1
|
Wang L, Shi F, Cao Y, Xie L. Multiple roles of branched-chain amino acid metabolism in tumour progression. J Biomed Sci 2025; 32:41. [PMID: 40205401 PMCID: PMC11983764 DOI: 10.1186/s12929-025-01132-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: 07/18/2024] [Accepted: 03/09/2025] [Indexed: 04/11/2025] Open
Abstract
Metabolic reprogramming enables tumour cells to sustain their continuous proliferation and adapt to the ever-changing microenvironment. Branched-chain amino acids (BCAAs) and their metabolites are involved in intracellular protein synthesis and catabolism, signal transduction, epigenetic modifications, and the maintenance of oxidative homeostasis. Alterations in BCAA metabolism can influence the progression of various tumours. However, how BCAA metabolism is dysregulated differs among depending on tumour type; for example, it can manifest as decreased BCAA metabolism leading to BCAA accumulation, or as enhanced BCAA uptake and increased catabolism. In this review, we describe the role of BCAA metabolism in the progression of different tumours. As well as discuss how BCAA metabolic reprogramming drives tumour therapy resistance and evasion of the antitumour immune response, and how these pro-cancer effects are achieved in part by activating the mTORC signalling pathway. In-depth investigations into the potential mechanisms by which BCAA metabolic reprogramming affects tumorigenesis and tumour progression can enhance our understanding of the relationship between metabolism and cancer and provide new strategies for cancer therapy.
Collapse
Affiliation(s)
- Lin Wang
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, XiangYa Hospital, Central South University, Changsha, 410078, China
| | - Feng Shi
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, XiangYa Hospital, Central South University, Changsha, 410078, China
- Key Laboratory of Carcinogenesis of National Health Commission, Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, 410078, China
| | - Ya Cao
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, XiangYa Hospital, Central South University, Changsha, 410078, China
- Key Laboratory of Carcinogenesis of National Health Commission, Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, 410078, China
| | - Longlong Xie
- Department of Radiology, The Affiliated Children's Hospital of Xiangya School of Medicine (Hunan Children's Hospital), Central South University, Changsha, 410078, China.
| |
Collapse
|
2
|
Wu Q, Wang D, Zhou J, Li S, Niu X. The Essence of Nature Can Be the Simplest (1)-Warburg Effect: Transition From Intracellular ATP to Extracellular Fenton Chemistry. Chem Biodivers 2025:e202500313. [PMID: 40197846 DOI: 10.1002/cbdv.202500313] [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: 01/24/2025] [Revised: 03/28/2025] [Accepted: 04/08/2025] [Indexed: 04/10/2025]
Abstract
Here, we explain the energy mechanism behind the Warburg effect of aerobic glycolysis, which has been unsolved for a hundred years. We found that fungal cells that can engage in extracellular Fenton reactions share central carbon metabolism with cancer cells that can produce the Warburg effect. Fungal cells also undergo aerobic glycolysis, significantly reducing intracellular ATP levels and allocating large amounts of oxygen for the extracellular Fenton reactions. The use of aerobic glycolysis for the extracellular Fenton reaction can be a common phenomenon in nature, as glycolysis is a metabolic pathway that occurs in every cell. The development of extracellular Fenton reaction can be divided into rapid and slow formation. Rapid extracellular Fenton reactions occur predominantly in organisms that contain the key biosynthetic genes for secondary metabolite biosynthesis, while endotherms have limited capacity for slow extracellular reactions due to a lack of these critical genes. Endogenous aromatic metabolites can initiate strong extracellular Fenton reactions, and siderophores can sequester and recycle iron and protect the host from extracellular Fenton reactions. Most exogenous aromatics can induce an extracellular Fenton reaction reflux, thereby inhibiting cancer cells and pathogenetic microorganisms that exhibit stronger extracellular Fenton reactions than normal cells and non-pathogenetic microorganisms.
