1
|
Li Z, Zou Y, Niu J, Zhang Y, Yang A, Lin W, Guo J, Wang S, Liu R. IMPDH2's Central Role in Cellular Growth and Diseases: A Potential Therapeutic Target. Cell Prolif 2025:e70031. [PMID: 40251939 DOI: 10.1111/cpr.70031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/13/2025] [Accepted: 03/18/2025] [Indexed: 04/21/2025] Open
Abstract
IMPDH2 is a rate-limiting enzyme in guanine nucleotide biosynthesis. It plays diverse roles in various physiological and pathological processes: nucleotide metabolism, inflammation, immune function, ribosomal stress. Structural or regulatory alterations in IMPDH2 are linked to significant health issues, and critical relevance in disease progression. We aim to underscore the potential of IMPDH2 as a promising therapeutic target for clinical applications.
Collapse
Affiliation(s)
- Zheng Li
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yunpeng Zou
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Jiayao Niu
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Ying Zhang
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Aohua Yang
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wenyu Lin
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Jie Guo
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Shuya Wang
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Ronghan Liu
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
2
|
Huang D, Wang J, Chen L, Jiang W, Inuzuka H, Simon DK, Wei W. Molecular Subtypes and Targeted Therapeutic Strategies in Small Cell Lung Cancer: Advances, Challenges, and Future Perspectives. Molecules 2025; 30:1731. [PMID: 40333678 PMCID: PMC12029361 DOI: 10.3390/molecules30081731] [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: 02/21/2025] [Revised: 04/09/2025] [Accepted: 04/10/2025] [Indexed: 05/09/2025] Open
Abstract
Small cell lung cancer (SCLC) is a highly aggressive malignancy characterized by rapid progression, early metastasis, and high recurrence rates. Historically considered a homogeneous disease, recent multi-omic studies have revealed distinct molecular subtypes driven by lineage-defining transcription factors, including ASCL1, NEUROD1, POU2F3, and YAP1, as well as an inflamed subtype (SCLC-I). These subtypes exhibit unique therapeutic vulnerabilities, thereby paving the way for precision medicine and targeted therapies. Despite recent advances in molecular classification, tumor heterogeneity, plasticity, and therapy resistance continue to hinder clinical success in treating SCLC patients. To this end, novel therapeutic strategies are being explored, including BCL2 inhibitors, DLL3-targeting agents, Aurora kinase inhibitors, PARP inhibitors, and epigenetic modulators. Additionally, immune checkpoint inhibitors (ICIs) show promise, particularly in immune-enriched subtypes of SCLC patients. Hence, a deeper understanding of SCLC subtype characteristics, evolution, and the regulatory mechanisms of subtype-specific transcription factors is crucial for rationally optimizing precision therapy. This knowledge not only facilitates the identification of subtype-specific therapeutic targets, but also provides a foundation for overcoming resistance and developing personalized combination treatment strategies. In the future, the integration of multi-omic data, dynamic molecular monitoring, and precision medicine approaches are expected to further advance the clinical translation of SCLC subtype-specific therapies, ultimately improving patient survival and outcomes.
Collapse
Affiliation(s)
- Daoyuan Huang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Jingchao Wang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Li Chen
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Weiwei Jiang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Hiroyuki Inuzuka
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - David K. Simon
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA;
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
3
|
Simpson KL, Rothwell DG, Blackhall F, Dive C. Challenges of small cell lung cancer heterogeneity and phenotypic plasticity. Nat Rev Cancer 2025:10.1038/s41568-025-00803-0. [PMID: 40211072 DOI: 10.1038/s41568-025-00803-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/19/2025] [Indexed: 04/12/2025]
Abstract
Small cell lung cancer (SCLC) is an aggressive neuroendocrine malignancy with ~7% 5-year overall survival reflecting early metastasis and rapid acquired chemoresistance. Immunotherapy briefly extends overall survival in ~15% cases, yet predictive biomarkers are lacking. Targeted therapies are beginning to show promise, with a recently approved delta-like ligand 3 (DLL3)-targeted therapy impacting the treatment landscape. The increased availability of patient-faithful models, accumulating human tumour biobanks and numerous comprehensive molecular profiling studies have collectively facilitated the mapping and understanding of substantial intertumoural and intratumoural heterogeneity. Beyond the almost ubiquitous loss of wild-type p53 and RB1, SCLC is characterized by heterogeneously mis-regulated expression of MYC family members, yes-associated protein 1 (YAP1), NOTCH pathway signalling, anti-apoptotic BCL2 and epigenetic regulators. Molecular subtypes are based on the neurogenic transcription factors achaete-scute homologue 1 (ASCL1) and neurogenic differentiation factor 1 (NEUROD1), the rarer non-neuroendocrine transcription factor POU class 2 homeobox 3 (POU2F3), and immune- and inflammation-related signatures. Furthermore, SCLC shows phenotypic plasticity, including neuroendocrine-to-non-neuroendocrine transition driven by NOTCH signalling, which is associated with disease progression, chemoresistance and immune modulation and, in mouse models, with metastasis. Although these features pose substantial challenges, understanding the molecular vulnerabilities of transcription factor subtypes, the functional relevance of plasticity and cell cooperation offer opportunities for personalized therapies informed by liquid and tissue biomarkers.
Collapse
Affiliation(s)
- Kathryn L Simpson
- SCLC Biology Group, Cancer Research UK Manchester Institute, Manchester, UK
- CRUK National Biomarker Centre, University of Manchester, Manchester, UK
- CRUK Lung Cancer Centre of Excellence, Manchester, UK
| | - Dominic G Rothwell
- CRUK National Biomarker Centre, University of Manchester, Manchester, UK
- CRUK Lung Cancer Centre of Excellence, Manchester, UK
| | - Fiona Blackhall
- CRUK Lung Cancer Centre of Excellence, Manchester, UK
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Medical Oncology, Christie Hospital National Health Service, Foundation Trust, Manchester, UK
| | - Caroline Dive
- SCLC Biology Group, Cancer Research UK Manchester Institute, Manchester, UK.
- CRUK National Biomarker Centre, University of Manchester, Manchester, UK.
- CRUK Lung Cancer Centre of Excellence, Manchester, UK.
| |
Collapse
|
4
|
Chang CC, Peng M, Keppeke GD, Tsai LK, Zhang Z, Pai LM, Sung LY, Liu JL. Y12C mutation disrupts IMPDH cytoophidia and alters cancer metabolism. FEBS J 2025. [PMID: 40186514 DOI: 10.1111/febs.70086] [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: 03/06/2024] [Revised: 12/17/2024] [Accepted: 03/25/2025] [Indexed: 04/07/2025]
Abstract
Guanosine triphosphate (GTP) is a building block for DNA and RNA, and plays a pivotal role in various cellular functions, serving as an energy source, enzyme cofactor and a key component of signal transduction. The activity of the rate-limiting enzyme in de novo GTP synthesis, inosine monophosphate dehydrogenase (IMPDH), is regulated by nucleotide binding. Recent studies have illuminated that IMPDH octamers can assemble into linear polymers, adding another dimension to its enzymatic regulation. This polymerisation reduces IMPDH's sensitivity to the inhibitory effects of GTP binding, thereby augmenting its activity under conditions with elevated GTP levels. Within cells, IMPDH polymers may cluster to form the distinctive structure known as the cytoophidium, which is postulated to reflect the cellular demand for increased GTP concentrations. Nevertheless, the functional significance of IMPDH polymerisation in in vivo metabolic regulation remains unclear. In this study, we report the widespread presence of IMPDH cytoophidia in various human cancer tissues. Utilising the ABEmax base editor, we introduced a Y12C point mutation into IMPDH2 across multiple cancer cell lines. This mutation disrupts the polymerisation interface of IMPDH and prevents cytoophidium assembly. In some cancer xenografts, the absence of IMPDH polymers led to a downregulation of IMPDH, as well as the glycolytic and pentose phosphate pathways. Furthermore, mutant HeLa-cell-derived xenografts were notably smaller than their wild-type counterparts. Our data suggest that IMPDH polymerisation and cytoophidium assembly could be instrumental in modulating metabolic homeostasis in certain cancers, offering insights into the clinical relevance of IMPDH cytoophidium.
Collapse
Affiliation(s)
- Chia-Chun Chang
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Min Peng
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Gerson Dierley Keppeke
- School of Life Science and Technology, ShanghaiTech University, China
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
| | - Li-Kuang Tsai
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Ziheng Zhang
- School of Life Science and Technology, ShanghaiTech University, China
| | - Li-Mei Pai
- Department of Biochemistry and Molecular Biology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Li-Ying Sung
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
- Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan
- Center for Biotechnology, National Taiwan University, Taipei, Taiwan
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan
| | - Ji-Long Liu
- School of Life Science and Technology, ShanghaiTech University, China
- Department of Physiology, Anatomy and Genetics, University of Oxford, UK
| |
Collapse
|
5
|
Solta A, Ernhofer B, Boettiger K, Lang C, Megyesfalvi Z, Mendrina T, Kirchhofer D, Timelthaler G, Szeitz B, Rezeli M, Aigner C, Haschemi A, Unger LW, Dome B, Schelch K. Unveiling the powerhouse: ASCL1-driven small cell lung cancer is characterized by higher numbers of mitochondria and enhanced oxidative phosphorylation. Cancer Metab 2025; 13:16. [PMID: 40165271 PMCID: PMC11959836 DOI: 10.1186/s40170-025-00382-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 03/01/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Small cell lung cancer (SCLC) is an aggressive malignancy with distinct molecular subtypes defined by transcription factors and inflammatory characteristics. This follow-up study aimed to validate the unique metabolic phenotype in achaete-scute homologue 1 (ASCL1)-driven SCLC cell lines and human tumor tissue. METHODS Metabolic alterations were analyzed using proteomic data. Structural and functional differences of mitochondria were investigated using qPCR, flow cytometry, confocal imaging, and transmission electron microscopy and seahorse assays. Several metabolic inhibitors were tested using MTT-based and clonogenic assays. Single-cell enzyme activity assays were conducted on cell lines and tumor tissue samples of SCLC patients. RESULTS We found increased mitochondrial numbers correlating with higher oxidative phosphorylation activity in ASCL1-dominant cells compared to other SCLC subtypes. Metabolic inhibitors targeting mitochondrial respiratory complex-I or carnitine palmitoyltransferase 1 revealed higher responsiveness in SCLC-A. Conversely, we demonstrated that non-ASCL1-driven SCLCs with lower oxidative signatures show dependence on glutaminolysis as evidenced by the enhanced susceptibility to glutaminase inhibition. Accordingly, we detected increased glutamate-dehydrogenase activity in non-ASCL1-dominant cell lines as well as in human SCLC tissue samples. CONCLUSIONS Distinct SCLC subtypes exhibit unique metabolic vulnerabilities, suggesting potential for subtype-specific therapies targeting the respiratory chain, fatty acid transport, or glutaminolysis.
Collapse
Grants
- FWF No. T 1062-B33, FWF I3522, FWF I3977 and I4677, Sonderforschungsbereich F83 Austrian Science Fund
- FWF No. T 1062-B33, FWF I3522, FWF I3977 and I4677, Sonderforschungsbereich F83 Austrian Science Fund
- FWF No. T 1062-B33, FWF I3522, FWF I3977 and I4677, Sonderforschungsbereich F83 Austrian Science Fund
- FWF No. T 1062-B33, FWF I3522, FWF I3977 and I4677, Sonderforschungsbereich F83 Austrian Science Fund
- 2020-1.1.6-JÖVŐ, TKP2021-EGA-33, FK-143751 and FK-147045, UNKP-20-3, UNKP-21-3 and UNKP-23-5, ÚNKP-22-3-II Nemzeti Kutatási Fejlesztési és Innovációs Hivatal
- 2020-1.1.6-JÖVŐ, TKP2021-EGA-33, FK-143751 and FK-147045, UNKP-20-3, UNKP-21-3 and UNKP-23-5, ÚNKP-22-3-II Nemzeti Kutatási Fejlesztési és Innovációs Hivatal
- 2020-1.1.6-JÖVŐ, TKP2021-EGA-33, FK-143751 and FK-147045, UNKP-20-3, UNKP-21-3 and UNKP-23-5, ÚNKP-22-3-II Nemzeti Kutatási Fejlesztési és Innovációs Hivatal
- EFOP-3.6.3-VEKOP-16-2017-00009 Semmelweis Egyetem
- FBKS-2020-22-(291) Fru Berta Kamprads Stiftelse
- 101131228-BIOSMALL HORIZON EUROPE Framework Programme
Collapse
Affiliation(s)
- Anna Solta
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, A-1090, Austria
| | - Büsra Ernhofer
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, A-1090, Austria
| | - Kristiina Boettiger
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, A-1090, Austria
| | - Christian Lang
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, A-1090, Austria
- Division of Pulmonology, Department of Medicine II, Medical University of Vienna, Vienna, Austria
| | - Zsolt Megyesfalvi
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, A-1090, Austria
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary
- National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - Theresa Mendrina
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Dominik Kirchhofer
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Gerald Timelthaler
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Beata Szeitz
- National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - Melinda Rezeli
- Department of Biomedical Engineering, Lund University, Lund, Sweden
| | - Clemens Aigner
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, A-1090, Austria
| | - Arvand Haschemi
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Lukas W Unger
- Deptartment of Colorectal Surgery, Oxford University Hospitals, Oxford, UK
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Balazs Dome
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, A-1090, Austria.
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary.
- National Koranyi Institute of Pulmonology, Budapest, Hungary.
- Department of Translational Medicine, Lund University, Lund, Sweden.
| | - Karin Schelch
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, A-1090, Austria.
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
6
|
An L, Peng W, Yang Y, Chen G, Luo QT, Ni M, Wang X, Fu Y, Zhou Y, Liu X. Emilia sonchifolia (L.) DC. inhibits the growth of Methicillin-Resistant Staphylococcus epidermidis by modulating its physiology through multiple mechanisms. Sci Rep 2025; 15:9779. [PMID: 40119097 PMCID: PMC11928732 DOI: 10.1038/s41598-025-93561-w] [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: 09/11/2024] [Accepted: 03/07/2025] [Indexed: 03/24/2025] Open
Abstract
Bloodstream infections (BSIs) are a public health concern, causing substantial morbidity and mortality. Staphylococcus epidermidis (S. epidermidis) is a leading cause BSIs. Antibiotics targeting S. epidermidis have been the mainstay of treatment for BSIs, however their efficacy is diminishing in combating with drug-resistant bacteria. Therefore, alternative treatments for antibiotic-resistant infections are urgently required. Studies have demonstrated that certain traditional Chinese medicine (TCM) exhibit notable antimicrobial activity and can help mitigate bacterial resistance. Among these, The ethanol extract of Emilia sonchifolia (L.) DC (E. sonchifolia) (10 g crude drug/1 g extract ) exhibits a noteworthy anti-methicillin-resistant S. epidermidis (MRSE) effect. This study explores antibacterial activity and underlying mechanisms of E. sonchifolia against MRSE. The antibacterial activity of E. sonchifolia against MRSE was assessed in vitro by measuring the minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC). The MRSE-induced mouse BSIs model was used to evaluate the antibacterial activity of E. sonchifolia in vivo. Proteomic and transcriptomic analyses were performed to elucidate the underlying antibacterial mechanisms. The MIC and MBC values of E. sonchifolia against MRSE were 5 mg/mL and 20 mg/mL, respectively. In vivo, E. sonchifolia effectively treated MRSE-induced BSIs. Additionally, proteomic and transcriptomic analyses revealed considerable down-regulation of purine metabolism, that were associated with oxidative stress and cell wall synthesis. The enzyme linked immunosorbent assay(ELISA) results showed decreased levels of inosine monophosphate (IMP), Adenosine monophosphate(AMP) and guanine monophosphate (GMP), indicating inhibited purine metabolism. Scanning electron microscopy (SEM) and transmission electron microscopy (TEM) analysis confirmed bacterial cell wall damage. E. sonchifolia exerts antibacterial effects by inhibiting purine metabolism, promoting bacterial oxidative stress, and impairing cell wall synthesis. These findings provide novel insights into the mechanistic understanding of E. sonchifolia's efficacy against MRSE, offering potential strategies for managing MRSE infections.
