zhejiang, China
| | - Dailiang Fei
- Department of General Surgery, Zhoushan Hospital, Wenzhou Medical University, No. 739 Dingshen Road, Lincheng New District, Zhoushan, Zhejiang, China
| | - Ting Le
- The Laboratory of Cytobiology and Molecular Biology, Zhoushan Hospital, Wenzhou Medical University, No. 739 Dingshen Road, Lincheng New District, Zhoushan, Zhejiang, China
| | - Liang Xiao
- The Department of Surgery and Oncology, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Jia Liu
- School of Agriculture, Sun Yat-Sen University, No. 66 Gongchang Road, Guangming District, Shenzhen, Guangdong, China
- Shenzhen Zhongjia Bio-Medical Technology Co., Ltd, No. 66 Gongchang Road, Guangming District, Shenzhen, Guangdong, China
| | - Ze Yu
- Department of General Surgery, Zhoushan Hospital, Wenzhou Medical University, No. 739 Dingshen Road, Lincheng New District, Zhoushan, Zhejiang, China.
- The Laboratory of Cytobiology and Molecular Biology, Zhoushan Hospital, Wenzhou Medical University, No. 739 Dingshen Road, Lincheng New District, Zhoushan, Zhejiang, China.
- The Second Affiliated Hospital of Zhejiang University, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
20
|
Gonzalez-Gutierrez L, Motiño O, Barriuso D, de la Puente-Aldea J, Alvarez-Frutos L, Kroemer G, Palacios-Ramirez R, Senovilla L. Obesity-Associated Colorectal Cancer. Int J Mol Sci 2024; 25:8836. [PMID: 39201522 PMCID: PMC11354800 DOI: 10.3390/ijms25168836] [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: 07/12/2024] [Revised: 08/02/2024] [Accepted: 08/07/2024] [Indexed: 09/02/2024] Open
Abstract
Colorectal cancer (CRC) affects approximately 2 million people worldwide. Obesity is the major risk factor for CRC. In addition, obesity contributes to a chronic inflammatory stage that enhances tumor progression through the secretion of proinflammatory cytokines. In addition to an increased inflammatory response, obesity-associated cancer presents accrued molecular factors related to cancer characteristics, such as genome instability, sustained cell proliferation, telomere dysfunctions, angiogenesis, and microbial alteration, among others. Despite the evidence accumulated over the last few years, the treatments for obesity-associated CRC do not differ from the CRC treatments in normal-weight individuals. In this review, we summarize the current knowledge on obesity-associated cancer, including its epidemiology, risk factors, molecular factors, and current treatments. Finally, we enumerate possible new therapeutic targets that may improve the conditions of obese CRC patients. Obesity is key for the development of CRC, and treatments resulting in the reversal of obesity should be considered as a strategy for improving antineoplastic CRC therapies.
Collapse
Affiliation(s)
- Lucia Gonzalez-Gutierrez
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid–CSIC, 47003 Valladolid, Spain; (L.G.-G.); (O.M.); (D.B.); (J.d.l.P.-A.); (L.A.-F.); (R.P.-R.)
| | - Omar Motiño
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid–CSIC, 47003 Valladolid, Spain; (L.G.-G.); (O.M.); (D.B.); (J.d.l.P.-A.); (L.A.-F.); (R.P.-R.)
| | - Daniel Barriuso
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid–CSIC, 47003 Valladolid, Spain; (L.G.-G.); (O.M.); (D.B.); (J.d.l.P.-A.); (L.A.-F.); (R.P.-R.)
| | - Juan de la Puente-Aldea
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid–CSIC, 47003 Valladolid, Spain; (L.G.-G.); (O.M.); (D.B.); (J.d.l.P.-A.); (L.A.-F.); (R.P.-R.)
| | - Lucia Alvarez-Frutos
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid–CSIC, 47003 Valladolid, Spain; (L.G.-G.); (O.M.); (D.B.); (J.d.l.P.-A.); (L.A.-F.); (R.P.-R.)
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, 75006 Paris, France;
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, 94805 Villejuif, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, 75015 Paris, France
| | - Roberto Palacios-Ramirez
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid–CSIC, 47003 Valladolid, Spain; (L.G.-G.); (O.M.); (D.B.); (J.d.l.P.-A.); (L.A.-F.); (R.P.-R.)
| | - Laura Senovilla
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid–CSIC, 47003 Valladolid, Spain; (L.G.-G.); (O.M.); (D.B.); (J.d.l.P.-A.); (L.A.-F.); (R.P.-R.)
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, 75006 Paris, France;
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, 94805 Villejuif, France
| |
Collapse
|
21
|
Tang F, Cui Q. Diverse roles of aldolase enzymes in cancer development, drug resistance and therapeutic approaches as moonlighting enzymes. Med Oncol 2024; 41:224. [PMID: 39120781 DOI: 10.1007/s12032-024-02470-x] [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: 06/26/2024] [Accepted: 08/01/2024] [Indexed: 08/10/2024]
Abstract
Aldolase enzymes, particularly ALDOA, ALDOB, and ALDOC, play a crucial role in the development and progression of cancer. While the aldolase family is mainly known for its involvement in the glycolysis pathway, these enzymes also have various pathological and physiological functions through distinct signaling pathways such as Wnt/β-catenin, EGFR/MAPK, Akt, and HIF-1α. This has garnered increased attention in recent years and shed light on other sides of this enzyme. Potential therapeutic strategies targeting aldolases include using siRNA, inhibitors like naphthol AS-E phosphate and TX-2098, and natural compounds such as HDPS-4II and L-carnosine. Additionally, anticancer peptides derived from ALDOA, like P04, can potentially increase cancer cells' sensitivity to chemotherapy. Aldolases also affect cancer drug resistance by different approaches, making them good therapeutic targets. In this review, we extensively explore the role of aldolase enzymes in various types of cancers in proliferation, invasion, migration, and drug resistance; we also significantly explore the possible treatment considering aldolase function.
Collapse
Affiliation(s)
- Fan Tang
- General Surgery Department, Xinhua Hospital of Yili Kazak Autonomous Prefecture, YiLi, 835000, China
| | - Qingyang Cui
- Department of Interventional Oncology, Xinhua Hospital of Yili Kazak Autonomous Prefecture, YiLi, 835000, China.
| |
Collapse
|
22
|
Pranzini E, Ippolito L, Pardella E, Giannoni E, Chiarugi P. Adapt and shape: metabolic features within the metastatic niche. Trends Endocrinol Metab 2024:S1043-2760(24)00197-8. [PMID: 39122599 DOI: 10.1016/j.tem.2024.07.016] [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: 05/31/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 08/12/2024]
Abstract
The success of disseminating cancer cells (DTCs) at specific metastatic sites is influenced by several metabolic factors. Even before DTCs arrival, metabolic conditioning from the primary tumor participates in creating a favorable premetastatic niche at distant organs. In addition, DTCs adjust their metabolism to better survive along the metastatic journey and successfully colonize their ultimate destination. However, the idea that the environment of the target organs may metabolically impact the metastatic fate is often underestimated. Here, we review the coexistence of two distinct strategies by which cancer cells shape and/or adapt to the metabolic profile of colonized tissues, ultimately creating a proper soil for their seeding and proliferation.
Collapse
Affiliation(s)
- Erica Pranzini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale Morgagni, 50, 50134 Firenze, (FI), Italy
| | - Luigi Ippolito
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale Morgagni, 50, 50134 Firenze, (FI), Italy
| | - Elisa Pardella
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale Morgagni, 50, 50134 Firenze, (FI), Italy
| | - Elisa Giannoni
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale Morgagni, 50, 50134 Firenze, (FI), Italy
| | - Paola Chiarugi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale Morgagni, 50, 50134 Firenze, (FI), Italy.
| |
Collapse
|
23
|
Lodge M, Dykes R, Kennedy A. Regulation of Fructose Metabolism in Nonalcoholic Fatty Liver Disease. Biomolecules 2024; 14:845. [PMID: 39062559 PMCID: PMC11274671 DOI: 10.3390/biom14070845] [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: 06/17/2024] [Revised: 07/02/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Elevations in fructose consumption have been reported to contribute significantly to an increased incidence of obesity and metabolic diseases in industrial countries. Mechanistically, a high fructose intake leads to the dysregulation of glucose, triglyceride, and cholesterol metabolism in the liver, and causes elevations in inflammation and drives the progression of nonalcoholic fatty liver disease (NAFLD). A high fructose consumption is considered to be toxic to the body, and there are ongoing measures to develop pharmaceutical therapies targeting fructose metabolism. Although a large amount of work has summarized the effects fructose exposure within the intestine, liver, and kidney, there remains a gap in our knowledge regarding how fructose both indirectly and directly influences immune cell recruitment, activation, and function in metabolic tissues, which are essential to tissue and systemic inflammation. The most recent literature demonstrates that direct fructose exposure regulates oxidative metabolism in macrophages, leading to inflammation. The present review highlights (1) the mechanisms by which fructose metabolism impacts crosstalk between tissues, nonparenchymal cells, microbes, and immune cells; (2) the direct impact of fructose on immune cell metabolism and function; and (3) therapeutic targets of fructose metabolism to treat NAFLD. In addition, the review highlights how fructose disrupts liver tissue homeostasis and identifies new therapeutic targets for treating NAFLD and obesity.