Collapse
Affiliation(s)
- Qunfu Wu
- School of Life Sciences, State Key Laboratory for Conservation and Utilization of Bio-Resources & Key Laboratory for Microbial Resources of the Ministry of Education, Yunnan University, Kunming, People's Republic of China
| | - Donglou Wang
- School of Life Sciences, State Key Laboratory for Conservation and Utilization of Bio-Resources & Key Laboratory for Microbial Resources of the Ministry of Education, Yunnan University, Kunming, People's Republic of China
| | - Jiao Zhou
- School of Life Sciences, State Key Laboratory for Conservation and Utilization of Bio-Resources & Key Laboratory for Microbial Resources of the Ministry of Education, Yunnan University, Kunming, People's Republic of China
| | - Shenghong Li
- State Key Laboratory of Phytochemistry and Natural Medicines, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, People's Republic of China
| | - Xuemei Niu
- School of Life Sciences, State Key Laboratory for Conservation and Utilization of Bio-Resources & Key Laboratory for Microbial Resources of the Ministry of Education, Yunnan University, Kunming, People's Republic of China
| |
Collapse
|
3
|
Wang H, Xu F, Wang C. Metabolic reprogramming of tumor microenviroment by engineered bacteria. Semin Cancer Biol 2025; 112:58-70. [PMID: 40157514 DOI: 10.1016/j.semcancer.2025.03.003] [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: 01/31/2025] [Revised: 03/16/2025] [Accepted: 03/21/2025] [Indexed: 04/01/2025]
Abstract
The tumor microenvironment (TME) is a complex ecosystem that plays a crucial role in tumor progression and response to therapy. The metabolic characteristics of the TME are fundamental to its function, influencing not only cancer cell proliferation and survival but also the behavior of immune cells within the tumor. Metabolic reprogramming-where cancer cells adapt their metabolic pathways to support rapid growth and immune evasion-has emerged as a key factor in cancer immunotherapy. Recently, the potential of engineered bacteria in cancer immunotherapy has gained increasing recognition, offering a novel strategy to modulate TME metabolism and enhance antitumor immunity. This review summarizes the metabolic properties and adaptations of tumor and immune cells within the TME and summarizes the strategies by which engineered bacteria regulate tumor metabolism. We discuss how engineered bacteria can overcome the immunosuppressive TME by reprogramming its metabolism to improve antitumor therapy. Furthermore, we examine the advantages, potential challenges, and future clinical translation of engineered bacteria in reshaping TME metabolism.
Collapse
Affiliation(s)
- Heng Wang
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China
| | - Fang Xu
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China
| | - Chao Wang
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China.
| |
Collapse
|
4
|
Han M, Si Y, Sun S, Hu J, Han Y, Liu X, Zhai Y, Su T, Cao F. Metabolism Plasticity on Account of Aspartate aminotransferase 10 Promotes Poplar Growth under Altered Nitrogen Regimes. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:6468-6485. [PMID: 40045927 DOI: 10.1021/acs.jafc.4c09107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Improving poplar productivity across a wide spectrum of nitrogen conditions is a primary objective in poplar breeding. In this research, we engineered transgenic poplars to overexpress the aspartate aminotransferase 10 (AspAT10) gene. The results showed that these transgenic plantlets significantly outperformed the wild-type control in terms of growth under both nitrogen-poor and nitrogen-rich conditions, exhibiting increased biomass, height, and root development. This improvement was linked to changes in internal nitrogen pools (including NO3-, NH4+, and total free amino acids) and sugar content. In line with the metabolic results, notable alterations in genes related to nitrogen and carbon metabolism as well as hormone signaling pathways were identified. Our findings highlight the versatile role of AspAT10 in regulating poplar's adaptation to variable nitrogen availability, attributed to the reversible nature of its catalytic reaction, which allows for the flexible reprogramming of nitrogen and carbon metabolism to align nitrogen supply with plant demand.
Collapse
Affiliation(s)
- Mei Han
- Co-Innovation Center for Sustainable Forestry in Southern China, College of Life Sciences, Nanjing Forestry University, Nanjing 210037, People's Republic of China
| | - Yujia Si
- Co-Innovation Center for Sustainable Forestry in Southern China, College of Life Sciences, Nanjing Forestry University, Nanjing 210037, People's Republic of China
| | - Shuyue Sun
- Co-Innovation Center for Sustainable Forestry in Southern China, College of Life Sciences, Nanjing Forestry University, Nanjing 210037, People's Republic of China
| | - Jinghan Hu
- College of Forestry, Nanjing Forestry University, Nanjing 210037, China
| | - Yirong Han
- Co-Innovation Center for Sustainable Forestry in Southern China, College of Life Sciences, Nanjing Forestry University, Nanjing 210037, People's Republic of China
| | - Xiaoning Liu
- Co-Innovation Center for Sustainable Forestry in Southern China, College of Life Sciences, Nanjing Forestry University, Nanjing 210037, People's Republic of China
| | - Yujie Zhai
- Co-Innovation Center for Sustainable Forestry in Southern China, College of Life Sciences, Nanjing Forestry University, Nanjing 210037, People's Republic of China
| | - Tao Su
