1
|
Fang H, Yu E, Liu C, Eapen C, Cheng C, Hu T. Metabolic landscape and rewiring in normal hematopoiesis, leukemia and aging. Semin Cancer Biol 2025; 111:1-15. [PMID: 39933639 DOI: 10.1016/j.semcancer.2025.02.003] [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: 10/23/2024] [Revised: 01/06/2025] [Accepted: 02/04/2025] [Indexed: 02/13/2025]
Abstract
Recent advancements in metabolism research have demonstrated its critical roles in a lot of critical biological processes, including stemness maintenance, cell differentiation, proliferation, and function. Hematopoiesis is the fundamental cell differentiation process with the production of millions of red blood cells per second in carrying oxygen and white blood cells in fighting infection and cancers. The differentiation processes of hematopoietic stem and progenitor cells (HSPCs) are accompanied by significant metabolic reprogramming. In hematological malignancy, metabolic reprogramming is also essential to the malignant hematopoiesis processes. The metabolic rewiring is driven by distinct molecular mechanisms that meet the specific demands of different target cells. Leukemic cells, for instance, adopt unique metabolic profiles to support their heightened energy needs for survival and proliferation. Moreover, aging HSPCs exhibit altered energy consumption compared to their younger counterparts, often triggering protective mechanisms at the cellular level. In this review, we provide a comprehensive analysis of the metabolic processes involved in hematopoiesis and the metabolic rewiring that occurs under adverse conditions. In addition, we highlight current research directions and discuss the potential of targeting metabolic pathways for the management of hematological malignancies and aging.
Collapse
Affiliation(s)
- Hui Fang
- Georgia Cancer Center, 1410 Laney Walker Blvd, Augusta, GA 30912, United States; Department of Stomatology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Enze Yu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa 999078, Macao
| | - Chang Liu
- Georgia Cancer Center, 1410 Laney Walker Blvd, Augusta, GA 30912, United States; Department of Stomatology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Christy Eapen
- Georgia Cancer Center, 1410 Laney Walker Blvd, Augusta, GA 30912, United States
| | - Chunming Cheng
- Stephenson Cancer Center at Oklahoma University, Oklahoma City, OK 73104, United States.
| | - Tianxiang Hu
- Georgia Cancer Center, 1410 Laney Walker Blvd, Augusta, GA 30912, United States.
| |
Collapse
|
2
|
Sekimoto K, Kinjo H, Murakami M, Ohashi A, Fukui R, Nagasaki-Maeoka E, Inagaki Y, Takayama T, Ikeda K, Takayama KI, Inoue S, Tsuji M, Otsuki J, Fujiwara K. Effects of the number of ethylene glycol units on the efficacy of novel complex I inhibitor 9bw. Biochem Biophys Rep 2025; 42:101981. [PMID: 40207083 PMCID: PMC11979437 DOI: 10.1016/j.bbrep.2025.101981] [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: 12/27/2024] [Revised: 03/14/2025] [Accepted: 03/14/2025] [Indexed: 04/11/2025] Open
Abstract
4'-Iodobiphenyl nonaethylene glycol ether (9bw) is a novel small molecule, composed of a biphenyl unit and 9 ethylene glycol (EG) units. Recently, we found that 9bw induces apoptosis in cancer cells by inhibiting mitochondrial respiratory complex I (CI) and accordingly reducing cellular ATP level. In addition, 9bw shows little effect on normal cells, suggesting that 9bw is a potential antitumor agent with few adverse effects. However, the exact molecular mechanisms by which 9bw acts on CI are still elusive. To clarify the molecular structure critical for 9bw's function, we tested the function of 9bw analogues on human oral squamous cell carcinoma lines HSC4 and Ca9-22. The analogues were 4-hydroxy-4'-iodobiphenyl (HIOP), I-BP-EG3, I-BP-EG6, and I-BP-EG12 containing 0, 3, 6, and 12 EG units, respectively. Our results demonstrated that I-BP-EG6 and I-BP-EG12 inhibited CI to a similar extent as 9bw, whereas I-BP3 and HIOP showed no effect on CI activity. These observations indicate that the number of EG units is crucial for the activity of 9bw and its analogues. As high-performance liquid chromatography (HPLC) analysis demonstrated that both HIOP and I-BP-EG3 could be incorporated into mitochondria abundantly, the number of EG units probably affects CI inhibitory function of 9bw and its analogues rather than their efficacy to enter cell and mitochondria.
Collapse
Affiliation(s)
- Kazuaki Sekimoto
- Department of Anatomy, Nihon University School of Dentistry, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Hanaka Kinjo
- Department of Materials and Applied Chemistry, College of Science and Technology, Nihon University, Chiyoda-ku, Tokyo, 101-8308, Japan
| | - Mizuki Murakami
- Department of Materials and Applied Chemistry, College of Science and Technology, Nihon University, Chiyoda-ku, Tokyo, 101-8308, Japan
| | - Akiko Ohashi
- Department of Anatomy, Nihon University School of Dentistry, Chiyoda-ku, Tokyo, 101-8310, Japan
- Division of Functional Morphology, Dental Research Center, Nihon University School of Dentistry, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Rei Fukui
- Department of Pathology, Nihon University School of Dentistry, Chiyoda-ku, 101-8310, Japan
| | - Eri Nagasaki-Maeoka
- Department of Pediatric Surgery, Jichi Medical University, Saitama Medical Center, Saitama, 330-8503, Japan
| | - Yoshinori Inagaki
- Division of General Medicine, Department of Medicine, Nihon University School of Medicine, Itabashi-ku, Tokyo, 173-0032, Japan
| | - Tadateru Takayama
- Division of General Medicine, Department of Medicine, Nihon University School of Medicine, Itabashi-ku, Tokyo, 173-0032, Japan
| | - Kazuhiro Ikeda
- Division of Systems Medicine & Gene Therapy, Saitama Medical University, Hidaka, Saitama, 350-1241, Japan
| | - Ken-ichi Takayama
- Department of Systems Aging Science and Medicine, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Satoshi Inoue
- Division of Systems Medicine & Gene Therapy, Saitama Medical University, Hidaka, Saitama, 350-1241, Japan
- Department of Systems Aging Science and Medicine, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Motonori Tsuji
- Institute of Molecular Function, 2-105-14, Takasu, Misato-shi, Saitama, 341-0037, Japan
| | - Joe Otsuki
- Department of Materials and Applied Chemistry, College of Science and Technology, Nihon University, Chiyoda-ku, Tokyo, 101-8308, Japan
| | - Kyoko Fujiwara
- Department of Anatomy, Nihon University School of Dentistry, Chiyoda-ku, Tokyo, 101-8310, Japan
- Division of Functional Morphology, Dental Research Center, Nihon University School of Dentistry, Chiyoda-ku, Tokyo, 101-8310, Japan
| |
Collapse
|
3
|
Zhang L, Luo X, Wu D, Zhou Q, Qiu J, Yan M, Wang Y. CD40 promotes AML survival via non-canonical NF-κB signaling and aberrant lipid metabolism. Int Immunopharmacol 2025; 156:114665. [PMID: 40252467 DOI: 10.1016/j.intimp.2025.114665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/27/2025] [Accepted: 04/09/2025] [Indexed: 04/21/2025]
Abstract
Despite the identification of several pathogenic drivers, the molecular mechanisms underlying the development of acute myeloid leukemia (AML) remain largely unknown. Therefore, we sought to explore the key genes associated with leukemia and identified cluster of differentiation 40 (CD40) as a key mediator linked to the incidence and progression of AML. Higher levels of CD40 were detected in patients with AML compared to healthy donors. Moreover, elevated CD40 expression was associated with lower overall survival rates. Furthermore, anti-CD40 antibody significantly induced apoptosis and enhanced drug sensitivity in human AML cell lines. Conversely, ex vivo treatment of primary AML samples with a CD40 agonist significantly decreased cell apoptosis and drug sensitivity. In Kasumi-1 AML cells, CD40 knockout (KO) significantly impaired the engraftment ability of leukemia cells and reduced the leukemia burden in NSG mice compared to wild-type mice. RNA sequencing showed that differentially expressed genes were significantly enriched in the nuclear factor-kB (NF-kB) signaling pathway in CD40-KO cells, which was confirmed through Western blotting. Untargeted metabolomic analysis revealed 179 metabolites with differential expression between WT and CD40 KO cells. Subsequent analysis revealed significant changes in the main metabolic pathways, particularly the biosynthesis of unsaturated fatty acids and lipid metabolism. A targeted metabolomics study of fatty acid metabolism demonstrated that cis-5, 8, 11, 14, 17-eicosapentaenoic acid (EPA) was markedly downregulated in CD40-KO cells compared to wild-type cells. Remarkably, EPA reversed the apoptosis and cell cycle arrest induced by CD40 deletion, simultaneously reducing the drug sensitivity of CD40-KO cells. Together, our study highlights the potential of CD40 as a target in the treatment of AML.
Collapse
Affiliation(s)
- Li Zhang
- Department of Hematology, Guangzhou Women and Children's Medical Center, GMU-GIBH Joint School of Life Sciences of Guangzhou Medical University, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, China
| | - Xin Luo
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, China
| | - Dongyan Wu
- Department of Hematology, Guangzhou Women and Children's Medical Center, GMU-GIBH Joint School of Life Sciences of Guangzhou Medical University, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, China
| | - Qiang Zhou
- The People's Hospital of Le'an County, Fuzhou, China
| | - Jiachun Qiu
- Department of Hematology, Guangzhou Women and Children's Medical Center, GMU-GIBH Joint School of Life Sciences of Guangzhou Medical University, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, China
| | - Muxia Yan
- Department of Hematology, Guangzhou Women and Children's Medical Center, GMU-GIBH Joint School of Life Sciences of Guangzhou Medical University, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, China
| | - Yiqian Wang
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
4
|
Addanki S, Kim L, Stevens A. Understanding and Targeting Metabolic Vulnerabilities in Acute Myeloid Leukemia: An Updated Comprehensive Review. Cancers (Basel) 2025; 17:1355. [PMID: 40282531 PMCID: PMC12025543 DOI: 10.3390/cancers17081355] [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: 02/21/2025] [Revised: 04/05/2025] [Accepted: 04/15/2025] [Indexed: 04/29/2025] Open
Abstract
Acute Myeloid Leukemia (AML) is characterized by aggressive proliferation and metabolic reprogramming that support its survival and resistance to therapy. This review explores the metabolic distinctions between AML cells and normal hematopoietic stem cells (HSCs), emphasizing the role of altered mitochondrial function, oxidative phosphorylation (OXPHOS), and biosynthetic pathways in leukemic progression. AML cells exhibit distinct metabolic vulnerabilities, including increased mitochondrial biogenesis, reliance on glycolysis and amino acid metabolism, and unique signaling interactions that sustain leukemic stem cells (LSCs). These dependencies provide potential therapeutic targets, as metabolic inhibitors have demonstrated efficacy in disrupting AML cell survival while sparing normal hematopoietic cells. We examine current and emerging metabolic therapies, such as inhibitors targeting glycolysis, amino acid metabolism, and lipid biosynthesis, highlighting their potential in overcoming drug resistance. However, challenges remain in translating these strategies into clinical practice due to AML's heterogeneity and adaptability. Further research into AML's metabolic plasticity and precision medicine approaches is crucial for improving treatment outcomes. Understanding and exploiting AML's metabolic vulnerabilities could pave the way for novel, more effective therapeutic strategies.
Collapse
Affiliation(s)
- Sridevi Addanki
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Alexandra Stevens
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
5
|
Deng R, Fu L, Liang H, Ai X, Liu F, Li N, Wu L, Li S, Yang X, Lin Y, Huang Y, Yun J. Inhibition of mitochondrial complex I induces mitochondrial ferroptosis by regulating CoQH2 levels in cancer. Cell Death Dis 2025; 16:254. [PMID: 40185704 PMCID: PMC11971431 DOI: 10.1038/s41419-025-07510-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 02/16/2025] [Accepted: 03/10/2025] [Indexed: 04/07/2025]
Abstract
Ferroptosis, a novel form of regulated cell death induced by the excessive accumulation of lipid peroxidation products, plays a pivotal role in the suppression of tumorigenesis. Two prominent mitochondrial ferroptosis defense systems are glutathione peroxidase 4 (GPX4) and dihydroorotate dehydrogenase (DHODH), both of which are localized within the mitochondria. However, the existence of supplementary cellular defense mechanisms against mitochondrial ferroptosis remains unclear. Our findings unequivocally demonstrate that inactivation of mitochondrial respiratory chain complex I (MCI) induces lipid peroxidation and consequently invokes ferroptosis across GPX4 low-expression cancer cells. However, in GPX4 high expression cancer cells, the MCI inhibitor did not induce ferroptosis, but increased cell sensitivity to ferroptosis induced by the GPX4 inhibitor. Overexpression of the MCI alternative protein yeast NADH-ubiquinone reductase (NDI1) not only quells ferroptosis induced by MCI inhibitors but also confers cellular protection against ferroptosis inducers. Mechanically, MCI inhibitors actuate an elevation in the NADH level while concomitantly diminishing the CoQH2 level. The manifestation of MCI inhibitor-induced ferroptosis can be reversed by supplementation with mitochondrial-specific analogues of CoQH2. Notably, MCI operates in parallel with mitochondrial-localized GPX4 and DHODH to inhibit mitochondrial ferroptosis, but independently of cytosolically localized GPX4 or ferroptosis suppressor protein 1(FSP1). The MCI inhibitor IACS-010759, is endowed with the ability to induce ferroptosis while concurrently impeding tumor proliferation in vivo. Our results identified a ferroptosis defense mechanism mediated by MCI within the mitochondria and suggested a therapeutic strategy for targeting ferroptosis in cancer treatment.
Collapse
Affiliation(s)
- Ru Deng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Radiation Oncology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Lingyi Fu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Haoyu Liang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xixiong Ai
- Reproductive Medicine Center, The Affiliated Shenzhen Maternity and Child Healthcare Hospital of the South Medical University, Shenzhen, Guangdong, China
| | - Fangyi Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Nai Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Liyan Wu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Shuo Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xia Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yansong Lin
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yuhua Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China.
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China.
| | - Jingping Yun
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China.
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
6
|
Cai L, Hammond NG, Tasdogan A, Alsamraae M, Yang C, Cameron RB, Quan P, Solmonson A, Gu W, Pachnis P, Kaur M, Chang BK, Zhou Q, Hensley CT, Do QN, Martins Nascentes Melo L, Ubellacker JM, Kaushik A, Clare MG, Alcazar IN, Kurylowicz K, Marcuccilli JD, Allies G, Kutritz A, Klode J, Ramesh V, Rogers TJ, Rao AD, Crentsil HE, Li H, Brister F, McDaniel P, Xu X, Evers BM, Zacharias LG, Sudderth J, Xu J, Mathews TP, Oliver D, Minna JD, Waters J, Morrison SJ, Kernstine KH, Faubert B, DeBerardinis RJ. High Glucose Contribution to the TCA Cycle Is a Feature of Aggressive Non-Small Cell Lung Cancer in Patients. Cancer Discov 2025; 15:702-716. [PMID: 39960461 PMCID: PMC11962397 DOI: 10.1158/2159-8290.cd-23-1319] [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: 11/07/2023] [Revised: 11/22/2024] [Accepted: 01/23/2025] [Indexed: 03/01/2025]
Abstract
SIGNIFICANCE Intraoperative 13C-glucose infusions in patients with NSCLC show that tumors with high labeling of TCA cycle intermediates progress rapidly, resulting in metastasis and early death. Blocking this pathway suppresses metastasis of human NSCLC cells in mice.