Collapse
Affiliation(s)
| | | | - Arion Kennedy
- Department of Molecular and Structural Biochemistry, North Carolina State University, 128 Polk Hall Campus, Box 7622, Raleigh, NC 27695, USA
| |
Collapse
|
24
|
Lane AN, Higashi RM, Fan TWM. Challenges of Spatially Resolved Metabolism in Cancer Research. Metabolites 2024; 14:383. [PMID: 39057706 PMCID: PMC11278851 DOI: 10.3390/metabo14070383] [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: 05/26/2024] [Revised: 06/28/2024] [Accepted: 07/07/2024] [Indexed: 07/28/2024] Open
Abstract
Stable isotope-resolved metabolomics comprises a critical set of technologies that can be applied to a wide variety of systems, from isolated cells to whole organisms, to define metabolic pathway usage and responses to perturbations such as drugs or mutations, as well as providing the basis for flux analysis. As the diversity of stable isotope-enriched compounds is very high, and with newer approaches to multiplexing, the coverage of metabolism is now very extensive. However, as the complexity of the model increases, including more kinds of interacting cell types and interorgan communication, the analytical complexity also increases. Further, as studies move further into spatially resolved biology, new technical problems have to be overcome owing to the small number of analytes present in the confines of a single cell or cell compartment. Here, we review the overall goals and solutions made possible by stable isotope tracing and their applications to models of increasing complexity. Finally, we discuss progress and outstanding difficulties in high-resolution spatially resolved tracer-based metabolic studies.
Collapse
Affiliation(s)
- Andrew N. Lane
- Department of Toxicology and Cancer Biology and Markey Cancer Center, University of Kentucky, 789 S. Limestone St., Lexington, KY 40536, USA; (R.M.H.); (T.W.-M.F.)
| | | | | |
Collapse
|
25
|
Peng C, Yang P, Zhang D, Jin C, Peng W, Wang T, Sun Q, Chen Z, Feng Y, Sun Y. KHK-A promotes fructose-dependent colorectal cancer liver metastasis by facilitating the phosphorylation and translocation of PKM2. Acta Pharm Sin B 2024; 14:2959-2976. [PMID: 39027256 PMCID: PMC11252482 DOI: 10.1016/j.apsb.2024.04.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 03/01/2024] [Accepted: 04/15/2024] [Indexed: 07/20/2024] Open
Abstract
Excessive fructose diet is closely associated with colorectal cancer (CRC) progression. Nevertheless, fructose's specific function and precise mechanism in colorectal cancer liver metastasis (CRLM) is rarely known. Here, this study reported that the fructose absorbed by primary colorectal cancer could accelerate CRLM, and the expression of KHK-A, not KHK-C, in liver metastasis was higher than in paired primary tumors. Furthermore, KHK-A facilitated fructose-dependent CRLM in vitro and in vivo by phosphorylating PKM2 at Ser37. PKM2 phosphorylated by KHK-A inhibited its tetramer formation and pyruvic acid kinase activity but promoted the nuclear accumulation of PKM2. EMT and aerobic glycolysis activated by nuclear PKM2 enhance CRC cells' migration ability and anoikis resistance during CRLM progression. TEPP-46 treatment, targeting the phosphorylation of PKM2, inhibited the pro-metastatic effect of KHK-A. Besides, c-myc activated by nuclear PKM2 promotes alternative splicing of KHK-A, forming a positive feedback loop.
Collapse
Affiliation(s)
- Chaofan Peng
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Colorectal Institute of Nanjing Medical University, Nanjing 210029, China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Peng Yang
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Colorectal Institute of Nanjing Medical University, Nanjing 210029, China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Dongsheng Zhang
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Colorectal Institute of Nanjing Medical University, Nanjing 210029, China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Chi Jin
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Colorectal Institute of Nanjing Medical University, Nanjing 210029, China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Wen Peng
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Colorectal Institute of Nanjing Medical University, Nanjing 210029, China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Tuo Wang
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Colorectal Institute of Nanjing Medical University, Nanjing 210029, China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Qingyang Sun
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Colorectal Institute of Nanjing Medical University, Nanjing 210029, China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Zhihao Chen
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Colorectal Institute of Nanjing Medical University, Nanjing 210029, China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Yifei Feng
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Colorectal Institute of Nanjing Medical University, Nanjing 210029, China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing 210029, China
| | - Yueming Sun
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Colorectal Institute of Nanjing Medical University, Nanjing 210029, China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing 210029, China
| |
Collapse
|
26
|
Wang DX, Liu H, Tian JC, Zhang DL, Yan LJ, Ding ZN, Li H, Yan YC, Dong ZR, Li T. Neoadjuvant immunotherapy based on PD-1/L1 inhibitors for gastrointestinal tumors: a review of the rationale and clinical advances. Int J Surg 2024; 110:3707-3722. [PMID: 38518083 PMCID: PMC11175801 DOI: 10.1097/js9.0000000000001357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 03/03/2024] [Indexed: 03/24/2024]
Abstract
The landscape of current tumor treatment has been revolutionized by the advent of immunotherapy based on PD-1/PD-L1 inhibitors. Leveraging its capacity to mobilize systemic antitumor immunity, which is primarily mediated by T cells, there is growing exploration and expansion of its potential value in various stages of clinical tumor treatment. Neoadjuvant immunotherapy induces a robust immune response against tumors prior to surgery, effectively facilitating tumor volume reduction, early eradication or suppression of tumor cell activity, and control of potential metastatic spread, to improve curative surgical resection rates, and prevent tumor recurrence. This review delineates the theoretical basis of neoadjuvant immunotherapy from preclinical research evidence, discusses specific challenges in clinical application, and provides a comprehensive overview of clinical research progress in neoadjuvant immunotherapy for gastrointestinal tumors. These findings suggest that neoadjuvant immunotherapy has the potential to ameliorate immunosuppressive states and enhance cytotoxic T cell function while preserving lymphatic drainage in the preoperative period. However, further investigations are needed on specific treatment regimens, suitable patient populations, and measurable endpoints. Despite numerous studies demonstrating the promising efficacy and manageable adverse events of neoadjuvant immunotherapy in gastrointestinal tumors, the availability of high-quality randomized controlled trials is limited, which highlights the necessity for further research.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Tao Li
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, People’s Republic of China
| |
Collapse
|
27
|
Liu W, Wu Y, Ma R, Zhu X, Wang R, He L, Shu M. Multi-omics analysis of a case of congenital microtia reveals aldob and oxidative stress associated with microtia etiology. Orphanet J Rare Dis 2024; 19:218. [PMID: 38802922 PMCID: PMC11129396 DOI: 10.1186/s13023-024-03149-2] [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: 04/04/2023] [Accepted: 03/27/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND Microtia is reported to be one of the most common congenital craniofacial malformations. Due to the complex etiology and the ethical barrier of embryonic study, the precise mechanisms of microtia remain unclear. Here we report a rare case of microtia with costal chondrodysplasia based on bioinformatics analysis and further verifications on other sporadic microtia patients. RESULTS One hundred fourteen deleterious insert and deletion (InDel) and 646 deleterious SNPs were screened out by WES, candidate genes were ranked in descending order according to their relative impact with microtia. Label-free proteomic analysis showed that proteins significantly different between the groups were related with oxidative stress and energy metabolism. By real-time PCR and immunohistochemistry, we further verified the candidate genes between other sporadic microtia and normal ear chondrocytes, which showed threonine aspartase, cadherin-13, aldolase B and adiponectin were significantly upregulated in mRNA levels but were significantly lower in protein levels. ROS detection and mitochondrial membrane potential (∆ Ψ m) detection proved that oxidative stress exists in microtia chondrocytes. CONCLUSIONS Our results not only spot new candidate genes by WES and label-free proteomics, but also speculate for the first time that metabolism and oxidative stress may disturb cartilage development and this might become therapeutic targets and potential biomarkers with clinical usefulness in the future.
Collapse
Affiliation(s)
- Wenbo Liu
- The First Affiliated Hospital of Xi'an Jiao Tong University, No.277 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Yi Wu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Rulan Ma
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiao Tong University Medical College, Xi'an, Shaanxi, China
| | - Xinxi Zhu
- The First Affiliated Hospital of Xi'an Jiao Tong University, No.277 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Rui Wang
- The First Affiliated Hospital of Xi'an Jiao Tong University, No.277 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Lin He
- The First Affiliated Hospital of Xi'an Jiao Tong University, No.277 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Maoguo Shu
- The First Affiliated Hospital of Xi'an Jiao Tong University, No.277 Yanta West Road, Xi'an, Shaanxi, 710061, China.
| |
Collapse
|
28
|
Chen J, Liu X, Zou Y, Gong J, Ge Z, Lin X, Zhang W, Huang H, Zhao J, Saw PE, Lu Y, Hu H, Song E. A high-fat diet promotes cancer progression by inducing gut microbiota-mediated leucine production and PMN-MDSC differentiation. Proc Natl Acad Sci U S A 2024; 121:e2306776121. [PMID: 38709933 PMCID: PMC11098111 DOI: 10.1073/pnas.2306776121] [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: 05/09/2023] [Accepted: 02/16/2024] [Indexed: 05/08/2024] Open
Abstract
A high-fat diet (HFD) is a high-risk factor for the malignant progression of cancers through the disruption of the intestinal microbiota. However, the role of the HFD-related gut microbiota in cancer development remains unclear. This study found that obesity and obesity-related gut microbiota were associated with poor prognosis and advanced clinicopathological status in female patients with breast cancer. To investigate the impact of HFD-associated gut microbiota on cancer progression, we established various models, including HFD feeding, fecal microbiota transplantation, antibiotic feeding, and bacterial gavage, in tumor-bearing mice. HFD-related microbiota promotes cancer progression by generating polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs). Mechanistically, the HFD microbiota released abundant leucine, which activated the mTORC1 signaling pathway in myeloid progenitors for PMN-MDSC differentiation. Clinically, the elevated leucine level in the peripheral blood induced by the HFD microbiota was correlated with abundant tumoral PMN-MDSC infiltration and poor clinical outcomes in female patients with breast cancer. These findings revealed that the "gut-bone marrow-tumor" axis is involved in HFD-mediated cancer progression and opens a broad avenue for anticancer therapeutic strategies by targeting the aberrant metabolism of the gut microbiota.