- Co-Innovation Center for Sustainable Forestry in Southern China, College of Life Sciences, Nanjing Forestry University, Nanjing 210037, People's Republic of China
- Key Laboratory of State Forestry Administration on Subtropical Forest Biodiversity Conservation, Nanjing Forestry University, Nanjing 210037, People's Republic of China
| | - Fuliang Cao
- College of Forestry, Nanjing Forestry University, Nanjing 210037, China
| |
Collapse
|
5
|
Khemchandani R, Pardhi E, Goud S, Tomar DS, Medtiya P, Khushwaha BS, Dhiman V, Golla VM, Shaikh N, Godugu C, Mehra NK, Samanthula G. Multi-method coamorphous systems of lumefantrine with alpha ketoglutaric acid: Comprehensive characterization, biological evaluation and stability analysis. Int J Pharm 2025; 669:125056. [PMID: 39653294 DOI: 10.1016/j.ijpharm.2024.125056] [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: 09/07/2024] [Revised: 12/04/2024] [Accepted: 12/06/2024] [Indexed: 12/15/2024]
Abstract
The coamorphous systems (CAM) of lumefantrine (LMF) and alpha-ketoglutaric acid (KGA) were developed using three different methods to address the solubility and bioavailability limitations of LMF. Powder X-ray diffraction spectroscopy (PXRD) and the differential scanning calorimetry (DSC) confirmed the complete amorphization of the three CAM by the halo pattern and glass transition temperature (Tg), respectively. Attenuated total reflectance-Fourier transform infrared spectroscopy (ATR-FTIR) and nuclear magnetic resonance spectroscopy (NMR) results indicated that intermolecular interactions existed in the three CAM. Solid-state molecular dynamics simulations revealed the different intermolecular disordered environments present in all three CAM. Solubility analysis showed 14.73×, 12.8× and 9.81× improvement in the liquid-assisted grinding-based coamorphous system (CAM LAG), solvent evaporation-based coamorphous system (CAM SE) and quench-cool-based coamorphous system (CAM QC), respectively, compared to LMF crystalline (LMF CRY). In the in-vitro dissolution experiment 2.63-, 2.16- and 2.17-times increments were observed in CAM LAG, CAM SE and CAM QC compared to LMF CRY, respectively. In-vivo pharmacokinetics study revealed 10.86-, 9.24- and 8.46- folds increments in Cmax of CAM LAG, CAM SE and CAM QC, respectively, compared to LMF CRY. All three CAM illustrated anti-cancer activity in the A549 lung cancer cell line. Molecular dynamics simulation of LMF and KGA with Epidermal growth factor receptor (EGFR), a target for lung cancer treatment, revealed good binding affinity and stability. Stability studies performed under accelerated storage (40°C/75% RH) and extreme environmental conditions indicated that all three CAM have good stability. Different methods based prepared CAM have shown different physicochemical properties, bioavailability and stability profiles. These findings suggest that LMF-KGA CAM effectively addresses the solubility and bioavailability challenges of LMF, potentially leading to better therapeutic outcomes in lung cancer treatment.
Collapse
Affiliation(s)
- Rahul Khemchandani
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Ekta Pardhi
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Sridivya Goud
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Devendra Singh Tomar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Pravin Medtiya
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Bhoopendra Singh Khushwaha
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Vivek Dhiman
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Vijaya Madhyanapu Golla
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Nadeem Shaikh
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Chandraiah Godugu
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Neelesh Kumar Mehra
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Gananadhamu Samanthula
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India.
| |
Collapse
|
6
|
Evans L, Trinder S, Dodgshun A, Eisenstat DD, Whittle JR, Hansford JR, Valvi S. IDH-mutant gliomas in children and adolescents - from biology to clinical trials. Front Oncol 2025; 14:1515538. [PMID: 39876890 PMCID: PMC11773619 DOI: 10.3389/fonc.2024.1515538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 12/10/2024] [Indexed: 01/31/2025] Open
Abstract
Gliomas account for nearly 30% of all primary central nervous system (CNS) tumors in children and adolescents and young adults (AYA), contributing to significant morbidity and mortality. The updated molecular classification of gliomas defines molecularly diverse subtypes with a spectrum of tumors associated with age-distinct incidence. In adults, gliomas are characterized by the presence or absence of mutations in isocitrate dehydrogenase (IDH), with mutated IDH (mIDH) gliomas providing favorable outcomes and avenues for targeted therapy with the emergence of mIDH inhibitors. Despite their rarity, IDH mutations have been reported in 5-15% of pediatric glioma cases. Those with primary mismatch-repair deficient mIDH astrocytomas (PMMRDIA) have a particularly poor prognosis. Here, we describe the biology of mIDH gliomas and review the literature regarding the emergence of mIDH inhibitors, including clinical trials in adults. Given the paucity of clinical trial data from pediatric patients with mIDH glioma, we propose guidelines for the inclusion of pediatric and AYA patients with gliomas onto prospective trials and expanded access programs as well as the potential of combined mIDH inhibition and immunotherapy in the treatment of patients with PMMRDIA at high risk of progression.