Collapse
Affiliation(s)
- Lili An
- Dermatology Department, First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Wei Peng
- College of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Yuqi Yang
- College of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Gongzhen Chen
- Dermatology Department, First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Qian Tonghan Luo
- College of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Meng Ni
- Dermatology Department, First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Xuebing Wang
- College of Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Yufeng Fu
- College of Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Yonghui Zhou
- College of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Xin Liu
- College of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China.
| |
Collapse
|
7
|
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
|
8
|
Schneider JL, Kurmi K, Dai Y, Dhiman I, Joshi S, Gassaway BM, Johnson CW, Jones N, Li Z, Joschko CP, Fujino T, Paulo JA, Yoda S, Baquer G, Ruiz D, Stopka SA, Kelley L, Do A, Mino-Kenudson M, Sequist LV, Lin JJ, Agar NYR, Gygi SP, Haigis KM, Hata AN, Haigis MC. GUK1 activation is a metabolic liability in lung cancer. Cell 2025; 188:1248-1264.e23. [PMID: 39919745 DOI: 10.1016/j.cell.2025.01.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/14/2024] [Accepted: 01/15/2025] [Indexed: 02/09/2025]
Abstract
Little is known about metabolic vulnerabilities in oncogene-driven lung cancer. Here, we perform a phosphoproteomic screen in anaplastic lymphoma kinase (ALK)-rearranged ("ALK+") patient-derived cell lines and identify guanylate kinase 1 (GUK1), a guanosine diphosphate (GDP)-synthesizing enzyme, as a target of ALK signaling in lung cancer. We demonstrate that ALK binds to and phosphorylates GUK1 at tyrosine 74 (Y74), resulting in increased GDP biosynthesis. Spatial imaging of ALK+ patient tumor specimens shows enhanced phosphorylation of GUK1 that significantly correlates with guanine nucleotides in situ. Abrogation of GUK1 phosphorylation reduces intracellular GDP and guanosine triphosphate (GTP) pools and decreases mitogen-activated protein kinase (MAPK) signaling and Ras-GTP loading. A GUK1 variant that cannot be phosphorylated (Y74F) decreases tumor proliferation in vitro and in vivo. Beyond ALK, other oncogenic fusion proteins in lung cancer also regulate GUK1 phosphorylation. These studies may pave the way for the development of new therapeutic approaches by exploiting metabolic dependencies in oncogene-driven lung cancers.
Collapse
Affiliation(s)
- Jaime L Schneider
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA; Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA, USA.
| | - Kiran Kurmi
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Yutong Dai
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA; Department of Chemistry and Chemical Biology, Harvard University, Boston, MA, USA
| | - Ishita Dhiman
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Shakchhi Joshi
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Brandon M Gassaway
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | | | - Nicole Jones
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Zongyu Li
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Christian P Joschko
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Toshio Fujino
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Joao A Paulo
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Satoshi Yoda
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Gerard Baquer
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Daniela Ruiz
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sylwia A Stopka
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Liam Kelley
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Andrew Do
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Mari Mino-Kenudson
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Lecia V Sequist
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Jessica J Lin
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Nathalie Y R Agar
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Steven P Gygi
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Kevin M Haigis
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Aaron N Hata
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Marcia C Haigis
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
9
|
Xu Y, Jiang X, Hu Z. Synergizing metabolomics and artificial intelligence for advancing precision oncology. Trends Mol Med 2025:S1471-4914(25)00016-4. [PMID: 39956738 DOI: 10.1016/j.molmed.2025.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 02/18/2025]
Abstract
Metabolomics has emerged as a transformative tool in precision oncology, with substantial potential for advancing biomarker discovery, monitoring treatment responses, and aiding drug development. Integrating artificial intelligence (AI) into metabolomics optimizes data acquisition and analysis, facilitating the interpretation of complex metabolic networks and enabling more effective multiomics integration. In this opinion, we explore recent advances in the application of metabolomics within precision oncology, emphasizing the unique advantages that AI-driven metabolomics offers. We propose that AI not only complements but also amplifies the potential of current platforms, accelerating research progress and ultimately improving patient outcomes. Finally, we discuss the opportunities and challenges involved in translating AI-driven metabolomics into clinical practice for precision oncology.
Collapse
Affiliation(s)
- Yipeng Xu
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Xiaojuan Jiang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Zeping Hu
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China; Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
10
|
Tasca CI, Zuccarini M, Di Iorio P, Ciruela F. Lessons from the physiological role of guanosine in neurodegeneration and cancer: Toward a multimodal mechanism of action? Purinergic Signal 2025; 21:133-148. [PMID: 39004650 PMCID: PMC11958862 DOI: 10.1007/s11302-024-10033-y] [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: 03/07/2024] [Accepted: 06/18/2024] [Indexed: 07/16/2024] Open
Abstract
Neurodegenerative diseases and brain tumours represent important health challenges due to their severe nature and debilitating consequences that require substantial medical care. Interestingly, these conditions share common physiological characteristics, namely increased glutamate, and adenosine transmission, which are often associated with cellular dysregulation and damage. Guanosine, an endogenous nucleoside, is safe and exerts neuroprotective effects in preclinical models of excitotoxicity, along with cytotoxic effects on tumour cells. However, the lack of well-defined mechanisms of action for guanosine hinders a comprehensive understanding of its physiological effects. In fact, the absence of specific receptors for guanosine impedes the development of structure-activity research programs to develop guanosine derivatives for therapeutic purposes. Alternatively, given its apparent interaction with the adenosinergic system, it is plausible that guanosine exerts its neuroprotective and anti-tumorigenic effects by modulating adenosine transmission through undisclosed mechanisms involving adenosine receptors, transporters, and purinergic metabolism. Here, several potential molecular mechanisms behind the protective actions of guanosine will be discussed. First, we explore its potential interaction with adenosine receptors (A1R and A2AR), including the A1R-A2AR heteromer. In addition, we consider the impact of guanosine on extracellular adenosine levels and the role of guanine-based purine-converting enzymes. Collectively, the diverse cellular functions of guanosine as neuroprotective and antiproliferative agent suggest a multimodal and complementary mechanism of action.
Collapse
Affiliation(s)
- Carla Inês Tasca
- Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, SC, Brazil.
- Laboratory of Neurochemistry-4, Neuroscience Program/Biochemistry Program, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, SC, Brazil.
| | - Mariachiara Zuccarini
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, 66100, Chieti, Italy
- Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, 66100, Chieti, Italy
| | - Patrizia Di Iorio
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, 66100, Chieti, Italy
- Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, 66100, Chieti, Italy
| | - Francisco Ciruela
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08907 L'Hospitalet de Llobregat, Barcelona, Spain
- Neuropharmacology & Pain Group, Neuroscience Program, Bellvitge Institute for Biomedical Research, 08907L'Hospitalet de Llobregat, Bellvitge, Spain
| |
Collapse
|
11
|
Zhou B, Zhao Q, Hou G, He J, Sha N, Zheng K, Peng H, Wang W, Zhou Y, Chen T, Jiang Y. IMPDH2 dephosphorylation under FGFR signaling promotes S-phase progression and tumor growth. Cell Rep 2025; 44:115116. [PMID: 39739531 DOI: 10.1016/j.celrep.2024.115116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 07/06/2024] [Accepted: 12/05/2024] [Indexed: 01/02/2025] Open
Abstract
Inosine monophosphate dehydrogenase 2 (IMPDH2) is highly expressed in human cancers; however, its physiological relevance under growth signaling remains to be investigated. Here, we show that IMPDH2 serine 122 is phosphorylated by CDK1, and this modification attenuates the catalytic activity of IMPDH2 for IMP oxidation and simultaneously represses its allosteric modulation by purine nucleotides. Fibroblast growth factor receptor (FGFR) signaling activation triggers IMPDH2-Ser122 dephosphorylation mediated by protein phosphatase 2A (PP2A), which is dependent on FGFR3-mediated PPP2R1A-Tyr261 phosphorylation leading to PPP2CA-PPP2R1A-IMPDH2 interactions. In turn, Ser122 dephosphorylation positively modulates IMPDH2 activity and contributes to guanine nucleotide synthesis and purine homeostasis, thereby facilitating S-phase completion and cell proliferation. Accordingly, IMPDH2 dephosphorylation is implicated in FGFR activation-enhanced tumorigenesis, and the low level of IMPDH2-Ser122 phosphorylation predicts the poor prognosis of patients with colorectal cancer. These findings illustrate a regulatory mechanism of purine nucleotide production under FGFR signaling, in which the oncogenic effect of reinforced IMPDH2 activity is underscored.
Collapse
Affiliation(s)
- Bei Zhou
- Department of Liver Surgery and Shanghai Cancer Institute, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qin Zhao
- Department of Liver Surgery and Shanghai Cancer Institute, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guofang Hou
- Department of Liver Surgery and Shanghai Cancer Institute, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing He
- Department of Liver Surgery and Shanghai Cancer Institute, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Nannan Sha
- Department of Liver Surgery and Shanghai Cancer Institute, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ke Zheng
- Department of Liver Surgery and Shanghai Cancer Institute, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongyu Peng
- Department of Liver Surgery and Shanghai Cancer Institute, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wang Wang
- Department of Liver Surgery and Shanghai Cancer Institute, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yue Zhou
- Department of Liver Surgery and Shanghai Cancer Institute, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tao Chen
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuhui Jiang
- Department of Liver Surgery and Shanghai Cancer Institute, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China.
| |
Collapse
|
12
|
da Silva Fernandes T, Gillard BM, Dai T, Martin JC, Chaudhry KA, Dugas SM, Fisher AA, Sharma P, Wu R, Attwood KM, Dasgupta S, Takabe K, Rosario SR, Bianchi-Smiraglia A. Inosine monophosphate dehydrogenase 2 (IMPDH2) modulates response to therapy and chemo-resistance in triple negative breast cancer. Sci Rep 2025; 15:1061. [PMID: 39774345 PMCID: PMC11707137 DOI: 10.1038/s41598-024-85094-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 12/31/2024] [Indexed: 01/11/2025] Open
Abstract
Triple negative breast cancer (TNBC) is one of the deadliest subtypes of breast cancer, whose high frequency of relapse is often due to resistance to chemotherapy. Here, we identify inosine monophosphate dehydrogenase 2 (IMPDH2) as a contributor to doxorubicin resistance, in multiple TNBC models. Analysis of publicly available datasets reveals elevated IMPDH2 expression to associate with worse overall TNBC prognosis in the clinic, including lower recurrence-free survival post adjuvant/neoadjuvant therapy. Importantly, both genetic depletion and pharmacological inhibition of IMPDH2 leads to reduction of pro-tumorigenic phenotypes in multiple doxorubicin-resistant TNBC models, both in vitro and in vivo. Overall, we propose IMPDH2 as a novel vulnerability that could be leveraged therapeutically to suppress and/or prevent the growth of chemo-resistant lesions.
Collapse
Affiliation(s)
- Tatiane da Silva Fernandes
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, CGP L3-317, Buffalo, NY, 14263, USA
| | - Bryan M Gillard
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Tao Dai
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, CGP L3-317, Buffalo, NY, 14263, USA
| | - Jeffrey C Martin
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, CGP L3-317, Buffalo, NY, 14263, USA
| | - Kanita A Chaudhry
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, CGP L3-317, Buffalo, NY, 14263, USA
| | - Scott M Dugas
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, CGP L3-317, Buffalo, NY, 14263, USA
| | - Alyssa A Fisher
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, CGP L3-317, Buffalo, NY, 14263, USA
| | - Pia Sharma
- Department of Breast Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - RongRong Wu
- Department of Breast Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Kristopher M Attwood
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, RSC R-410, Buffalo, NY, 14263, USA
| | - Subhamoy Dasgupta
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, CGP L3-317, Buffalo, NY, 14263, USA
| | - Kazuaki Takabe
- Department of Breast Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Spencer R Rosario
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, RSC R-410, Buffalo, NY, 14263, USA.
| | - Anna Bianchi-Smiraglia
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, CGP L3-317, Buffalo, NY, 14263, USA.
| |
Collapse
|
13
|
Aihemaiti A, Liu Y, Zou P, Liu H, Zhu L, Tang Y. Simultaneous determination of canonical purine metabolism using a newly developed HILIC-MS/MS in cultured cells. J Pharm Biomed Anal 2025; 252:116468. [PMID: 39278159 DOI: 10.1016/j.jpba.2024.116468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 09/02/2024] [Accepted: 09/05/2024] [Indexed: 09/17/2024]
Abstract
Purine metabolism acts as the core role in human metabolic network. It offers purine metabolites as raw material for building blocks in cell survival and proliferation. Purine metabolites are the most abundant metabolic substrates in organisms. There are few reports to simultaneously quantify canonical purine metabolism in cells. A novel hydrophilic interaction liquid chromatography coupled with mass spectrometry (HILIC-MS/MS) method was developed to simultaneously determine purines profile in biological samples. Chromatographic separation was achieved using a HILIC (Waters Xbridge™ Amide) column. Different optimizing chromatographic conditions and mass spectrometric parameters were tested in order to provide the best separation and the lowest limit of quantification (LLOQ) values for targeted metabolites. The validation was evaluated according to the Food and Drug Administration guidelines. The limit of determination (LOD) and the LOQ values were in the range of 0.02-8.33 ng mL-1 and 0.1-24.5 ng mL-1, respectively. All calibration curves displayed good linear relationship of with excellent correlation coefficient (r) ranging from 0.9943 to 0.9999. Both intra-day and inter-day variability were below 15 %, respectively. Trueness, expressed as relative error, was always within ±15 %. In addition, no derivatization procedure and ion-pair reagents are in need. The innovated approach demonstrates high sensitivity, strong specificity, and good repeatability, making it suitable for absolute quantitative studies of canonical purine metabolism in cultured cells.
Collapse
Affiliation(s)
- Ayinazhaer Aihemaiti
- Department of Pharmacology and Chemical Biology, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yuqing Liu
- Department of Pharmacology and Chemical Biology, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Peichen Zou
- Department of Pharmacology and Chemical Biology, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hongyu Liu
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Liang Zhu
- Department of Pharmacology and Chemical Biology, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Yabin Tang
- Department of Pharmacology and Chemical Biology, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
14
|
Schneider JL, Han S, Nabel CS. Fuel for thought: targeting metabolism in lung cancer. Transl Lung Cancer Res 2024; 13:3692-3717. [PMID: 39830762 PMCID: PMC11736591 DOI: 10.21037/tlcr-24-662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 11/22/2024] [Indexed: 01/22/2025]
Abstract
For over a century, we have appreciated that the biochemical processes through which micro- and macronutrients are anabolized and catabolized-collectively referred to as "cellular metabolism"-are reprogrammed in malignancies. Cancer cells in lung tumors rewire pathways of nutrient acquisition and metabolism to meet the bioenergetic demands for unchecked proliferation. Advances in precision medicine have ushered in routine genotyping of patient lung tumors, enabling a deeper understanding of the contribution of altered metabolism to tumor biology and patient outcomes. This paradigm shift in thoracic oncology has spawned a new enthusiasm for dissecting oncogenotype-specific metabolic phenotypes and creates opportunity for selective targeting of essential tumor metabolic pathways. In this review, we discuss metabolic states across histologic and molecular subtypes of lung cancers and the additional changes in tumor metabolic pathways that occur during acquired therapeutic resistance. We summarize the clinical investigation of metabolism-specific therapies, addressing successes and limitations to guide the evaluation of these novel strategies in the clinic. Beyond changes in tumor metabolism, we also highlight how non-cellular autonomous processes merit particular consideration when manipulating metabolic processes systemically, such as efforts to disentangle how lung tumor cells influence immunometabolism. As the future of metabolic therapeutics hinges on use of models that faithfully recapitulate metabolic rewiring in lung cancer, we also discuss best practices for harmonizing workflows to capture patient specimens for translational metabolic analyses.