Collapse
Affiliation(s)
- Jiewen Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou510120, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou510120, China
- Department of Breast Medicine, Affiliated Foshan Maternity and Child Healthcare Hospital, Southern Medical University, Foshan528000, China
| | - Xiyuan Liu
- Run-ze Laboratory for Gastrointestinal Microbiome Study, School of Life Sciences, Sun Yat-Sen University, Guangzhou510275, China
| | - Yi Zou
- Run-ze Laboratory for Gastrointestinal Microbiome Study, School of Life Sciences, Sun Yat-Sen University, Guangzhou510275, China
| | - Junli Gong
- Run-ze Laboratory for Gastrointestinal Microbiome Study, School of Life Sciences, Sun Yat-Sen University, Guangzhou510275, China
| | - Zhenhuang Ge
- Run-ze Laboratory for Gastrointestinal Microbiome Study, School of Life Sciences, Sun Yat-Sen University, Guangzhou510275, China
| | - Xiaorong Lin
- Diagnosis and Treatment Center of Breast Diseases, Shantou Central Hospital, Shantou515000, China
| | - Wei Zhang
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou510120, China
| | - Hongyan Huang
- Department of Breast Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou510282, China
| | - Jianli Zhao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou510120, China
| | - Phei Er Saw
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou510120, China
| | - Yongjun Lu
- Run-ze Laboratory for Gastrointestinal Microbiome Study, School of Life Sciences, Sun Yat-Sen University, Guangzhou510275, China
| | - Hai Hu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou510120, China
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou510120, China
| | - Erwei Song
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou510120, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou510120, China
| |
Collapse
|
29
|
Zhang J, Wang QH, Miao BB, Wu RX, Li QQ, Tang BG, Liang ZB, Niu SF. Liver transcriptome analysis reveal the metabolic and apoptotic responses of Trachinotus ovatus under acute cold stress. FISH & SHELLFISH IMMUNOLOGY 2024; 148:109476. [PMID: 38447780 DOI: 10.1016/j.fsi.2024.109476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 02/07/2024] [Accepted: 03/03/2024] [Indexed: 03/08/2024]
Abstract
Trachinotus ovatus is an economically important fish and has been recommended as a high-quality aquaculture fish breed for the high-quality development of sea ranches in the South China Sea. However, T. ovatus shows intolerance to low temperature, greatly limiting the extension of farming scale, reducing production efficiency in winter, and increasing farming risks. In this study, liver transcriptome analysis was investigated in T. ovatus under acute low temperature conditions (20 and 15 °C) using RNA sequencing (RNA-Seq) technology. Inter-groups differential expression analysis and trend analysis screened 1219 DEGs and four significant profiles (profiles 0, 3, 4, and 7), respectively. GO enrichment analysis showed that these DEGs were mainly related to metabolic process and cell growth and death process. KEGG enrichment analysis found that DEGs were mainly associated with lipid metabolism, carbohydrate metabolism, and cell growth and death, such as gluconeogenesis, glycolysis, fatty acid oxidation, cholesterol biosynthesis, p53 signaling pathway, cell cycle arrest, and apoptotic cell death. Moreover, protein-protein interaction networks identified two hub genes (FOS and JUNB) and some important genes related to metabolic process and cell growth and death process, that corresponding to enrichment analysis. Overall, gluconeogenesis, lipid mobilization, and fatty acid oxidation in metabolic process and cell cycle arrest and apoptotic cell death in cell growth and death process were enhanced, while glycolysis, liver glycogen synthesis and cholesterol biosynthesis in metabolic process were inhibited. The enhancement or attenuatment of metabolic process and cell growth and death process is conducive to maintain energy balance, normal fluidity of cell membrane, normal physiological functions of liver cell, enhancing the tolerance of T. ovatus to cold stress. These results suggested that metabolic process and cell growth and death process play important roles in response to acute cold stress in the liver of T. ovatus. Gene expreesion level analysis showed that acute cold stress at 15 °C was identified as a critical temperature point for T. ovatus in term of cellular metabolism alteration and apoptosis inducement, and rewarming intervention should be timely implemented above 15 °C. Our study can provide theoretical support for breeding cold-tolerant cultivars of T. ovatus, which is contributed to high-quality productions fish production.
Collapse
Affiliation(s)
- Jing Zhang
- College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, China; Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang, 524025, China
| | - Qing-Hua Wang
- College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Ben-Ben Miao
- College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Ren-Xie Wu
- College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, China; Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang, 524025, China
| | - Qian-Qian Li
- College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Bao-Gui Tang
- College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, China; Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang, 524025, China
| | - Zhen-Bang Liang
- College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Su-Fang Niu
- College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, China; Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang, 524025, China.
| |
Collapse
|
30
|
Ting KKY. Fructose-induced metabolic reprogramming of cancer cells. Front Immunol 2024; 15:1375461. [PMID: 38711514 PMCID: PMC11070519 DOI: 10.3389/fimmu.2024.1375461] [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: 01/23/2024] [Accepted: 04/10/2024] [Indexed: 05/08/2024] Open
Abstract
Excess dietary fructose consumption has been long proposed as a culprit for the world-wide increase of incidence in metabolic disorders and cancer within the past decades. Understanding that cancer cells can gradually accumulate metabolic mutations in the tumor microenvironment, where glucose is often depleted, this raises the possibility that fructose can be utilized by cancer cells as an alternative source of carbon. Indeed, recent research has increasingly identified various mechanisms that show how cancer cells can metabolize fructose to support their proliferating and migrating needs. In light of this growing interest, this review will summarize the recent advances in understanding how fructose can metabolically reprogram different types of cancer cells, as well as how these metabolic adaptations can positively support cancer cells development and malignancy.
Collapse
Affiliation(s)
- Kenneth K. Y. Ting
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| |
Collapse
|
31
|
DeVito NC, Nguyen YV, Sturdivant M, Plebanek MP, Howell K, Yarla N, Jain V, Aksu M, Beasley G, Theivanthiran B, Hanks BA. Gli2 Facilitates Tumor Immune Evasion and Immunotherapeutic Resistance by Coordinating Wnt Ligand and Prostaglandin Signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.31.587500. [PMID: 38617347 PMCID: PMC11014473 DOI: 10.1101/2024.03.31.587500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Therapeutic resistance to immune checkpoint blockade has been commonly linked to the process of mesenchymal transformation (MT) and remains a prevalent obstacle across many cancer types. An improved mechanistic understanding for MT-mediated immune evasion promises to lead to more effective combination therapeutic regimens. Herein, we identify the Hedgehog transcription factor, Gli2, as a key node of tumor-mediated immune evasion and immunotherapy resistance during MT. Mechanistic studies reveal that Gli2 generates an immunotolerant tumor microenvironment through the upregulation of Wnt ligand production and increased prostaglandin synthesis. This pathway drives the recruitment, viability, and function of granulocytic myeloid-derived suppressor cells (PMN-MDSCs) while also impairing type I conventional dendritic cell, CD8 + T cell, and NK cell functionality. Pharmacologic EP2/EP4 prostaglandin receptor inhibition and Wnt ligand inhibition each reverses a subset of these effects, while preventing primary and adaptive resistance to anti-PD-1 immunotherapy, respectively. A transcriptional Gli2 signature correlates with resistance to anti-PD-1 immunotherapy in stage IV melanoma patients, providing a translational roadmap to direct combination immunotherapeutics in the clinic. SIGNIFICANCE Gli2-induced EMT promotes immune evasion and immunotherapeutic resistance via coordinated prostaglandin and Wnt signaling.