Collapse
Affiliation(s)
- Ling Cai
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas
- Quantitative Biomedical Research Center, Peter O’Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Nia G. Hammond
- Section of Hematology and Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Alpaslan Tasdogan
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Dermatology, University Hospital Essen & German Cancer Consortium, Essen, Germany
| | - Massar Alsamraae
- Section of Hematology and Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Chendong Yang
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Robert B. Cameron
- Section of Hematology and Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Peiran Quan
- Quantitative Biomedical Research Center, Peter O’Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ashley Solmonson
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Wen Gu
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Panayotis Pachnis
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Mayher Kaur
- Section of Hematology and Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Brianna K. Chang
- Section of Hematology and Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Qin Zhou
- Quantitative Biomedical Research Center, Peter O’Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Christopher T. Hensley
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Quyen N. Do
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | - Jessalyn M. Ubellacker
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Akash Kaushik
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Maia G. Clare
- Section of Hematology and Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Isabel N. Alcazar
- Section of Hematology and Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Katarzyna Kurylowicz
- Section of Hematology and Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Joseph D. Marcuccilli
- Section of Hematology and Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Gabriele Allies
- Department of Dermatology, University Hospital Essen & German Cancer Consortium, Essen, Germany
| | - Andrea Kutritz
- Department of Dermatology, University Hospital Essen & German Cancer Consortium, Essen, Germany
| | - Joachim Klode
- Department of Dermatology, University Hospital Essen & German Cancer Consortium, Essen, Germany
| | - Vijayashree Ramesh
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Thomas J. Rogers
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Aparna D. Rao
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Hannah E. Crentsil
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Hong Li
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas
- Clinical Research Unit, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Fang Brister
- Clinical Research Unit, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Phyllis McDaniel
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas
- Clinical Research Unit, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Xiaohong Xu
- Clinical Research Unit, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Bret M. Evers
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Lauren G. Zacharias
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jessica Sudderth
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jian Xu
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Thomas P. Mathews
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Dwight Oliver
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - John D. Minna
- Hamon Center for Therapeutic Oncology Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - John Waters
- Department of Cardiovascular and Thoracic Surgery, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Sean J. Morrison
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Kemp H. Kernstine
- Department of Cardiovascular and Thoracic Surgery, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Brandon Faubert
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas
- Section of Hematology and Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Ralph J. DeBerardinis
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
7
|
Gopalkrishnan A, Wang N, Cruz-Rangel S, Yassin-Kassab A, Shiva S, Kurukulasuriya C, Monga SP, DeBerardinis RJ, Skinner HD, Kiselyov K, Duvvuri U. Lysosomal-Mitochondrial Interaction Promotes Tumor Growth in Squamous Cell Carcinoma of the Head and Neck. Mol Cancer Res 2025; 23:339-349. [PMID: 39699311 PMCID: PMC11961326 DOI: 10.1158/1541-7786.mcr-24-0337] [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: 04/14/2024] [Revised: 05/26/2024] [Accepted: 12/16/2024] [Indexed: 12/20/2024]
Abstract
Communication between intracellular organelles including lysosomes and mitochondria has recently been shown to regulate cellular proliferation and fitness. The way lysosomes and mitochondria communicate with each other [lysosomal-mitochondrial interaction (LMI)] is emerging as a major determinant of tumor proliferation and growth. About 30% of squamous carcinomas [including squamous cell carcinoma of the head and neck (SCCHN)] overexpress transmembrane member 16A (TMEM16A), a calcium-activated chloride channel, which promotes cellular growth and negatively correlates with patient survival. We have recently shown that TMEM16A drives lysosomal biogenesis; however, its impact on mitochondrial function has not been explored. In this study, we show that in the context of high-TMEM16A SCCHN, (i) patients display increased mitochondrial content, specifically complex I; (ii) in vitro and in vivo models uniquely depend on mitochondrial complex I activity for growth and survival; (iii) NRF2 signaling is a critical linchpin that drives mitochondrial function, and (iv) mitochondrial complex I and lysosomal function are codependent for proliferation. Taken together, our data demonstrate that coordinated lysosomal and mitochondrial activity and biogenesis via LMI drive tumor proliferation and facilitate a functional interaction between lysosomal and mitochondrial networks. Therefore, inhibition of LMI instauration may serve as a therapeutic strategy for patients with SCCHN. Implications: Intervention of LMI may serve as a therapeutic approach for patients with high TMEM16A-expressing SCCHN.
Collapse
Affiliation(s)
- Avani Gopalkrishnan
- Department of Radiation Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- UPMC Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Nathaniel Wang
- Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- UPMC Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Silvia Cruz-Rangel
- Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- UPMC Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Abdul Yassin-Kassab
- Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- UPMC Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Sruti Shiva
- Dept of Pharmacology and Chemical Biology, Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | | | - Satdarshan P. Monga
- Division of Experimental Pathology, Department of Pathology, Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Ralph J DeBerardinis
- Children’s Medical Research Institute and Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Heath D. Skinner
- Department of Radiation Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- UPMC Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Kirill Kiselyov
- Department of Biological Sciences, University of Pittsburgh, PA
| | - Umamaheswar Duvvuri
- Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Department of Otolaryngology-Head and Neck Surgery, NYU Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
8
|
Varney SD, Erkes DA, Mersky GL, Mustafa MU, Chua V, Chervoneva I, Purwin TJ, Alnemri E, Aplin AE. Metabolic Inhibition Induces Pyroptosis in Uveal Melanoma. Mol Cancer Res 2025; 23:350-362. [PMID: 39670827 PMCID: PMC11961327 DOI: 10.1158/1541-7786.mcr-24-0508] [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: 06/05/2024] [Revised: 11/02/2024] [Accepted: 12/11/2024] [Indexed: 12/14/2024]
Abstract
Few treatment options are available for patients with metastatic uveal melanoma. Although the bispecific tebentafusp is FDA approved, immunotherapy has largely failed, likely given the poorly immunogenic nature of uveal melanoma. Treatment options that improve the recognition of uveal melanoma by the immune system may be key to reducing disease burden. We investigated whether uveal melanoma has the ability to undergo pyroptosis, a form of immunogenic cell death. Publicly available patient data and cell line analysis showed that uveal melanoma expressed the machinery needed for pyroptosis, including gasdermins D and E (GSDMD and E), caspases 1, 3, 4, and 8, and ninjurin-1. We induced cleavage of GSDMs in uveal melanoma cell lines treated with metabolic inhibitors. In particular, the carnitine palmitoyltransferase 1 (CPT1) inhibitor, etomoxir, induced propidium iodide uptake, caspase 3 cleavage, and the release of HMGB1 and IL-1β, indicating that the observed cleavage of GSDMs led to pyroptosis. Importantly, a gene signature reflecting CPT1A activity correlated with poor prognosis in patients with uveal melanoma and knockdown of CPT1A also induced pyroptosis. Etomoxir-induced pyroptosis was dependent on GSDME but not on GSDMD, and a pyroptosis gene signature correlated with immune infiltration and improved response to immune checkpoint blockade in a set of patients with uveal melanoma. Together, these data show that metabolic inhibitors can induce pyroptosis in uveal melanoma cell lines, potentially offering an approach to enhance inflammation-mediated immune targeting in patients with metastatic uveal melanoma. Implications: Induction of pyroptosis by metabolic inhibition may alter the tumor immune microenvironment and improve the efficacy of immunotherapy in uveal melanoma.
Collapse
Affiliation(s)
- Scott D. Varney
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107 USA
| | - Dan A. Erkes
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107 USA
| | - Glenn L. Mersky
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107 USA
| | - Manal U. Mustafa
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107 USA
| | - Vivian Chua
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107 USA
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Perth, WA 6027, Australia
- Centre for Precision Health, Edith Cowan University, Joondalup, Perth, WA 6027, Australia
| | - Inna Chervoneva
- Division of Biostatistics, Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Timothy J. Purwin
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107 USA
| | - Emad Alnemri
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107 USA
- Sidney Kimmel Comprehensive Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107 USA
| | - Andrew E. Aplin
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107 USA
- Sidney Kimmel Comprehensive Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107 USA
| |
Collapse
|
9
|
Shangguan F, Ma N, Chen Y, Zheng Y, Ma T, An J, Lin J, Yang H. Fucoxanthin suppresses pancreatic cancer progression by inducing bioenergetics metabolism crisis and promoting SLC31A1‑mediated sensitivity to DDP. Int J Oncol 2025; 66:31. [PMID: 40052552 PMCID: PMC11900939 DOI: 10.3892/ijo.2025.5737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 01/16/2025] [Indexed: 03/14/2025] Open
Abstract
Pancreatic cancer (PC) is one of the most malignant tumors, with a 5‑year survival rate <10%. Chemosynthetic drugs are widely used in the treatment of PC; however, their toxicity and side effects often reduce the quality of life for patients. MTT and colony formation assay were performed to detect cell growth and viability in PC cells. Levels of ROS in whole cell and mitochondria were analyzed through flow cytometry. ATP production was evaluated using an ATP Assay Kit. Cellular bioenergetics were analyzed with a Seahorse XFe96 Analyzer, and changes in target molecules were monitored by western blotting. The present study reports that fucoxanthin (FX), a carotenoid derived from aquatic brown seaweed, significantly inhibits PC by inhibiting cell proliferation and inducing cell death via the non‑classical pathway. FX switches mitochondrial respiration to aerobic glycolysis in PC cells. Furthermore, FX decreases whole‑cell ATP levels, which indicates that promotion of glycolysis does not compensate for FX‑induced ATP depletion in mitochondria. Moreover, FX decreased the reduced glutathione/oxidized glutathione ratio observed under glucose‑limited conditions. These alterations caused by FX may decrease metabolic flexibility, indicating higher sensitivity to glucose‑limited (GL) conditions. FX increased the cytotoxicity of cisplatin (DDP) and the expression of solute carrier family 31 member 1 (SLC31A1) in PC cells. Furthermore, the knockdown of SLC31A1 can attenuate cytotoxicity caused by the combination of FX and DDP. It was inferred that FX increased the sensitivity of PC cells to DDP), potentially by upregulating SLC31A1 expression. In conclusion, FX exhibited potent antitumor effects by reprogramming energy metabolism and inducing a distinct form of regulated cell death. Therefore, combining FX with GL treatment or DDP presents a promising therapeutic strategy for PC.
Collapse
Affiliation(s)
- Fugen Shangguan
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Nengfang Ma
- College of Life and Environmental Science, Wenzhou University, Wenzhou, Zhejiang 325000, P.R. China
| | - Yang Chen
- College of Life and Environmental Science, Wenzhou University, Wenzhou, Zhejiang 325000, P.R. China
| | - Yuansi Zheng
- Department of Pathology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Ting Ma
- College of Life and Environmental Science, Wenzhou University, Wenzhou, Zhejiang 325000, P.R. China
| | - Jing An
- Division of Infectious Diseases and Global Health, School of Medicine, University of California San Diego, La Jolla, CA 92037, USA
| | - Jianhu Lin
- Department of Trauma Surgery and Emergency Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Hailong Yang
- College of Life and Environmental Science, Wenzhou University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
10
|
Woytash JA, Kumar R, Chaudhary AK, Donnelly C, Wojtulski A, Bethu M, Wang J, Spernyak J, Bross P, Yadav N, Inigo JR, Chandra D. Mitochondrial unfolded protein response-dependent β-catenin signaling promotes neuroendocrine prostate cancer. Oncogene 2025; 44:820-834. [PMID: 39690273 DOI: 10.1038/s41388-024-03261-4] [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: 06/19/2024] [Revised: 12/02/2024] [Accepted: 12/09/2024] [Indexed: 12/19/2024]
Abstract
The mitochondrial unfolded protein response (UPRmt) maintains mitochondrial quality control and proteostasis under stress conditions. However, the role of UPRmt in aggressive and resistant prostate cancer is not clearly defined. We show that castration-resistant neuroendocrine prostate cancer (CRPC-NE) harbored highly dysfunctional oxidative phosphorylation (OXPHOS) Complexes. However, biochemical and protein analyses of CRPC-NE tumors showed upregulation of nuclear-encoded OXPHOS proteins and UPRmt in this lethal subset of prostate cancer suggestive of compensatory upregulation of stress signaling. Genetic deletion and pharmacological inhibition of the main chaperone of UPRmt heat shock protein 60 (HSP60) reduced neuroendocrine prostate cancer (NEPC) growth in vivo as well as reverted NEPC cells to a more epithelial-like state. HSP60-dependent aggressive NEPC phenotypes was associated with upregulation of β-catenin signaling both in cancer cells and in vivo tumors. HSP60 expression rendered enrichment of aggressive prostate cancer signatures and metastatic potential were inhibited upon suppression of UPRmt. We discovered that UPRmt promoted OXPHOS functions including mitochondrial bioenergetics in CRPC-NE via regulation of β-catenin signaling. Mitochondrial biogenesis facilitated cisplatin resistance and inhibition of UPRmt resensitizes CRPC-NE cells to cisplatin. Together, our findings demonstrated that UPRmt promotes mitochondrial health via upregulating β-catenin signaling and UPRmt represents viable therapeutic target for NEPC.
Collapse
Affiliation(s)
- Jordan Alyse Woytash
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Rahul Kumar
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Ajay K Chaudhary
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Cullan Donnelly
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Adam Wojtulski
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Murali Bethu
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Jianmin Wang
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Joseph Spernyak
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Peter Bross
- Research Unit for Molecular Medicine, Aarhus University and Aarhus University Hospital, 8200, Aarhus N, Denmark
| | - Neelu Yadav
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Joseph R Inigo
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Dhyan Chandra
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA.
| |
Collapse
|
11
|
Millman SE, Chaves-Perez A, Janaki-Raman S, Ho YJ, Morris JP, Narendra V, Chen CC, Jackson BT, Yashinskie JJ, Mezzadra R, Devine TI, Barthet VJA, Saoi M, Baslan T, Tian S, Sachs Z, Finley LWS, Cross JR, Lowe SW. α-Ketoglutarate dehydrogenase is a therapeutic vulnerability in acute myeloid leukemia. Blood 2025; 145:1422-1436. [PMID: 39791576 PMCID: PMC11969269 DOI: 10.1182/blood.2024025245] [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: 05/06/2024] [Revised: 10/22/2024] [Accepted: 11/19/2024] [Indexed: 01/12/2025] Open
Abstract
ABSTRACT Perturbations in intermediary metabolism contribute to the pathogenesis of acute myeloid leukemia (AML) and can produce therapeutically actionable dependencies. Here, we probed whether α-ketoglutarate (αKG) metabolism represents a specific vulnerability in AML. Using functional genomics, metabolomics, and mouse models, we identified the αKG dehydrogenase complex, which catalyzes the conversion of αKG to succinyl coenzyme A, as a molecular dependency across multiple models of adverse-risk AML. Inhibition of 2-oxoglutarate dehydrogenase (OGDH), the E1 subunit of the αKG dehydrogenase complex, impaired AML progression and drove differentiation. Mechanistically, hindrance of αKG flux through the tricarboxylic acid (TCA) cycle resulted in rapid exhaustion of aspartate pools and blockade of de novo nucleotide biosynthesis, whereas cellular bioenergetics was largely preserved. Additionally, increased αKG levels after OGDH inhibition affected the biosynthesis of other critical amino acids. Thus, this work has identified a previously undescribed, functional link between certain TCA cycle components and nucleotide biosynthesis enzymes across AML. This metabolic node may serve as a cancer-specific vulnerability, amenable to therapeutic targeting in AML and perhaps in other cancers with similar metabolic wiring.
Collapse
MESH Headings
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/enzymology
- Leukemia, Myeloid, Acute/genetics
- Ketoglutarate Dehydrogenase Complex/metabolism
- Animals
- Mice
- Humans
- Citric Acid Cycle
- Ketoglutaric Acids/metabolism
Collapse
Affiliation(s)
- Scott E. Millman
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Almudena Chaves-Perez
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Sudha Janaki-Raman
- Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Yu-Jui Ho
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - John P. Morris
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Varun Narendra
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Chi-Chao Chen
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | | | - Riccardo Mezzadra
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Tessa I. Devine
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Valentin J. A. Barthet
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Michelle Saoi
- Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Timour Baslan
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Sha Tian
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Zohar Sachs
- Division of Hematology, Oncology, and Transplantation, Department of Medicine and Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - Lydia W. S. Finley
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Justin R. Cross
- Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Scott W. Lowe
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY
- Howard Hughes Medical Institute, Chevy Chase, MD
| |
Collapse
|
12
|
Hernández-Esquivel L, Del Mazo-Monsalvo I, Pacheco-Velázquez SC, Feregrino-Mondragón RD, Robledo-Cadena DX, Sánchez-Thomas R, Jasso-Chávez R, Saavedra E, Marín-Hernández Á. Modeling Krebs cycle from liver, heart and hepatoma mitochondria, supported Complex I as target for specific inhibition of cancer cell proliferation. Front Oncol 2025; 15:1557638. [PMID: 40206582 PMCID: PMC11979947 DOI: 10.3389/fonc.2025.1557638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 03/04/2025] [Indexed: 04/11/2025] Open
Abstract
Introduction The Krebs cycle (KC) is an important pathway for cancer cells because it produces reduced coenzymes for ATP synthesis and precursors for cellular proliferation. Described changes in cancer KC enzyme activities suggested modifications in the reactions that control the KC flux compared to normal cells. Methods In this work, kinetic metabolic models of KC of mitochondria from cancer (HepM), liver (RLM) and heart (RHM) to identify targets to decrease the KC flux were constructed from kinetic parameters (Vmax and Km) of enzymes here determined. Results The enzymes Vmax values were higher in the following order: RHM > HepM > RLM; meanwhile, Km values were similar. Kinetic modeling indicated that the NADH consumption reaction (complex I) exerted higher control on the Krebs cycle flux in HepM versus RLM and to a lesser extent in RHM. These results suggested that cancer cells may be more sensitive to complex I inhibition than heart and other non-cancer cells. Indeed, cancer cell proliferation was more sensitive to rotenone (a complex I inhibitor) than heart and non-cancer cells. In contrast, cell proliferation had similar sensitivities to malonate, an inhibitor of succinate dehydrogenase, an enzyme that does not exert control. Discussion Our results showed that kinetic modeling and metabolic control analysis allow the identification of high flux-controlling targets in cancer cells that help to design strategies to specifically inhibit their proliferation. This can minimize the toxic effects in normal cells, such as the cardiac ones that are highly sensitive to conventional chemotherapy.