Collapse
Affiliation(s)
- Jaime L. Schneider
- Department of Medicine and Cancer Center, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | | | - Christopher S. Nabel
- Department of Medicine and Cancer Center, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
15
|
Mao Y, Xia Z, Xia W, Jiang P. Metabolic reprogramming, sensing, and cancer therapy. Cell Rep 2024; 43:115064. [PMID: 39671294 DOI: 10.1016/j.celrep.2024.115064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 10/30/2024] [Accepted: 11/21/2024] [Indexed: 12/15/2024] Open
Abstract
The metabolic reprogramming of tumor cells is a crucial strategy for their survival and proliferation, involving tissue- and condition-dependent remodeling of certain metabolic pathways. While it has become increasingly clear that tumor cells integrate extracellular and intracellular signals to adapt and proliferate, nutrient and metabolite sensing also exert direct or indirect influences, although the underlying mechanisms remain incompletely understood. Furthermore, metabolic changes not only support the rapid growth and dissemination of tumor cells but also promote immune evasion by metabolically "educating" immune cells in the tumor microenvironment (TME). Recent studies have highlighted the profound impact of metabolic reprogramming on the TME and the potential of targeting metabolic pathways as a therapeutic strategy, with several enzyme inhibitors showing promising results in clinical trials. Thus, understanding how tumor cells alter their metabolic pathways and metabolically remodel the TME to support their survival and proliferation may offer new strategies for metabolic therapy and immunotherapy.
Collapse
Affiliation(s)
- Youxiang Mao
- State Key Laboratory of Molecular Oncology, School of Life Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Ziyan Xia
- State Key Laboratory of Molecular Oncology, School of Life Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Wenjun Xia
- State Key Laboratory of Molecular Oncology, School of Life Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Peng Jiang
- State Key Laboratory of Molecular Oncology, School of Life Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China.
| |
Collapse
|
16
|
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
|
17
|
Fernández-Maestre I, Cai SF, Levine RL. A View of Myeloid Transformation through the Hallmarks of Cancer. Blood Cancer Discov 2024; 5:377-387. [PMID: 39422551 PMCID: PMC11528188 DOI: 10.1158/2643-3230.bcd-24-0009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 05/30/2024] [Accepted: 08/30/2024] [Indexed: 10/19/2024] Open
Abstract
The development of myeloid malignancies is influenced by a range of cell-intrinsic and cell-extrinsic factors, which can be conceptualized using the hallmarks of cancer. Although many facets of myeloid transformation are similar to those in solid tumors, there are also notable differences. Unlike solid tumors, hematologic malignancies typically exhibit fewer genetic mutations, which have been well characterized. However, understanding the cell-extrinsic factors contributing to myeloid malignancies can be challenging due to the complex interactions in the hematopoietic microenvironment. Researchers need to focus on these intricate factors to prevent the early onset of myeloid transformation and develop appropriate interventions. Significance: Myeloid malignancies are common in the elderly, and acute myeloid leukemia has an adverse prognosis in older patients. Investigating cell-extrinsic factors influencing myeloid malignancies is crucial to developing approaches for preventing or halting disease progression and predicting clinical outcomes in patients with advanced disease. Whereas successful intervention may require targeting various mechanisms, understanding the contribution of each cell-extrinsic factor will help prioritize clinical targets.
Collapse
Affiliation(s)
- Inés Fernández-Maestre
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Louis V. Gerstner Jr Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sheng F. Cai
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Leukemia Service, Department of Medicine and Center for Hematologic Malignancies, Memorial Sloan Kettering, Cancer Center, New York, New York
| | - Ross L. Levine
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Leukemia Service, Department of Medicine and Center for Hematologic Malignancies, Memorial Sloan Kettering, Cancer Center, New York, New York
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
18
|
Xu C, Wei Z, Lv L, Dong X, Xia W, Xing J, Liu H, Zhao X, Liu Y, Wang W, Jiang H, Gong Y, Liu C, Xu K, Wang S, Akimoto Y, Hu Z. Impdh2 deficiency suppresses osteoclastogenesis through mitochondrial oxidative phosphorylation and alleviates ovariectomy-induced osteoporosis. Biochem Biophys Res Commun 2024; 727:150317. [PMID: 38959733 DOI: 10.1016/j.bbrc.2024.150317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 06/25/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024]
Abstract
Abnormalities in osteoclastic generation or activity disrupt bone homeostasis and are highly involved in many pathologic bone-related diseases, including rheumatoid arthritis, osteopetrosis, and osteoporosis. Control of osteoclast-mediated bone resorption is crucial for treating these bone diseases. However, the mechanisms of control of osteoclastogenesis are incompletely understood. In this study, we identified that inosine 5'-monophosphate dehydrogenase type II (Impdh2) positively regulates bone resorption. By histomorphometric analysis, Impdh2 deletion in mouse myeloid lineage cells (Impdh2LysM-/- mice) showed a high bone mass due to the reduced osteoclast number. qPCR and western blotting results demonstrated that the expression of osteoclast marker genes, including Nfatc1, Ctsk, Calcr, Acp5, Dcstamp, and Atp6v0d2, was significantly decreased in the Impdh2LysM-/- mice. Furthermore, the Impdh inhibitor MPA treatment inhibited osteoclast differentiation and induced Impdh2-cytoophidia formation. The ability of osteoclast differentiation was recovered after MPA deprivation. Interestingly, genome-wide analysis revealed that the osteoclastic mitochondrial biogenesis and functions, such as oxidative phosphorylation, were impaired in the Impdh2LysM-/- mice. Moreover, the deletion of Impdh2 alleviated ovariectomy-induced bone loss. In conclusion, our findings revealed a previously unrecognized function of Impdh2, suggesting that Impdh2-mediated mechanisms represent therapeutic targets for osteolytic diseases.
Collapse
Affiliation(s)
- Cheng Xu
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, Hubei, 430056, China.
| | - Zhixin Wei
- Hubei Engineering Research Center for Protection and Utilization of Special Biological Resources in the Hanjiang River Basin, School of Life Sciences, Jianghan University, Wuhan, Hubei, 430056, China
| | - Longfei Lv
- Hubei Engineering Research Center for Protection and Utilization of Special Biological Resources in the Hanjiang River Basin, School of Life Sciences, Jianghan University, Wuhan, Hubei, 430056, China
| | - Xiaoyu Dong
- Hubei Engineering Research Center for Protection and Utilization of Special Biological Resources in the Hanjiang River Basin, School of Life Sciences, Jianghan University, Wuhan, Hubei, 430056, China
| | - Wenwen Xia
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, Hubei, 430056, China
| | - Junqiao Xing
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, Hubei, 430056, China
| | - Hongni Liu
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, Hubei, 430056, China
| | - Xue Zhao
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, Hubei, 430056, China
| | - Yuan Liu
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, Hubei, 430056, China
| | - Weihua Wang
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, Hubei, 430056, China
| | - Haochen Jiang
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, Hubei, 430056, China
| | - Yeli Gong
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, Hubei, 430056, China
| | - Cong Liu
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Kai Xu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Siyuan Wang
- Department of Medicinal Chemistry, College of Pharmacy, Shenzhen Technology University, Shenzhen, Guangdong, 518118, China
| | | | - Zhangfeng Hu
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, Hubei, 430056, China; Hubei Engineering Research Center for Protection and Utilization of Special Biological Resources in the Hanjiang River Basin, School of Life Sciences, Jianghan University, Wuhan, Hubei, 430056, China; Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances, Jianghan University, Wuhan, Hubei, 430056, China.
| |
Collapse
|
19
|
Manning BD, Dibble CC. Growth Signaling Networks Orchestrate Cancer Metabolic Networks. Cold Spring Harb Perspect Med 2024; 14:a041543. [PMID: 38438221 PMCID: PMC11444256 DOI: 10.1101/cshperspect.a041543] [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: 03/06/2024]
Abstract
Normal cells grow and divide only when instructed to by signaling pathways stimulated by exogenous growth factors. A nearly ubiquitous feature of cancer cells is their capacity to grow independent of such signals, in an uncontrolled, cell-intrinsic manner. This property arises due to the frequent oncogenic activation of core growth factor signaling pathway components, including receptor tyrosine kinases, PI3K-AKT, RAS-RAF, mTORC1, and MYC, leading to the aberrant propagation of pro-growth signals independent of exogenous growth factors. The growth of both normal and cancer cells requires the acquisition of nutrients and their anabolic conversion to the primary macromolecules underlying biomass production (protein, nucleic acids, and lipids). The core growth factor signaling pathways exert tight regulation of these metabolic processes and the oncogenic activation of these pathways drive the key metabolic properties of cancer cells and tumors. Here, we review the molecular mechanisms through which these growth signaling pathways control and coordinate cancer metabolism.
Collapse
Affiliation(s)
- Brendan D Manning
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Christian C Dibble
- Department of Pathology, Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
20
|
Meena JK, Wang JH, Neill NJ, Keough D, Putluri N, Katsonis P, Koire AM, Lee H, Bowling EA, Tyagi S, Orellana M, Dominguez-Vidaña R, Li H, Eagle K, Danan C, Chung HC, Yang AD, Wu W, Kurley SJ, Ho BM, Zoeller JR, Olson CM, Meerbrey KL, Lichtarge O, Sreekumar A, Dacso CC, Guddat LW, Rejman D, Hocková D, Janeba Z, Simon LM, Lin CY, Pillon MC, Westbrook TF. MYC Induces Oncogenic Stress through RNA Decay and Ribonucleotide Catabolism in Breast Cancer. Cancer Discov 2024; 14:1699-1716. [PMID: 39193992 PMCID: PMC11372365 DOI: 10.1158/2159-8290.cd-22-0649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 09/12/2023] [Accepted: 05/06/2024] [Indexed: 08/29/2024]
Abstract
Upregulation of MYC is a hallmark of cancer, wherein MYC drives oncogenic gene expression and elevates total RNA synthesis across cancer cell transcriptomes. Although this transcriptional anabolism fuels cancer growth and survival, the consequences and metabolic stresses induced by excess cellular RNA are poorly understood. Herein, we discover that RNA degradation and downstream ribonucleotide catabolism is a novel mechanism of MYC-induced cancer cell death. Combining genetics and metabolomics, we find that MYC increases RNA decay through the cytoplasmic exosome, resulting in the accumulation of cytotoxic RNA catabolites and reactive oxygen species. Notably, tumor-derived exosome mutations abrogate MYC-induced cell death, suggesting excess RNA decay may be toxic to human cancers. In agreement, purine salvage acts as a compensatory pathway that mitigates MYC-induced ribonucleotide catabolism, and inhibitors of purine salvage impair MYC+ tumor progression. Together, these data suggest that MYC-induced RNA decay is an oncogenic stress that can be exploited therapeutically. Significance: MYC is the most common oncogenic driver of poor-prognosis cancers but has been recalcitrant to therapeutic inhibition. We discovered a new vulnerability in MYC+ cancer where MYC induces cell death through excess RNA decay. Therapeutics that exacerbate downstream ribonucleotide catabolism provide a therapeutically tractable approach to TNBC (Triple-negative Breast Cancer) and other MYC-driven cancers.
Collapse
Affiliation(s)
- Jitendra K. Meena
- Therapeutic Innovation Center (THINC), Baylor College of Medicine, Houston, Texas.
- Verna & Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas.
| | - Jarey H. Wang
- Verna & Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas.
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas.
| | - Nicholas J. Neill
- Therapeutic Innovation Center (THINC), Baylor College of Medicine, Houston, Texas.
- Verna & Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas.
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas.
| | - Dianne Keough
- The School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia.
| | - Nagireddy Putluri
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas.
| | - Panagiotis Katsonis
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas.
| | - Amanda M. Koire
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas.
| | - Hyemin Lee
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Elizabeth A. Bowling
- Therapeutic Innovation Center (THINC), Baylor College of Medicine, Houston, Texas.
- Verna & Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas.
| | - Siddhartha Tyagi
- Verna & Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas.
| | - Mayra Orellana
- Verna & Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas.
| | - Rocio Dominguez-Vidaña
- Verna & Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas.
| | - Heyuan Li
- Verna & Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas.
| | - Kenneth Eagle
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas.
| | - Charles Danan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas.
| | - Hsiang-Ching Chung
- Verna & Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas.
| | - Andrew D. Yang
- Therapeutic Innovation Center (THINC), Baylor College of Medicine, Houston, Texas.
- Verna & Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas.
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas.
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas.
| | - William Wu
- Verna & Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas.
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas.
| | - Sarah J. Kurley
- Verna & Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas.
| | - Brian M. Ho
- Verna & Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas.
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas.
| | - Joseph R. Zoeller
- Therapeutic Innovation Center (THINC), Baylor College of Medicine, Houston, Texas.
- Verna & Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas.
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas.
| | - Calla M. Olson
- Therapeutic Innovation Center (THINC), Baylor College of Medicine, Houston, Texas.
- Verna & Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas.
| | - Kristen L. Meerbrey
- Therapeutic Innovation Center (THINC), Baylor College of Medicine, Houston, Texas.
- Verna & Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas.
| | - Olivier Lichtarge
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas.
| | - Arun Sreekumar
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas.
| | - Clifford C. Dacso
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas.
| | - Luke W. Guddat
- The School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia.
| | - Dominik Rejman
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic.
| | - Dana Hocková
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic.
| | - Zlatko Janeba
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic.
| | - Lukas M. Simon
- Therapeutic Innovation Center (THINC), Baylor College of Medicine, Houston, Texas.
| | - Charles Y. Lin
- Therapeutic Innovation Center (THINC), Baylor College of Medicine, Houston, Texas.
- Verna & Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas.
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas.
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.
| | - Monica C. Pillon
- Therapeutic Innovation Center (THINC), Baylor College of Medicine, Houston, Texas.
- Verna & Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas.
| | - Thomas F. Westbrook
- Therapeutic Innovation Center (THINC), Baylor College of Medicine, Houston, Texas.
- Verna & Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas.
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas.
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
21
|
Zhu Z, Zhang Y, Wang L, Geng H, Li M, Chen S, Wang X, Chen P, Sun C, Zhang C. Spatial Metabolomics Profiling Reveals Curcumin Induces Metabolic Reprogramming in Three-Dimensional Tumor Spheroids. Metabolites 2024; 14:482. [PMID: 39330489 PMCID: PMC11433860 DOI: 10.3390/metabo14090482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/28/2024] Open
Abstract
Curcumin is widely recognized for its diverse antitumor properties, ranging from breast cancer to many other types of cancers. However, its role in the tumor microenvironment remains to be elucidated. In this study, we established a 3D tumor spheroids model that can simulate the growth environment of tumor cells and visualized the antitumor metabolic alteration caused by curcumin using mass spectrometry imaging technology. Our results showed that curcumin not only exerts a profound impact on the growth and proliferation of breast cancer cells but in situ multivariate statistical analysis also reveals the significant effect on the overall metabolic profile of tumor spheroids. Meanwhile, our visualization map characterized curcumin metabolic processes of reduction and glucuronidation in tumor spheroids. More importantly, abnormal metabolic pathways related to lipid metabolism and polyamine metabolism were also remodeled at the metabolite and gene levels after curcumin intervention. These insights deepen our comprehension of the regulatory mechanism of curcumin on the tumor metabolic network, furnishing powerful references for antitumor treatment.