Collapse
|
32
|
Xiang K, Wang E, Mantyh J, Rupprecht G, Negrete M, Sanati G, Hsu C, Randon P, Dohlman A, Kretzschmar K, Bose S, Giroux N, Ding S, Wang L, Balcazar JP, Huang Q, Sundaramoorthy P, Xi R, McCall SJ, Wang Z, Jiang C, Kang Y, Kopetz S, Crawford GE, Lipkin SM, Wang XF, Clevers H, Hsu D, Shen X. Chromatin Remodeling in Patient-Derived Colorectal Cancer Models. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2303379. [PMID: 38380561 DOI: 10.1002/advs.202303379] [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: 05/24/2023] [Revised: 11/22/2023] [Indexed: 02/22/2024]
Abstract
Patient-Derived Organoids (PDO) and Xenografts (PDX) are the current gold standards for patient-derived models of cancer (PDMC). Nevertheless, how patient tumor cells evolve in these models and the impact on drug response remains unclear. Herein, the transcriptomic and chromatin accessibility landscapes of matched colorectal cancer (CRC) PDO, PDX, PDO-derived PDX (PDOX), and original patient tumors (PT) are compared. Two major remodeling axes are discovered. The first axis delineates PDMC from PT, and the second axis distinguishes PDX and PDO. PDOX are more similar to PDX than PDO, indicating the growth environment is a driving force for chromatin adaptation. Transcription factors (TF) that differentially bind to open chromatins between matched PDO and PDOX are identified. Among them, KLF14 and EGR2 footprints are enriched in PDOX relative to matched PDO, and silencing of KLF14 or EGR2 promoted tumor growth. Furthermore, EPHA4, a shared downstream target gene of KLF14 and EGR2, altered tumor sensitivity to MEK inhibitor treatment. Altogether, patient-derived CRC cells undergo both common and distinct chromatin remodeling in PDO and PDX/PDOX, driven largely by their respective microenvironments, which results in differences in growth and drug sensitivity and needs to be taken into consideration when interpreting their ability to predict clinical outcome.
Collapse
Affiliation(s)
- Kun Xiang
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, 27708, USA
| | - Ergang Wang
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, 27708, USA
| | - John Mantyh
- Department of Medicine, School of Medicine, Duke University, Durham, NC, 27710, USA
| | - Gabrielle Rupprecht
- Department of Medicine, School of Medicine, Duke University, Durham, NC, 27710, USA
| | - Marcos Negrete
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, 27708, USA
| | - Golshid Sanati
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, 27708, USA
| | - Carolyn Hsu
- Department of Medicine, School of Medicine, Duke University, Durham, NC, 27710, USA
| | - Peggy Randon
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, 27709, USA
| | - Anders Dohlman
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, 27708, USA
| | - Kai Kretzschmar
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center (UMC) Utrecht, Uppsalalaan 8, Utrecht, CT, 3584, The Netherlands
- Mildred Scheel Early Career Centre (MSNZ) for Cancer Research Würzburg, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Shree Bose
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, 27708, USA
| | - Nicholas Giroux
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, 27708, USA
| | - Shengli Ding
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, 27708, USA
| | - Lihua Wang
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, 27708, USA
| | - Jorge Prado Balcazar
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, 27708, USA
| | - Qiang Huang
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, 27708, USA
- Terasaki Institute, Los Angeles, CA, 90024, USA
| | | | - Rui Xi
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, 27708, USA
| | - Shannon Jones McCall
- Department of Pathology, School of Medicine, Duke University, Durham, NC, 27710, USA
| | - Zhaohui Wang
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, 27708, USA
| | | | - Yubin Kang
- Department of Medicine, School of Medicine, Duke University, Durham, NC, 27710, USA
| | - Scott Kopetz
- Department of Gastrointestinal (GI) Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Gregory E Crawford
- Department of Pediatrics, Division of Medical Genetics, School of Medicine, Duke University, Durham, NC, 27710, USA
| | - Steven M Lipkin
- Department of Medicine and Program in Mendelian Genetics, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Xiao-Fan Wang
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Hans Clevers
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center (UMC) Utrecht, Uppsalalaan 8, Utrecht, CT, 3584, The Netherlands
| | - David Hsu
- Department of Medicine, School of Medicine, Duke University, Durham, NC, 27710, USA
| | - Xiling Shen
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, 27708, USA
- Terasaki Institute, Los Angeles, CA, 90024, USA
| |
Collapse
|
33
|
Zhou X, Qian Y, Ling C, He Z, Shi P, Gao Y, Sui X. An integrated framework for prognosis prediction and drug response modeling in colorectal liver metastasis drug discovery. J Transl Med 2024; 22:321. [PMID: 38555418 PMCID: PMC10981831 DOI: 10.1186/s12967-024-05127-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 03/23/2024] [Indexed: 04/02/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is the third most prevalent cancer globally, and liver metastasis (CRLM) is the primary cause of death. Hence, it is essential to discover novel prognostic biomarkers and therapeutic drugs for CRLM. METHODS This study developed two liver metastasis-associated prognostic signatures based on differentially expressed genes (DEGs) in CRLM. Additionally, we employed an interpretable deep learning model utilizing drug sensitivity databases to identify potential therapeutic drugs for high-risk CRLM patients. Subsequently, in vitro and in vivo experiments were performed to verify the efficacy of these compounds. RESULTS These two prognostic models exhibited superior performance compared to previously reported ones. Obatoclax, a BCL-2 inhibitor, showed significant differential responses between high and low risk groups classified by prognostic models, and demonstrated remarkable effectiveness in both Transwell assay and CT26 colorectal liver metastasis mouse model. CONCLUSIONS This study highlights the significance of developing specialized prognostication approaches and investigating effective therapeutic drugs for patients with CRLM. The application of a deep learning drug response model provides a new drug discovery strategy for translational medicine in precision oncology.
Collapse
Affiliation(s)
- Xiuman Zhou
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong Province, 518107, China
| | - Yuzhen Qian
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Chen Ling
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong Province, 518107, China
| | - Zhuoying He
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong Province, 518107, China
| | - Peishang Shi
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yanfeng Gao
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong Province, 518107, China.
| | - Xinghua Sui
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong Province, 518107, China.
| |
Collapse
|
34
|
Kopec M, Beton-Mysur K. The role of glucose and fructose on lipid droplet metabolism in human normal bronchial and cancer lung cells by Raman spectroscopy. Chem Phys Lipids 2024; 259:105375. [PMID: 38159659 DOI: 10.1016/j.chemphyslip.2023.105375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/18/2023] [Accepted: 12/27/2023] [Indexed: 01/03/2024]
Abstract
Fructose is one of the most important monosaccharides in the human diet that the human body needs for proper metabolism. This paper presents an approach to study biochemical changes caused by sugars in human normal bronchial cells (BEpiC) and human cancer lung cells (A549) by Raman spectroscopy and Raman imaging. Results after supplementation of human bronchial and lung cells with fructose are also discussed and compared with results obtained for pure human bronchial and lung cells. Based on Raman techniques we have proved that peaks at 750 cm-1, 1126 cm-1, 1444 cm-1, 1584 cm-1 and 2845 cm-1 can be treated as biomarkers to monitor fructose changes in cells. Results for fructose have been compared with results for glucose. Raman analysis of the bands at 750 cm-1, 1126 cm-1, 1584 cm-1 and 2845 cm-1 for pure BEpiC and A549 cells and BEpiC and A549 after supplementation with fructose and glucose are higher after supplementation with fructose in comparison to glucose. The obtained results shed light on the uninvestigated influence of glucose and fructose on lipid droplet metabolism by Raman spectroscopy methods.
Collapse
Affiliation(s)
- Monika Kopec
- Lodz University of Technology, Institute of Applied Radiation Chemistry, Laboratory of Laser Molecular Spectroscopy, Wroblewskiego 15, 93-590 Lodz, Poland.
| | - Karolina Beton-Mysur
- Lodz University of Technology, Institute of Applied Radiation Chemistry, Laboratory of Laser Molecular Spectroscopy, Wroblewskiego 15, 93-590 Lodz, Poland
| |
Collapse
|
35
|
Menyhárt O, Győrffy B. Dietary approaches for exploiting metabolic vulnerabilities in cancer. Biochim Biophys Acta Rev Cancer 2024; 1879:189062. [PMID: 38158024 DOI: 10.1016/j.bbcan.2023.189062] [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: 06/20/2023] [Revised: 12/20/2023] [Accepted: 12/20/2023] [Indexed: 01/03/2024]
Abstract
Renewed interest in tumor metabolism sparked an enthusiasm for dietary interventions to prevent and treat cancer. Changes in diet impact circulating nutrient levels in the plasma and the tumor microenvironment, and preclinical studies suggest that dietary approaches, including caloric and nutrient restrictions, can modulate tumor initiation, progression, and metastasis. Cancers are heterogeneous in their metabolic dependencies and preferred energy sources and can be addicted to glucose, fructose, amino acids, or lipids for survival and growth. This dependence is influenced by tumor type, anatomical location, tissue of origin, aberrant signaling, and the microenvironment. This review summarizes nutrient dependencies and the related signaling pathway activations that provide targets for nutritional interventions. We examine popular dietary approaches used as adjuvants to anticancer therapies, encompassing caloric restrictions, including time-restricted feeding, intermittent fasting, fasting-mimicking diets (FMDs), and nutrient restrictions, notably the ketogenic diet. Despite promising results, much of the knowledge on dietary restrictions comes from in vitro and animal studies, which may not accurately reflect real-life situations. Further research is needed to determine the optimal duration, timing, safety, and efficacy of dietary restrictions for different cancers and treatments. In addition, well-designed human trials are necessary to establish the link between specific metabolic vulnerabilities and targeted dietary interventions. However, low patient compliance in clinical trials remains a significant challenge.