Collapse
Affiliation(s)
- Luz Hernández-Esquivel
- Departamento de Bioquímica, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Isis Del Mazo-Monsalvo
- Departamento de Bioquímica, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | | | | | | | - Rosina Sánchez-Thomas
- Departamento de Bioquímica, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Ricardo Jasso-Chávez
- Departamento de Bioquímica, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Emma Saavedra
- Departamento de Bioquímica, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Álvaro Marín-Hernández
- Departamento de Bioquímica, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| |
Collapse
|
13
|
Xue Q, Wang W, Liu J, Wang D, Zhang T, Shen T, Liu X, Wang X, Shao X, Zhou W, Fang X. LRPPRC confers enhanced oxidative phosphorylation metabolism in triple-negative breast cancer and represents a therapeutic target. J Transl Med 2025; 23:372. [PMID: 40133967 PMCID: PMC11938637 DOI: 10.1186/s12967-024-05946-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 12/06/2024] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is a highly malignant tumor that requires effective therapeutic targets and drugs. Oxidative phosphorylation (OXPHOS) is a metabolic vulnerability of TNBC, but the molecular mechanism responsible for the enhanced OXPHOS remains unclear. The current strategies that target the electronic transfer function of OXPHOS cannot distinguish tumor cells from normal cells. Investigating the mechanism underlying OXPHOS regulation and developing corresponding therapy strategies for TNBC is of great significance. METHODS Immunohistochemistry and sequencing data reanalysis were used to investigate LRPPRC expression in TNBC. In vitro and in vivo assays were applied to investigate the roles of LRPPRC in TNBC progression. RT-qPCR, immunoblotting, and Seahorse XF assay were used to examine LRPPRC's functions in the expression of OXPHOS subunits and energy metabolism. In vitro and in vivo functional assays were used to test the therapeutic effect of gossypol acetate (GAA), a traditional gynecological drug, on LRPPRC suppression and OXPOHS inhibition. RESULTS LRPPRC was specifically overexpressed in TNBC. LRPPRC knockdown suppressed the proliferation, metastasis, and tumor formation of TNBC cells. LRPPRC enhanced OXPHOS metabolism by increasing the expression of OXPHOS complex subunits encoded by the mitochondrial genome. GAA inhibited OXPHOS metabolism by directly binding LRPPRC, causing LRPPRC degradation, and downregulating the expression of OXPHOS complex subunits encoded by the mitochondrial genome. GAA administration suppressed TNBC cell proliferation, metastasis in vitro, and tumor formation in vivo. CONCLUSIONS This work demonstrated a new regulatory pathway of TNBC to promote the expression of mitochondrial genes by upregulating the nuclear gene LRPPRC, resulting in increased OXPHOS. We also suggested a promising therapeutic target LRPPRC for TNBC, and its inhibitor, the traditional gynecological medicine GAA, presented significant antitumor activity.
Collapse
Affiliation(s)
- Qiqi Xue
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310018, Zhejiang, China
- Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Wenxi Wang
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310018, Zhejiang, China
| | - Jie Liu
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310018, Zhejiang, China
| | - Dachi Wang
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310018, Zhejiang, China
| | - Tianyu Zhang
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310018, Zhejiang, China
| | - Tingting Shen
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310018, Zhejiang, China
| | - Xiangsheng Liu
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310018, Zhejiang, China
| | - Xiaojia Wang
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310018, Zhejiang, China
- Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Xiying Shao
- Zhejiang Cancer Hospital, Hangzhou, 310022, China.
| | - Wei Zhou
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310018, Zhejiang, China.
| | - Xiaohong Fang
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310018, Zhejiang, China.
- Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China.
| |
Collapse
|
14
|
Lorenz NI, Sauer B, Urban H, Weinem JB, Parmar BS, Zeiner PS, Strecker MI, Schulte D, Mittelbronn M, Alekseeva T, Sevenich L, Harter PN, Münch C, Steinbach JP, Luger AL, Heiland DH, Ronellenfitsch MW. AMP-activated protein kinase mediates adaptation of glioblastoma cells to conditions of the tumor microenvironment. J Exp Clin Cancer Res 2025; 44:104. [PMID: 40122814 PMCID: PMC11931870 DOI: 10.1186/s13046-025-03346-2] [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: 12/27/2024] [Accepted: 02/22/2025] [Indexed: 03/25/2025] Open
Abstract
AMP-activated protein kinase (AMPK) is an energy sensor that regulates cellular metabolic activity. We hypothesized that in glioblastoma (GB), AMPK plays a pivotal role in balancing metabolism under conditions of the tumor microenvironment with fluctuating and often low nutrient and oxygen availability. Impairment of this network could thus interfere with tumor progression. AMPK activity was modulated genetically by CRISPR/Cas9-based double knockout (DKO) of the catalytic α1 and α2 subunits in human GB cells and effects were confirmed by pharmacological AMPK inhibition using BAY3827 and an inactive control compound in primary GB cell cultures. We found that metabolic adaptation of GB cells under energy stress conditions (hypoxia, glucose deprivation) was dependent on AMPK and accordingly that AMPK DKO cells were more vulnerable to glucose deprivation or inhibition of glycolysis and sensitized to hypoxia-induced cell death. This effect was rescued by reexpression of the AMPK α2 subunit. Similar results were observed using the selective pharmacological AMPK inhibitor BAY3827. Mitochondrial biogenesis was regulated AMPK-dependently with a reduced mitochondrial mass and mitochondrial membrane potential in AMPK DKO GB cells. In vivo, AMPK DKO GB cells showed impaired tumor growth and tumor formation in CAM assays as well as in an orthotopic glioma mouse model. Our study highlights the importance of AMPK for GB cell adaptation towards energy depletion and emphasizes the role of AMPK for tumor formation in vivo. Moreover, we identified mitochondria as central downstream effectors of AMPK signaling. The development of AMPK inhibitors could open opportunities for the treatment of hypoxic tumors.
Collapse
Affiliation(s)
- Nadja I Lorenz
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- University Cancer Center Frankfurt (UCT), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Benedikt Sauer
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- University Cancer Center Frankfurt (UCT), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- Institute of Molecular Systems Medicine, Goethe University, Frankfurt am Main, Germany
| | - Hans Urban
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- University Cancer Center Frankfurt (UCT), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Jan-Béla Weinem
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- University Cancer Center Frankfurt (UCT), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Bhavesh S Parmar
- Institute of Molecular Systems Medicine, Goethe University, Frankfurt am Main, Germany
| | - Pia S Zeiner
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- University Cancer Center Frankfurt (UCT), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- Department of Neurology, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Maja I Strecker
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- University Cancer Center Frankfurt (UCT), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Dorothea Schulte
- Institute of Neurology (Edinger Institute), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Michel Mittelbronn
- Luxembourg Centre of Neuropathology (LCNP), Dudelange, Luxembourg
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
- National Center of Pathology (NCP), Laboratoire National de Santé (LNS), Dudelange, Luxembourg
- Department of Cancer Research (DoCR), Luxembourg Institute of Health (LIH), Strassen, Luxembourg
- Faculty of Science, Technology and Medicine (FSTM), University of Luxembourg, Esch-sur- Alzette, Luxembourg
- Department of Life Science and Medicine (DLSM), University of Luxembourg, Esch-sur- Alzette, Luxembourg
| | - Tijna Alekseeva
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
| | - Lisa Sevenich
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
| | - Patrick N Harter
- Center for Neuropathology and Prion Research, Faculty of Medicine, Ludwig-Maximilians- University of Munich, Munich, Germany
| | - Christian Münch
- Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- Institute of Molecular Systems Medicine, Goethe University, Frankfurt am Main, Germany
- Cardio-Pulmonary Institute, Frankfurt am Main, Germany
| | - Joachim P Steinbach
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- University Cancer Center Frankfurt (UCT), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Anna-Luisa Luger
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- University Cancer Center Frankfurt (UCT), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Dieter Henrik Heiland
- Microenvironment and Immunology Research Laboratory, Medical Center, University of Freiburg, Freiburg, Germany
- Department of Neurosurgery, Medical Center, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Comprehensive Cancer Center Freiburg (CCCF), Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner site Freiburg, Freiburg, Germany
- Department of Neurosurgery, University Clinic, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Michael W Ronellenfitsch
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany.
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany.
- Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany.
- University Cancer Center Frankfurt (UCT), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany.
| |
Collapse
|
15
|
Tessier C, Toujas M, Pagano Zottola AC, Bikfalvi A, Mathivet T, Daubon T, Brisson L, Burban A, Sharanek A. Protocol for real-time measurement of mitochondrial bioenergetics in 3D-cultured brain tumor stem cells using the Resipher system. STAR Protoc 2025; 6:103651. [PMID: 39998952 PMCID: PMC11907464 DOI: 10.1016/j.xpro.2025.103651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/07/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Aberrant mitochondrial function can lead to severe human diseases, including neurodegenerative diseases and cancer. Here, we describe a cell-based protocol for measuring different mitochondrial respiratory parameters using the high-resolution real-time Resipher system. We optimized this protocol on brain tumor stem cells cultured in three-dimensional spheroids. We provide essential optimization steps for cell seeding density, mitochondrial respiration modulator concentrations, running the assay, and data analysis. For complete details on the use and execution of this protocol, please refer to Burban et al.1.
Collapse
Affiliation(s)
- Cloé Tessier
- University of Bordeaux, INSERM UMR1312, BRIC BoRdeaux Institute of Oncology, Bordeaux, France
| | - Maxime Toujas
- University of Bordeaux, INSERM UMR1312, BRIC BoRdeaux Institute of Oncology, Bordeaux, France
| | | | - Andreas Bikfalvi
- University of Bordeaux, INSERM UMR1312, BRIC BoRdeaux Institute of Oncology, Bordeaux, France
| | - Thomas Mathivet
- University of Bordeaux, INSERM UMR1312, BRIC BoRdeaux Institute of Oncology, Bordeaux, France
| | - Thomas Daubon
- University of Bordeaux, CNRS, IBGC, UMR5095, Bordeaux, France
| | - Lucie Brisson
- University of Bordeaux, INSERM UMR1312, BRIC BoRdeaux Institute of Oncology, Bordeaux, France
| | - Audrey Burban
- University of Bordeaux, CNRS, IBGC, UMR5095, Bordeaux, France.
| | - Ahmad Sharanek
- University of Bordeaux, INSERM UMR1312, BRIC BoRdeaux Institute of Oncology, Bordeaux, France.
| |
Collapse
|
16
|
Shi X, Li M, Liu Z, Tiessen J, Li Y, Zhou J, Zhu Y, Mahesula S, Ding Q, Tan L, Feng M, Kageyama Y, Hara Y, Tao JJ, Luo X, Patras KA, Lorenzi PL, Huang S, Stevens AM, Takahashi K, Issa GC, Samee MAH, Agathocleous M, Nakada D. Guanine nucleotide biosynthesis blockade impairs MLL complex formation and sensitizes leukemias to menin inhibition. Nat Commun 2025; 16:2641. [PMID: 40102405 PMCID: PMC11920272 DOI: 10.1038/s41467-025-57544-9] [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: 06/13/2024] [Accepted: 02/26/2025] [Indexed: 03/20/2025] Open
Abstract
Targeting the dependency of MLL-rearranged (MLLr) leukemias on menin with small molecule inhibitors has opened new therapeutic strategies for these poor-prognosis diseases. However, the rapid development of menin inhibitor resistance calls for combinatory strategies to improve responses and prevent resistance. Here we show that leukemia stem cells (LSCs) of MLLr acute myeloid leukemia (AML) exhibit enhanced guanine nucleotide biosynthesis, the inhibition of which leads to myeloid differentiation and sensitization to menin inhibitors. Mechanistically, targeting inosine monophosphate dehydrogenase 2 (IMPDH2) reduces guanine nucleotides and rRNA transcription, leading to reduced protein expression of LEDGF and menin. Consequently, the formation and chromatin binding of the MLL-fusion complex is impaired, reducing the expression of MLL target genes. Inhibition of guanine nucleotide biosynthesis or rRNA transcription further suppresses MLLr AML when combined with a menin inhibitor. Our findings underscore the requirement of guanine nucleotide biosynthesis in maintaining the function of the LEDGF/menin/MLL-fusion complex and provide a rationale to target guanine nucleotide biosynthesis to sensitize MLLr leukemias to menin inhibitors.
Collapse
MESH Headings
- Myeloid-Lymphoid Leukemia Protein/metabolism
- Myeloid-Lymphoid Leukemia Protein/genetics
- Proto-Oncogene Proteins/metabolism
- Proto-Oncogene Proteins/genetics
- Histone-Lysine N-Methyltransferase/metabolism
- Histone-Lysine N-Methyltransferase/genetics
- Histone-Lysine N-Methyltransferase/antagonists & inhibitors
- Humans
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/pathology
- Animals
- Mice
- IMP Dehydrogenase/metabolism
- IMP Dehydrogenase/antagonists & inhibitors
- IMP Dehydrogenase/genetics
- Cell Line, Tumor
- Gene Expression Regulation, Leukemic/drug effects
Collapse
Affiliation(s)
- Xiangguo Shi
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA.
- Department of Molecular and Precision Medicine, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA.
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA.
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA.
| | - Minhua Li
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Zian Liu
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jonathan Tiessen
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yuan Li
- Department of Biostatistics and Data Science, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Jing Zhou
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yudan Zhu
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Swetha Mahesula
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Qing Ding
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Lin Tan
- Metabolomics Core Facility, Department of Bioinformatics and Computational Biology, Division of Basic Science Research, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Mengdie Feng
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yuki Kageyama
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yusuke Hara
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jacob J Tao
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Xuan Luo
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Kathryn A Patras
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Philip L Lorenzi
- Metabolomics Core Facility, Department of Bioinformatics and Computational Biology, Division of Basic Science Research, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Suming Huang
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Alexandra M Stevens
- Section of Hematology/Oncology, Department of Pediatrics, Texas Children's Cancer and Hematology Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Koichi Takahashi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Ghayas C Issa
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Md Abul Hassan Samee
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Michalis Agathocleous
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Daisuke Nakada
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA.
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX, 77030, USA.
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
17
|
Tau S, Chamberlin MD, Yang H, Marotti JD, Muskus PC, Roberts AM, Carmichael MM, Cressey L, Dragnev CPC, Demidenko E, Hampsch RA, Soucy SM, Kolling FW, Samkoe KS, Alvarez JV, Kettenbach AN, Miller TW. Oxidative Phosphorylation Is a Metabolic Vulnerability of Endocrine Therapy-Tolerant Persister Cells in ER+ Breast Cancer. Cancer Res 2025; 85:1145-1161. [PMID: 39777474 PMCID: PMC11908958 DOI: 10.1158/0008-5472.can-24-1204] [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: 04/11/2024] [Revised: 11/05/2024] [Accepted: 12/27/2024] [Indexed: 01/11/2025]
Abstract
Despite adjuvant treatment with endocrine therapies, estrogen receptor-positive (ER+) breast cancers recur in a significant proportion of patients. Recurrences are attributable to clinically undetectable endocrine-tolerant persister cancer cells that retain tumor-forming potential. Therefore, strategies targeting such persister cells may prevent recurrent disease. Using CRISPR-Cas9 genome-wide knockout screening in ER+ breast cancer cells, we identified a survival mechanism involving metabolic reprogramming with reliance upon mitochondrial respiration in endocrine-tolerant persister cells. Quantitative proteomic profiling showed reduced levels of glycolytic proteins in persisters. Metabolic tracing of glucose revealed an energy-depleted state in persisters, in which oxidative phosphorylation was required to generate ATP. A phase II clinical trial was conducted to evaluate changes in mitochondrial markers in primary ER+/HER2- breast tumors induced by neoadjuvant endocrine therapy (NCT04568616). In an analysis of tumor specimens from 32 patients, tumors exhibiting residual cell proliferation after aromatase inhibitor-induced estrogen deprivation with letrozole showed increased mitochondrial content. Genetic profiling and barcode lineage tracing showed that endocrine-tolerant persistence occurred stochastically without genetic predisposition. Pharmacologic inhibition of mitochondrial complex I suppressed the tumor-forming potential of persisters in mice and synergized with the antiestrogen drug fulvestrant to induce regression of patient-derived xenografts. These findings indicate that mitochondrial metabolism is essential in endocrine-tolerant persister ER+ breast cancer cells and warrant the development of treatment strategies to leverage this vulnerability for treating breast cancer. Significance: Persister cancer cells that survive endocrine therapy exhibit increased energetic dependence upon mitochondria for survival and tumor regrowth potential, indicating that therapies targeting this metabolic dependency could help prevent disease recurrence.
Collapse
Affiliation(s)
- Steven Tau
- Department of Molecular and Systems Biology, Dartmouth Geisel School of Medicine, Lebanon, NH, USA
| | - Mary D. Chamberlin
- Department of Medicine, Dartmouth Geisel School of Medicine, Lebanon, NH, USA
| | - Huijuan Yang
- Department of Molecular and Systems Biology, Dartmouth Geisel School of Medicine, Lebanon, NH, USA
| | - Jonathan D. Marotti
- Department of Pathology and Laboratory Medicine, Dartmouth Geisel School of Medicine, Lebanon, NH, USA
| | - Patricia C. Muskus
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Alyssa M. Roberts
- Department of Molecular and Systems Biology, Dartmouth Geisel School of Medicine, Lebanon, NH, USA
| | - Melissa M. Carmichael
- Department of Molecular and Systems Biology, Dartmouth Geisel School of Medicine, Lebanon, NH, USA
| | - Lauren Cressey
- Department of Biochemistry and Cell Biology, Dartmouth Geisel School of Medicine, Lebanon, NH, USA
| | | | - Eugene Demidenko
- Department of Biomedical Data Science, Dartmouth Geisel School of Medicine, Lebanon, NH, USA
| | - Riley A. Hampsch
- Department of Molecular and Systems Biology, Dartmouth Geisel School of Medicine, Lebanon, NH, USA
| | - Shannon M. Soucy
- Department of Biomedical Data Science, Dartmouth Geisel School of Medicine, Lebanon, NH, USA
- Center for Quantitative Biology, Dartmouth Geisel School of Medicine, Lebanon, NH, USA
| | - Fred W. Kolling
- Center for Quantitative Biology, Dartmouth Geisel School of Medicine, Lebanon, NH, USA
| | | | - James V. Alvarez
- Translational Research Program, Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Arminja N. Kettenbach
- Department of Biochemistry and Cell Biology, Dartmouth Geisel School of Medicine, Lebanon, NH, USA
| | - Todd W. Miller
- Department of Molecular and Systems Biology, Dartmouth Geisel School of Medicine, Lebanon, NH, USA
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, USA
- Lead Contact
| |
Collapse
|
18
|
Simoes RV, Henriques RN, Olesen JL, Cardoso BM, Fernandes FF, Monteiro MAV, Jespersen SN, Carvalho T, Shemesh N. Deuterium metabolic imaging phenotypes mouse glioblastoma heterogeneity through glucose turnover kinetics. eLife 2025; 13:RP100570. [PMID: 40035743 DOI: 10.7554/elife.100570] [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] [Indexed: 03/06/2025] Open
Abstract
Glioblastomas are aggressive brain tumors with dismal prognosis. One of the main bottlenecks for developing more effective therapies for glioblastoma stems from their histologic and molecular heterogeneity, leading to distinct tumor microenvironments and disease phenotypes. Effectively characterizing these features would improve the clinical management of glioblastoma. Glucose flux rates through glycolysis and mitochondrial oxidation have been recently shown to quantitatively depict glioblastoma proliferation in mouse models (GL261 and CT2A tumors) using dynamic glucose-enhanced (DGE) deuterium spectroscopy. However, the spatial features of tumor microenvironment phenotypes remain hitherto unresolved. Here, we develop a DGE Deuterium Metabolic Imaging (DMI) approach for profiling tumor microenvironments through glucose conversion kinetics. Using a multimodal combination of tumor mouse models, novel strategies for spectroscopic imaging and noise attenuation, and histopathological correlations, we show that tumor lactate turnover mirrors phenotype differences between GL261 and CT2A mouse glioblastoma, whereas recycling of the peritumoral glutamate-glutamine pool is a potential marker of invasion capacity in pooled cohorts, linked to secondary brain lesions. These findings were validated by histopathological characterization of each tumor, including cell density and proliferation, peritumoral invasion and distant migration, and immune cell infiltration. Our study bodes well for precision neuro-oncology, highlighting the importance of mapping glucose flux rates to better understand the metabolic heterogeneity of glioblastoma and its links to disease phenotypes.