Collapse
Affiliation(s)
- Zihan Zhu
- Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
- Key Laboratory for Applied Technology of Sophisticated Analytical Instruments of Shandong Province, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Yaqi Zhang
- Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
- Key Laboratory for Applied Technology of Sophisticated Analytical Instruments of Shandong Province, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Lei Wang
- Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
- Key Laboratory for Applied Technology of Sophisticated Analytical Instruments of Shandong Province, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Haoyuan Geng
- Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
- Key Laboratory for Applied Technology of Sophisticated Analytical Instruments of Shandong Province, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Min Li
- Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
- Key Laboratory for Applied Technology of Sophisticated Analytical Instruments of Shandong Province, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Shiping Chen
- Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
- Key Laboratory for Applied Technology of Sophisticated Analytical Instruments of Shandong Province, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Xiao Wang
- Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
- Key Laboratory for Applied Technology of Sophisticated Analytical Instruments of Shandong Province, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Panpan Chen
- Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
- Key Laboratory for Applied Technology of Sophisticated Analytical Instruments of Shandong Province, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Chenglong Sun
- Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
- Key Laboratory for Applied Technology of Sophisticated Analytical Instruments of Shandong Province, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Chao Zhang
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan 250012, China
| |
Collapse
|
22
|
Zhang J, Zeng X, Guo Q, Sheng Z, Chen Y, Wan S, Zhang L, Zhang P. Small cell lung cancer: emerging subtypes, signaling pathways, and therapeutic vulnerabilities. Exp Hematol Oncol 2024; 13:78. [PMID: 39103941 DOI: 10.1186/s40164-024-00548-w] [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: 06/26/2024] [Accepted: 07/27/2024] [Indexed: 08/07/2024] Open
Abstract
Small cell lung cancer (SCLC) is a recalcitrant cancer characterized by early metastasis, rapid tumor growth and poor prognosis. In recent decades, the epidemiology, initiation and mutation characteristics of SCLC, as well as abnormal signaling pathways contributing to its progression, have been widely studied. Despite extensive investigation, fewer drugs have been approved for SCLC. Recent advancements in multi-omics studies have revealed diverse classifications of SCLC that are featured by distinct characteristics and therapeutic vulnerabilities. With the accumulation of SCLC samples, different subtypes of SCLC and specific treatments for these subtypes were further explored. The identification of different molecular subtypes has opened up novel avenues for the treatment of SCLC; however, the inconsistent and uncertain classification of SCLC has hindered the translation from basic research to clinical applications. Therefore, a comprehensives review is essential to conclude these emerging subtypes and related drugs targeting specific therapeutic vulnerabilities within abnormal signaling pathways. In this current review, we summarized the epidemiology, risk factors, mutation characteristics of and classification, related molecular pathways and treatments for SCLC. We hope that this review will facilitate the translation of molecular subtyping of SCLC from theory to clinical application.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China.
| | - Xiaoping Zeng
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Qiji Guo
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Zhenxin Sheng
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Yan Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Shiyue Wan
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Lele Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Peng Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China.
| |
Collapse
|
23
|
Plotnik JP, Zha Z, Feng W, Lee I, Riehm J, McClure RA, Sandoval S, Uziel T, Murphy E, Lu X, Lam LT. MYC Family Amplification Dictates Sensitivity to BET Bromodomain Protein Inhibitor Mivebresib (ABBV075) in Small-Cell Lung Cancer. Mol Cancer Res 2024; 22:689-698. [PMID: 38747975 PMCID: PMC11294817 DOI: 10.1158/1541-7786.mcr-23-0599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 03/28/2024] [Accepted: 05/10/2024] [Indexed: 08/03/2024]
Abstract
Small-cell lung cancer (SCLC) accounts for nearly 15% of all lung cancers. Although patients respond to first-line therapy readily, rapid relapse is inevitable, with few treatment options in the second-line setting. Here, we describe SCLC cell lines harboring amplification of MYC and MYCN but not MYCL1 or non-amplified MYC cell lines exhibit superior sensitivity to treatment with the pan-BET bromodomain protein inhibitor mivebresib (ABBV075). Silencing MYC and MYCN partially rescued SCLC cell lines harboring these respective amplifications from the antiproliferative effects of mivebresib. Further characterization of genome-wide binding of MYC, MYCN, and MYCL1 uncovered unique enhancer and epigenetic preferences. Implications: Our study suggests that chromatin landscapes can establish cell states with unique gene expression programs, conveying sensitivity to epigenetic inhibitors such as mivebresib.
Collapse
Affiliation(s)
| | - Zheng Zha
- Genomic Research Center, AbbVie Inc., North Chicago, Illinois.
| | - Weiguo Feng
- Genomic Research Center, AbbVie Inc., North Chicago, Illinois.
| | - Irene Lee
- Precision Medicine, Translational Oncology, AbbVie Inc., North Chicago, Illinois.
| | - Jacob Riehm
- Precision Medicine, Translational Oncology, AbbVie Inc., North Chicago, Illinois.
| | - Ryan A. McClure
- Physical Chemistry, Discovery, AbbVie Inc., North Chicago, Illinois.
| | | | - Tamar Uziel
- Precision Medicine, Translational Oncology, AbbVie Inc., North Chicago, Illinois.
| | - Erin Murphy
- Genomic Research Center, AbbVie Inc., North Chicago, Illinois.
| | - Xin Lu
- Genomic Research Center, AbbVie Inc., North Chicago, Illinois.
| | - Lloyd T. Lam
- Precision Medicine, Translational Oncology, AbbVie Inc., North Chicago, Illinois.
| |
Collapse
|
24
|
Li TW, Park Y, Watters EG, Wang X, Zhou D, Fiches GN, Wu Z, Badley AD, Sacha JB, Ho WZ, Santoso NG, Qi J, Zhu J. KDM5A/B contribute to HIV-1 latent infection and survival of HIV-1 infected cells. Antiviral Res 2024; 228:105947. [PMID: 38925368 PMCID: PMC11927087 DOI: 10.1016/j.antiviral.2024.105947] [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: 12/26/2023] [Revised: 06/22/2024] [Accepted: 06/23/2024] [Indexed: 06/28/2024]
Abstract
Combinational antiretroviral therapy (cART) suppresses human immunodeficiency virus type 1 (HIV-1) viral replication and pathogenesis in acquired immunodeficiency syndrome (AIDS) patients. However, HIV-1 remains in the latent stage of infection by suppressing viral transcription, which hinders an HIV-1 cure. One approach for an HIV-1 cure is the "shock and kill" strategy. The strategy focuses on reactivating latent HIV-1, inducing the viral cytopathic effect and facilitating the immune clearance for the elimination of latent HIV-1 reservoirs. Here, we reported that the H3K4 trimethylation (H3K4me3)-specific demethylase KDM5A/B play a role in suppressing HIV-1 Tat/LTR-mediated viral transcription in HIV-1 latent cells. Furthermore, we evaluated the potential of KDM5-specific inhibitor JQKD82 as an HIV-1 "shock and kill" agent. Our results showed that JQKD82 increases the H3K4me3 level at HIV-1 5' LTR promoter regions, HIV-1 reactivation, and the cytopathic effects in an HIV-1-latent T cell model. In addition, we identified that the combination of JQKD82 and AZD5582, a non-canonical NF-κB activator, generates a synergistic impact on inducing HIV-1 lytic reactivation and cell death in the T cell. The latency-reversing potency of the JQKD82 and AZD5582 pair was also confirmed in peripheral blood mononuclear cells (PBMCs) isolated from HIV-1 aviremic patients and in an HIV-1 latent monocyte. In latently infected microglia (HC69) of the brain, either deletion or inhibition of KDM5A/B results in a reversal of the HIV-1 latency. Overall, we concluded that KDM5A/B function as a host repressor of the HIV-1 lytic reactivation and thus promote the latency and the survival of HIV-1 infected reservoirs.
Collapse
Affiliation(s)
- Tai-Wei Li
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Youngmin Park
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Emily G Watters
- Department of Microbiology, College of Arts and Sciences, The Ohio State University, Columbus, OH, 43210, USA
| | - Xu Wang
- Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140, USA
| | - Dawei Zhou
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Guillaume N Fiches
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Zhenyu Wu
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Andrew D Badley
- Division of Infectious Diseases, Mayo Clinic, Rochester, MN, 55902, USA
| | - Jonah B Sacha
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA; Vaccine and Gene Therapy Institute, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Wen-Zhe Ho
- Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140, USA
| | - Netty G Santoso
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Jun Qi
- Dana-Farber Cancer Institute, Boston, MA, 02215, USA.
| | - Jian Zhu
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA; Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
25
|
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 D, Lee JK, Graeber T, Shirihai O, Witte O. PGC-1α drives small cell neuroendocrine cancer progression towards an ASCL1-expressing subtype with increased mitochondrial capacity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.09.588489. [PMID: 38645232 PMCID: PMC11030384 DOI: 10.1101/2024.04.09.588489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Adenocarcinomas from multiple tissues can evolve into lethal, treatment-resistant small cell neuroendocrine (SCN) cancers comprising multiple subtypes with poorly defined metabolic characteristics. The role of metabolism in directly driving subtype determination remains unclear. Through bioinformatics analyses of thousands of patient tumors, we identified enhanced PGC-1α-a potent regulator of oxidative phosphorylation (OXPHOS)-in various SCN cancers (SCNCs), closely linked with neuroendocrine differentiation. In a patient-derived prostate tissue SCNC transformation system, the ASCL1-expressing neuroendocrine subtype showed elevated PGC-1α expression and increased OXPHOS activity. Inhibition of PGC-1α and OXPHOS reduced the proliferation of SCN lung and prostate cancer cell lines and blocked SCN prostate tumor formation. Conversely, enhancing PGC- 1α and 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 novel metabolic vulnerabilities in SCNCs across different tissues. STATEMENT OF SIGNIFICANCE Our study provides functional evidence that metabolic reprogramming can directly impact cancer phenotypes and establishes PGC-1α-induced mitochondrial metabolism as a driver of SCNC progression and lineage determination. These mechanistic insights reveal common metabolic vulnerabilities across SCNCs originating from multiple tissues, opening new avenues for pan-SCN cancer therapeutic strategies.
Collapse
|
26
|
Hu J, Zheng S, Hua M, Ding M, Hu Z, Jiang H. Deletion of Impdh2 in adipocyte precursors limits the expansion of white adipose tissue and enhances metabolic health with overnutrition. Biochem Biophys Res Commun 2024; 716:149998. [PMID: 38692012 DOI: 10.1016/j.bbrc.2024.149998] [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: 04/11/2024] [Accepted: 04/23/2024] [Indexed: 05/03/2024]
Abstract
The equilibrium between the hypertrophic growth of existing adipocytes and adipogenesis is vital in managing metabolic stability in white adipocytes when faced with overnutrition. Adipogenesis has been established as a key player in combating metabolic irregularities caused by various factors. However, the benefits of increasing adipogenesis-mediated white adipose tissue (WAT) expansion for metabolic health regulation remain uncertain. Our findings reveal an increase in Impdh2 expression during the adipogenesis phase, both in vivo and in vitro. Xmp enhances adipogenic potential by fostering mitotic clonal expansion (MCE). The conditional knockout of Impdh2 in adipocyte progenitor cells(APCs) in adult and aged mice effectively curbs white adipose tissue expansion, ameliorates glucose tolerance, and augments energy expenditure under high-fat diet (HFD). However, no significant difference is observed under normal chow diet (NCD). Concurrently, the knockout of Impdh2 in APCs significantly reduces the count of new adipocytes induced by HFD, without affecting adipocyte size. Mechanistically, Impdh2 regulates the proliferation of APCs during the MCE phase via Xmp. Exogenous Xmp can significantly offset the reduction in adipogenic abilities of APCs due to Impdh2 deficiency. In summary, we discovered that adipogenesis-mediated WAT expansion, induced by overnutrition, also contributes to metabolic abnormalities. Moreover, the pivotal role of Impdh2 in regulating adipogenesis in APCs offers a novel therapeutic approach to combat obesity.
Collapse
Affiliation(s)
- Jiaqi Hu
- Hubei Engineering Research Center for Protection and Utilization of Special Biological Resources in the Hanjiang River Basin, School of Life Sciences, Jianghan University, Wuhan, 430056, Hubei, China
| | - Shiqiong Zheng
- Hubei Engineering Research Center for Protection and Utilization of Special Biological Resources in the Hanjiang River Basin, School of Life Sciences, Jianghan University, Wuhan, 430056, Hubei, China
| | - Mengting Hua
- Hubei Engineering Research Center for Protection and Utilization of Special Biological Resources in the Hanjiang River Basin, School of Life Sciences, Jianghan University, Wuhan, 430056, Hubei, China
| | - Mei Ding
- Hubei Engineering Research Center for Protection and Utilization of Special Biological Resources in the Hanjiang River Basin, School of Life Sciences, Jianghan University, Wuhan, 430056, Hubei, China
| | - Zhangfeng Hu
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Bio-medical Sciences, School of Medicine, Jianghan University, Wuhan, Hubei, 430056, China; Hubei Engineering Research Center for Protection and Utilization of Special Biological Resources in the Hanjiang River Basin, School of Life Sciences, Jianghan University, Wuhan, 430056, Hubei, China.
| | - Haochen Jiang
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Bio-medical Sciences, School of Medicine, Jianghan University, Wuhan, Hubei, 430056, China.
| |
Collapse
|
27
|
Wang L, Yang R, Kong Y, Zhou J, Chen Y, Li R, Chen C, Tang X, Chen X, Xia J, Chen X, Cheng B, Ren X. Integrative single-cell and bulk transcriptomes analyses reveals heterogeneity of serine-glycine-one-carbon metabolism with distinct prognoses and therapeutic vulnerabilities in HNSCC. Int J Oral Sci 2024; 16:44. [PMID: 38886346 PMCID: PMC11183126 DOI: 10.1038/s41368-024-00310-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 04/03/2024] [Accepted: 04/28/2024] [Indexed: 06/20/2024] Open
Abstract
Metabolic heterogeneity plays a central role in sustaining uncontrolled cancer cell proliferation and shaping the tumor microenvironment (TME), which significantly compromises the clinical outcomes and responses to therapy in head and neck squamous cell carcinoma (HNSCC) patients. This highlights the urgent need to delineate the intrinsic heterogeneity and biological roles of metabolic vulnerabilities to advance precision oncology. The metabolic heterogeneity of malignant cells was identified using single-cell RNA sequencing (scRNA-seq) profiles and validated through bulk transcriptomes. Serine-glycine-one-carbon (SGOC) metabolism was screened out to be responsible for the aggressive malignant properties and poor prognosis in HNSCC patients. A 4-SGOC gene prognostic signature, constructed by LASSO-COX regression analysis, demonstrated good predictive performance for overall survival and therapeutic responses. Patients in the low-risk group exhibited greater infiltration of exhausted CD8+ T cells, and demonstrated better clinical outcomes after receiving immunotherapy and chemotherapy. Conversely, high-risk patients exhibited characteristics of cold tumors, with enhanced IMPDH1-mediated purine biosynthesis, resulting in poor responses to current therapies. IMPDH1 emerged as a potential therapeutic metabolic target. Treatment with IMPDH inhibitors effectively suppressed HNSCC cell proliferation and metastasis and induced apoptosis in vitro and in vivo by triggering GTP-exhaustion nucleolar stress. Our findings underscore the metabolic vulnerabilities of HNSCC in facilitating accurate patient stratification and individualized precise metabolic-targeted treatment.