Collapse
Affiliation(s)
- Otília Menyhárt
- Semmelweis University, Department of Bioinformatics, Tűzoltó u. 7-9, H-1094 Budapest, Hungary; Research Centre for Natural Sciences, Cancer Biomarker Research Group, Institute of Enzymology, Magyar tudósok krt. 2, H-1117 Budapest, Hungary; National Laboratory for Drug Research and Development, Magyar tudósok krt. 2, H-1117 Budapest, Hungary
| | - Balázs Győrffy
- Semmelweis University, Department of Bioinformatics, Tűzoltó u. 7-9, H-1094 Budapest, Hungary; Research Centre for Natural Sciences, Cancer Biomarker Research Group, Institute of Enzymology, Magyar tudósok krt. 2, H-1117 Budapest, Hungary; National Laboratory for Drug Research and Development, Magyar tudósok krt. 2, H-1117 Budapest, Hungary.
| |
Collapse
|
36
|
Zhang M, Duan C, Lin W, Wu H, Chen L, Guo H, Yu M, Liu Q, Nie Y, Wang H, Wang S. Levistilide A Exerts a Neuroprotective Effect by Suppressing Glucose Metabolism Reprogramming and Preventing Microglia Polarization Shift: Implications for Parkinson's Disease. Molecules 2024; 29:912. [PMID: 38398662 PMCID: PMC10893236 DOI: 10.3390/molecules29040912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/16/2024] [Accepted: 02/17/2024] [Indexed: 02/25/2024] Open
Abstract
The microglia, displaying diverse phenotypes, play a significant regulatory role in the development, progression, and prognosis of Parkinson's disease. Research has established that glycolytic reprogramming serves as a critical regulator of inflammation initiation in pro-inflammatory macrophages. Furthermore, the modulation of glycolytic reprogramming has the potential to reverse the polarized state of these macrophages. Previous studies have shown that Levistilide A (LA), a phthalide component derived from Angelica sinensis, possesses a range of pharmacological effects, including anti-inflammatory, antioxidant, and neuroprotective properties. In our study, we have examined the impact of LA on inflammatory cytokines and glucose metabolism in microglia induced by lipopolysaccharide (LPS). Furthermore, we explored the effects of LA on the AMPK/mTOR pathway and assessed its neuroprotective potential both in vitro and in vivo. The findings revealed that LA notably diminished the expression of M1 pro-inflammatory factors induced by LPS in microglia, while leaving M2 anti-inflammatory factor expression unaltered. Additionally, it reduced ROS production and suppressed IκB-α phosphorylation levels as well as NF-κB p65 nuclear translocation. Notably, LA exhibited the ability to reverse microglial glucose metabolism reprogramming and modulate the phosphorylation levels of AMPK/mTOR. In vivo experiments further corroborated these findings, demonstrating that LA mitigated the death of TH-positive dopaminergic neurons and reduced microglia activation in the ventral SNpc brain region of the midbrain and the striatum. In summary, LA exhibited neuroprotective benefits by modulating the polarization state of microglia and altering glucose metabolism, highlighting its therapeutic potential.
Collapse
Affiliation(s)
- Mingjie Zhang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China; (M.Z.); (C.D.); (W.L.); (M.Y.); (Q.L.); (Y.N.)
| | - Congyan Duan
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China; (M.Z.); (C.D.); (W.L.); (M.Y.); (Q.L.); (Y.N.)
| | - Weifang Lin
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China; (M.Z.); (C.D.); (W.L.); (M.Y.); (Q.L.); (Y.N.)
| | - Honghua Wu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China; (H.W.); (L.C.); (H.G.)
| | - Lu Chen
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China; (H.W.); (L.C.); (H.G.)
| | - Hong Guo
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China; (H.W.); (L.C.); (H.G.)
| | - Minyu Yu
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China; (M.Z.); (C.D.); (W.L.); (M.Y.); (Q.L.); (Y.N.)
| | - Qi Liu
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China; (M.Z.); (C.D.); (W.L.); (M.Y.); (Q.L.); (Y.N.)
| | - Yaling Nie
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China; (M.Z.); (C.D.); (W.L.); (M.Y.); (Q.L.); (Y.N.)
| | - Hong Wang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China; (M.Z.); (C.D.); (W.L.); (M.Y.); (Q.L.); (Y.N.)
| | - Shaoxia Wang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China; (M.Z.); (C.D.); (W.L.); (M.Y.); (Q.L.); (Y.N.)
| |
Collapse
|
37
|
Yeh CY, Cai HY, Kuo HH, Lin YY, He ZJ, Cheng HC, Yang CJ, Huang CYF, Chang YC. ALDOA coordinates PDE3A through the β-catenin/ID3 axis to stimulate cancer metastasis and M2 polarization in lung cancer with EGFR mutations. Biochem Biophys Res Commun 2024; 696:149489. [PMID: 38244313 DOI: 10.1016/j.bbrc.2024.149489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/21/2023] [Accepted: 01/05/2024] [Indexed: 01/22/2024]
Abstract
Lung cancer has a high incidence rate and requires more effective treatment strategies and drug options for clinical patients. EGFR is a common genetic alteration event in lung cancer that affects patient survival and drug strategy. Our study discovered aberrant aldolase A (ALDOA) expression and dysfunction in lung cancer patients with EGFR mutations. In addition to investigating relevant metabolic processes like glucose uptake, lactate production, and ATPase activity, we examined multi-omics profiles (transcriptomics, proteomics, and pull-down assays). It was observed that phosphodiesterase 3A (PDE3A) enzyme and ALDOA exhibit correlation, and furthermore, they impact M2 macrophage polarization through β-catenin and downstream ID3. In addition to demonstrating the aforementioned mechanism of action, our experiments discovered that the PDE3 inhibitor trequinsin has a substantial impact on lung cancer cell lines with EGFR mutants. The trequinsin medication was found to decrease the M2 macrophage polarization status and several cancer phenotypes, in addition to transduction. These findings have potential prognostic and therapeutic applications for clinical patients with EGFR mutation and lung cancer.
Collapse
Affiliation(s)
- Chia-Ying Yeh
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Huei Yu Cai
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Han-His Kuo
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - You-Yu Lin
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Zhao-Jing He
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hsiao-Chen Cheng
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chih-Jen Yang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; School of Post-Baccalaureate Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chi-Ying F Huang
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yu-Chan Chang
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
38
|
Kang YL, Kim J, Kwak SB, Kim YS, Huh J, Park JW. The polyol pathway and nuclear ketohexokinase A signaling drive hyperglycemia-induced metastasis of gastric cancer. Exp Mol Med 2024; 56:220-234. [PMID: 38200154 PMCID: PMC10834943 DOI: 10.1038/s12276-023-01153-3] [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: 01/10/2023] [Revised: 10/20/2023] [Accepted: 11/07/2023] [Indexed: 01/12/2024] Open
Abstract
Diabetes might be associated with increased cancer risk, with several studies reporting hyperglycemia as a primary oncogenic stimulant. Since glucose metabolism is linked to numerous metabolic pathways, it is difficult to specify the mechanisms underlying hyperglycemia-induced cancer progression. Here, we focused on the polyol pathway, which is dramatically activated under hyperglycemia and causes diabetic complications. We investigated whether polyol pathway-derived fructose facilitates hyperglycemia-induced gastric cancer metastasis. We performed bioinformatics analysis of gastric cancer datasets and immunohistochemical analyses of gastric cancer specimens, followed by transcriptomic and proteomic analyses to evaluate phenotypic changes in gastric cancer cells. Consequently, we found a clinical association between the polyol pathway and gastric cancer progression. In gastric cancer cell lines, hyperglycemia enhanced cell migration and invasion, cytoskeletal rearrangement, and epithelial-mesenchymal transition (EMT). The hyperglycemia-induced acquisition of metastatic potential was mediated by increased fructose derived from the polyol pathway, which stimulated the nuclear ketohexokinase-A (KHK-A) signaling pathway, thereby inducing EMT by repressing the CDH1 gene. In two different xenograft models of cancer metastasis, gastric cancers overexpressing AKR1B1 were found to be highly metastatic in diabetic mice, but these effects of AKR1B1 were attenuated by KHK-A knockdown. In conclusion, hyperglycemia induces fructose formation through the polyol pathway, which in turn stimulates the KHK-A signaling pathway, driving gastric cancer metastasis by inducing EMT. Thus, the polyol and KHK-A signaling pathways could be potential therapeutic targets to decrease the metastatic risk in gastric cancer patients with diabetes.
Collapse
Affiliation(s)
- Ye-Lim Kang
- Department of Biomedical Science, BK21-Plus Education Program, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea
- Department of Pharmacology, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Jiyoung Kim
- Department of Biomedical Science, BK21-Plus Education Program, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea
- Department of Pharmacology, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Su-Bin Kwak
- Department of Biomedical Science, BK21-Plus Education Program, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea
- Department of Pharmacology, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Yi-Sook Kim
- Department of Biomedical Science, BK21-Plus Education Program, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea
- Department of Pharmacology, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - June Huh
- Department of Chemical and Biological Engineering, Korea University, Anam-ro, Seongbuk-gu, Seoul, 02841, Korea
| | - Jong-Wan Park
- Department of Biomedical Science, BK21-Plus Education Program, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea.
- Department of Pharmacology, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea.