Collapse
Affiliation(s)
- Rui Vasco Simoes
- Preclinical MRI, Champalimaud Research, Champalimaud Foundation, Lisbon, Portugal
- Neuroengineering and Computational Neuroscience, Institute for Research and Innovation in Health (i3S), Porto, Portugal
| | | | - Jonas L Olesen
- Center of Functionally Integrative Neuroscience (CFIN) and MINDLab, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Physics and Astronomy, Aarhus University, Aarhus, Denmark
| | - Beatriz M Cardoso
- Preclinical MRI, Champalimaud Research, Champalimaud Foundation, Lisbon, Portugal
| | | | - Mariana A V Monteiro
- Histopathology Platform, Champalimaud Research, Champalimaud Foundation, Lisbon, Portugal
| | - Sune N Jespersen
- Center of Functionally Integrative Neuroscience (CFIN) and MINDLab, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Physics and Astronomy, Aarhus University, Aarhus, Denmark
| | - Tânia Carvalho
- Histopathology Platform, Champalimaud Research, Champalimaud Foundation, Lisbon, Portugal
| | - Noam Shemesh
- Preclinical MRI, Champalimaud Research, Champalimaud Foundation, Lisbon, Portugal
| |
Collapse
|
19
|
Rubinstein JC, Domanskyi S, Sheridan TB, Sanderson B, Park S, Kaster J, Li H, Anczukow O, Herlyn M, Chuang JH. Spatiotemporal Profiling Defines Persistence and Resistance Dynamics during Targeted Treatment of Melanoma. Cancer Res 2025; 85:987-1002. [PMID: 39700408 PMCID: PMC11875961 DOI: 10.1158/0008-5472.can-24-0690] [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: 07/16/2024] [Revised: 10/03/2024] [Accepted: 12/11/2024] [Indexed: 12/21/2024]
Abstract
Resistance of BRAF-mutant melanomas to targeted therapy arises from the ability of cells to enter a persister state, evade treatment with relative dormancy, and repopulate the tumor when reactivated. A better understanding of the temporal dynamics and specific pathways leading into and out of the persister state is needed to identify strategies to prevent treatment failure. Using spatial transcriptomics in patient-derived xenograft models, we captured clonal lineage evolution during treatment. The persister state showed increased oxidative phosphorylation, decreased proliferation, and increased invasive capacity, with central-to-peripheral gradients. Phylogenetic tracing identified intrinsic and acquired resistance mechanisms (e.g., dual-specific phosphatases, reticulon-4, and cyclin-dependent kinase 2) and suggested specific temporal windows of potential therapeutic susceptibility. Deep learning-enabled analysis of histopathologic slides revealed morphologic features correlating with specific cell states, demonstrating that juxtaposition of transcriptomics and histologic data enabled identification of phenotypically distinct populations from using imaging data alone. In summary, this study defined state change and lineage selection during melanoma treatment with spatiotemporal resolution, elucidating how choice and timing of therapeutic agents will impact the ability to eradicate resistant clones. Significance: Tracking clonal progression during treatment uncovers conserved, global transcriptional changes and local clone-clone and spatial patterns underlying the emergence of resistance, providing insights into therapy-induced tumor evolution.
Collapse
Affiliation(s)
- Jill C. Rubinstein
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
- Department of Surgery, Hartford Healthcare, Hartford, CT, USA
- Department of Surgery, UCONN School of Medicine, Farmington, CT, USA
- These authors provided equal contribution to this work
- Further information and requests for resources should be directed to and will be fulfilled by the lead contact, Jill Rubinstein
| | - Sergii Domanskyi
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
- These authors provided equal contribution to this work
| | - Todd B. Sheridan
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
- Department of Surgery, Hartford Healthcare, Hartford, CT, USA
| | - Brian Sanderson
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - SungHee Park
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Jessica Kaster
- The Wistar Institute, Philadelphia, PA, USA
- Saint Joseph’s University, Philadelphia, PA, USA
| | - Haiyin Li
- The Wistar Institute, Philadelphia, PA, USA
| | - Olga Anczukow
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | | | | |
Collapse
|
20
|
Gritsch D, Brastianos PK. Molecular evolution of central nervous system metastasis and therapeutic implications. Trends Mol Med 2025; 31:240-251. [PMID: 39424530 PMCID: PMC11908961 DOI: 10.1016/j.molmed.2024.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 09/27/2024] [Accepted: 09/30/2024] [Indexed: 10/21/2024]
Abstract
The increasing prevalence and poor prognosis of central nervous system (CNS) metastases pose a significant challenge in oncology, necessitating improved therapeutic strategies. Recent research has shed light on the complex genomic landscape of brain metastases, identifying unique and potentially actionable genetic alterations. These insights offer new avenues for targeted therapy, highlighting the potential of precision medicine approaches in treating CNS metastases. However, translating these discoveries into clinical practice requires overcoming challenges such as availability of tissue for characterization, access to molecular testing, drug delivery across the blood-brain barrier (BBB) and addressing intra- and intertumoral genetic heterogeneity. This review explores novel insights into the evolution of CNS metastases, the molecular mechanisms underlying their development, and implications for therapeutic interventions.
Collapse
Affiliation(s)
- David Gritsch
- Massachusetts General Hospital Cancer Center, Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA
| | - Priscilla K Brastianos
- Massachusetts General Hospital Cancer Center, Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA.
| |
Collapse
|
21
|
Burban A, Tessier C, Larroquette M, Guyon J, Lubiato C, Pinglaut M, Toujas M, Galvis J, Dartigues B, Georget E, Luchman HA, Weiss S, Cappellen D, Nicot N, Klink B, Nikolski M, Brisson L, Mathivet T, Bikfalvi A, Daubon T, Sharanek A. Exploiting metabolic vulnerability in glioblastoma using a brain-penetrant drug with a safe profile. EMBO Mol Med 2025; 17:469-503. [PMID: 39901019 PMCID: PMC11903783 DOI: 10.1038/s44321-025-00195-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 01/13/2025] [Accepted: 01/14/2025] [Indexed: 02/05/2025] Open
Abstract
Glioblastoma is one of the most treatment-resistant and lethal cancers, with a subset of self-renewing brain tumour stem cells (BTSCs), driving therapy resistance and relapse. Here, we report that mubritinib effectively impairs BTSC stemness and growth. Mechanistically, bioenergetic assays and rescue experiments showed that mubritinib targets complex I of the electron transport chain, thereby impairing BTSC self-renewal and proliferation. Gene expression profiling and Western blot analysis revealed that mubritinib disrupts the AMPK/p27Kip1 pathway, leading to cell-cycle impairment. By employing in vivo pharmacokinetic assays, we established that mubritinib crosses the blood-brain barrier. Using preclinical patient-derived and syngeneic models, we demonstrated that mubritinib delays glioblastoma progression and extends animal survival. Moreover, combining mubritinib with radiotherapy or chemotherapy offers survival advantage to animals. Notably, we showed that mubritinib alleviates hypoxia, thereby enhancing ROS generation, DNA damage, and apoptosis in tumours when combined with radiotherapy. Encouragingly, toxicological and behavioural studies revealed that mubritinib is well tolerated and spares normal cells. Our findings underscore the promising therapeutic potential of mubritinib, warranting its further exploration in clinic for glioblastoma therapy.
Collapse
Affiliation(s)
- Audrey Burban
- University of Bordeaux, CNRS, IBGC, UMR5095, Bordeaux, France
| | - Cloe Tessier
- University of Bordeaux, INSERM, UMR1312, BRIC, BoRdeaux Institute of onCology, Bordeaux, France
| | | | - Joris Guyon
- CHU of Bordeaux, Service de Pharmacologie Médicale, Bordeaux, France
- University of Bordeaux, INSERM, BPH, U1219, Bordeaux, France
| | - Cloe Lubiato
- University of Bordeaux, INSERM, UMR1312, BRIC, BoRdeaux Institute of onCology, Bordeaux, France
| | - Mathis Pinglaut
- University of Bordeaux, CNRS, IBGC, UMR5095, Bordeaux, France
| | - Maxime Toujas
- University of Bordeaux, INSERM, UMR1312, BRIC, BoRdeaux Institute of onCology, Bordeaux, France
| | - Johanna Galvis
- University of Bordeaux, CNRS, IBGC, UMR5095, Bordeaux, France
| | - Benjamin Dartigues
- Bordeaux Bioinformatic Center CBiB, University of Bordeaux, Bordeaux, France
| | - Emmanuelle Georget
- University of Bordeaux, INSERM, UMR1312, BRIC, BoRdeaux Institute of onCology, Bordeaux, France
| | - H Artee Luchman
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada
- Arnie Charbonneau Cancer Institute and Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Samuel Weiss
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada
- Arnie Charbonneau Cancer Institute and Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - David Cappellen
- University of Bordeaux, INSERM, UMR1312, BRIC, BoRdeaux Institute of onCology, Bordeaux, France
| | - Nathalie Nicot
- LuxGen Genome Center, Luxembourg Institute of Health, Laboratoire national de santé, Dudelange, Luxembourg
| | - Barbara Klink
- LuxGen Genome Center, Luxembourg Institute of Health, Laboratoire national de santé, Dudelange, Luxembourg
- National Center of Genetics (NCG), Laboratoire National de Santé (LNS), Dudelange, Luxembourg
- Department of Cancer Research (DoCR), Luxembourg Institute of Health (LIH), Luxembourg, 1526, Luxembourg
- Department of Life Sciences and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Macha Nikolski
- University of Bordeaux, CNRS, IBGC, UMR5095, Bordeaux, France
- Bordeaux Bioinformatic Center CBiB, University of Bordeaux, Bordeaux, France
| | - Lucie Brisson
- University of Bordeaux, INSERM, UMR1312, BRIC, BoRdeaux Institute of onCology, Bordeaux, France
| | - Thomas Mathivet
- University of Bordeaux, INSERM, UMR1312, BRIC, BoRdeaux Institute of onCology, Bordeaux, France
| | - Andreas Bikfalvi
- University of Bordeaux, INSERM, UMR1312, BRIC, BoRdeaux Institute of onCology, Bordeaux, France.
| | - Thomas Daubon
- University of Bordeaux, CNRS, IBGC, UMR5095, Bordeaux, France.
| | - Ahmad Sharanek
- University of Bordeaux, INSERM, UMR1312, BRIC, BoRdeaux Institute of onCology, Bordeaux, France.
| |
Collapse
|
22
|
Mizushina Y, Sun L, Nishio M, Nagata S, Kamakura T, Fukuda M, Tanaka K, Toguchida J, Jin Y. Hydroxycitric acid reconstructs damaged articular cartilages by modifying the metabolic cascade in chondrogenic cells. OSTEOARTHRITIS AND CARTILAGE OPEN 2025; 7:100564. [PMID: 39835169 PMCID: PMC11743121 DOI: 10.1016/j.ocarto.2024.100564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 12/19/2024] [Indexed: 01/22/2025] Open
Abstract
Objective Osteoarthritis, a degenerative joint disease, requires innovative therapies due to the limited ability of cartilage to regenerate. Since mesenchymal stem cells (MSCs) provide a cell source for chondrogenic cells, we hypothesize that chemicals capable of enhancing the chondrogenic potential of MSCs with transforming growth factor-beta (TGFβ) in vitro may similarly promote chondrogenesis in articular cartilage in vivo. Design Chemical compounds that enhance the TGFβ signaling for chondrogenesis were investigated utilizing mesenchymal stem cells derived from human induced pluripotent stem cells. The mechanisms of action underlying the identified compound were explored in vitro, and its therapeutic effects were validated in vivo using a mouse model of exercise-induced osteoarthritis. Results Hydroxycitric acid (HCA) emerged as the lead compound. In vitro, HCA effectively enhanced chondrogenesis by inhibiting ATP citrate lyase, inducing citrate and alpha-ketoglutarate (α-KG), while reducing cytosolic acetyl coenzyme A (Ac-CoA). This induction of α-KG promoted collagen prolyl-4-hydroxylase activity, boosting hydroxyproline production and matrix formation. The reduction of Ac-CoA attenuated the inhibitory effect of β-catenin on mitochondrial activity by diminishing its acetylation. In vivo, orally administered HCA accumulated in joint tissues of mice and histological examination demonstrated newly synthesized cartilage tissues in damaged area. Analysis of joint tissue extracts revealed a downregulation of Ac-CoA and an upregulation of citrate and α-KG, accompanied by a systemic increase in an anabolic biomarker. Conclusions HCA demonstrates promise as an osteoarthritis therapy by enhancing chondrogenic differentiation. Its ability to modulate crucial metabolic pathways and facilitate cartilage repair suggests potential for clinical translation, addressing a critical need in the treatment of osteoarthritis.
Collapse
Affiliation(s)
- Yoshiyuki Mizushina
- Department of Regeneration Sciences and Engineering, Institute for Life and Medical Sciences, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-Ku, Kyoto, 606-8507, Japan
- Central R & D Laboratory, Kobayashi Pharmaceutical Co., Ltd., 1-30-3 Toyokawa, Ibaraki, 567-0057, Japan
| | - Liping Sun
- Department of Regeneration Sciences and Engineering, Institute for Life and Medical Sciences, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Megumi Nishio
- Department of Fundamental Cell Technology, Center for iPS Cell Research and Application, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Sanae Nagata
- Department of Fundamental Cell Technology, Center for iPS Cell Research and Application, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Takeshi Kamakura
- Department of Regeneration Sciences and Engineering, Institute for Life and Medical Sciences, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Masayuki Fukuda
- Department of Regeneration Sciences and Engineering, Institute for Life and Medical Sciences, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Kousuke Tanaka
- Central R & D Laboratory, Kobayashi Pharmaceutical Co., Ltd., 1-30-3 Toyokawa, Ibaraki, 567-0057, Japan
| | - Junya Toguchida
- Department of Fundamental Cell Technology, Center for iPS Cell Research and Application, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-Ku, Kyoto, 606-8507, Japan
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Yonghui Jin
- Department of Regeneration Sciences and Engineering, Institute for Life and Medical Sciences, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| |
Collapse
|
23
|
D’Aprile S, Denaro S, Gervasi A, Vicario N, Parenti R. Targeting metabolic reprogramming in glioblastoma as a new strategy to overcome therapy resistance. Front Cell Dev Biol 2025; 13:1535073. [PMID: 40078366 PMCID: PMC11897528 DOI: 10.3389/fcell.2025.1535073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 02/04/2025] [Indexed: 03/14/2025] Open
Abstract
Glioblastoma (GBM) is one of the deadliest tumors due to its high aggressiveness and resistance to standard therapies, resulting in a dismal prognosis. This lethal tumor carries out metabolic reprogramming in order to modulate specific pathways, providing metabolites that promote GBM cells proliferation and limit the efficacy of standard treatments. Indeed, GBM remodels glucose metabolism and undergoes Warburg effect, fuelling glycolysis even when oxygen is available. Moreover, recent evidence revealed a rewiring in nucleotide, lipid and iron metabolism, resulting not only in an increased tumor growth, but also in radio- and chemo-resistance. Thus, while on the one hand metabolic reprogramming is an advantage for GBM, on the other hand it may represent an exploitable target to hamper GBM progression. Lately, a number of studies focused on drugs targeting metabolism to uncover their effects on tumor proliferation and therapy resistance, demonstrating that some of these are effective, in combination with conventional treatments, sensitizing GBM to radiotherapy and chemotherapy. However, GBM heterogeneity could lead to a plethora of metabolic alterations among subtypes, hence a metabolic treatment might be effective for proneural tumors but not for mesenchymal ones, which are more aggressive and resistant to conventional approaches. This review explores key mechanisms of GBM metabolic reprogramming and their involvement in therapy resistance, highlighting how metabolism acts as a double-edged sword for GBM, taking into account metabolic pathways that seem to offer promising treatment options for GBM.