Collapse
Affiliation(s)
- Lixuan Wang
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Rongchun Yang
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Yue Kong
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Jing Zhou
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Yingyao Chen
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Rui Li
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chuwen Chen
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Xinran Tang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaobing Chen
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Juan Xia
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Xijuan Chen
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Bin Cheng
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China.
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China.
| | - Xianyue Ren
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China.
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
28
|
Li Y, Yang X, Geng C, Liu Y, Tang T, Zhang L, Liu F, Zhang M, Hao J, Ma L. Identification of molecular subtypes based on chromatin regulator-related genes and experimental verification of the role of ASCL1 in conferring chemotherapy resistance to breast cancer. Front Immunol 2024; 15:1390261. [PMID: 38726001 PMCID: PMC11079216 DOI: 10.3389/fimmu.2024.1390261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 04/08/2024] [Indexed: 05/12/2024] Open
Abstract
Objective The aim of this study was to identify the molecular subtypes of breast cancer based on chromatin regulator-related genes. Methods The RNA sequencing data of The Cancer Genome Atlas-Breast Cancer cohort were obtained from the official website, while the single-cell data were downloaded from the Gene Expression Omnibus database (GSE176078). Validation was performed using the Molecular Taxonomy of Breast Cancer International Consortium dataset. Furthermore, the immune characteristics, tumor stemness, heterogeneity, and clinical characteristics of these molecular subtypes were analyzed. The correlation between chromatin regulators and chemotherapy resistance was examined in vitro using the quantitative real-time polymerase chain reaction (qRT-PCR) and Cell Counting Kit-8 (CCK8) assays. Results This study identified three stable molecular subtypes with different prognostic and pathological features. Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, and protein-protein interaction analyses revealed that the differentially expressed genes were associated with disease processes, such as mitotic nuclear division, chromosome segregation, condensed chromosome, and specific chromosome region. The T stage and subtypes were correlated with the clinical features. Tumor heterogeneity (mutant-allele tumor heterogeneity, tumor mutational burden, purity, and homologous recombination deficiency) and tumor stemness (RNA expression-based stemness score, epigenetically regulated RNA expression-based stemness score, DNA methylation-based stemness score, and epigenetically regulated DNA methylation-based stemness score) significantly varied between the three subtypes. Furthermore, Western blotting, qRT-PCR, and CCK8 assays demonstrated that the expression of ASCL1 was positively correlated with chemotherapy resistance in breast cancer. Conclusion This study identified the subtypes of breast cancer based on chromatin regulators and analyzed their clinical features, gene mutation status, immunophenotype, and drug sensitivity. The results of this study provide effective strategies for assessing clinical prognosis and developing personalized treatment strategies.
Collapse
Affiliation(s)
- Yilun Li
- Department of Breast Disease Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Xiaolu Yang
- Department of Breast Disease Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Cuizhi Geng
- Department of Breast Disease Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yunjiang Liu
- Department of Breast Disease Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Tiantian Tang
- Department of Breast Disease Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lina Zhang
- Department of Breast Disease Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Fei Liu
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Meng Zhang
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jun Hao
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Li Ma
- Department of Breast Disease Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
29
|
Grenda A, Krawczyk P, Obara A, Gajek Ł, Łomża-Łaba A, Milanowski J. Transitioning to a Personalized Approach in Molecularly Subtyped Small-Cell Lung Cancer (SCLC). Int J Mol Sci 2024; 25:4208. [PMID: 38673793 PMCID: PMC11050005 DOI: 10.3390/ijms25084208] [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: 02/29/2024] [Revised: 04/08/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Lung cancer has become a major public health concern, standing as the leading cause of cancer-related deaths worldwide. Among its subtypes, small-cell lung cancer (SCLC) is characterized by aggressive and rapid growth, poor differentiation, and neuroendocrine features. Typically, SCLC is diagnosed at an advanced stage (extensive disease, ED-SCLC), with distant metastases, and is strongly associated with tobacco smoking and has a poor prognosis. Recent clinical trials, such as CASPIAN and IMpower133, have demonstrated promising outcomes with the incorporation of immune checkpoint inhibitors in first-line chemotherapy, leading to prolonged progression-free survival and overall survival in patients with ED-SCLC compared to standard chemotherapy. Other studies have emphasized the potential for future development of molecularly targeted therapies in SCLC patients, including inhibitors of IGF-1R, DLL3, BCL-2, MYC, or PARP. The molecular subdivision of SCLC based on transcriptomic and immunohistochemical analyses represents a significant advancement in both diagnostic and clinical approaches in SCLC patients. Specific molecular pathways are activated within distinct transcriptome subtypes of SCLC, offering the potential for personalized treatment strategies, such as targeted therapies and immunotherapies. Such tailored approaches hold promise for significantly improving outcomes in SCLC patients.
Collapse
Affiliation(s)
- Anna Grenda
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, 20-950 Lublin, Poland; (P.K.); (A.Ł.-Ł.); (J.M.)
| | - Paweł Krawczyk
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, 20-950 Lublin, Poland; (P.K.); (A.Ł.-Ł.); (J.M.)
| | - Adrian Obara
- Institute of Genetics and Immunology Genim LCC, Filaretów 27/2, 20-609 Lublin, Poland; (A.O.); (Ł.G.)
| | - Łukasz Gajek
- Institute of Genetics and Immunology Genim LCC, Filaretów 27/2, 20-609 Lublin, Poland; (A.O.); (Ł.G.)
| | - Aleksandra Łomża-Łaba
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, 20-950 Lublin, Poland; (P.K.); (A.Ł.-Ł.); (J.M.)
| | - Janusz Milanowski
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, 20-950 Lublin, Poland; (P.K.); (A.Ł.-Ł.); (J.M.)
| |
Collapse
|
30
|
Cunha PDS, de Miranda MC, de Melo MIA, Ferreira ADF, Barbosa JL, Oliveira JADC, Goes TDS, Gomes DA, de Goes AM. Selection of internalizing RNA aptamers into human breast cancer cells derived from primary sites. J Cell Biochem 2024; 125:e30540. [PMID: 38372191 DOI: 10.1002/jcb.30540] [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: 10/11/2023] [Revised: 01/30/2024] [Accepted: 02/06/2024] [Indexed: 02/20/2024]
Abstract
Breast cancer is the most common cancer in women. Although chemotherapy is still broadly used in its treatment, adverse effects remain a challenge. In this scenario, aptamers emerge as a promising alternative for theranostic applications. Studies using breast cancer cell lines provide useful information in laboratory and preclinical investigations, most of which use cell lines established from metastatic sites. However, these cell lines correspond to cell populations of the late stage of tumor progression. On the other hand, studies using breast cancer cells established from primary sites make it possible to search for new theranostic approaches in the early stages of the disease. Therefore, this work aimed to select RNA aptamers internalized by MGSO-3 cells, a human breast cancer cell line, derived from a primary site previously established in our laboratory. Using the Cell-Internalization SELEX method, we have selected two candidate aptamers (ApBC1 and ApBC2). We evaluated their internalization efficiencies, specificities, cellular localization by Reverse Transcription-qPCR (RT-qPCR) and confocal microscopy assays. The results suggest that both aptamers were efficiently internalized by human breast cancer cells, MACL-1, MDA-MB-231, and especially by MGSO-3 cells. Furthermore, both aptamers could effectively distinguish human breast cancer cells derived from normal human mammary cell (MCF 10A) and prostate cancer cell (PC3) lines. Therefore, ApBC1 and ApBC2 could be promising candidate molecules for theranostic applications, even in the early stages of tumor progression.
Collapse
Affiliation(s)
- Pricila da Silva Cunha
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Marcelo Coutinho de Miranda
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Mariane Izabella Abreu de Melo
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Andrea da Fonseca Ferreira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Joana Lobato Barbosa
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Tércio de Souza Goes
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Dawidson Assis Gomes
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Alfredo Miranda de Goes
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
31
|
Pillai R, LeBoeuf SE, Hao Y, New C, Blum JLE, Rashidfarrokhi A, Huang SM, Bahamon C, Wu WL, Karadal-Ferrena B, Herrera A, Ivanova E, Cross M, Bossowski JP, Ding H, Hayashi M, Rajalingam S, Karakousi T, Sayin VI, Khanna KM, Wong KK, Wild R, Tsirigos A, Poirier JT, Rudin CM, Davidson SM, Koralov SB, Papagiannakopoulos T. Glutamine antagonist DRP-104 suppresses tumor growth and enhances response to checkpoint blockade in KEAP1 mutant lung cancer. SCIENCE ADVANCES 2024; 10:eadm9859. [PMID: 38536921 PMCID: PMC10971495 DOI: 10.1126/sciadv.adm9859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 02/15/2024] [Indexed: 04/04/2024]
Abstract
Loss-of-function mutations in KEAP1 frequently occur in lung cancer and are associated with poor prognosis and resistance to standard of care treatment, highlighting the need for the development of targeted therapies. We previously showed that KEAP1 mutant tumors consume glutamine to support the metabolic rewiring associated with NRF2-dependent antioxidant production. Here, using preclinical patient-derived xenograft models and antigenic orthotopic lung cancer models, we show that the glutamine antagonist prodrug DRP-104 impairs the growth of KEAP1 mutant tumors. We find that DRP-104 suppresses KEAP1 mutant tumors by inhibiting glutamine-dependent nucleotide synthesis and promoting antitumor T cell responses. Using multimodal single-cell sequencing and ex vivo functional assays, we demonstrate that DRP-104 reverses T cell exhaustion, decreases Tregs, and enhances the function of CD4 and CD8 T cells, culminating in an improved response to anti-PD1 therapy. Our preclinical findings provide compelling evidence that DRP-104, currently in clinical trials, offers a promising therapeutic approach for treating patients with KEAP1 mutant lung cancer.
Collapse
Affiliation(s)
- Ray Pillai
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, VA New York Harbor Healthcare System, New York, NY 10016, USA
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Sarah E. LeBoeuf
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Yuan Hao
- Applied Bioinformatics Laboratories, New York University Langone Health, New York, NY 10016, USA
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| | - Connie New
- Departments of Biological Engineering and Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jenna L. E. Blum
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, The Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Ali Rashidfarrokhi
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Shih Ming Huang
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Christian Bahamon
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Warren L. Wu
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Burcu Karadal-Ferrena
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Alberto Herrera
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Ellie Ivanova
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Michael Cross
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Jozef P. Bossowski
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Hongyu Ding
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Makiko Hayashi
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Sahith Rajalingam
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Triantafyllia Karakousi
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Volkan I. Sayin
- Institute of Clinical Sciences, Department of Surgery, Sahlgrenska Center for Cancer Research, University of Gothenburg, 41345 Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, 41345 Gothenburg, Sweden
| | - Kamal M. Khanna
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
- Department of Microbiology, New York University Langone Health, New York, NY 10016, USA
| | - Kwok-Kin Wong
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| | - Robert Wild
- Dracen Pharmaceuticals Inc., San Diego, CA 92121, USA
| | - Aristotelis Tsirigos
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| | - John T. Poirier
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| | - Charles M. Rudin
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10655, USA
| | - Shawn M. Davidson
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, The Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
| | - Sergei B. Koralov
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| | - Thales Papagiannakopoulos
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| |
Collapse
|
32
|
Xie Q, Cao Z, You W, Cai X, Shen M, Yin Z, Jiang Y, Wang X, Ye S. Ganodermanontriol Suppresses the Progression of Lung Adenocarcinoma by Activating CES2 to Enhance the Metabolism of Mycophenolate Mofetil. J Microbiol Biotechnol 2024; 34:249-261. [PMID: 38419324 PMCID: PMC10940751 DOI: 10.4014/jmb.2306.06020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 03/02/2024]
Abstract
New anti-lung cancer therapies are urgently required to improve clinical outcomes. Since ganodermanontriol (GDNT) has been identified as a potential antineoplastic agent, its role in lung adenocarcinoma (LUAD) is investigated in this study. Concretely, lung cancer cells were treated with GDNT and/or mycophenolate mofetil (MMF), after which MTT assay, flow cytometry and Western blot were conducted. Following bioinformatics analysis, carboxylesterase 2 (CES2) was knocked down and rescue assays were carried out in vitro. Xenograft experiment was performed on mice, followed by drug administration, measurement of tumor growth and determination of CES2, IMPDH1 and IMPDH2 expressions. As a result, the viability of lung cancer cells was reduced by GDNT or MMF. GDNT enhanced the effects of MMF on suppressing viability, promoting apoptosis and inducing cell cycle arrest in lung cancer cells. GDNT up-regulated CES2 level, and strengthened the effects of MMF on down-regulating IMPDH1 and IMPDH2 levels in the cells. IMPDH1 and IMPDH2 were highly expressed in LUAD samples. CES2 was a potential target for GDNT. CES2 knockdown reversed the synergistic effect of GDNT and MMF against lung cancer in vitro. GDNT potentiated the role of MMF in inhibiting tumor growth and expressions of CES2 and IMPDH1/2 in lung cancer in vivo. Collectively, GDNT suppresses the progression of LUAD by activating CES2 to enhance the metabolism of MMF.
Collapse
Affiliation(s)
- Qingfeng Xie
- Respiratory Department, Longquan People’s Hospital, No. 699, Dongcha Road, Longquan City, Zhejiang Province, 323000, P.R. China
| | - Zhuo Cao
- Respiratory Department, The Sixth Affiliated Hospital of Wenzhou Medical University, No. 15 Dazhong Street, Liandu District, Lishui City, Zhejiang Province, 323000, P.R. China
| | - Weiling You
- Respiratory Department, Longquan People’s Hospital, No. 699, Dongcha Road, Longquan City, Zhejiang Province, 323000, P.R. China
| | - Xiaoping Cai
- Respiratory Department, The Sixth Affiliated Hospital of Wenzhou Medical University, No. 15 Dazhong Street, Liandu District, Lishui City, Zhejiang Province, 323000, P.R. China
| | - Mei Shen
- Longquan People’s Hospital, No. 699, Dongcha Road, Longquan City, Zhejiang Province, 323000, P.R. China
| | - Zhangyong Yin
- Respiratory Department, The Sixth Affiliated Hospital of Wenzhou Medical University, No. 15 Dazhong Street, Liandu District, Lishui City, Zhejiang Province, 323000, P.R. China
| | - Yiwei Jiang
- Wenzhou Medical University, Wenzhou Chashan Higher Education Park, Wenzhou, Zhejiang Province, 325006, P.R. China
| | - Xin Wang
- Wenzhou Medical University, Wenzhou Chashan Higher Education Park, Wenzhou, Zhejiang Province, 325006, P.R. China
| | - Siyu Ye
- School of Public Administration, Wenzhou Medical University, Wenzhou Chashan Higher Education Park, Wenzhou, Zhejiang Province, 325006, P.R. China
| |
Collapse
|
33
|
Long R, Abulimiti N, Wang X. Proteomics-based clustering of lung adenocarcinoma identifies three subtypes with significantly different clinical and molecular features. Clin Transl Oncol 2024; 26:538-548. [PMID: 37603150 DOI: 10.1007/s12094-023-03275-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 07/03/2023] [Indexed: 08/22/2023]
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) is a predominant subtype of lung cancer. Although molecular classification of LUAD has been widely explored, proteomics-based subtyping of LUAD remains scarce. METHODS We proposed a subtyping method for LUAD based on the expression profiles of 500 proteins with the largest expression variability across LUAD. Furthermore, we comprehensively compared molecular and clinical features among the LUAD subtypes. RESULTS Consensus clustering identified three subtypes of LUAD, namely MtE, DrE, and StE. We demonstrated this subtyping method to be reproducible by analyzing two independent LUAD cohorts. MtE was characterized by high enrichment of metabolic pathways, high EGFR mutation rate, low stemness, proliferation, invasion, metastasis and inflammation signatures, favorable prognosis; DrE was characterized by high enrichment of DNA repair pathways, high TP53 mutation rate, and high levels of genomic instability, stemness, proliferation, and intratumor heterogeneity (ITH); and StE was characterized by high enrichment of stroma-related pathways, high KRAS mutation rate, and low levels of genomic instability. CONCLUSIONS The proteomics-based clustering analysis identified three LUAD subtypes with significantly different molecular and clinical properties. The novel subtyping method offers new perspectives on the cancer biology and holds promise in improving the clinical management of LUAD.