- Cancer Research Institute and Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea.
| |
Collapse
|
39
|
Zhang C, Zhang Y, Dong Y, Zi R, Wang Y, Chen Y, Liu C, Wang J, Wang X, Li J, Liang H, Ou J. Non-alcoholic fatty liver disease promotes liver metastasis of colorectal cancer via fatty acid synthase dependent EGFR palmitoylation. Cell Death Discov 2024; 10:41. [PMID: 38263401 PMCID: PMC10805926 DOI: 10.1038/s41420-023-01770-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 12/02/2023] [Accepted: 12/07/2023] [Indexed: 01/25/2024] Open
Abstract
Liver metastasis is the major reason for most of colorectal cancer (CRC) related deaths. Accumulating evidence indicates that CRC patients with non-alcoholic fatty liver disease (NAFLD) are at a greater risk of developing liver metastasis. With the growing prevalence of NAFLD, a better understanding of the molecular mechanism in NAFLD-driven CRC liver metastasis is needed. In this study, we demonstrated that NAFLD facilitated CRC liver metastasis as a metabolic disorder and promoted the stemness of metastatic CRC cells for their colonization and outgrowth in hepatic niches. Metabolically, the lipid-rich microenvironment in NAFLD activated de novo palmitate biosynthesis in metastatic CRC cells via upregulating fatty acid synthase (FASN). Moreover, increased intracellular palmitate bioavailability promoted EGFR palmitoylation to enhance its protein stability and plasma membrane localization. Furthermore, we demonstrated that the FDA-approved FASN inhibitor orlistat could reduce NAFLD-activated endogenous palmitate production, thus inhibiting palmitoylation of EGFR to suppress CRC cell stemness and restrict liver metastasis in synergy with conventional chemotherapy. These findings reveal that the NAFLD metabolic microenvironment boosts endogenous palmitate biosynthesis in metastatic CRC cells and promotes cell stemness via EGFR palmitoylation, and FASN inhibitor orlistat could be a candidate adjuvant drug to suppress liver metastasis in CRC patients with NAFLD.
Collapse
Affiliation(s)
- Chi Zhang
- Department of Oncology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Yue Zhang
- Department of Oncology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Yan Dong
- Department of Oncology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Ruiyang Zi
- Department of Oncology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Yijie Wang
- Department of Oncology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Yanrong Chen
- Department of Oncology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Chengxiang Liu
- Department of Oncology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Junyi Wang
- Department of Oncology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Xuesong Wang
- Department of Oncology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Jianjun Li
- Department of Oncology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China.
| | - Houjie Liang
- Department of Oncology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China.
| | - Juanjuan Ou
- Department of Oncology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China.
- Jinfeng Laboratory, 401329, Chongqing, China.
| |
Collapse
|
40
|
Echeverría CE, Oyarzún VI, López-Cortés A, Cancino J, Sotomayor PC, Goncalves MD, Godoy AS. Biological role of fructose in the male reproductive system: Potential implications for prostate cancer. Prostate 2024; 84:8-24. [PMID: 37888416 PMCID: PMC10872645 DOI: 10.1002/pros.24631] [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: 03/03/2023] [Revised: 08/21/2023] [Accepted: 09/20/2023] [Indexed: 10/28/2023]
Abstract
BACKGROUND Over the last 20 years, fructose has gradually emerged as a potential metabolic substrate capable of promoting the growth and progression of various cancers, including prostate cancer (PCa). The biological and molecular mechanisms that underlie the effects of fructose on cancer are beginning to be elucidated. METHODS This review summarizes the biological function of fructose as a potential carbon source for PCa cells and its role in the functionality of the male reproductive tract under normal conditions. RESULTS The most recent biological advances related to fructose transport and metabolism as well as their implications in PCa growth and progression suggest that fructose represent a potential carbon source for PCa cells. Consequently, fructose derivatives may represent efficient radiotracers for obtaining PCa images via positron emission tomography and fructose transporters/fructose-metabolizing enzymes could be utilized as potential diagnostic and/or predictive biomarkers for PCa. CONCLUSION The existing data suggest that restriction of fructose from the diet could be a useful therapeutic strategy for patients with PCa.
Collapse
Affiliation(s)
- Carolina E. Echeverría
- Division of Endocrinology, Department of Medicine, Weill Cornell Medical, New York, NY, USA
| | - Vanessa I. Oyarzún
- Laboratory of Ocular and Systemic Autoimmune Diseases, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Andrés López-Cortés
- Cancer Research Group (CRG), Faculty of Medicine, Universidad de Las Américas, Quito, Ecuador
| | - Jorge Cancino
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Paula C. Sotomayor
- Departamento de Urología, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Marcus D. Goncalves
- Division of Endocrinology, Department of Medicine, Weill Cornell Medical, New York, NY, USA
| | - Alejandro S. Godoy
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo New York, USA
| |
Collapse
|
41
|
Pascual G, Majem B, Benitah SA. Targeting lipid metabolism in cancer metastasis. Biochim Biophys Acta Rev Cancer 2024; 1879:189051. [PMID: 38101461 DOI: 10.1016/j.bbcan.2023.189051] [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: 08/06/2023] [Revised: 11/29/2023] [Accepted: 12/04/2023] [Indexed: 12/17/2023]
Abstract
This review delves into the most recent research on the metabolic adaptability of cancer cells and examines how their metabolic functions can impact their progression into metastatic forms. We emphasize the growing significance of lipid metabolism and dietary lipids within the tumor microenvironment, underscoring their influence on tumor progression. Additionally, we present an outline of the interplay between metabolic processes and the epigenome of cancer cells, underscoring the importance regarding the metastatic process. Lastly, we examine the potential of targeting metabolism as a therapeutic approach in combating cancer progression, shedding light on innovative drugs/targets currently undergoing preclinical evaluation.
Collapse
Affiliation(s)
- Gloria Pascual
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
| | - Blanca Majem
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Salvador Aznar Benitah
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain; Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain.
| |
Collapse
|
42
|
Chen H, Zhai C, Xu X, Wang H, Han W, Shen J. Multilevel Heterogeneity of Colorectal Cancer Liver Metastasis. Cancers (Basel) 2023; 16:59. [PMID: 38201487 PMCID: PMC10778489 DOI: 10.3390/cancers16010059] [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: 11/03/2023] [Revised: 12/19/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
Colorectal cancer liver metastasis (CRLM) is a highly heterogeneous disease. Therapies that target both primary foci and liver metastasis are severely lacking. Therefore, understanding the features of metastatic tumor cells in the liver is valuable for the overall control of CRLM patients. In this review, we summarize the heterogeneity exhibited in CRLM from five aspects (gene, transcriptome, protein, metabolism, and immunity). In addition to genetic heterogeneity, the other four aspects exhibit significant heterogeneity. Compared to primary CRC, the dysregulation of epithelial-mesenchymal transition (EMT)-related proteins, the enhanced metabolic activity, and the increased infiltration of immunosuppressive cells are detected in CRLM. Preclinical evidence shows that targeting the EMT process or enhancing cellular metabolism may represent a novel approach to increasing the therapeutic efficacy of CRLM.
Collapse
Affiliation(s)
| | | | | | | | - Weidong Han
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China; (H.C.); (C.Z.); (X.X.); (H.W.)
| | - Jiaying Shen
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China; (H.C.); (C.Z.); (X.X.); (H.W.)
| |
Collapse
|
43
|
Cui Y, Tian J, Wang Z, Guo H, Zhang H, Wang Z, Liu H, Song W, Liu L, Tian R, Zuo X, Ren S, Niu R, Zhang F. Fructose-Induced mTORC1 Activation Promotes Pancreatic Cancer Progression through Inhibition of Autophagy. Cancer Res 2023; 83:4063-4079. [PMID: 37738413 PMCID: PMC10722142 DOI: 10.1158/0008-5472.can-23-0464] [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: 02/14/2023] [Revised: 08/02/2023] [Accepted: 09/19/2023] [Indexed: 09/24/2023]
Abstract
Excessive fructose intake is associated with the occurrence, progression, and poor prognosis of various tumors. A better understanding of the mechanisms underlying the functions of fructose in cancer could facilitate the development of better treatment and prevention strategies. In this study, we investigated the functional association between fructose utilization and pancreatic ductal adenocarcinoma (PDAC) progression. Fructose could be taken up and metabolized by PDAC cells and provided an adaptive survival mechanism for PDAC cells under glucose-deficient conditions. GLUT5-mediated fructose metabolism maintained the survival, proliferation, and invasion capacities of PDAC cells in vivo and in vitro. Fructose metabolism not only provided ATP and biomass to PDAC cells but also conferred metabolic plasticity to the cells, making them more adaptable to the tumor microenvironment. Mechanistically, fructose activated the AMP-activated protein kinase (AMPK)-mTORC1 signaling pathway to inhibit glucose deficiency-induced autophagic cell death. Moreover, the fructose-specific transporter GLUT5 was highly expressed in PDAC tissues and was an independent marker of disease progression in patients with PDAC. These findings provide mechanistic insights into the role of fructose in promoting PDAC progression and offer potential strategies for targeting metabolism to treat PDAC. SIGNIFICANCE Fructose activates AMPK-mTORC1 signaling to inhibit autophagy-mediated cell death in pancreatic cancer cells caused by glucose deficiency, facilitating metabolic adaptation to the tumor microenvironment and supporting tumor growth.