Collapse
Affiliation(s)
| | | | | | | | - Rosalba Parenti
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| |
Collapse
|
24
|
Wang Y, Xiu Y, Dong Q, Zhao J, Neumbo K, Miyagi M, Borcherding N, Fu L, De Celis H, Pintozzi N, Starczynowski DT, Zhao C. TIFAB modulates metabolic pathways in KMT2A::MLLT3-induced AML through HNF4A. Blood Adv 2025; 9:844-855. [PMID: 39626355 PMCID: PMC11872587 DOI: 10.1182/bloodadvances.2024013446] [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: 04/22/2024] [Accepted: 11/11/2024] [Indexed: 03/05/2025] Open
Abstract
ABSTRACT Tumor necrosis factor (TNF) receptor-associated factor (TRAF)-interacting protein with forkhead-associated domain B (TIFAB), an inhibitor of NF-κB signaling, plays critical roles in hematopoiesis, myelodysplastic neoplasms, and leukemia. We previously demonstrated that Tifab enhances KMT2A::MLLT3-driven acute myeloid leukemia (AML) by either upregulating Hoxa9 or through ubiquitin-specific peptidase 15-mediated downregulation of p53 signaling. In this study, we show that Tifab deletion in KMT2A::MLLT3-induced AML impairs leukemia stem/progenitor cell (LSPC) engraftment, glucose uptake, and mitochondrial function. Gene set enrichment analysis reveals that Tifab deletion downregulates MYC, HOXA9/MEIS1, mTORC1 signaling, and genes involved in glycolysis and oxidative phosphorylation. By comparing genes upregulated in TIFAB-overexpressing LSPCs with those downregulated upon Tifab deletion, we identify hepatocyte nuclear factor 4 alpha (Hnf4a) as a key TIFAB target, regulated through the inhibition of NF-κB component RelB, which suppresses Hnf4a in leukemia cells. HNF4A, a nuclear receptor involved in organ development, metabolism, and tumorigenesis, rescues the metabolic defects caused by Tifab deletion and enhances leukemia cell engraftment. Conversely, Hnf4a knockdown attenuates TIFAB-mediated enhancement of LSPC function. These findings highlight the critical role of the TIFAB-HNF4A axis in KMT2A::MLLT3-induced AML and uncover a novel regulator in leukemia biology.
Collapse
Affiliation(s)
- Yang Wang
- Department of Pathology, Case Western Reserve University, Cleveland, OH
| | - Yan Xiu
- Department of Pathology, Case Western Reserve University, Cleveland, OH
| | - Qianze Dong
- Department of Pathology, Case Western Reserve University, Cleveland, OH
| | - Jinming Zhao
- Department of Pathology, Case Western Reserve University, Cleveland, OH
- Department of Pathology, China Medical University, Shenyang, China
| | - Kelao Neumbo
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH
| | - Masaru Miyagi
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH
| | - Nicholas Borcherding
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Lin Fu
- Department of Pathology, China Medical University, Shenyang, China
| | - Havana De Celis
- Department of Biology, Case Western Reserve University, Cleveland, OH
| | - Nicolas Pintozzi
- Department of Biology, Case Western Reserve University, Cleveland, OH
| | - Daniel T. Starczynowski
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- University of Cincinnati Cancer Center, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH
| | - Chen Zhao
- Department of Pathology, Case Western Reserve University, Cleveland, OH
- Department of Pathology, University Hospitals Case Medical Center, Cleveland, OH
- Department of Pathology, Louis Stokes Veterans Affairs Medical Center, Cleveland, OH
| |
Collapse
|
25
|
Liu H, Wang S, Wang J, Guo X, Song Y, Fu K, Gao Z, Liu D, He W, Yang LL. Energy metabolism in health and diseases. Signal Transduct Target Ther 2025; 10:69. [PMID: 39966374 PMCID: PMC11836267 DOI: 10.1038/s41392-025-02141-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 11/08/2024] [Accepted: 12/25/2024] [Indexed: 02/20/2025] Open
Abstract
Energy metabolism is indispensable for sustaining physiological functions in living organisms and assumes a pivotal role across physiological and pathological conditions. This review provides an extensive overview of advancements in energy metabolism research, elucidating critical pathways such as glycolysis, oxidative phosphorylation, fatty acid metabolism, and amino acid metabolism, along with their intricate regulatory mechanisms. The homeostatic balance of these processes is crucial; however, in pathological states such as neurodegenerative diseases, autoimmune disorders, and cancer, extensive metabolic reprogramming occurs, resulting in impaired glucose metabolism and mitochondrial dysfunction, which accelerate disease progression. Recent investigations into key regulatory pathways, including mechanistic target of rapamycin, sirtuins, and adenosine monophosphate-activated protein kinase, have considerably deepened our understanding of metabolic dysregulation and opened new avenues for therapeutic innovation. Emerging technologies, such as fluorescent probes, nano-biomaterials, and metabolomic analyses, promise substantial improvements in diagnostic precision. This review critically examines recent advancements and ongoing challenges in metabolism research, emphasizing its potential for precision diagnostics and personalized therapeutic interventions. Future studies should prioritize unraveling the regulatory mechanisms of energy metabolism and the dynamics of intercellular energy interactions. Integrating cutting-edge gene-editing technologies and multi-omics approaches, the development of multi-target pharmaceuticals in synergy with existing therapies such as immunotherapy and dietary interventions could enhance therapeutic efficacy. Personalized metabolic analysis is indispensable for crafting tailored treatment protocols, ultimately providing more accurate medical solutions for patients. This review aims to deepen the understanding and improve the application of energy metabolism to drive innovative diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Hui Liu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuo Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianhua Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xin Guo
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yujing Song
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kun Fu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenjie Gao
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Danfeng Liu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Wei He
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Lei-Lei Yang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
26
|
Bischoff ME, Shamsaei B, Yang J, Secic D, Vemuri B, Reisz JA, D’Alessandro A, Bartolacci C, Adamczak R, Schmidt L, Wang J, Martines A, Venkat J, Tcheuyap VT, Biesiada J, Behrmann CA, Vest KE, Brugarolas J, Scaglioni PP, Plas DR, Patra KC, Gulati S, Landero Figueroa JA, Meller J, Cunningham JT, Czyzyk-Krzeska MF. Copper Drives Remodeling of Metabolic State and Progression of Clear Cell Renal Cell Carcinoma. Cancer Discov 2025; 15:401-426. [PMID: 39476412 PMCID: PMC11803400 DOI: 10.1158/2159-8290.cd-24-0187] [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: 02/05/2024] [Revised: 09/23/2024] [Accepted: 10/30/2024] [Indexed: 11/02/2024]
Abstract
SIGNIFICANCE The work establishes a requirement for glucose-dependent coordination between energy production and redox homeostasis, which is fundamental for the survival of cancer cells that accumulate Cu and contributes to tumor growth.
Collapse
Affiliation(s)
- Megan E. Bischoff
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Behrouz Shamsaei
- Department of Biostatistics, Health Informatics and Data Sciences, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Biostatistics and Bioinformatics, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Juechen Yang
- Department of Biostatistics, Health Informatics and Data Sciences, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Biostatistics and Bioinformatics, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Dina Secic
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Bhargav Vemuri
- Department of Biostatistics, Health Informatics and Data Sciences, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Julie A. Reisz
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado
| | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado
| | - Caterina Bartolacci
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Rafal Adamczak
- Institute of Engineering and Technology, Faculty of Physics, Astronomy and Informatics, Nicolaus Copernicus University, Torun, Poland
| | - Lucas Schmidt
- Trace Elements Group, Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jiang Wang
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Amelia Martines
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Jahnavi Venkat
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Vanina Toffessi Tcheuyap
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jacek Biesiada
- Division of Biostatistics and Bioinformatics, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Catherine A. Behrmann
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Katherine E. Vest
- Department of Molecular and Cellular Biosciences, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - James Brugarolas
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Pier Paolo Scaglioni
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - David R. Plas
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Krushna C. Patra
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Shuchi Gulati
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Oncology and Hematology, Department of Internal Medicine, University of California Davis Comprehensive Cancer Center, Sacramento, California
| | - Julio A. Landero Figueroa
- Trace Elements Group, Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jarek Meller
- Department of Biostatistics, Health Informatics and Data Sciences, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Biostatistics and Bioinformatics, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
- Institute of Engineering and Technology, Faculty of Physics, Astronomy and Informatics, Nicolaus Copernicus University, Torun, Poland
- Department of Computer Science, University of Cincinnati College of Engineering and Applied Sciences, Cincinnati, Ohio
| | - John T. Cunningham
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Maria F. Czyzyk-Krzeska
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Department of Veterans Affairs, Veteran Affairs Medical Center, Cincinnati, Ohio
- Department of Pharmacology and System Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
27
|
Liang J, Vitale T, Zhang X, Jackson TD, Yu D, Jedrychowski M, Gygi SP, Widlund HR, Wucherpfennig KW, Puigserver P. Selective deficiency of mitochondrial respiratory complex I subunits Ndufs4/6 causes tumor immunogenicity. NATURE CANCER 2025; 6:323-337. [PMID: 39824999 DOI: 10.1038/s43018-024-00895-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 12/10/2024] [Indexed: 01/20/2025]
Abstract
Cancer cells frequently rewire their metabolism to support proliferation and evade immune surveillance, but little is known about metabolic targets that could increase immune surveillance. Here we show a specific means of mitochondrial respiratory complex I (CI) inhibition that improves tumor immunogenicity and sensitivity to immune checkpoint blockade (ICB). Targeted genetic deletion of either Ndufs4 or Ndufs6, but not other CI subunits, induces an immune-dependent growth attenuation in melanoma and breast cancer models. We show that deletion of Ndufs4 induces expression of the major histocompatibility complex (MHC) class I co-activator Nlrc5 and antigen presentation machinery components, most notably H2-K1. This induction of MHC-related genes is driven by a pyruvate dehydrogenase-dependent accumulation of mitochondrial acetyl-CoA, which leads to an increase in histone H3K27 acetylation within the Nlrc5 and H2-K1 promoters. Taken together, this work shows that selective CI inhibition restricts tumor growth and that specific targeting of Ndufs4 or Ndufs6 increases T cell surveillance and ICB responsiveness.
Collapse
Affiliation(s)
- Jiaxin Liang
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Tevis Vitale
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Xixi Zhang
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Thomas D Jackson
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Deyang Yu
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Steve P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Hans R Widlund
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kai W Wucherpfennig
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Pere Puigserver
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA.
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
28
|
Komza M, Khatun J, Gelles JD, Trotta AP, Abraham-Enachescu I, Henao J, Elsaadi A, Kotini AG, Clementelli C, Arandela J, Ghaity-Beckley SE, Barua A, Chen Y, Berisa M, Marcellino BK, Papapetrou EP, Poyurovsky MV, Chipuk JE. Metabolic adaptations to acute glucose uptake inhibition converge upon mitochondrial respiration for leukemia cell survival. Cell Commun Signal 2025; 23:47. [PMID: 39863913 PMCID: PMC11762851 DOI: 10.1186/s12964-025-02044-y] [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: 11/20/2024] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
One hallmark of cancer is the upregulation and dependency on glucose metabolism to fuel macromolecule biosynthesis and rapid proliferation. Despite significant pre-clinical effort to exploit this pathway, additional mechanistic insights are necessary to prioritize the diversity of metabolic adaptations upon acute loss of glucose metabolism. Here, we investigated a potent small molecule inhibitor to Class I glucose transporters, KL-11743, using glycolytic leukemia cell lines and patient-based model systems. Our results reveal that while several metabolic adaptations occur in response to acute glucose uptake inhibition, the most critical is increased mitochondrial oxidative phosphorylation. KL-11743 treatment efficiently blocks the majority of glucose uptake and glycolysis, yet markedly increases mitochondrial respiration via enhanced Complex I function. Compared to partial glucose uptake inhibition, dependency on mitochondrial respiration is less apparent suggesting robust blockage of glucose uptake is essential to create a metabolic vulnerability. When wild-type and oncogenic RAS patient-derived induced pluripotent stem cell acute myeloid leukemia (AML) models were examined, KL-11743 mediated induction of mitochondrial respiration and dependency for survival associated with oncogenic RAS. Furthermore, we examined the therapeutic potential of these observations by treating a cohort of primary AML patient samples with KL-11743 and witnessed similar dependency on mitochondrial respiration for sustained cellular survival. Together, these data highlight conserved adaptations to acute glucose uptake inhibition in diverse leukemic models and AML patient samples, and position mitochondrial respiration as a key determinant of treatment success.
Collapse
MESH Headings
- Humans
- Mitochondria/metabolism
- Mitochondria/drug effects
- Glucose/metabolism
- Cell Survival/drug effects
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/drug therapy
- Cell Respiration/drug effects
- Glycolysis/drug effects
- Oxidative Phosphorylation/drug effects
- Cell Line, Tumor
- Adaptation, Physiological
Collapse
Affiliation(s)
- Monika Komza
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Present Address: Louis V. Gerstner, Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 441, New York, NY, 10065, USA
| | - Jesminara Khatun
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Jesse D Gelles
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Andrew P Trotta
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Ioana Abraham-Enachescu
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Juan Henao
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Ahmed Elsaadi
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Andriana G Kotini
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Cara Clementelli
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - JoAnn Arandela
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Sebastian El Ghaity-Beckley
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Agneesh Barua
- Department of Ecology and Evolution, University of Lausanne, Biophore, 1015, Lausanne, CH, Switzerland
| | - Yiyang Chen
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Mirela Berisa
- Metabolomics Core, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Bridget K Marcellino
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Eirini P Papapetrou
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Masha V Poyurovsky
- Kadmon Pharmaceuticals, 450 East 29th Street, New York, NY, 10016, USA
- Present address: PMV Pharmaceuticals, Inc., 1 Research Way, Princeton, NJ, 08540, USA
| | - Jerry Edward Chipuk
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA.
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA.
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA.
- The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA.
- The Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA.
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, Box 1130, 1425 Madison Avenue, New York, NY, 10029, USA.
| |
Collapse
|
29
|
Tjahjono E, Daneman MR, Meika B, Revtovich AV, Kirienko NV. Mitochondrial abnormalities as a target of intervention in acute myeloid leukemia. Front Oncol 2025; 14:1532857. [PMID: 39902131 PMCID: PMC11788353 DOI: 10.3389/fonc.2024.1532857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 12/27/2024] [Indexed: 02/05/2025] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive hematological malignancy; it is the most common acute leukemia in adults. AML prognosis is often poor, and relapse often occurs after initial remission. Recurrent genetic abnormalities underlying this disease and the presence of leukemic stem cells complicate disease treatment. However, the complex metabolic reprogramming that enables the unrestrained cell growth seen in these cells may also be their Achilles' heel. In these cells, mitophagy operates as a double-edged sword. On one hand, it provides a source of building blocks for further cell division and serves as a method for removing damaged organelles, promoting cell survival. However, the profound metabolic changes to mitochondria also render these organelles more sensitive to damage and place them precariously close to excess mitophagic activation. This review discusses the dual role mitophagy plays in AML survival, the importance of targeting mitophagy to treat AML, and current progress in the area. The discovery and mechanism of action of multiple compounds that were used to inhibit or stimulate mitophagy and their effects on AML survival are also described. Further, we explore the combination strategy of mitophagy-targeting compounds with existing and/or novel chemotherapeutics to eradicate AML and discuss strategies to uncover new drug targets and novel mitochondria-targeting drugs.
Collapse
|
30
|
Zhao XH, Han MM, Yan QQ, Yue YM, Ye K, Zhang YY, Teng L, Xu L, Shi XJ, La T, Feng YC, Xu R, Narayana VK, De Souza DP, Quek LE, Holst J, Liu T, Baker MA, Thorne RF, Zhang XD, Jin L. DNA replication stress underpins the vulnerability to oxidative phosphorylation inhibition in colorectal cancer. Cell Death Dis 2025; 16:16. [PMID: 39809754 PMCID: PMC11733219 DOI: 10.1038/s41419-025-07334-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 12/07/2024] [Accepted: 01/07/2025] [Indexed: 01/16/2025]
Abstract
Mitochondrial oxidative phosphorylation (OXPHOS) is a therapeutic vulnerability in glycolysis-deficient cancers. Here we show that inhibiting OXPHOS similarly suppresses the proliferation and tumorigenicity of glycolytically competent colorectal cancer (CRC) cells in vitro and in patient-derived CRC xenografts. While the increased glycolytic activity rapidly replenished the ATP pool, it did not restore the reduced production of aspartate upon OXPHOS inhibition. This shortage in aspartate, in turn, caused nucleotide deficiencies, leading to S phase cell cycle arrest, replication fork stalling, and enrichment of the p53 pathway, manifestations of replication stress. The addition of purine nucleobases adenine and guanine along with the pyrimidine nucleoside uridine restored replication fork progression and cell proliferation, whereas the supplementation of exogenous aspartate recovered the nucleotide pool, demonstrating a causal role of the aspartate shortage in OXPHOS inhibition-induced nucleotide deficiencies and consequently replication stress and reductions in proliferation. Moreover, we demonstrate that glutamic-oxaloacetic transaminase 1 (GOT1) is critical for maintaining the minimum aspartate pool when OXPHOS is inhibited, as knockdown of GOT1 further reduced aspartate levels and rendered CRC cells more sensitive to OXPHOS inhibition both in vitro and in vivo. These results propose GOT1 targeting as a potential avenue to sensitize cancer cells to OXPHOS inhibitors, thus lowering the necessary doses to efficiently inhibit cancer growth while alleviating their adverse effects.