Collapse
Affiliation(s)
- Rongzhuo Long
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
- Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
- Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198, China
| | - Nayila Abulimiti
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
- Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
- Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198, China
| | - Xiaosheng Wang
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
- Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
- Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
34
|
Zhou W, Zhao Z, Lin A, Yang JZ, Xu J, Wilder-Romans K, Yang A, Li J, Solanki S, Speth JM, Walker N, Scott AJ, Wang L, Wen B, Andren A, Zhang L, Kothari AU, Yao Y, Peterson ER, Korimerla N, Werner CK, Ullrich A, Liang J, Jacobson J, Palavalasa S, O’Brien AM, Elaimy AL, Ferris SP, Zhao SG, Sarkaria JN, Győrffy B, Zhang S, Al-Holou WN, Umemura Y, Morgan MA, Lawrence TS, Lyssiotis CA, Peters-Golden M, Shah YM, Wahl DR. GTP Signaling Links Metabolism, DNA Repair, and Responses to Genotoxic Stress. Cancer Discov 2024; 14:158-175. [PMID: 37902550 PMCID: PMC10872631 DOI: 10.1158/2159-8290.cd-23-0437] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 09/07/2023] [Accepted: 10/19/2023] [Indexed: 10/31/2023]
Abstract
How cell metabolism regulates DNA repair is incompletely understood. Here, we define a GTP-mediated signaling cascade that links metabolism to DNA repair and has significant therapeutic implications. GTP, but not other nucleotides, regulates the activity of Rac1, a guanine nucleotide-binding protein, which promotes the dephosphorylation of serine 323 on Abl-interactor 1 (Abi-1) by protein phosphatase 5 (PP5). Dephosphorylated Abi-1, a protein previously not known to activate DNA repair, promotes nonhomologous end joining. In patients and mouse models of glioblastoma, Rac1 and dephosphorylated Abi-1 mediate DNA repair and resistance to standard-of-care genotoxic treatments. The GTP-Rac1-PP5-Abi-1 signaling axis is not limited to brain cancer, as GTP supplementation promotes DNA repair and Abi-1-S323 dephosphorylation in nonmalignant cells and protects mouse tissues from genotoxic insult. This unexpected ability of GTP to regulate DNA repair independently of deoxynucleotide pools has important implications for normal physiology and cancer treatment. SIGNIFICANCE A newly described GTP-dependent signaling axis is an unexpected link between nucleotide metabolism and DNA repair. Disrupting this pathway can overcome cancer resistance to genotoxic therapy while augmenting it can mitigate genotoxic injury of normal tissues. This article is featured in Selected Articles from This Issue, p. 5.
Collapse
Affiliation(s)
- Weihua Zhou
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Zitong Zhao
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
- Department of Oncology, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shan Xi, PR China
| | - Angelica Lin
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - John Z Yang
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Jie Xu
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Kari Wilder-Romans
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Annabel Yang
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Jing Li
- Cell Signaling Technology, Inc., Danvers, MA, USA
| | - Sumeet Solanki
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Jennifer M Speth
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Natalie Walker
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Andrew J Scott
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Lu Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, USA
| | - Bo Wen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, USA
| | - Anthony Andren
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Li Zhang
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Ayesha U Kothari
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Yangyang Yao
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
- Department of Oncology, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, PR China
| | - Erik R Peterson
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Navyateja Korimerla
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Christian K Werner
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Alexander Ullrich
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Jessica Liang
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Janna Jacobson
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Sravya Palavalasa
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Alexandra M O’Brien
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Ameer L Elaimy
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Sean P Ferris
- Department of Pathology, Division of Neuropathology, University of Michigan, Ann Arbor, MI, USA
| | - Shuang G Zhao
- Department of Human Oncology, University of Wisconsin Madison, WI, USA
| | | | - Balázs Győrffy
- Department of Bioinformatics, Semmelweis University, Budapest, Hungary; and TTK Cancer Biomarker Research Group, Institute of Enzymology, Budapest, Hungary
| | - Shuqun Zhang
- Department of Oncology, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shan Xi, PR China
| | - Wajd N Al-Holou
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, United States
| | - Yoshie Umemura
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| | - Meredith A Morgan
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Theodore S Lawrence
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Costas A Lyssiotis
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular and Integrative Physiology and Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Marc Peters-Golden
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Yatrik M Shah
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Daniel R Wahl
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
- Lead contact
| |
Collapse
|
35
|
Tabata S, Umemura S, Narita M, Udagawa H, Ishikawa T, Tsuboi M, Goto K, Ishii G, Tsuchihara K, Ochiai A, Kobayashi SS, Soga T, Makinoshima H. Metabolic Hallmarks for Purine Nucleotide Biosynthesis in Small Cell Lung Carcinoma. Mol Cancer Res 2024; 22:82-93. [PMID: 37773022 PMCID: PMC10758693 DOI: 10.1158/1541-7786.mcr-23-0386] [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: 05/19/2023] [Revised: 08/16/2023] [Accepted: 09/25/2023] [Indexed: 09/30/2023]
Abstract
Small cell lung cancer (SCLC) has a poor prognosis, emphasizing the necessity for developing new therapies. The de novo synthesis pathway of purine nucleotides, which is involved in the malignant growth of SCLC, has emerged as a novel therapeutic target. Purine nucleotides are supplied by two pathways: de novo and salvage. However, the role of the salvage pathway in SCLC and the differences in utilization and crosstalk between the two pathways remain largely unclear. Here, we found that deletion of the HPRT1 gene, which codes for the rate-limiting enzyme of the purine salvage pathway, significantly suppressed tumor growth in vivo in several SCLC cells. We also demonstrated that HPRT1 expression confers resistance to lemetrexol (LMX), an inhibitor of the purine de novo pathway. Interestingly, HPRT1-knockout had less effect on SCLC SBC-5 cells, which are more sensitive to LMX than other SCLC cell lines, suggesting that a preference for either the purine de novo or salvage pathway occurs in SCLC. Furthermore, metabolome analysis of HPRT1-knockout cells revealed increased intermediates in the pentose phosphate pathway and elevated metabolic flux in the purine de novo pathway, indicating compensated metabolism between the de novo and salvage pathways in purine nucleotide biosynthesis. These results suggest that HPRT1 has therapeutic implications in SCLC and provide fundamental insights into the regulation of purine nucleotide biosynthesis. IMPLICATIONS SCLC tumors preferentially utilize either the de novo or salvage pathway in purine nucleotide biosynthesis, and HPRT1 has therapeutic implications in SCLC.
Collapse
Affiliation(s)
- Sho Tabata
- Tsuruoka Metabolomics Laboratory, National Cancer Center, Tsuruoka, Japan
- Shonai Regional Industry Promotion Center, Tsuruoka, Japan
- Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Shigeki Umemura
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Miyu Narita
- Tsuruoka Metabolomics Laboratory, National Cancer Center, Tsuruoka, Japan
- Shonai Regional Industry Promotion Center, Tsuruoka, Japan
| | - Hibiki Udagawa
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takamasa Ishikawa
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
| | - Masahiro Tsuboi
- Department of Thoracic Surgery, National Cancer Center Hospital East, Kashiwa, Japan
| | - Koichi Goto
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Genichiro Ishii
- Division of Pathology, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan
| | - Katsuya Tsuchihara
- Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Atsushi Ochiai
- Division of Biomarker Discovery, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
- Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
- Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| | - Susumu S. Kobayashi
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
- Division of Translational Genomics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Tomoyoshi Soga
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hideki Makinoshima
- Tsuruoka Metabolomics Laboratory, National Cancer Center, Tsuruoka, Japan
- Shonai Regional Industry Promotion Center, Tsuruoka, Japan
- Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
- Yamagata University, Yamagata, Japan
| |
Collapse
|
36
|
Nomura M, Ohuchi M, Sakamoto Y, Kudo K, Yaku K, Soga T, Sugiura Y, Morita M, Hayashi K, Miyahara S, Sato T, Yamashita Y, Ito S, Kikuchi N, Sato I, Saito R, Yaegashi N, Fukuhara T, Yamada H, Shima H, Nakayama KI, Hirao A, Kawasaki K, Arai Y, Akamatsu S, Tanuma SI, Sato T, Nakagawa T, Tanuma N. Niacin restriction with NAMPT-inhibition is synthetic lethal to neuroendocrine carcinoma. Nat Commun 2023; 14:8095. [PMID: 38092728 PMCID: PMC10719245 DOI: 10.1038/s41467-023-43630-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 11/13/2023] [Indexed: 12/17/2023] Open
Abstract
Nicotinamide phosphoribosyltransferase (NAMPT) plays a major role in NAD biosynthesis in many cancers and is an attractive potential cancer target. However, factors dictating therapeutic efficacy of NAMPT inhibitors (NAMPTi) are unclear. We report that neuroendocrine phenotypes predict lung and prostate carcinoma vulnerability to NAMPTi, and that NAMPTi therapy against those cancers is enhanced by dietary modification. Neuroendocrine differentiation of tumor cells is associated with down-regulation of genes relevant to quinolinate phosphoribosyltransferase-dependent de novo NAD synthesis, promoting NAMPTi susceptibility in vitro. We also report that circulating nicotinic acid riboside (NAR), a non-canonical niacin absent in culture media, antagonizes NAMPTi efficacy as it fuels NAMPT-independent but nicotinamide riboside kinase 1-dependent NAD synthesis in tumors. In mouse transplantation models, depleting blood NAR by nutritional or genetic manipulations is synthetic lethal to tumors when combined with NAMPTi. Our findings provide a rationale for simultaneous targeting of NAR metabolism and NAMPT therapeutically in neuroendocrine carcinoma.
Collapse
Affiliation(s)
- Miyuki Nomura
- Division of Cancer Chemotherapy, Miyagi Cancer Center Research Institute, Natori, Japan
| | - Mai Ohuchi
- Division of Cancer Chemotherapy, Miyagi Cancer Center Research Institute, Natori, Japan
| | - Yoshimi Sakamoto
- Division of Cancer Chemotherapy, Miyagi Cancer Center Research Institute, Natori, Japan
| | - Kei Kudo
- Division of Cancer Chemotherapy, Miyagi Cancer Center Research Institute, Natori, Japan
- Department of Biochemical Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Keisuke Yaku
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
| | - Yuki Sugiura
- Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Mami Morita
- Division of Cancer Chemotherapy, Miyagi Cancer Center Research Institute, Natori, Japan
| | - Kayoko Hayashi
- Division of Cancer Chemotherapy, Miyagi Cancer Center Research Institute, Natori, Japan
| | - Shuko Miyahara
- Division of Cancer Chemotherapy, Miyagi Cancer Center Research Institute, Natori, Japan
- Department of Biochemical Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Taku Sato
- Division of Cancer Chemotherapy, Miyagi Cancer Center Research Institute, Natori, Japan
| | - Yoji Yamashita
- Division of Cancer Chemotherapy, Miyagi Cancer Center Research Institute, Natori, Japan
| | - Shigemi Ito
- Division of Cancer Chemotherapy, Miyagi Cancer Center Research Institute, Natori, Japan
| | - Naohiko Kikuchi
- Division of Cancer Chemotherapy, Miyagi Cancer Center Research Institute, Natori, Japan
| | - Ikuro Sato
- Department of Pathology, Miyagi Cancer Center Hospital, Natori, Japan
| | - Rintaro Saito
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
| | - Nobuo Yaegashi
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tatsuro Fukuhara
- Department of Respiratory Medicine, Miyagi Cancer Center Hospital, Natori, Japan
| | - Hidekazu Yamada
- Division of Cancer Chemotherapy, Miyagi Cancer Center Research Institute, Natori, Japan
| | - Hiroshi Shima
- Division of Cancer Chemotherapy, Miyagi Cancer Center Research Institute, Natori, Japan
| | - Keiichi I Nakayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyusyu University, Fukuoka, Japan
- TMDU Advanced Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Atsushi Hirao
- Division of Molecular Genetics, Cancer and Stem Cell Research Program, Cancer Research Institute and WPI Nano Life Science Institute, Kanazawa University, Kanazawa, Japan
| | - Kenta Kawasaki
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Yoichi Arai
- Department of Urology, Miyagi Cancer Center Hospital, Natori, Japan
| | - Shusuke Akamatsu
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Sei-Ichi Tanuma
- Meikai University Research Institute of Odontology, Sakado, Japan
- University of Human Arts and Sciences, Saitama, Japan
| | - Toshiro Sato
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Takashi Nakagawa
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Nobuhiro Tanuma
- Division of Cancer Chemotherapy, Miyagi Cancer Center Research Institute, Natori, Japan.
- Department of Biochemical Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan.
| |
Collapse
|
37
|
Gao G, Xue Q, He J, Wu M, Jiang Y, Li Q, Zhang Y, Shi W. Single-cell RNA sequencing in double-hit lymphoma: IMPDH2 induces the progression of lymphoma by activating the PI3K/AKT/mTOR signaling pathway. Int Immunopharmacol 2023; 125:111125. [PMID: 37907047 DOI: 10.1016/j.intimp.2023.111125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 10/14/2023] [Accepted: 10/20/2023] [Indexed: 11/02/2023]
Abstract
BACKGROUND IMPDH2 is the rate-limiting enzyme of the de novo GTP synthesis pathway and has a key role in tumors; however, the specific mechanism underlying IMPDH2 activity in diffuse large B cell lymphoma (DLBCL) is still undetermined. This study aims to explore the potential mechanism of IMPDH2 in DLBCL, and its possible involvement in double-hit lymphoma (DHL), i.e., cases with translocations involving MYC and BCL2 and/or BCL6. METHODS Using single-cell sequencing and bioinformatics analysis to screen for IMPDH2. Exploring the differential expression of IMPDH2 and its correlation with prognosis through multiplexed immunofluorescence analysis. Using CCK8, EdU, clone formation assay, and animal model to analyze biological behavior changes after inhibiting IMPDH2. Explaining the potential mechanism of IMPDH2 in DLBCL by Western blot and multiplexed immunofluorescence. RESULTS Prognostic risk model was constructed by single-cell sequencing, which identified IMPDH2 as a DHL-related gene. IMPDH2 was highly expressed in cell lines and tissues, associated with poor patient prognosis and an independent prognostic factor. In vitro and in vivo experiments showed that IMPDH2 inhibition significantly inhibited DHL cell proliferation. Flow cytometry showed apoptosis and cycle arrest. Western blot results suggested that c-Myc regulated the activation of PI3K/AKT/mTOR signaling pathway by IMPDH2 to promote tumor development in DHL. Moreover, multiplex immunofluorescence revealed decreased T-cell infiltration within the tumor microenvironment exhibiting concurrent high expression of IMPDH2 and PD-L1. CONCLUSIONS Our results suggest that IMPDH2 functions as a tumor-promoting factor in DHL. This finding is expected to generate novel insights into the pathogenesis of these patients, thereby identifying potential therapeutic targets.