Collapse
Affiliation(s)
- Yanfen Cui
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Jianfei Tian
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Zhaosong Wang
- Laboratory Animal Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Hui Guo
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - He Zhang
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Zhiyong Wang
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Hui Liu
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Weijie Song
- Laboratory Animal Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Liming Liu
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Ruinan Tian
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Xiaoyan Zuo
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Sixin Ren
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Ruifang Niu
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Fei Zhang
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
44
|
Zhang Y, Yu X, Bao R, Huang H, Gu C, Lv Q, Han Q, Du X, Zhao XY, Ye Y, Zhao R, Sun J, Zou Q. Dietary fructose-mediated adipocyte metabolism drives antitumor CD8 + T cell responses. Cell Metab 2023; 35:2107-2118.e6. [PMID: 37863051 DOI: 10.1016/j.cmet.2023.09.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/14/2023] [Accepted: 09/25/2023] [Indexed: 10/22/2023]
Abstract
Fructose consumption is associated with tumor growth and metastasis in mice, yet its impact on antitumor immune responses remains unclear. Here, we show that dietary fructose modulates adipocyte metabolism to enhance antitumor CD8+ T cell immune responses and control tumor growth. Transcriptional profiling of tumor-infiltrating CD8+ T cells reveals that dietary fructose mediates attenuated transition of CD8+ T cells to terminal exhaustion, leading to a superior antitumor efficacy. High-fructose feeding initiates adipocyte-derived leptin production in an mTORC1-dependent manner, thereby triggering leptin-boosted antitumor CD8+ T cell responses. Importantly, high plasma leptin levels are correlated with elevated plasma fructose concentrations and improved antitumor CD8+ T cell responses in patients with lung cancer. Our study characterizes a critical role for dietary fructose in shaping adipocyte metabolism to prime antitumor CD8+ T cell responses and highlights that the fructose-leptin axis may be harnessed for cancer immunotherapy.
Collapse
Affiliation(s)
- Yuerong Zhang
- Shanghai Chest Hospital & Shanghai Institute of Immunology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Xiaoyan Yu
- Shanghai Chest Hospital & Shanghai Institute of Immunology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Rujuan Bao
- Shanghai Chest Hospital & Shanghai Institute of Immunology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Haiyan Huang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Chuanjia Gu
- Department of Respiratory Endoscopy, Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Qianming Lv
- Shanghai Chest Hospital & Shanghai Institute of Immunology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Qiaoqiao Han
- Shanghai Chest Hospital & Shanghai Institute of Immunology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Xian Du
- Shanghai Chest Hospital & Shanghai Institute of Immunology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Xu-Yun Zhao
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Youqiong Ye
- Shanghai Chest Hospital & Shanghai Institute of Immunology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China.
| | - Ren Zhao
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Jiayuan Sun
- Department of Respiratory Endoscopy, Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China.
| | - Qiang Zou
- Shanghai Chest Hospital & Shanghai Institute of Immunology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China.
| |
Collapse
|
45
|
Huang S, Wang X, Zhu Y, Wang Y, Chen J, Zheng H. SOX2 promotes vasculogenic mimicry by accelerating glycolysis via the lncRNA AC005392.2-GLUT1 axis in colorectal cancer. Cell Death Dis 2023; 14:791. [PMID: 38044399 PMCID: PMC10694132 DOI: 10.1038/s41419-023-06274-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 10/30/2023] [Accepted: 11/06/2023] [Indexed: 12/05/2023]
Abstract
Vasculogenic mimicry (VM), a new model of angiogenesis, fulfills the metabolic demands of solid tumors and contributes to tumor aggressiveness. Our previous study demonstrated the effect of SOX2 in promoting VM in colorectal cancer (CRC). However, the underlying mechanisms behind this effect remain elusive. Here, we show that SOX2 overexpression enhanced glycolysis and sustained VM formation via the transcriptional activation of lncRNA AC005392.2. Suppression of either glycolysis or AC005392.2 expression curbed SOX2-driven VM formation in vivo and in vitro. Mechanistically, SOX2 combined with the promoter of AC005392.2, which decreased H3K27me3 enrichment and thus increased its transcriptional activity. Overexpression of AC005392.2 increased the stability of GLUT1 protein by enhancing its SUMOylation, leading to a decrease in the ubiquitination and degradation of GLUT1. Accumulation of GLUT1 contributed to SOX2-mediated glycolysis and VM. Additionally, clinical analyses showed that increased levels of AC005392.2, GLUT1, and EPHA2 expression were positively correlated with SOX2 and were also associated with poor prognoses in patients with CRC. Our study conclusively demonstrates that the SOX2-lncRNA AC005392.2-GLUT1 signaling axis regulates VM formation in CRC, offering a foundation for the development of new antiangiogenic drugs or new drug combination regimens.
Collapse
Affiliation(s)
- Shimiao Huang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China
| | - Xuan Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China
| | - Yin Zhu
- School of Laboratory Medicine and Biotechnology, Southern Medical University, 510515, Guangzhou, China
| | - Yadong Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China
| | - Jiaxuan Chen
- State Key Laboratory of Organ Failure Research, Guangdong Key Laboratory of Viral Hepatitis Research, Guangdong Institute of Liver Diseases, Department of Infectious Diseases and Hepatology Unit, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China
| | - Haoxuan Zheng
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China.
| |
Collapse
|
46
|
Wu Z, Zhang X, An Y, Ma K, Xue R, Ye G, Du J, Chen Z, Zhu Z, Shi G, Ding X, Wan M, Jiang B, Zhang P, Liu J, Bu P. CLMP is a tumor suppressor that determines all-trans retinoic acid response in colorectal cancer. Dev Cell 2023; 58:2684-2699.e6. [PMID: 37944525 DOI: 10.1016/j.devcel.2023.10.006] [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: 03/30/2023] [Revised: 08/16/2023] [Accepted: 10/11/2023] [Indexed: 11/12/2023]
Abstract
CAR-like membrane protein (CLMP) is a tight junction-associated protein whose mutation is associated with congenital short bowel syndrome (CSBS), but its functions in colorectal cancer (CRC) remain unknown. Here, we demonstrate that CLMP is rarely mutated but significantly decreased in CRC patients, and its deficiency accelerates CRC tumorigenesis, growth, and resistance to all-trans retinoic acid (ATRA). Mechanistically, CLMP recruits β-catenin to cell membrane, independent of cadherin proteins. CLMP-mediated β-catenin translocation inactivates Wnt(Wingless and INT-1)/β-catenin signaling, thereby suppressing CRC tumorigenesis and growth in ApcMin/+, azoxymethane/dextran sodium sulfate (AOM/DSS), and orthotopic CRC mouse models. As a direct target of Wnt/β-catenin, cytochrome P450 hydroxylase A1 (CYP26A1)-an enzyme that degrades ATRA to a less bioactive retinoid-is upregulated by CLMP deficiency, resulting in ATRA-resistant CRC that can be reversed by administering CYP26A1 inhibitor. Collectively, our data identify the anti-CRC role of CLMP and suggest that CYP26A1 inhibitor enable to boost ATRA's therapeutic efficiency.
Collapse
Affiliation(s)
- Zhenzhen Wu
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xuanxuan Zhang
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yunhe An
- Institute of Analysis and Testing, Beijing Academy of Science and Technology (Beijing Center for Physical & Chemical Analysis), Beijing 100089, China
| | - Kaiyue Ma
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ruixin Xue
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Gaoqi Ye
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Junfeng Du
- Department of General Surgery, the 7(th) Medical Center, Chinese PLA General Hospital, Beijing 100700, China
| | - Zhiyong Chen
- Department of Radiation Oncology Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou 510080, China
| | - Zijing Zhu
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guizhi Shi
- Laboratory Animal Research Center, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiang Ding
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; Laboratory of Proteomics, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Meng Wan
- Laboratory Animal Research Center, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Bing Jiang
- Nanozyme Medical Center, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Peng Zhang
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, MOE Key Laboratory of Major Diseases in Children, Rare Disease Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China.
| | - Jinbo Liu
- Department of Colorectal Surgery of the 1(st) Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Pengcheng Bu
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
47
|
Yuan M, Zhang X, Yue F, Zhang F, Jiang S, Zhou X, Lv J, Zhang Z, Sun Y, Chen Z, Wu H, Liu X, Yu X, Wei B, Jiang K, Lin F, Zuo Y, Ren S. CircNOLC1 Promotes Colorectal Cancer Liver Metastasis by Interacting with AZGP1 and Sponging miR-212-5p to Regulate Reprogramming of the Oxidative Pentose Phosphate Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205229. [PMID: 37870214 PMCID: PMC10667818 DOI: 10.1002/advs.202205229] [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: 09/12/2022] [Revised: 08/27/2023] [Indexed: 10/24/2023]
Abstract
Liver metastasis is a common cause of death in progressive colorectal cancer patients, but the molecular mechanisms remain unclear. Here, it is reported that a conserved and oxidative pentose phosphate pathway-associated circular RNA, circNOLC1, plays a crucial role in colorectal cancer liver metastasis. It is found that circNOLC1 silencing reduces the oxidative pentose phosphate pathway-related intermediate metabolites and elevates NADP+ /NADPH ratio and intracellular ROS levels, thereby attenuating colorectal cancer cell proliferation, migration, and liver metastasis. circNOLC1 interacting with AZGP1 to activate mTOR/SREBP1 signaling, or sponging miR-212-5p to upregulate c-Met expression, both of which can further induce G6PD to activate oxidative pentose phosphate pathway in colorectal cancer liver metastasis. Moreover, circNOLC1 is regulated by the transcription factor YY1 and specifically stabilized HuR induces its parental gene mRNA expression. The associations between circNOLC1 and these signaling molecules are validated in primary CRC and corresponding liver metastasis tissues. These findings reveal that circNOLC1 interacting with AZGP1 and circNOLC1/miR-212-5p/c-Met axis plays a key role in oxidative pentose phosphate pathway-mediated colorectal cancer liver metastasis, which may provide a novel target for precision medicine of colorectal cancer.