Collapse
Affiliation(s)
- Xiao Hong Zhao
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, NSW, Australia
| | - Man Man Han
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Translational Research Institute, Henan Provincial and Zhengzhou City Key Laboratory of Noncoding RNA and Cancer Metabolism, Henan International Join Laboratory of Noncoding RNA and Metabolism in Cancer, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Qian Qian Yan
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Translational Research Institute, Henan Provincial and Zhengzhou City Key Laboratory of Noncoding RNA and Cancer Metabolism, Henan International Join Laboratory of Noncoding RNA and Metabolism in Cancer, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Yi Meng Yue
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Translational Research Institute, Henan Provincial and Zhengzhou City Key Laboratory of Noncoding RNA and Cancer Metabolism, Henan International Join Laboratory of Noncoding RNA and Metabolism in Cancer, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Kaihong Ye
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, NSW, Australia
- Translational Research Institute, Henan Provincial and Zhengzhou City Key Laboratory of Noncoding RNA and Cancer Metabolism, Henan International Join Laboratory of Noncoding RNA and Metabolism in Cancer, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Yuan Yuan Zhang
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, NSW, Australia
| | - Liu Teng
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, NSW, Australia
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Translational Research Institute, Henan Provincial and Zhengzhou City Key Laboratory of Noncoding RNA and Cancer Metabolism, Henan International Join Laboratory of Noncoding RNA and Metabolism in Cancer, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Liang Xu
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, NSW, Australia
| | - Xiao-Jing Shi
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Ting La
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, NSW, Australia
- National-Local Joint Engineering Research Center of Biodiagnosis & Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yu Chen Feng
- School of Medicine and Public Health, The University of Newcastle, Newcastle, NSW, Australia
| | - Ran Xu
- School of Medicine and Public Health, The University of Newcastle, Newcastle, NSW, Australia
| | - Vinod K Narayana
- Bio21 Institute and Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, Australia
- Metabolomics Australia, University of Melbourne, Parkville, VIC, Australia
| | - David P De Souza
- Bio21 Institute and Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, Australia
- Metabolomics Australia, University of Melbourne, Parkville, VIC, Australia
| | - Lake-Ee Quek
- School of Mathematics and Statistics, The University of Sydney, Sydney, NSW, Australia
| | - Jeff Holst
- School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Tao Liu
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Translational Research Institute, Henan Provincial and Zhengzhou City Key Laboratory of Noncoding RNA and Cancer Metabolism, Henan International Join Laboratory of Noncoding RNA and Metabolism in Cancer, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, Henan, China
- Children's Cancer Institute Australia for Medical Research, University of New South Wales, Sydney, NSW, Australia
| | - Mark A Baker
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, NSW, Australia
| | - Rick F Thorne
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, NSW, Australia
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Translational Research Institute, Henan Provincial and Zhengzhou City Key Laboratory of Noncoding RNA and Cancer Metabolism, Henan International Join Laboratory of Noncoding RNA and Metabolism in Cancer, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Xu Dong Zhang
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, NSW, Australia.
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
- Translational Research Institute, Henan Provincial and Zhengzhou City Key Laboratory of Noncoding RNA and Cancer Metabolism, Henan International Join Laboratory of Noncoding RNA and Metabolism in Cancer, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, Henan, China.
| | - Lei Jin
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
- Translational Research Institute, Henan Provincial and Zhengzhou City Key Laboratory of Noncoding RNA and Cancer Metabolism, Henan International Join Laboratory of Noncoding RNA and Metabolism in Cancer, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, Henan, China.
- School of Medicine and Public Health, The University of Newcastle, Newcastle, NSW, Australia.
| |
Collapse
|
31
|
Cao Y, Qian R, Yao R, Zheng Q, Yang C, Yang X, Ji S, Zhang L, Zhan S, Wang Y, Wang T, Wang H, Wong CM, Yuan S, Heeschen C, Gao Q, Bernards R, Qin W, Wang C. DYRK1A-TGF-β signaling axis determines sensitivity to OXPHOS inhibition in hepatocellular carcinoma. Dev Cell 2025:S1534-5807(24)00775-5. [PMID: 39798576 DOI: 10.1016/j.devcel.2024.12.035] [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: 04/05/2024] [Revised: 09/14/2024] [Accepted: 12/17/2024] [Indexed: 01/15/2025]
Abstract
Intervening in mitochondrial oxidative phosphorylation (OXPHOS) has emerged as a potential therapeutic strategy for certain types of cancers. Employing kinome-based CRISPR screen, we find that knockout of dual-specificity tyrosine phosphorylation-regulated kinase 1A (DYRK1A) synergizes with OXPHOS inhibitor IACS-010759 in liver cancer cells. Targeting DYRK1A combined with OXPHOS inhibitors activates TGF-β signaling, which is crucial for OXPHOS-inhibition-triggered cell death. Mechanistically, upregulation of glutamine transporter solute carrier family 1 member 5 (SLC1A5) transcription compensates for the increased glutamine requirement upon OXPHOS inhibition. DYRK1A directly phosphorylates SMAD3 Thr132, thereby suppressing the negative impact of TGF-β signaling on transcription of SLC1A5, leading to intrinsic resistance of liver cancer cells to OXPHOS inhibition. Moreover, we demonstrate the therapeutic efficacy of IACS-010759 in combination with DYRK1A inhibition in multiple liver cancer models, including xenografts, patient-derived xenografts, and spontaneous tumor model. Our study elucidates how the DYRK1A-TGF-β signaling axis controls the response of tumor cells to OXPHOS inhibition and provides valuable insights into targeting OXPHOS for liver cancer therapy.
Collapse
Affiliation(s)
- Ying Cao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruolan Qian
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruilian Yao
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Quan Zheng
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Yang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xupeng Yang
- Department of Liver Surgery and Transplantation, Key Laboratory of Carcinogenesis and Cancer Invasion, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shuyi Ji
- Department of Liver Surgery and Transplantation, Key Laboratory of Carcinogenesis and Cancer Invasion, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Linmen Zhang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shujie Zhan
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiping Wang
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianshi Wang
- Department of Nephrology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chun-Ming Wong
- State Key Laboratory for Liver Research and Department of Pathology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Shengxian Yuan
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Christopher Heeschen
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiang Gao
- Department of Liver Surgery and Transplantation, Key Laboratory of Carcinogenesis and Cancer Invasion, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - René Bernards
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
| | - Wenxin Qin
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Cun Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
32
|
Gamma JM, Liu Q, Beauchamp E, Iyer A, Yap MC, Zak Z, Ekstrom C, Pain R, Kostiuk MA, Mackey JR, Brandwein J, Wang JCY, Berthiaume LG. Zelenirstat Inhibits N-Myristoyltransferases to Disrupt Src Family Kinase Signaling and Oxidative Phosphorylation, Killing Acute Myeloid Leukemia Cells. Mol Cancer Ther 2025; 24:69-80. [PMID: 39382188 DOI: 10.1158/1535-7163.mct-24-0307] [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: 05/01/2024] [Revised: 08/14/2024] [Accepted: 09/27/2024] [Indexed: 10/10/2024]
Abstract
Acute myeloid leukemia (AML) is a hematologic malignancy with limited treatment options and a high likelihood of recurrence after chemotherapy. We studied N-myristoylation, the myristate modification of proteins linked to survival signaling and metabolism, as a potential therapeutic target for AML. N-myristoylation is catalyzed by two N-myristoyltransferases (NMT), NMT1 and NMT2, with varying expressions in AML cell lines and patient samples. We identified NMT2 expression as a marker for survival of patients with AML, and low NMT2 expression was associated with poor outcomes. We used the first-in-class pan-NMT inhibitor, zelenirstat, to investigate the role of N-myristoylation in AML. Zelenirstat effectively inhibits myristoylation in AML cell lines and patient samples, leading to degradation of Src family kinases, induction of endoplasmic reticulum stress, apoptosis, and cell death. Zelenirstat was well tolerated in vivo and reduced the leukemic burden in an ectopic AML cell line and in multiple orthotopic AML patient-derived xenograft models. The leukemia stem cell-enriched fractions of the hierarchical OCI-AML22 model were highly sensitive to myristoylation inhibition. Zelenirstat also impairs mitochondrial complex I and oxidative phosphorylation, which are critical for leukemia stem cell survival. These findings suggest that targeting N-myristoylation with zelenirstat represents a novel therapeutic approach for AML, with promise in patients with currently poor outcomes.
Collapse
Affiliation(s)
- Jay M Gamma
- Department of Medicine, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada
- Department of Cell Biology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada
| | - Qiang Liu
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Department of Medicine, University of Toronto, Toronto, Canada
| | | | - Aishwarya Iyer
- Department of Cell Biology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada
| | - Megan C Yap
- Department of Cell Biology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada
| | - Zoulika Zak
- Department of Cell Biology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada
| | - Cassidy Ekstrom
- Department of Cell Biology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada
| | - Rony Pain
- Department of Cell Biology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada
| | - Morris A Kostiuk
- Department of Cell Biology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada
| | | | - Joseph Brandwein
- Department of Medicine, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada
| | - Jean C Y Wang
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Department of Medicine, University of Toronto, Toronto, Canada
- Division of Medical Oncology and Hematology, Department of Medicine, University Health Network, Toronto, Canada
| | - Luc G Berthiaume
- Department of Cell Biology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada
- Pacylex Pharmaceuticals Inc., Edmonton, Canada
| |
Collapse
|
33
|
Magnitov MD, Maresca M, Alonso Saiz N, Teunissen H, Dong J, Sathyan KM, Braccioli L, Guertin MJ, de Wit E. ZNF143 is a transcriptional regulator of nuclear-encoded mitochondrial genes that acts independently of looping and CTCF. Mol Cell 2025; 85:24-41.e11. [PMID: 39708805 PMCID: PMC11687419 DOI: 10.1016/j.molcel.2024.11.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 09/23/2024] [Accepted: 11/22/2024] [Indexed: 12/23/2024]
Abstract
Gene expression is orchestrated by transcription factors, which function within the context of a three-dimensional genome. Zinc-finger protein 143 (ZNF143/ZFP143) is a transcription factor that has been implicated in both gene activation and chromatin looping. To study the direct consequences of ZNF143/ZFP143 loss, we generated a ZNF143/ZFP143 depletion system in mouse embryonic stem cells. Our results show that ZNF143/ZFP143 degradation has no effect on chromatin looping. Systematic analysis of ZNF143/ZFP143 occupancy data revealed that a commonly used antibody cross-reacts with CTCF, leading to its incorrect association with chromatin loops. Nevertheless, ZNF143/ZFP143 specifically activates nuclear-encoded mitochondrial genes, and its loss leads to severe mitochondrial dysfunction. Using an in vitro embryo model, we find that ZNF143/ZFP143 is an essential regulator of organismal development. Our results establish ZNF143/ZFP143 as a conserved transcriptional regulator of cell proliferation and differentiation by safeguarding mitochondrial activity.
Collapse
Affiliation(s)
- Mikhail D Magnitov
- Division of Gene Regulation, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Michela Maresca
- Division of Gene Regulation, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands; Department of Clinical Genetics, Erasmus University MC, Dr. Molewaterplein 40, 3015 GD Rotterdam, the Netherlands
| | - Noemí Alonso Saiz
- Division of Gene Regulation, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Hans Teunissen
- Division of Gene Regulation, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Jinhong Dong
- Center for Cell Analysis and Modeling, University of Connecticut, 400 Farmington Avenue, Farmington, CT, USA
| | - Kizhakke M Sathyan
- Center for Cell Analysis and Modeling, University of Connecticut, 400 Farmington Avenue, Farmington, CT, USA; Department of Genetics and Genome Sciences, University of Connecticut, 400 Farmington Avenue, Farmington, CT, USA
| | - Luca Braccioli
- Division of Gene Regulation, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Michael J Guertin
- Center for Cell Analysis and Modeling, University of Connecticut, 400 Farmington Avenue, Farmington, CT, USA; Department of Genetics and Genome Sciences, University of Connecticut, 400 Farmington Avenue, Farmington, CT, USA
| | - Elzo de Wit
- Division of Gene Regulation, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands.
| |
Collapse
|
34
|
Komza M, Chipuk JE. Mitochondrial metabolism: A moving target in hepatocellular carcinoma therapy. J Cell Physiol 2025; 240:e31441. [PMID: 39324415 PMCID: PMC11732733 DOI: 10.1002/jcp.31441] [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: 05/01/2024] [Revised: 08/21/2024] [Accepted: 09/10/2024] [Indexed: 09/27/2024]
Abstract
Mitochondria are pivotal contributors to cancer mechanisms due to their homeostatic and pathological roles in cellular bioenergetics, biosynthesis, metabolism, signaling, and survival. During transformation and tumor initiation, mitochondrial function is often disrupted by oncogenic mutations, leading to a metabolic profile distinct from precursor cells. In this review, we focus on hepatocellular carcinoma, a cancer arising from metabolically robust and nutrient rich hepatocytes, and discuss the mechanistic impact of altered metabolism in this setting. We provide distinctions between normal mitochondrial activity versus disease-related function which yielded therapeutic opportunities, along with highlighting recent preclinical and clinical efforts focused on targeting mitochondrial metabolism. Finally, several novel strategies for exploiting mitochondrial programs to eliminate hepatocellular carcinoma cells in metabolism-specific contexts are presented to integrate these concepts and gain foresight into the future of mitochondria-focused therapeutics.
Collapse
Affiliation(s)
- Monika Komza
- Louis V. Gerstner, Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jerry Edward Chipuk
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Icahn School of Medicine at Mount Sinai, The Tisch Cancer Institute, New York, New York, USA
- The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Icahn School of Medicine at Mount Sinai, The Diabetes, Obesity, and Metabolism Institute, New York, New York, USA
| |
Collapse
|
35
|
Zhang M, Dong Y, Meng R, Zhang D. SMARCA4 Deficiency in Lung Cancer: From Signaling Pathway to Potential Therapeutic Targets. Genes Chromosomes Cancer 2025; 64:e70022. [PMID: 39812394 DOI: 10.1002/gcc.70022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Accepted: 12/27/2024] [Indexed: 01/16/2025] Open
Abstract
SMARCA4-deficient lung cancer, including thoracic SMARCA4-deficient undifferentiated tumors and SMARCA4-deficient nonsmall-cell lung carcinomas, is a rare and aggressive disease characterized by rapid progression and poor prognosis. This cancer was identified as a distinct entity with specific morphologic and molecular features in the 2021 WHO Classification of Thoracic Tumors. Molecular alterations in SMARCA4 are specific to this type of lung cancer. Deficiency in SMARCA4 suppresses the SWI/SNF tumor suppressor complex, driving tumor progression. Due to the lack of standardized treatment, most patients succumb to the disease within 6 months. This study provides a detailed analysis of the SMARCA4 pathway and its role in lung cancer. We analyzed potential therapeutic targets and agents to offer insights for the precise and effective treatment of SMARCA4-deficient lung cancer.
Collapse
Affiliation(s)
- Mingzhu Zhang
- Department of Oncology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, China
| | - Youhong Dong
- Department of Oncology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, China
| | - Rui Meng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dongdong Zhang
- Department of Oncology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, China
| |
Collapse
|
36
|
Corrêa-Ferreira ML, do Rocio Andrade Pires A, Miranda JV, de Freitas Montin E, Barbosa IR, Lima AEAN, Rocha MEM, Martinez GR, Cadena SMSC. The Mesoionic 1,3,4-thiadiazolium Derivative, MI-D, is a Potential Drug for Treating Glioblastoma by Impairing Mitochondrial Functions Linked to Energy Provision in Glioma Cells. Anticancer Agents Med Chem 2025; 25:411-419. [PMID: 39440773 DOI: 10.2174/0118715206329159241010052746] [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: 06/13/2024] [Revised: 09/15/2024] [Accepted: 09/19/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND Mesoionic compound MI-D possesses important biological activities, such as antiinflammatory and antitumoral against melanoma and hepatocarcinoma. Glioblastoma is the most aggressive and common central nervous system tumor in adults. Currently, chemotherapies are not entirely effective, and the survival of patients diagnosed with glioblastoma is extremely short. OBJECTIVE In this study, we aimed to evaluate the cytotoxicity of MI-D in noninvasive A172 glioblastoma cells and establish which changes in functions linked to energy provision are associated with this effect. METHODS Cells A172 were cultured under glycolysis and phosphorylation oxidative conditions and evaluated: viability by the MTT method, oxygen consumption by high-resolution respirometry, levels of pyruvate, lactate, citrate, and ATP, and glutaminase and citrate synthase activities by spectrophotometric methods. RESULTS Under glycolysis-dependent conditions, MI-D caused significant cytotoxic effects with impaired cell respiration, reducing the maximal capacity of the electron transport chain. However, A172 cells were more susceptible to MI-D effects under oxidative phosphorylation-dependent conditions. At the IC25, inhibition of basal and maximal respiration of A172 cells was observed, without stimulation of the glycolytic pathway or Krebs cycle, along with inhibition of the activity of glutaminase enzyme, resulting in a 30% ATP deficit. Additionally, independent of metabolic conditions, MI-D treatment induced cell death in A172 cells by apoptosis machinery/ processes. CONCLUSION The impairment of mitochondrial respiration by MI-D under the condition sustained by oxidative phosphorylation may enhance the cytotoxic effect on A172 glioma cells, although the mechanism of cell death relies on apoptosis.