Collapse
Affiliation(s)
- Guangcan Gao
- Department of Oncology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong 226001, Jiangsu, China; Nantong University Medical School, 19, Qixiu Road, Nantong 226001, Jiangsu, China; Department of Clinical Biobank & Institute of Oncology, Nantong University Affiliated Hospital, Nantong 226001, Jiangsu, China
| | - Qingfeng Xue
- Department of Oncology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong 226001, Jiangsu, China; Nantong University Medical School, 19, Qixiu Road, Nantong 226001, Jiangsu, China; Department of Clinical Biobank & Institute of Oncology, Nantong University Affiliated Hospital, Nantong 226001, Jiangsu, China
| | - Jing He
- Department of Oncology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong 226001, Jiangsu, China; Nantong University Medical School, 19, Qixiu Road, Nantong 226001, Jiangsu, China; Department of Clinical Biobank & Institute of Oncology, Nantong University Affiliated Hospital, Nantong 226001, Jiangsu, China
| | - Meng Wu
- Department of Oncology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong 226001, Jiangsu, China
| | - Yongning Jiang
- Department of Oncology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong 226001, Jiangsu, China; Nantong University Medical School, 19, Qixiu Road, Nantong 226001, Jiangsu, China; Department of Clinical Biobank & Institute of Oncology, Nantong University Affiliated Hospital, Nantong 226001, Jiangsu, China
| | - Quanqing Li
- Department of Oncology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong 226001, Jiangsu, China; Nantong University Medical School, 19, Qixiu Road, Nantong 226001, Jiangsu, China; Department of Clinical Biobank & Institute of Oncology, Nantong University Affiliated Hospital, Nantong 226001, Jiangsu, China
| | - Yaping Zhang
- Nantong University Medical School, 19, Qixiu Road, Nantong 226001, Jiangsu, China; Department of Clinical Biobank & Institute of Oncology, Nantong University Affiliated Hospital, Nantong 226001, Jiangsu, China; Department of Hematology, Affiliated Hospital of Nantong University, 20, Xisi Road, Nantong 226001, Jiangsu, China.
| | - Wenyu Shi
- Department of Oncology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong 226001, Jiangsu, China; Nantong University Medical School, 19, Qixiu Road, Nantong 226001, Jiangsu, China; Department of Clinical Biobank & Institute of Oncology, Nantong University Affiliated Hospital, Nantong 226001, Jiangsu, China.
| |
Collapse
|
38
|
He X, Cui J, Ma H, Abuduaini N, Huang Y, Tang L, Wang W, Zhang Y, Wang Y, Lu W, Feng B, Huang J. Berberrubine is a novel and selective IMPDH2 inhibitor that impairs the growth of colorectal cancer. Biochem Pharmacol 2023; 218:115868. [PMID: 37871880 DOI: 10.1016/j.bcp.2023.115868] [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: 08/14/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 10/25/2023]
Abstract
Inosine monophosphate dehydrogenase (IMPDH) catalyzes the rate-limiting reaction in the de novo synthesis pathway of guanine nucleotides that is highly required for cancer cell outgrowth. Herein, we found that IMPDH isoform 2 (IMPDH2) is highly expressed in colorectal cancer (CRC) and is correlated with poor patient prognosis. Via structure-based virtual screening, we identified berberrubine, a critical ingredient of the medical plant Coptis chinensis, as a novel, selective, and competitive inhibitor of IMPDH2, which demonstrated over 15-fold selectivity to IMPDH2 than IMPDH1. Besides, we also confirmed the interaction between berberrubine and IMPDH2. Of note, berberrubine treatment significantly impairs the growth of human CRC cells in a dose-dependent manner, which can be rescued by supplementing with guanosine. Furthermore, oral administration of berberrubine remarkably reduced tumor volume and weight in a human cell line-derived xenograft model. Importantly, the anti-cancer activity of berberrubine was also confirmed by using the azoxymethane (AOM) / dextran sulfate sodium (DSS)-induced spontaneous CRC mouse model. Taken together, our study highlights that berberrubine acts as a novel IMPDH2 inhibitor, suppressing the growth of CRC in vitro and in vivo, providing a fresh perspective for its potential application in the treatment of CRC.
Collapse
Affiliation(s)
- Xiangli He
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Jiayan Cui
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Hui Ma
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Naijipu Abuduaini
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Huang
- Drug Inspection Technology, Guangdong Institute For Drug Control, 766 Shenzhou Road, Guangzhou 510663, China
| | - Lu Tang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Wanyan Wang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Yuanyuan Zhang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Yang Wang
- Department of Urology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Weiqiang Lu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China.
| | - Bo Feng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jin Huang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China.
| |
Collapse
|
39
|
Ali ES, Ben-Sahra I. Regulation of nucleotide metabolism in cancers and immune disorders. Trends Cell Biol 2023; 33:950-966. [PMID: 36967301 PMCID: PMC10518033 DOI: 10.1016/j.tcb.2023.03.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/05/2023] [Accepted: 03/08/2023] [Indexed: 04/03/2023]
Abstract
Nucleotides are the foundational elements of life. Proliferative cells acquire nutrients for energy production and the synthesis of macromolecules, including proteins, lipids, and nucleic acids. Nucleotides are continuously replenished through the activation of the nucleotide synthesis pathways. Despite the importance of nucleotides in cell physiology, there is still much to learn about how the purine and pyrimidine synthesis pathways are regulated in response to intracellular and exogenous signals. Over the past decade, evidence has emerged that several signaling pathways [Akt, mechanistic target of rapamycin complex I (mTORC1), RAS, TP53, and Hippo-Yes-associated protein (YAP) signaling] alter nucleotide synthesis activity and influence cell function. Here, we examine the mechanisms by which these signaling networks affect de novo nucleotide synthesis in mammalian cells. We also discuss how these molecular links can be targeted in diseases such as cancers and immune disorders.
Collapse
Affiliation(s)
- Eunus S Ali
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
| | - Issam Ben-Sahra
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
40
|
Lu Y, Wang S, Jiao Y. The Effects of Deregulated Ribosomal Biogenesis in Cancer. Biomolecules 2023; 13:1593. [PMID: 38002277 PMCID: PMC10669593 DOI: 10.3390/biom13111593] [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: 09/13/2023] [Revised: 10/04/2023] [Accepted: 10/22/2023] [Indexed: 11/26/2023] Open
Abstract
Ribosomes are macromolecular ribonucleoprotein complexes assembled from RNA and proteins. Functional ribosomes arise from the nucleolus, require ribosomal RNA processing and the coordinated assembly of ribosomal proteins (RPs), and are frequently hyperactivated to support the requirement for protein synthesis during the self-biosynthetic and metabolic activities of cancer cells. Studies have provided relevant information on targeted anticancer molecules involved in ribosome biogenesis (RiBi), as increased RiBi is characteristic of many types of cancer. The association between unlimited cell proliferation and alterations in specific steps of RiBi has been highlighted as a possible critical driver of tumorigenesis and metastasis. Thus, alterations in numerous regulators and actors involved in RiBi, particularly in cancer, significantly affect the rate and quality of protein synthesis and, ultimately, the transcriptome to generate the associated proteome. Alterations in RiBi in cancer cells activate nucleolar stress response-related pathways that play important roles in cancer-targeted interventions and immunotherapies. In this review, we focus on the association between alterations in RiBi and cancer. Emphasis is placed on RiBi deregulation and its secondary consequences, including changes in protein synthesis, loss of RPs, adaptive transcription and translation, nucleolar stress regulation, metabolic changes, and the impaired ribosome biogenesis checkpoint.
Collapse
Affiliation(s)
| | - Shizhuo Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110055, China;
| | - Yisheng Jiao
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110055, China;
| |
Collapse
|
41
|
Kodama M, Toyokawa G, Sugahara O, Sugiyama S, Haratake N, Yamada Y, Wada R, Takamori S, Shimokawa M, Takenaka T, Tagawa T, Kittaka H, Tsuruda T, Tanaka K, Komatsu Y, Nakata K, Imado Y, Yamazaki K, Okamoto I, Oda Y, Takahashi M, Izumi Y, Bamba T, Shimizu H, Yoshizumi T, Nakayama KI. Modulation of host glutamine anabolism enhances the sensitivity of small cell lung cancer to chemotherapy. Cell Rep 2023; 42:112899. [PMID: 37531252 DOI: 10.1016/j.celrep.2023.112899] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 06/22/2023] [Accepted: 07/14/2023] [Indexed: 08/04/2023] Open
Abstract
Small cell lung cancer (SCLC) is one of the deadliest human cancers, with a 5-year survival rate of ∼7%. Here, we performed a targeted proteomics analysis of human SCLC samples and thereby identified hypoxanthine phosphoribosyltransferase 1 (HPRT1) in the salvage purine synthesis pathway as a factor that contributes to SCLC malignancy by promoting cell survival in a glutamine-starved environment. Inhibition of HPRT1 by 6-mercaptopurine (6-MP) in combination with methotrexate (MTX), which blocks the de novo purine synthesis pathway, attenuated the growth of SCLC in mouse xenograft models. Moreover, modulation of host glutamine anabolism with the glutamine synthetase inhibitor methionine sulfoximine (MSO) in combination with 6-MP and MTX treatment resulted in marked tumor suppression and prolongation of host survival. Our results thus suggest that modulation of host glutamine anabolism combined with simultaneous inhibition of the de novo and salvage purine synthesis pathways may be of therapeutic benefit for SCLC.
Collapse
Affiliation(s)
- Manabu Kodama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Gouji Toyokawa
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; Department of Thoracic Surgery, NHO Kyushu Medical Center, 1-8-1 Jigyohama, Chuo-ku, Fukuoka 810-0065, Japan
| | - Osamu Sugahara
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Shigeaki Sugiyama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Naoki Haratake
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yuichi Yamada
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Reona Wada
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Shinkichi Takamori
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Mototsugu Shimokawa
- Department of Biostatistics, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, Yamaguchi 755-8505, Japan
| | - Tomoyoshi Takenaka
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Tetsuzo Tagawa
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Hiroki Kittaka
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; LSI Medience Corporation, 1-13-4 Uchikanda, Chiyoda-ku, Tokyo 101-8517, Japan
| | - Takeshi Tsuruda
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Kentaro Tanaka
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yushiro Komatsu
- Department of AI Systems Medicine, M&D Data Science Center, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Keisuke Nakata
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yuri Imado
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Koji Yamazaki
- Department of Thoracic Surgery, NHO Kyushu Medical Center, 1-8-1 Jigyohama, Chuo-ku, Fukuoka 810-0065, Japan
| | - Isamu Okamoto
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Masatomo Takahashi
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yoshihiro Izumi
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Takeshi Bamba
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Hideyuki Shimizu
- Department of AI Systems Medicine, M&D Data Science Center, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Tomoharu Yoshizumi
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Keiichi I Nakayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; Anticancer Strategies Laboratory, TMDU Advanced Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan.
| |
Collapse
|
42
|
Sugimoto A, Watanabe T, Matsuoka K, Okuno Y, Yanagi Y, Narita Y, Mabuchi S, Nobusue H, Sugihara E, Hirayama M, Ide T, Onouchi T, Sato Y, Kanda T, Saya H, Iwatani Y, Kimura H, Murata T. Growth Transformation of B Cells by Epstein-Barr Virus Requires IMPDH2 Induction and Nucleolar Hypertrophy. Microbiol Spectr 2023; 11:e0044023. [PMID: 37409959 PMCID: PMC10433962 DOI: 10.1128/spectrum.00440-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 06/20/2023] [Indexed: 07/07/2023] Open
Abstract
The in vitro growth transformation of primary B cells by Epstein-Barr virus (EBV) is the initial step in the development of posttransplant lymphoproliferative disorder (PTLD). We performed electron microscopic analysis and immunostaining of primary B cells infected with wild-type EBV. Interestingly, the nucleolar size was increased by two days after infection. A recent study found that nucleolar hypertrophy, which is caused by the induction of the IMPDH2 gene, is required for the efficient promotion of growth in cancers. In the present study, RNA-seq revealed that the IMPDH2 gene was significantly induced by EBV and that its level peaked at day 2. Even without EBV infection, the activation of primary B cells by the CD40 ligand and interleukin-4 increased IMPDH2 expression and nucleolar hypertrophy. Using EBNA2 or LMP1 knockout viruses, we found that EBNA2 and MYC, but not LMP1, induced the IMPDH2 gene during primary infections. IMPDH2 inhibition by mycophenolic acid (MPA) blocked the growth transformation of primary B cells by EBV, leading to smaller nucleoli, nuclei, and cells. Mycophenolate mofetil (MMF), which is a prodrug of MPA that is approved for use as an immunosuppressant, was tested in a mouse xenograft model. Oral MMF significantly improved the survival of mice and reduced splenomegaly. Taken together, these results indicate that EBV induces IMPDH2 expression through EBNA2-dependent and MYC-dependent mechanisms, leading to the hypertrophy of the nucleoli, nuclei, and cells as well as efficient cell proliferation. Our results provide basic evidence that IMPDH2 induction and nucleolar enlargement are crucial for B cell transformation by EBV. In addition, the use of MMF suppresses PTLD. IMPORTANCE EBV infections cause nucleolar enlargement via the induction of IMPDH2, which are essential for B cell growth transformation by EBV. Although the significance of IMPDH2 induction and nuclear hypertrophy in the tumorigenesis of glioblastoma has been reported, EBV infection brings about the change quickly by using its transcriptional cofactor, EBNA2, and MYC. Moreover, we present here, for the novel, basic evidence that an IMPDH2 inhibitor, namely, MPA or MMF, can be used for EBV-positive posttransplant lymphoproliferative disorder (PTLD).
Collapse
Affiliation(s)
- Atsuko Sugimoto
- Department of Virology, Fujita Health University School of Medicine, Toyoake, Japan
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
- Department of Virology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takahiro Watanabe
- Department of Virology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazuhiro Matsuoka
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Yusuke Okuno
- Department of Virology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yusuke Yanagi
- Department of Virology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yohei Narita
- Division of Infectious Disease, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Seiyo Mabuchi
- Department of Virology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Pathology and Laboratory Medicine, Nagoya University Hospital, Nagoya, Japan
| | - Hiroyuki Nobusue
- Division of Gene Regulation, Cancer Center, Research Promotion Headquarters, Fujita Health University, Toyoake, Japan
| | - Eiji Sugihara
- Division of Gene Regulation, Cancer Center, Research Promotion Headquarters, Fujita Health University, Toyoake, Japan
- Open Facility Center, Research Promotion Headquarters, Fujita Health University, Toyoake, Japan
| | - Masaya Hirayama
- Department of Morphology and Diagnostic Pathology, School of Medical Sciences, Fujita Health University, Toyoake, Japan
- Department of Biomedical Molecular Sciences, Graduate School of Medicine, Fujita Health University, Toyoake, Japan
| | - Tomihiko Ide
- Department of Virology, Fujita Health University School of Medicine, Toyoake, Japan
- Open Facility Center, Research Promotion Headquarters, Fujita Health University, Toyoake, Japan
| | - Takanori Onouchi
- Open Facility Center, Research Promotion Headquarters, Fujita Health University, Toyoake, Japan
| | - Yoshitaka Sato
- Department of Virology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Teru Kanda
- Department of Microbiology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Hideyuki Saya
- Division of Gene Regulation, Cancer Center, Research Promotion Headquarters, Fujita Health University, Toyoake, Japan
| | - Yasumasa Iwatani
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Hiroshi Kimura
- Department of Virology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takayuki Murata
- Department of Virology, Fujita Health University School of Medicine, Toyoake, Japan
- Department of Virology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
43
|
Pang H, Hu Z. Metabolomics in drug research and development: The recent advances in technologies and applications. Acta Pharm Sin B 2023; 13:3238-3251. [PMID: 37655318 PMCID: PMC10465962 DOI: 10.1016/j.apsb.2023.05.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 03/21/2023] [Accepted: 04/28/2023] [Indexed: 09/02/2023] Open
Abstract
Emerging evidence has demonstrated the vital role of metabolism in various diseases or disorders. Metabolomics provides a comprehensive understanding of metabolism in biological systems. With advanced analytical techniques, metabolomics exhibits unprecedented significant value in basic drug research, including understanding disease mechanisms, identifying drug targets, and elucidating the mode of action of drugs. More importantly, metabolomics greatly accelerates the drug development process by predicting pharmacokinetics, pharmacodynamics, and drug response. In addition, metabolomics facilitates the exploration of drug repurposing and drug-drug interactions, as well as the development of personalized treatment strategies. Here, we briefly review the recent advances in technologies in metabolomics and update our knowledge of the applications of metabolomics in drug research and development.