Collapse
Affiliation(s)
- Menglang Yuan
- Department of General SurgeryThe Second Hospital of Dalian Medical University116023DalianChina
- Department of OncologySidney Kimmel Comprehensive Cancer CenterSchool of MedicineJohns Hopkins UniversityBaltimoreMD21287USA
| | - Xinsheng Zhang
- Department of General SurgeryThe Second Hospital of Dalian Medical University116023DalianChina
| | - Fangxia Yue
- Department of Clinical BiochemistryCollege of Laboratory Diagnostic MedicineDalian Medical University116044DalianChina
| | - Feifan Zhang
- Department of General SurgeryThe Second Hospital of Dalian Medical University116023DalianChina
| | - Sufen Jiang
- Department of Clinical BiochemistryCollege of Laboratory Diagnostic MedicineDalian Medical University116044DalianChina
| | - Xu Zhou
- Department of Clinical BiochemistryCollege of Laboratory Diagnostic MedicineDalian Medical University116044DalianChina
| | - Jinjuan Lv
- Department of Clinical BiochemistryCollege of Laboratory Diagnostic MedicineDalian Medical University116044DalianChina
| | - Zhenyu Zhang
- Department of Clinical BiochemistryCollege of Laboratory Diagnostic MedicineDalian Medical University116044DalianChina
| | - Yuzhu Sun
- Department of Clinical BiochemistryCollege of Laboratory Diagnostic MedicineDalian Medical University116044DalianChina
| | - Zihao Chen
- Department of General SurgeryThe Second Hospital of Dalian Medical University116023DalianChina
| | - Han Wu
- Department of Clinical BiochemistryCollege of Laboratory Diagnostic MedicineDalian Medical University116044DalianChina
| | - Xiaoqian Liu
- Department of Clinical BiochemistryCollege of Laboratory Diagnostic MedicineDalian Medical University116044DalianChina
| | - Xiaoqi Yu
- Department of Clinical BiochemistryCollege of Laboratory Diagnostic MedicineDalian Medical University116044DalianChina
| | - Bowen Wei
- Department of Clinical BiochemistryCollege of Laboratory Diagnostic MedicineDalian Medical University116044DalianChina
| | - Kexin Jiang
- Department of Clinical BiochemistryCollege of Laboratory Diagnostic MedicineDalian Medical University116044DalianChina
| | - Fang Lin
- Department of Clinical BiochemistryCollege of Laboratory Diagnostic MedicineDalian Medical University116044DalianChina
| | - Yunfei Zuo
- Department of Clinical BiochemistryCollege of Laboratory Diagnostic MedicineDalian Medical University116044DalianChina
| | - Shuangyi Ren
- Department of General SurgeryThe Second Hospital of Dalian Medical University116023DalianChina
| |
Collapse
|
48
|
Song A, Mao Y, Wei H. GLUT5: structure, functions, diseases and potential applications. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1519-1538. [PMID: 37674366 PMCID: PMC10582729 DOI: 10.3724/abbs.2023158] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 05/19/2023] [Indexed: 09/08/2023] Open
Abstract
Glucose transporter 5 (GLUT5) is a membrane transporter that specifically transports fructose and plays a key role in dietary fructose uptake and metabolism. In recent years, a high fructose diet has occupied an important position in the daily intake of human beings, resulting in a significant increase in the incidence of obesity and metabolic diseases worldwide. Over the past few decades, GLUT5 has been well understood to play a significant role in the pathogenesis of human digestive diseases. Recently, the role of GLUT5 in human cancer has received widespread attention, and a large number of studies have focused on exploring the effects of changes in GLUT5 expression levels on cancer cell survival, metabolism and metastasis. However, due to various difficulties and shortcomings, the molecular structure and mechanism of GLUT5 have not been fully elucidated, which to some extent prevents us from revealing the relationship between GLUT5 expression and cell carcinogenesis at the protein molecular level. In this review, we summarize the current understanding of the structure and function of mammalian GLUT5 and its relationship to intestinal diseases and cancer and suggest that GLUT5 may be an important target for cancer therapy.
Collapse
Affiliation(s)
- Aqian Song
- Department of GastroenterologyBeijing Ditan HospitalCapital Medical UniversityBeijing100015China
| | - Yuanpeng Mao
- Department of GastroenterologyPeking University Ditan Teaching HospitalBeijing100015China
| | - Hongshan Wei
- Department of GastroenterologyBeijing Ditan HospitalCapital Medical UniversityBeijing100015China
- Department of GastroenterologyPeking University Ditan Teaching HospitalBeijing100015China
| |
Collapse
|
49
|
Chu YD, Cheng LC, Lim SN, Lai MW, Yeh CT, Lin WR. Aldolase B-driven lactagenesis and CEACAM6 activation promote cell renewal and chemoresistance in colorectal cancer through the Warburg effect. Cell Death Dis 2023; 14:660. [PMID: 37816733 PMCID: PMC10564793 DOI: 10.1038/s41419-023-06187-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 09/13/2023] [Accepted: 09/28/2023] [Indexed: 10/12/2023]
Abstract
Colorectal cancer (CRC) is a prevalent malignancy worldwide and is associated with a high mortality rate. Changes in bioenergy metabolism, such as the Warburg effect, are often observed in CRC. Aldolase B (ALDOB) has been identified as a potential regulator of these changes, but its exact role in CRC cell behavior and bioenergetic homeostasis is not fully understood. To investigate this, two cohorts of CRC patients were analyzed independently. The results showed that higher ALDOB expression was linked to unfavorable prognosis, increased circulating carcinoembryonic antigen (CEA) levels, and altered bioenergetics in CRC. Further analysis using cell-based assays demonstrated that ALDOB promoted cell proliferation, chemoresistance, and increased expression of CEA in CRC cells. The activation of pyruvate dehydrogenase kinase-1 (PDK1) by ALDOB-induced lactagenesis and secretion, which in turn mediated the effects on CEA expression. Secreted lactate was found to enhance lactate dehydrogenase B (LDHB) expression in adjacent cells and to be a crucial modulator of ALDOB-mediated phenotypes. Additionally, the effect of ALDOB on CEA expression was downstream of the bioenergetic changes mediated by secreted lactate. The study also identified CEA cell adhesion molecule-6 (CEACAM6) as a downstream effector of ALDOB that controlled CRC cell proliferation and chemoresistance. Notably, CEACAM6 activation was shown to enhance protein stability through lysine lactylation, downstream of ALDOB-mediated lactagenesis. The ALDOB/PDK1/lactate/CEACAM6 axis plays an essential role in CRC cell behavior and bioenergetic homeostasis, providing new insights into the involvement of CEACAM6 in CRC and the Warburg effect. These findings may lead to the development of new treatment strategies for CRC patients.
Collapse
Affiliation(s)
- Yu-De Chu
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan
| | - Li-Chun Cheng
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan
| | - Siew-Na Lim
- Department of Neurology, Linkou Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan
| | - Ming-Wei Lai
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan
- Division of Pediatric Gastroenterology Department of Pediatrics, Linkou Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, 333, Taiwan
| | - Chau-Ting Yeh
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan.
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, 333, Taiwan.
- College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan.
- Department of Hepatology and Gastroenterology, Linkou Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan.
| | - Wey-Ran Lin
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan.
- College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan.
- Department of Hepatology and Gastroenterology, Linkou Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan.
| |
Collapse
|
50
|
He K, Wang Z, Luo M, Li B, Ding N, Li L, He B, Wang H, Cao J, Huang C, Yang J, Chen HN. Metastasis organotropism in colorectal cancer: advancing toward innovative therapies. J Transl Med 2023; 21:612. [PMID: 37689664 PMCID: PMC10493031 DOI: 10.1186/s12967-023-04460-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 08/19/2023] [Indexed: 09/11/2023] Open
Abstract
Distant metastasis remains a leading cause of mortality among patients with colorectal cancer (CRC). Organotropism, referring to the propensity of metastasis to target specific organs, is a well-documented phenomenon in CRC, with the liver, lungs, and peritoneum being preferred sites. Prior to establishing premetastatic niches within host organs, CRC cells secrete substances that promote metastatic organotropism. Given the pivotal role of organotropism in CRC metastasis, a comprehensive understanding of its molecular underpinnings is crucial for biomarker-based diagnosis, innovative treatment development, and ultimately, improved patient outcomes. In this review, we focus on metabolic reprogramming, tumor-derived exosomes, the immune system, and cancer cell-organ interactions to outline the molecular mechanisms of CRC organotropic metastasis. Furthermore, we consider the prospect of targeting metastatic organotropism for CRC therapy.
Collapse
Affiliation(s)
- Kai He
- School of Basic Medical Sciences and State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Zhihan Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Maochao Luo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Bowen Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Ning Ding
- School of Basic Medical Sciences and State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Lei Li
- School of Basic Medical Sciences and State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Bo He
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Han Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Jiangjun Cao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Canhua Huang
- School of Basic Medical Sciences and State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Jun Yang
- Department of Oncology, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China.
| | - Hai-Ning Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
- Department of General Surgery, State Key Laboratory of Biotherapy and Cancer Center, Colorectal Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China.
| |
Collapse
|