Collapse
Affiliation(s)
| | | | - Juan Vitor Miranda
- Department of Biochemistry and Molecular Biology, Federal University of Paraná, Curitiba, Brazil
| | | | - Igor Resendes Barbosa
- Department of Chemistry, Federal Rural University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | - Glaucia Regina Martinez
- Department of Biochemistry and Molecular Biology, Federal University of Paraná, Curitiba, Brazil
| | | |
Collapse
|
37
|
Ngoi NYL, Gallo D, Torrado C, Nardo M, Durocher D, Yap TA. Synthetic lethal strategies for the development of cancer therapeutics. Nat Rev Clin Oncol 2025; 22:46-64. [PMID: 39627502 DOI: 10.1038/s41571-024-00966-z] [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: 11/01/2024] [Indexed: 12/20/2024]
Abstract
Synthetic lethality is a genetic phenomenon whereby the simultaneous presence of two different genetic alterations impairs cellular viability. Importantly, targeting synthetic lethal interactions offers potential therapeutic strategies for cancers with alterations in pathways that might otherwise be considered undruggable. High-throughput screening methods based on modern CRISPR-Cas9 technologies have emerged and become crucial for identifying novel synthetic lethal interactions with the potential for translation into biologically rational cancer therapeutic strategies as well as associated predictive biomarkers of response capable of guiding patient selection. Spurred by the clinical success of PARP inhibitors in patients with BRCA-mutant cancers, novel agents targeting multiple synthetic lethal interactions within DNA damage response pathways are in clinical development, and rational strategies targeting synthetic lethal interactions spanning alterations in epigenetic, metabolic and proliferative pathways have also emerged and are in late preclinical and/or early clinical testing. In this Review, we provide a comprehensive overview of established and emerging technologies for synthetic lethal drug discovery and development and discuss promising therapeutic strategies targeting such interactions.
Collapse
Affiliation(s)
- Natalie Y L Ngoi
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - David Gallo
- Repare Therapeutics, Inc., Montreal, Quebec, Canada
| | - Carlos Torrado
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mirella Nardo
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Daniel Durocher
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Timothy A Yap
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Therapeutics Discovery Division, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Khalifa Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
38
|
Tejero JD, Hesterberg RS, Drapela S, Ilter D, Raizada D, Lazure F, Kashfi H, Liu M, Silvane L, Avram D, Fernández-García J, Asara JM, Fendt SM, Cleveland JL, Gomes AP. Methylmalonic acid induces metabolic abnormalities and exhaustion in CD8 + T cells to suppress anti-tumor immunity. Oncogene 2025; 44:105-114. [PMID: 39472497 DOI: 10.1038/s41388-024-03191-1] [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: 02/28/2024] [Revised: 10/04/2024] [Accepted: 10/08/2024] [Indexed: 01/11/2025]
Abstract
Systemic levels of methylmalonic acid (MMA), a byproduct of propionate metabolism, increase with age and MMA promotes tumor progression via its direct effects in tumor cells. However, the role of MMA in modulating the tumor ecosystem remains to be investigated. The proliferation and function of CD8+ T cells, key anti-tumor immune cells, declines with age and in conditions of vitamin B12 deficiency, which are the two most well-established conditions that lead to increased systemic levels of MMA. Thus, we hypothesized that increased circulatory levels of MMA would lead to a suppression of CD8+ T cell immunity. Treatment of primary CD8+ T cells with MMA induced a dysfunctional phenotype characterized by robust immunosuppressive transcriptional reprogramming and marked increases in the expression of the exhaustion regulator, TOX. Accordingly, MMA treatment upregulated exhaustion markers in CD8+ T cells and decreased their effector functions, which drove the suppression of anti-tumor immunity in vitro and in vivo. Mechanistically, MMA-induced CD8+ T cell exhaustion was associated with a suppression of NADH-regenerating reactions in the TCA cycle and concomitant defects in mitochondrial function. Thus, MMA has immunomodulatory roles, thereby highlighting MMA as an important link between aging, immune dysfunction, and cancer.
Collapse
Affiliation(s)
- Joanne D Tejero
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
- Department of Molecular Medicine, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA
| | - Rebecca S Hesterberg
- Department of Tumor Microenvironment and Metastasis, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Stanislav Drapela
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Didem Ilter
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Devesh Raizada
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Felicia Lazure
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Hossein Kashfi
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Min Liu
- Proteomics and Metabolomics Core, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Leonardo Silvane
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Dorina Avram
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Juan Fernández-García
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Herestraat 49, 3000, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Herestraat 49, 3000, Leuven, Belgium
| | - John M Asara
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, 02215, USA
| | - Sarah-Maria Fendt
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Herestraat 49, 3000, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Herestraat 49, 3000, Leuven, Belgium
| | - John L Cleveland
- Department of Tumor Microenvironment and Metastasis, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Ana P Gomes
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA.
| |
Collapse
|
39
|
Zhao Z, Zhu L, Luo Y, Xu H, Zhang Y. Collateral lethality: A unique type of synthetic lethality in cancers. Pharmacol Ther 2025; 265:108755. [PMID: 39581504 DOI: 10.1016/j.pharmthera.2024.108755] [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: 08/10/2024] [Revised: 10/31/2024] [Accepted: 11/19/2024] [Indexed: 11/26/2024]
Abstract
Genetic interactions play crucial roles in cell-essential functions. Intrinsic genetic defects in tumors typically involve gain-of- and loss-of-function mutations in tumor suppressor genes (TSGs) and oncogenes, respectively, providing potential antitumor vulnerabilities. Moreover, tumor cells with TSG deficiencies exhibit heightened sensitivity to the inhibition of compensatory pathways. Synthetic and collateral lethality are two strategies used for exploiting novel drug targets in multiple types of cancer. Collateral lethality is a unique type of synthetic lethality that occurs when passenger genes are co-deleted in neighboring TSGs. Although synthetic lethality has already been successfully demonstrated in clinical practice, antitumor therapeutics based on collateral lethality are predominantly still in the preclinical phase. Therefore, screening for potential genetic interactions within the cancer genome has emerged as a promising approach for drug development. Here, the two conceptual therapeutic strategies that involve the deletion or inactivation of cancer-specific TSGs are discussed. Moreover, existing approaches for screening and identifying potential gene partners are also discussed. Particularly, this review highlights the current advances of "collateral lethality" in the preclinical phase and addresses the challenges involved in translating them into therapeutic applications. This review provides insights into these strategies as new opportunities for the development of personalized antitumor therapies.
Collapse
Affiliation(s)
- Zichen Zhao
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China; Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Lingling Zhu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China; Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Luo
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Heng Xu
- Institute of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Laboratory Medicine/Research Center of Clinical Laboratory Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yan Zhang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China; Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
40
|
Rumiano L, Manzo T. Lipids guide T cell antitumor immunity by shaping their metabolic and functional fitness. Trends Endocrinol Metab 2024:S1043-2760(24)00321-7. [PMID: 39743401 DOI: 10.1016/j.tem.2024.11.014] [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: 10/01/2024] [Revised: 11/15/2024] [Accepted: 11/27/2024] [Indexed: 01/04/2025]
Abstract
Lipids are metabolic messengers essential for energy production, membrane structure, and signal transduction. Beyond their recognized role, lipids have emerged as metabolic rheostats of T cell responses, with distinct species differentially modulating CD8+ T cell (CTL) fate and function. Indeed, lipids can influence T cell signaling by altering their membrane composition; in addition, they can affect the differentiation path of T cells through cellular metabolism. This Review discusses the ability of lipids to shape T cell phenotypes and functions. Based on this link between lipid metabolism, metabolic fitness and immunosurveillance, we suggest that lipid could be rationally integrated in the context of immunotherapies to fine-tune fitness and function of adoptive T cell therapy (ACT) products.
Collapse
Affiliation(s)
- Letizia Rumiano
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Teresa Manzo
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy.
| |
Collapse
|
41
|
Zhu S, Chen C, Wang M, Liu Y, Li B, Qi X, Song M, Liu X, Feng J, Liu J. Pan-cancer association of a mitochondrial function score with genomic alterations and clinical outcome. Sci Rep 2024; 14:31430. [PMID: 39733076 PMCID: PMC11682264 DOI: 10.1038/s41598-024-83022-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 12/11/2024] [Indexed: 12/30/2024] Open
Abstract
Mitochondria are pivotal in cellular energy metabolism and have garnered significant attention for their roles in cancer progression and therapy resistance. Despite this, the functional diversity of mitochondria across various cancer types remains inadequately characterized. This study seeks to fill this knowledge gap by introducing and validating MitoScore-a novel metric designed to quantitatively assess mitochondrial function across a wide array of cancers. Our investigation evaluates the capacity of MitoScore not only to distinguish between tumor and adjacent normal tissues but also to serve as a predictive marker for clinical outcomes. We analyzed gene expression data from 24 cancer types and corresponding normal tissues using the TCGA database. MitoScore was calculated by summing the normalized expression levels of six mitochondrial genes known to be consistently altered across multiple cancers. Differential gene expression was assessed using DESeq2, with a focus on identifying significant changes in mitochondrial function. MitoScore's associations with tumor proliferation, hypoxia, aneuploidy, and clinical outcomes were evaluated using Spearman's correlation, linear regression, and Kaplan-Meier survival analyses. MitoScore was significantly higher in tumor tissues compared to normal tissues across most cancer types (p < 0.001). It positively correlated with tumor proliferation rates (r = 0.46), hypoxia scores (r = 0.61), and aneuploidy (r = 0.44), indicating its potential as a marker of aggressive tumor behavior. High MitoScore was also associated with poorer prognosis in several cancer types, suggesting its utility as a predictive biomarker for clinical outcomes. This study introduces MitoScore, a metric for mitochondrial activity often elevated in tumors and linked to poor prognosis. It correlates positively with hypoxia and negatively with stromal and immune infiltration, highlighting mitochondria's role in the tumor microenvironment. MitoScore's association with genomic instability, such as aneuploidy, suggests mitochondrial dysfunction contributes to cancer progression. Despite challenges in mitochondrial-targeted therapies, MitoScore may identify tumors responsive to such treatments, warranting further research for clinical application.
Collapse
Affiliation(s)
- Shikun Zhu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, China
| | - Chen Chen
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, China
| | - Min Wang
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, China
| | - Yue Liu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, China
| | - Baolin Li
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, China
| | - Xing Qi
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, China
- Ziyang People's Hospital, Ziyang, Sichaun, China
| | - Miao Song
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, China
| | - Xuexue Liu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, China
| | - Jia Feng
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, China.
| | - Jinbo Liu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, China.
| |
Collapse
|
42
|
Wang Y, Hu W, Zhou B, Zhao Y, Tang Y, Deng Z, Chen M. Mitochondrial transcription elongation factor TEFM promotes malignant progression of gliomas. Cancer Cell Int 2024; 24:429. [PMID: 39719635 DOI: 10.1186/s12935-024-03617-6] [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: 09/17/2024] [Accepted: 12/13/2024] [Indexed: 12/26/2024] Open
Abstract
Gliomas are the most common tumors of the central nervous system, with glioblastoma (GBM) being particularly aggressive and fatal. Current treatments for GBM, including surgery and chemotherapy, are limited by tumor aggressiveness and the blood-brain barrier. Therefore, understanding the molecular mechanisms driving GBM growth is essential. Mitochondria, key players in cellular energy production, have been implicated in cancer development. In this study, we investigated the expression of mitochondrial transcription elongation factor (TEFM) in gliomas and its potential role in tumor progression. Analysis of data from The Cancer Genome Atlas (TCGA) revealed that TEFM transcript levels were significantly higher in glioma tissues compared to adjacent normal tissues. High TEFM expression was associated with poor survival outcomes in glioma patients. Furthermore, TEFM was notably upregulated in glioma tissue and in primary glioma cells derived from local patients, while its expression was relatively low in normal tissues and astrocytes. Silencing or knockout of TEFM significantly inhibited glioma cell growth, proliferation, clonogenicity, migration, and invasion, while inducing apoptosis and activating caspases. In contrast, ectopic overexpression of TEFM promoted tumorigenic activity, enhancing the malignant behavior of glioma cells. Co-expression analysis identified a strong correlation between TEFM and the epithelial-mesenchymal transition (EMT) pathway in gliomas. Notably, the expression of EMT markers, such as N-cadherin and Vimentin, decreased upon TEFM knockdown or knockout. Additionally, TEFM depletion impaired mitochondrial function, disrupting the mitochondrial respiratory chain in glioma cells. In vivo experiments demonstrated that TEFM knockout effectively suppressed the growth of subcutaneous glioma xenografts in nude mice. Collectively, these findings highlight the critical role of TEFM in GBM growth and invasion, suggesting that it could serve as a promising therapeutic target for glioma treatment.
Collapse
Affiliation(s)
- Yin Wang
- Institute for Excellence in Clinical Medicine of Kunshan First People's Hospital, Soochow University, Suzhou, China
| | - Wenxuan Hu
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Boya Zhou
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Yu Zhao
- Department of Soochow Medical College, Soochow University, Suzhou, China
| | - Yufei Tang
- Department of Soochow Medical College, Soochow University, Suzhou, China
| | - Zhiyong Deng
- Institute for Excellence in Clinical Medicine of Kunshan First People's Hospital, Soochow University, Suzhou, China.
| | - Minbin Chen
- Institute for Excellence in Clinical Medicine of Kunshan First People's Hospital, Soochow University, Suzhou, China.
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China.
| |
Collapse
|
43
|
Reczek CR, Chakrabarty RP, D'Alessandro KB, Sebo ZL, Grant RA, Gao P, Budinger GR, Chandel NS. Metformin targets mitochondrial complex I to lower blood glucose levels. SCIENCE ADVANCES 2024; 10:eads5466. [PMID: 39693440 DOI: 10.1126/sciadv.ads5466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 11/19/2024] [Indexed: 12/20/2024]
Abstract
Metformin is among the most prescribed antidiabetic drugs, but the primary molecular mechanism by which metformin lowers blood glucose levels is unknown. Previous studies have proposed numerous mechanisms by which acute metformin lowers blood glucose, including the inhibition of mitochondrial complex I of the electron transport chain (ETC). Here, we used transgenic mice that globally express the Saccharomyces cerevisiae internal alternative NADH dehydrogenase (NDI1) protein to determine whether the glucose-lowering effect of acute oral administration of metformin requires inhibition of mitochondrial complex I of the ETC in vivo. NDI1 is a yeast NADH dehydrogenase enzyme that complements the loss of mammalian mitochondrial complex I electron transport function and is insensitive to pharmacologic mitochondrial complex I inhibitors including metformin. We demonstrate that NDI1 expression attenuates metformin's ability to lower blood glucose levels under standard chow and high-fat diet conditions. Our results indicate that acute oral administration of metformin targets mitochondrial complex I to lower blood glucose.
Collapse
Affiliation(s)
- Colleen R Reczek
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ram P Chakrabarty
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Karis B D'Alessandro
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Zachary L Sebo
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Rogan A Grant
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Peng Gao
- Robert H. Lurie Cancer Center Metabolomics Core, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - G R Budinger
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Navdeep S Chandel
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Chan Zuckerberg Biohub, Chicago, IL, USA
| |
Collapse
|
44
|
Gao W, Song Y, Wu F, Xu S, Liu B, Zeng L, Zheng E, Song H, Zhang Q. Tumor-Targeted Metal-Organic Framework for Improved Photodynamic Therapy and Inhibited Tumor Metastasis in Melanoma. ACS APPLIED MATERIALS & INTERFACES 2024; 16:69769-69788. [PMID: 39652639 DOI: 10.1021/acsami.4c18058] [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: 12/20/2024]
Abstract
Tumor hypoxia and elevated intracellular glutathione (GSH) levels significantly compromise the effectiveness of photodynamic therapy (PDT) in treating melanoma. In this study, we synthesized positively charged nanoparticles through a self-assembly method, incorporating photosensitizer verteporfin (VER), mitochondrial respiratory inhibitor atovaquone (ATO), and Fe3+. Subsequently, the nanoparticles were modified with sodium hyaluronate (HA) to obtain HA-ATO-Fe3+-VER nanoparticles (HAFV NPs). The fabricated HAFV NPs demonstrated excellent stability and in vitro Fenton reaction activity. HA facilitated the cellular internalization of HAFV NPs by targeting CD44 receptors, hence relieving tumor hypoxia through the disruption of the mitochondrial respiratory chain and involvement in the Fenton reaction. Simultaneously, ATO directly impeded the biosynthesis of GSH by diminishing ATP levels, while Fe3+ was supposed to oxidate GSH to GSSG, thereby doubly depleting GSH. The integration of these multiple mechanisms markedly enhanced the PDT efficacy of VER. Following intravenous administration, HAFV NPs preferentially accumulated in tumor tissues with minimal accumulation in the skin, demonstrating favorable biocompatibility in vivo. Furthermore, HAFV NPs effectively inhibited tumor growth and lung metastasis, which presents a promising strategy for melanoma treatment.