Collapse
Affiliation(s)
| | - Zeping Hu
- School of Pharmaceutical Sciences, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing 100084, China
| |
Collapse
|
44
|
Shi DD, Savani MR, Abdullah KG, McBrayer SK. Emerging roles of nucleotide metabolism in cancer. Trends Cancer 2023; 9:624-635. [PMID: 37173188 PMCID: PMC10967252 DOI: 10.1016/j.trecan.2023.04.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/14/2023] [Accepted: 04/18/2023] [Indexed: 05/15/2023]
Abstract
Nucleotides are substrates for multiple anabolic pathways, most notably DNA and RNA synthesis. Since nucleotide synthesis inhibitors began to be used for cancer therapy in the 1950s, our understanding of how nucleotides function in tumor cells has evolved, prompting a resurgence of interest in targeting nucleotide metabolism for cancer therapy. In this review, we discuss recent advances that challenge the idea that nucleotides are mere building blocks for the genome and transcriptome and highlight ways that these metabolites support oncogenic signaling, stress resistance, and energy homeostasis in tumor cells. These findings point to a rich network of processes sustained by aberrant nucleotide metabolism in cancer and reveal new therapeutic opportunities.
Collapse
Affiliation(s)
- Diana D Shi
- Department of Radiation Oncology, Brigham and Women's Hospital and Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| | - Milan R Savani
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA; Medical Scientist Training Program, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Kalil G Abdullah
- Department of Neurosurgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Hillman Comprehensive Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA
| | - Samuel K McBrayer
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA; Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Harrold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA.
| |
Collapse
|
45
|
Vízkeleti L, Spisák S. Rewired Metabolism Caused by the Oncogenic Deregulation of MYC as an Attractive Therapeutic Target in Cancers. Cells 2023; 12:1745. [PMID: 37443779 PMCID: PMC10341379 DOI: 10.3390/cells12131745] [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: 04/10/2023] [Revised: 06/20/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
MYC is one of the most deregulated oncogenes on multiple levels in cancer. As a node transcription factor, MYC plays a diverse regulatory role in many cellular processes, including cell cycle and metabolism, both in physiological and pathological conditions. The relentless growth and proliferation of tumor cells lead to an insatiable demand for energy and nutrients, which requires the rewiring of cellular metabolism. As MYC can orchestrate all aspects of cellular metabolism, its altered regulation plays a central role in these processes, such as the Warburg effect, and is a well-established hallmark of cancer development. However, our current knowledge of MYC suggests that its spatial- and concentration-dependent contribution to tumorigenesis depends more on changes in the global or relative expression of target genes. As the direct targeting of MYC is proven to be challenging due to its relatively high toxicity, understanding its underlying regulatory mechanisms is essential for the development of tumor-selective targeted therapies. The aim of this review is to comprehensively summarize the diverse forms of MYC oncogenic deregulation, including DNA-, transcriptional- and post-translational level alterations, and their consequences for cellular metabolism. Furthermore, we also review the currently available and potentially attractive therapeutic options that exploit the vulnerability arising from the metabolic rearrangement of MYC-driven tumors.
Collapse
Affiliation(s)
- Laura Vízkeleti
- Department of Bioinformatics, Faculty of Medicine, Semmelweis University, 1094 Budapest, Hungary;
| | - Sándor Spisák
- Institute of Enzymology, Research Centre for Natural Sciences, Eötvös Loránd Research Network, 1117 Budapest, Hungary
| |
Collapse
|
46
|
Pillai R, LeBoeuf SE, Hao Y, New C, Blum JLE, Rashidfarrokhi A, Huang SM, Bahamon C, Wu WL, Karadal-Ferrena B, Herrera A, Ivanova E, Cross M, Bossowski JP, Ding H, Hayashi M, Rajalingam S, Karakousi T, Sayin VI, Khanna KM, Wong KK, Wild R, Tsirigos A, Poirier JT, Rudin CM, Davidson SM, Koralov SB, Papagiannakopoulos T. Glutamine antagonist DRP-104 suppresses tumor growth and enhances response to checkpoint blockade in KEAP1 mutant lung cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.27.546750. [PMID: 37425844 PMCID: PMC10327154 DOI: 10.1101/2023.06.27.546750] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Loss-of-function mutations in KEAP1 frequently occur in lung cancer and are associated with resistance to standard of care treatment, highlighting the need for the development of targeted therapies. We have previously shown that KEAP1 mutant tumors have increased glutamine consumption to support the metabolic rewiring associated with NRF2 activation. Here, using patient-derived xenograft models and antigenic orthotopic lung cancer models, we show that the novel glutamine antagonist DRP-104 impairs the growth of KEAP1 mutant tumors. We find that DRP-104 suppresses KEAP1 mutant tumor growth by inhibiting glutamine-dependent nucleotide synthesis and promoting anti-tumor CD4 and CD8 T cell responses. Using multimodal single-cell sequencing and ex vivo functional assays, we discover that DRP-104 reverses T cell exhaustion and enhances the function of CD4 and CD8 T cells culminating in an improved response to anti-PD1 therapy. Our pre-clinical findings provide compelling evidence that DRP-104, currently in phase 1 clinical trials, offers a promising therapeutic approach for treating patients with KEAP1 mutant lung cancer. Furthermore, we demonstrate that by combining DRP-104 with checkpoint inhibition, we can achieve suppression of tumor intrinsic metabolism and augmentation of anti-tumor T cell responses.
Collapse
|
47
|
Liu SS, Li JS, Xue M, Wu WJ, Li X, Chen W. LncRNA UCA1 Participates in De Novo Synthesis of Guanine Nucleotides in Bladder Cancer by Recruiting TWIST1 to Increase IMPDH1/2. Int J Biol Sci 2023; 19:2599-2612. [PMID: 37215997 PMCID: PMC10197894 DOI: 10.7150/ijbs.82875] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 04/19/2023] [Indexed: 05/24/2023] Open
Abstract
Metabolic dysregulation has been identified as one of the hallmarks of cancer biology. Based on metabolic heterogeneity between bladder cancer tissues and adjacent tissues, we discovered several potential driving factors for the bladder cancer occurrence and development. Metabolic genomics showed purine metabolism pathway was mainly accumulated in bladder cancer. Long noncoding RNA urothelial carcinoma-associated 1 (LncRNA UCA1) is a potential tumor biomarker for bladder cancer diagnosis and prognosis, and it increases bladder cancer cell proliferation, migration, and invasion via the glycolysis pathway. However, whether UCA1 plays a role in purine metabolism in bladder cancer is unknown. Our findings showed that UCA1 could increase the transcription activity of guanine nucleotide de novo synthesis rate limiting enzyme inosine monophosphate dehydrogenase 1 (IMPDH1) and inosine monophosphate dehydrogenase 2 (IMPDH2), triggering in guanine nucleotide metabolic reprogramming. This process was achieved by UCA1 recruiting the transcription factor TWIST1 which binds to the IMPDH1and IMPDH2 promoter region. Increased guanine nucleotide synthesis pathway products stimulate RNA polymerase-dependent production of pre-ribosomal RNA and GTPase activity in bladder cancer cells, hence increasing bladder cancer cell proliferation, migration, and invasion. We have demonstrated that UCA1 regulates IMPDH1/2-mediated guanine nucleotide production via TWIST1, providing additional evidence of metabolic reprogramming.
Collapse
Affiliation(s)
- Shan-Shan Liu
- Clinical Laboratory, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Jia-Shu Li
- Clinical Laboratory, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Mei Xue
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Wen-Jing Wu
- Clinical Laboratory, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Xu Li
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Wei Chen
- Clinical Laboratory, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| |
Collapse
|
48
|
Zhou W, Zhao Z, Lin A, Yang J, Xu J, Kari WR, Yang A, Li J, Solanki S, Speth J, Walker N, Scott AJ, Kothari AU, Yao Y, Peterson ER, Korimerla N, Werner CK, Liang J, Jacobson J, Palavalasa S, Obrien AM, Elaimy AL, Ferris SP, Zhao SG, Sarkaria JN, Győrffy B, Zhang S, Al-Holou WN, Umemura Y, Morgan MA, Lawrence TS, Lyssiotis CA, Peters-Golden M, Shah YM, Wahl DR. GTP signaling links metabolism, DNA repair, and responses to genotoxic stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.12.536297. [PMID: 37090571 PMCID: PMC10120670 DOI: 10.1101/2023.04.12.536297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
How cell metabolism regulates DNA repair is incompletely understood. Here, we define a GTP-mediated signaling cascade that links metabolism to DNA repair and has significant therapeutic implications. GTP, but not other nucleotides, regulates the activity of Rac1, a G protein, that promotes the dephosphorylation of serine 323 on Abl-interactor 1 (Abi-1) by protein phosphatase 5 (PP5). Dephosphorylated Abi-1, a protein previously not known to activate DNA repair, promotes non-homologous end joining. In patients and mouse models of glioblastoma, Rac1 and dephosphorylated Abi-1 mediate DNA repair and resistance to standard of care genotoxic treatments. The GTP-Rac1-PP5-Abi-1 signaling axis is not limited to brain cancer, as GTP supplementation promotes DNA repair and Abi-1-S323 dephosphorylation in non-malignant cells and protects mouse tissues from genotoxic insult. This unexpected ability of GTP to regulate DNA repair independently of deoxynucleotide pools has important implications for normal physiology and cancer treatment.
Collapse
|
49
|
Lafita-Navarro MC, Conacci-Sorrell M. Nucleolar stress: From development to cancer. Semin Cell Dev Biol 2023; 136:64-74. [PMID: 35410715 PMCID: PMC9883801 DOI: 10.1016/j.semcdb.2022.04.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/29/2022] [Accepted: 04/02/2022] [Indexed: 02/06/2023]
Abstract
The nucleolus is a large nuclear membraneless organelle responsible for ribosome biogenesis. Ribosomes are cytoplasmic macromolecular complexes comprising RNA and proteins that link amino acids together to form new proteins. The biogenesis of ribosomes is an intricate multistep process that involves the transcription of ribosomal DNA (rDNA), the processing of ribosomal RNA (rRNA), and the assembly of rRNA with ribosomal proteins to form active ribosomes. Nearly all steps necessary for ribosome production and maturation occur in the nucleolus. Nucleolar shape, size, and number are directly linked to ribosome biogenesis. Errors in the steps of ribosomal biogenesis are sensed by the nucleolus causing global alterations in nucleolar function and morphology. This phenomenon, known as nucleolar stress, can lead to molecular changes such as stabilization of p53, which in turn activates cell cycle arrest or apoptosis. In this review, we discuss recent work on the association of nucleolar stress with degenerative diseases and developmental defects. In addition, we highlight the importance of de novo nucleotide biosynthesis for the enhanced nucleolar activity of cancer cells and discuss targeting nucleotide biosynthesis as a strategy to activate nucleolar stress to specifically target cancer cells.
Collapse
Affiliation(s)
- M Carmen Lafita-Navarro
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Maralice Conacci-Sorrell
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.
| |
Collapse
|
50
|
Casado P, Rio-Machin A, Miettinen JJ, Bewicke-Copley F, Rouault-Pierre K, Krizsan S, Parsons A, Rajeeve V, Miraki-Moud F, Taussig DC, Bödör C, Gribben J, Heckman C, Fitzgibbon J, Cutillas PR. Integrative phosphoproteomics defines two biologically distinct groups of KMT2A rearranged acute myeloid leukaemia with different drug response phenotypes. Signal Transduct Target Ther 2023; 8:80. [PMID: 36843114 PMCID: PMC9968719 DOI: 10.1038/s41392-022-01288-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 11/18/2022] [Accepted: 12/03/2022] [Indexed: 02/28/2023] Open
Abstract
Acute myeloid leukaemia (AML) patients harbouring certain chromosome abnormalities have particularly adverse prognosis. For these patients, targeted therapies have not yet made a significant clinical impact. To understand the molecular landscape of poor prognosis AML we profiled 74 patients from two different centres (in UK and Finland) at the proteomic, phosphoproteomic and drug response phenotypic levels. These data were complemented with transcriptomics analysis for 39 cases. Data integration highlighted a phosphoproteomics signature that define two biologically distinct groups of KMT2A rearranged leukaemia, which we term MLLGA and MLLGB. MLLGA presented increased DOT1L phosphorylation, HOXA gene expression, CDK1 activity and phosphorylation of proteins involved in RNA metabolism, replication and DNA damage when compared to MLLGB and no KMT2A rearranged samples. MLLGA was particularly sensitive to 15 compounds including genotoxic drugs and inhibitors of mitotic kinases and inosine-5-monosphosphate dehydrogenase (IMPDH) relative to other cases. Intermediate-risk KMT2A-MLLT3 cases were mainly represented in a third group closer to MLLGA than to MLLGB. The expression of IMPDH2 and multiple nucleolar proteins was higher in MLLGA and correlated with the response to IMPDH inhibition in KMT2A rearranged leukaemia, suggesting a role of the nucleolar activity in sensitivity to treatment. In summary, our multilayer molecular profiling of AML with poor prognosis and KMT2A-MLLT3 karyotypes identified a phosphoproteomics signature that defines two biologically and phenotypically distinct groups of KMT2A rearranged leukaemia. These data provide a rationale for the potential development of specific therapies for AML patients characterised by the MLLGA phosphoproteomics signature identified in this study.
Collapse
Affiliation(s)
- Pedro Casado
- Cell Signalling and Proteomics Group, Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, EC1M6BQ, UK
| | - Ana Rio-Machin
- Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, EC1M6BQ, UK
| | - Juho J Miettinen
- Institute for Molecular Medicine Finland - FIMM, HiLIFE - Helsinki Institute of Life Science, iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Findlay Bewicke-Copley
- Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, EC1M6BQ, UK
| | - Kevin Rouault-Pierre
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, EC1M6BQ, UK
| | - Szilvia Krizsan
- HCEMM-SU Molecular Oncohematology Research Group, 1st Department of Pathology and Experimental Cancer Research, Semmelweis University Budapest, Budapest, Hungary
| | - Alun Parsons
- Institute for Molecular Medicine Finland - FIMM, HiLIFE - Helsinki Institute of Life Science, iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Vinothini Rajeeve
- Cell Signalling and Proteomics Group, Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, EC1M6BQ, UK
| | - Farideh Miraki-Moud
- Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, Sutton, UK
| | - David C Taussig
- Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, Sutton, UK
| | - Csaba Bödör
- HCEMM-SU Molecular Oncohematology Research Group, 1st Department of Pathology and Experimental Cancer Research, Semmelweis University Budapest, Budapest, Hungary
| | - John Gribben
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, EC1M6BQ, UK
| | - Caroline Heckman
- Institute for Molecular Medicine Finland - FIMM, HiLIFE - Helsinki Institute of Life Science, iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Jude Fitzgibbon
- Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, EC1M6BQ, UK
| | - Pedro R Cutillas
- Cell Signalling and Proteomics Group, Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, EC1M6BQ, UK.
- The Alan Turing Institute, The British Library, 2QR, 96 Euston Rd, London, NW1 2DB, UK.
| |
Collapse
|