Collapse
Affiliation(s)
- Wenhao Gao
- School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
- Department of Pharmacy, The 900th Hospital of Joint Logistic Support Force PLA, Fuzhou 350025, China
| | - Yutong Song
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Fei Wu
- School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
- Department of Pharmacy, The 900th Hospital of Joint Logistic Support Force PLA, Fuzhou 350025, China
| | - Shiting Xu
- School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
- Department of Pharmacy, The 900th Hospital of Joint Logistic Support Force PLA, Fuzhou 350025, China
| | - Bin Liu
- School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Lingjun Zeng
- Department of Pharmacy, The 900th Hospital of Joint Logistic Support Force PLA, Fuzhou 350025, China
| | - Enqin Zheng
- Department of Pharmacy, The 900th Hospital of Joint Logistic Support Force PLA, Fuzhou 350025, China
- School of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Hongtao Song
- School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
- Department of Pharmacy, The 900th Hospital of Joint Logistic Support Force PLA, Fuzhou 350025, China
| | - Qian Zhang
- School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| |
Collapse
|
45
|
Aisu Y, Oshima N, Hyodo F, Elhelaly AE, Masuo A, Okada T, Hisamori S, Tsunoda S, Hida K, Morimoto T, Miyoshi H, Taketo MM, Matsuo M, Neckers LM, Sakai Y, Obama K. Dual inhibition of oxidative phosphorylation and glycolysis exerts a synergistic antitumor effect on colorectal and gastric cancer by creating energy depletion and preventing metabolic switch. PLoS One 2024; 19:e0309700. [PMID: 39666615 PMCID: PMC11637386 DOI: 10.1371/journal.pone.0309700] [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: 04/03/2024] [Accepted: 08/18/2024] [Indexed: 12/14/2024] Open
Abstract
Pyruvate is situated at the intersection of oxidative phosphorylation (OXPHOS) and glycolysis, which are the primary energy-producing pathways in cells. Cancer therapies targeting these pathways have been previously documented, indicating that inhibiting one pathway may lead to functional compensation by the other, resulting in an insufficient antitumor effect. Thus, effective cancer treatment necessitates concurrent and comprehensive suppression of both. However, whether a metabolic switch between the metabolic pathways occurs in colorectal and gastric cancer cells and whether blocking it by inhibiting both pathways has an antitumor effect remain to be determined. In the present study, we used two small molecules, namely OXPHOS and glycolysis inhibitors, to target pyruvate metabolic pathways as a cancer treatment in these cancer cells. OXPHOS and glycolysis inhibition each augmented the other metabolic pathway in vitro and in vivo. OXPHOS inhibition alone enhanced glycolysis and showed antitumor effects on colorectal and gastric cancer cells in vitro and in vivo. Moreover, glycolysis inhibition in addition to OXPHOS inhibition blocked the metabolic switch from OXPHOS to glycolysis, causing an energy depletion and deterioration of the tumor microenvironment that synergistically enhanced the antitumor effect of OXPHOS inhibitors. In addition, using hyperpolarized 13C-magnetic resonance spectroscopic imaging (HP-MRSI), which enables real-time and in vivo monitoring of molecules containing 13C, we visualized how the inhibitors shifted the flux of pyruvate and how this dual inhibition in colorectal and gastric cancer mouse models altered the two pathways. Integrating dual inhibition of OXPHOS and glycolysis with HP-MRSI, this therapeutic model shows promise as a future "cancer theranostics" treatment option.
Collapse
Affiliation(s)
- Yuki Aisu
- Department of Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Nobu Oshima
- Department of Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Surgery, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Fuminori Hyodo
- Department of Radiology, Gifu University Hospital, Gifu, Japan
- Department of Radiology, Frontier Science for Imaging, Gifu University, Gifu, Japan
| | | | - Akihiko Masuo
- Department of Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tomoaki Okada
- Department of Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shigeo Hisamori
- Department of Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shigeru Tsunoda
- Department of Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Koya Hida
- Department of Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tomonori Morimoto
- Department of Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hiroyuki Miyoshi
- Colon Cancer Project, Kyoto University Hospital-iACT, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Makoto M. Taketo
- Colon Cancer Project, Kyoto University Hospital-iACT, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masayuki Matsuo
- Department of Radiology, Gifu University Hospital, Gifu, Japan
| | - Leonard M. Neckers
- National Cancer Institute, Urologic Oncology Branch, Center for Cancer Research, NIH, Bethesda, Maryland, United States of America
| | - Yoshiharu Sakai
- Department of Gastrointestinal Surgery, Osaka Red Cross Hospital, Osaka, Japan
| | - Kazutaka Obama
- Department of Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
46
|
Piao S, Kim S, Vu GH, Kim M, Lee EO, Jeon BH, Kim CS. The Downregulation of CRIF1 Exerts Antitumor Effects Partially via TP53-Induced Glycolysis and Apoptosis Regulator Induction in BT549 Breast Cancer Cells. Cancers (Basel) 2024; 16:4081. [PMID: 39682267 DOI: 10.3390/cancers16234081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/20/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND/OBJECTIVES Mitochondrial oxidative phosphorylation (OXPHOS) has been exploited as a therapeutic target in cancer treatments because of its crucial role in tumorigenesis. CR6-interacting factor 1 (CRIF1), a mitochondrial ribosomal subunit protein, is essential for the regulation of mitochondrial OXPHOS capacity. However, the mechanism of CRIF1 in triple-negative breast cancer (TNBC) cells remains unclear. METHODS/RESULTS We showed that the downregulation of CRIF1 reduced cell proliferation in the TNBC cell lines MDA-MB-468, MDA-MB-231, and, especially, BT549. In addition, wound scratch and Transwell assays showed that CRIF1 deficiency inhibited the migration and invasion of BT549 cells. CRIF1 downregulation resulted in the suppression of mitochondrial bioenergetics in BT549 cells, specifically affecting the inhibition of OXPHOS complexes I and II. This was evidenced by a decrease in the mitochondrial oxygen consumption rate and the depolarization of the mitochondrial membrane potential. Damage to mitochondria resulted in a lower adenosine triphosphate level and an elevated production of mitochondrial reactive oxygen species. In addition, CRIF1 deficiency decreased hypoxia-inducible factor 1α accumulation, NADPH synthesis, and TP53-induced glycolysis and apoptosis regulator (TIGAR) expression in BT549 cells. These events contributed to G0/G1-phase cell cycle inhibition and the upregulation of the cell cycle protein markers p53, p21, and p16. Transfection with a TIGAR overexpression plasmid reversed these effects and prevented CRIF1 downregulation-induced proliferation and migration reduction. CONCLUSIONS These results indicate that blocking mitochondrial OXPHOS synthesis via CRIF1 may have a therapeutic antitumor effect in BT549 TNBC cells.
Collapse
Affiliation(s)
- Shuyu Piao
- Department of Physiology & Medical Science, College of Medicine, Chungnam National University, Daejeon 301-747, Republic of Korea
| | - Seonhee Kim
- Department of Physiology & Medical Science, College of Medicine, Chungnam National University, Daejeon 301-747, Republic of Korea
| | - Giang-Huong Vu
- Department of Physiology & Medical Science, College of Medicine, Chungnam National University, Daejeon 301-747, Republic of Korea
| | - Minsoo Kim
- Department of Physiology & Medical Science, College of Medicine, Chungnam National University, Daejeon 301-747, Republic of Korea
| | - Eun-Ok Lee
- Department of Physiology & Medical Science, College of Medicine, Chungnam National University, Daejeon 301-747, Republic of Korea
| | - Byeong Hwa Jeon
- Department of Physiology & Medical Science, College of Medicine, Chungnam National University, Daejeon 301-747, Republic of Korea
| | - Cuk-Seong Kim
- Department of Physiology & Medical Science, College of Medicine, Chungnam National University, Daejeon 301-747, Republic of Korea
| |
Collapse
|
47
|
Varuzhanyan G, Chen CC, Freeland J, He T, Tran W, Song K, Wang L, Cheng D, Xu S, Dibernardo GA, Esedebe FN, Bhatia V, Han M, Abt ER, Park JW, Memarzadeh S, Shackelford DB, Lee JK, Graeber TG, Shirihai OS, Witte ON. PGC-1α drives small cell neuroendocrine cancer progression toward an ASCL1-expressing subtype with increased mitochondrial capacity. Proc Natl Acad Sci U S A 2024; 121:e2416882121. [PMID: 39589879 PMCID: PMC11626175 DOI: 10.1073/pnas.2416882121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 10/25/2024] [Indexed: 11/28/2024] Open
Abstract
Adenocarcinomas from multiple tissues can converge to treatment-resistant small cell neuroendocrine (SCN) cancers composed of ASCL1, POU2F3, NEUROD1, and YAP1 subtypes. We investigated how mitochondrial metabolism influences SCN cancer (SCNC) progression. Extensive bioinformatics analyses encompassing thousands of patient tumors and human cancer cell lines uncovered enhanced expression of proliferator-activatedreceptor gamma coactivator 1-alpha (PGC-1α), a potent regulator of mitochondrial oxidative phosphorylation (OXPHOS), across several SCNCs. PGC-1α correlated tightly with increased expression of the lineage marker Achaete-scute homolog 1, (ASCL1) through a positive feedback mechanism. Analyses using a human prostate tissue-based SCN transformation system showed that the ASCL1 subtype has heightened PGC-1α expression and OXPHOS activity. PGC-1α inhibition diminished OXPHOS, reduced SCNC cell proliferation, and blocked SCN prostate tumor formation. Conversely, PGC-1α overexpression enhanced OXPHOS, validated by small-animal Positron Emission Tomography mitochondrial imaging, tripled the SCN prostate tumor formation rate, and promoted commitment to the ASCL1 lineage. These results establish PGC-1α as a driver of SCNC progression and subtype determination, highlighting metabolic vulnerabilities in SCNCs across different tissues.
Collapse
Affiliation(s)
- Grigor Varuzhanyan
- Department of Microbiology Immunology and Molecular Genetics, University of California, Los Angeles, CA90095
| | - Chia-Chun Chen
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA90095
| | - Jack Freeland
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA90095
- Molecular Biology Interdepartmental Program, University of California, Los Angeles, CA90095
| | - Tian He
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA90095
| | - Wendy Tran
- Department of Microbiology Immunology and Molecular Genetics, University of California, Los Angeles, CA90095
| | - Kai Song
- Department of Bioengineering, University of California, Los Angeles, CA90095
| | - Liang Wang
- Department of Microbiology Immunology and Molecular Genetics, University of California, Los Angeles, CA90095
| | - Donghui Cheng
- Department of Microbiology Immunology and Molecular Genetics, University of California, Los Angeles, CA90095
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA90095
| | - Shili Xu
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA90095
- Crump Institute for Molecular Imaging, David Geffen School of Medicine, University of California, Los Angeles, CA90095
- Jonsson Comprehensive Cancer Center, the David Geffen School of Medicine, University of California, Los Angeles, CA90095
| | - Gabriella A. Dibernardo
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA90095
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, CA90095
| | - Favour N. Esedebe
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA90095
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, CA90095
| | - Vipul Bhatia
- Division of Hematology/Oncology, Department of Medicine University of California Los Angeles Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA90095
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA98109
| | - Mingqi Han
- Department of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA90095
| | - Evan R. Abt
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA90095
| | - Jung Wook Park
- Department of Pathology, Duke University School of Medicine, Durham, NC27710
| | - Sanaz Memarzadeh
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA90095
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA90095
- Jonsson Comprehensive Cancer Center, the David Geffen School of Medicine, University of California, Los Angeles, CA90095
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, CA90095
- The Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA90073
- Molecular Biology Institute, University of California, Los Angeles, CA90095
| | - David B. Shackelford
- Jonsson Comprehensive Cancer Center, the David Geffen School of Medicine, University of California, Los Angeles, CA90095
- Department of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA90095
| | - John K. Lee
- Division of Hematology/Oncology, Department of Medicine University of California Los Angeles Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA90095
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA98109
| | - Thomas G. Graeber
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA90095
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA90095
- Crump Institute for Molecular Imaging, David Geffen School of Medicine, University of California, Los Angeles, CA90095
- Jonsson Comprehensive Cancer Center, the David Geffen School of Medicine, University of California, Los Angeles, CA90095
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, CA90095
- California NanoSystems Institute, University of California, Los Angeles, CA90095
- UCLA Metabolomics Center, University of California, Los Angeles, CA90095
| | - Orian S. Shirihai
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA90095
- Molecular Biology Interdepartmental Program, University of California, Los Angeles, CA90095
- University of California Los Angeles Division of Endocrinology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA90095
- Department of Clinical Biochemistry, School of Medicine, Ben Gurion University of The Negev, Beer-Sheva8410501, Israel
| | - Owen N. Witte
- Department of Microbiology Immunology and Molecular Genetics, University of California, Los Angeles, CA90095
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA90095
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA90095
- Jonsson Comprehensive Cancer Center, the David Geffen School of Medicine, University of California, Los Angeles, CA90095
- Molecular Biology Institute, University of California, Los Angeles, CA90095
- Parker Institute for Cancer Immunotherapy, University of California, Los Angeles, CA90095
| |
Collapse
|
48
|
Liaghat M, Ferdousmakan S, Mortazavi SH, Yahyazadeh S, Irani A, Banihashemi S, Seyedi Asl FS, Akbari A, Farzam F, Aziziyan F, Bakhtiyari M, Arghavani MJ, Zalpoor H, Nabi-Afjadi M. The impact of epithelial-mesenchymal transition (EMT) induced by metabolic processes and intracellular signaling pathways on chemo-resistance, metastasis, and recurrence in solid tumors. Cell Commun Signal 2024; 22:575. [PMID: 39623377 PMCID: PMC11610171 DOI: 10.1186/s12964-024-01957-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 11/22/2024] [Indexed: 12/06/2024] Open
Abstract
The intricate cellular process, known as the epithelial-mesenchymal transition (EMT), significantly influences solid tumors development. Changes in cell shape, metabolism, and gene expression linked to EMT facilitate tumor cell invasion, metastasis, drug resistance, and recurrence. So, a better understanding of the intricate processes underlying EMT and its role in tumor growth may lead to the development of novel therapeutic approaches for the treatment of solid tumors. This review article focuses on the signals that promote EMT and metabolism, the intracellular signaling pathways leading to EMT, and the network of interactions between EMT and cancer cell metabolism. Furthermore, the functions of EMT in treatment resistance, recurrence, and metastasis of solid cancers are covered. Lastly, treatment approaches that focus on intracellular signaling networks and metabolic alterations brought on by EMT will be discussed.
Collapse
Affiliation(s)
- Mahsa Liaghat
- Department of Medical Laboratory Sciences, Faculty of Medical Sciences, Kazerun Branch, Islamic Azad University, Kazerun, Iran
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Saeid Ferdousmakan
- Department of Pharmacy Practice, Nargund College of Pharmacy, Bangalore, 560085, India
| | | | - Sheida Yahyazadeh
- Department of Immunology, Faculty of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Asrin Irani
- Department of Biology, Faculty of Basic Sciences, University of Guilan, Rasht, Iran
| | - Sara Banihashemi
- Department of Bioscience, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | | | - Abdullatif Akbari
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farnoosh Farzam
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Aziziyan
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Maryam Bakhtiyari
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
- Department of Medical Laboratory Sciences, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Mohammad Javad Arghavani
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Hamidreza Zalpoor
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran.
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mohsen Nabi-Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
49
|
Kenny TC, Birsoy K. Mitochondria and Cancer. Cold Spring Harb Perspect Med 2024; 14:a041534. [PMID: 38692736 PMCID: PMC11610758 DOI: 10.1101/cshperspect.a041534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2024]
Abstract
Mitochondria are semiautonomous organelles with diverse metabolic and cellular functions including anabolism and energy production through oxidative phosphorylation. Following the pioneering observations of Otto Warburg nearly a century ago, an immense body of work has examined the role of mitochondria in cancer pathogenesis and progression. Here, we summarize the current state of the field, which has coalesced around the position that functional mitochondria are required for cancer cell proliferation. In this review, we discuss how mitochondria influence tumorigenesis by impacting anabolism, intracellular signaling, and the tumor microenvironment. Consistent with their critical functions in tumor formation, mitochondria have become an attractive target for cancer therapy. We provide a comprehensive update on the numerous therapeutic modalities targeting the mitochondria of cancer cells making their way through clinical trials.
Collapse
Affiliation(s)
- Timothy C Kenny
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, New York, New York 10065, USA
| | - Kıvanç Birsoy
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, New York, New York 10065, USA
| |
Collapse
|
50
|
Tatarata QZ, Wang Z, Konopleva M. BCL-2 inhibition in acute myeloid leukemia: resistance and combinations. Expert Rev Hematol 2024; 17:935-946. [PMID: 39552410 DOI: 10.1080/17474086.2024.2429604] [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: 06/19/2024] [Accepted: 11/11/2024] [Indexed: 11/19/2024]
Abstract
INTRODUCTION The introduction of venetoclax has revolutionized the treatment landscape of acute myeloid leukemia, offering new therapeutic opportunities. However, the clinical response to venetoclax varies significantly between patients, with many experiencing limited duration of response. AREAS COVERED Identified resistance mechanisms include both intrinsic and acquired resistance to VEN. The former is associated with cell lineage and differentiation state. The latter includes dependency on alternative BCL-2 family anti-apoptotic protein(s) mediated by genetic, epigenetic, or post-translational mechanisms, mitochondrial and metabolic involvement, as well as microenvironment. Understanding these mechanisms is crucial for optimizing venetoclax-based therapies and enhancing treatment outcomes for patients with acute myeloid leukemia. This review aims to elucidate the primary mechanisms underlying resistance to venetoclax and explore current therapeutic strategies to overcome this challenge. EXPERT OPINION In patients with venetoclax resistance, alternative options include targeted combination therapies tailored to individual cases based on cytogenetics and prior treatments. Many of these therapies require further clinical investigation to validate their safety and efficacy.
Collapse
Affiliation(s)
- Qi Zhang Tatarata
- The Department of Leukemia, The University of Texas MD, Anderson Cancer Center, Houston, TX, USA
- The Department of Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Zhe Wang
- The Department of Leukemia, The University of Texas MD, Anderson Cancer Center, Houston, TX, USA
| | - Marina Konopleva
- The Department of Leukemia, The University of Texas MD, Anderson Cancer Center, Houston, TX, USA
- Department of Oncology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|