Collapse
Affiliation(s)
- Louise Evans
- Michael Rice Centre for Hematology and Oncology, Women’s and Children’s Hospital, North Adelaide, SA, Australia
| | - Sarah Trinder
- Kids Cancer Centre, Sydney Children’s Hospital, Sydney, NSW, Australia
- Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales Sydney, Sydney, NSW, Australia
| | - Andrew Dodgshun
- Department of Pediatrics, University of Otago, Christchurch, New Zealand
- Children’s Hematology/Oncology Centre, Christchurch Hospital, Christchurch, New Zealand
| | - David D. Eisenstat
- Children’s Cancer Centre, Royal Children’s Hospital, Melbourne, VIC, Australia
- Department of Stem Cell Medicine, Murdoch Children’s Research Institute, Melbourne, VIC, Australia
- Department of Pediatrics, University of Melbourne, Melbourne, VIC, Australia
| | - James R. Whittle
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Personalized Oncology Division, Walter and Eliza Hall Institute (WEHI), Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Jordan R. Hansford
- Michael Rice Centre for Hematology and Oncology, Women’s and Children’s Hospital, North Adelaide, SA, Australia
- Pediatric Neuro-Oncology, Precision Cancer Medicine, South Australia Health and Medical Reseach Institute, Adelaide, SA, Australia
- South Australia ImmunoGENomics Cancer Institute, University of Adelaide, Adelaide, SA, Australia
| | - Santosh Valvi
- Department of Pediatric and Adolescent Oncology/Hematology, Perth Children’s Hospital, Nedlands, WA, Australia
- Brain Tumor Research Program, Telethon Kids Institute, Nedlands, WA, Australia
- School of Medicine, Division of Pediatrics, The University of Western Australia, Perth, WA, Australia
| |
Collapse
|
7
|
Wang Y, He Z, Dong X, Yao Y, Chen Q, Shi Y, Deng Y, Zhang Q, Yu L, Wang C. Regulation and therapy: the role of ferroptosis in DLBCL. Front Pharmacol 2025; 15:1458412. [PMID: 39834804 PMCID: PMC11743434 DOI: 10.3389/fphar.2024.1458412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 12/09/2024] [Indexed: 01/22/2025] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common subtype of B-cell non-Hodgkin's lymphoma (NHL), up to 30%-40% of patients will relapse and 10%-15% of patients have primary refractory disease, so exploring new treatment options is necessary. Ferroptosis is a non-apoptotic cell death mode discovered in recent years. Its occurrence pathway plays an essential impact on the therapeutic effect of tumors. Numerous studies have shown that modulating critical factors in the ferroptosis pathway can influence the growth of tumor cells in hematological malignancies including DLBCL. This review highlights recent advances in ferroptosis-related genes (FRGs), including STAT3, Nrf2, and ZEB1, and focuses on the clinical potential of ferroptosis inducers such as IKE, α-KG, DMF, and APR-246, which are currently being explored in clinical studies for their therapeutic effects in DLBCL. Correlational studies provide a novel idea for the research and treatment of ferroptosis in DLBCL and other hematological malignancies and lay a solid foundation for future studies.
Collapse
Affiliation(s)
- Yifan Wang
- Department of Hematology, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huai’an, China
- Northern Jiangsu Institute of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Zhengmei He
- Department of Hematology, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huai’an, China
- Northern Jiangsu Institute of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Xinyu Dong
- Northern Jiangsu Institute of Clinical Medicine, Nanjing Medical University, Nanjing, China
- Department of Hematology, The Huaian Clinical College of Xuzhou Medical University, Huai’an, China
| | - Yiming Yao
- Northern Jiangsu Institute of Clinical Medicine, Nanjing Medical University, Nanjing, China
- Department of Hematology, The Huaian Clinical College of Xuzhou Medical University, Huai’an, China
| | - Qiuni Chen
- Department of Hematology, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huai’an, China
- Northern Jiangsu Institute of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Yuye Shi
- Department of Hematology, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huai’an, China
- Northern Jiangsu Institute of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Yuan Deng
- Department of Hematology, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huai’an, China
- Northern Jiangsu Institute of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Quane Zhang
- Department of Hematology, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huai’an, China
- Northern Jiangsu Institute of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Liang Yu
- Department of Hematology, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huai’an, China
- Northern Jiangsu Institute of Clinical Medicine, Nanjing Medical University, Nanjing, China
- Department of Hematology, The Huaian Clinical College of Xuzhou Medical University, Huai’an, China
| | - Chunling Wang
- Department of Hematology, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huai’an, China
- Northern Jiangsu Institute of Clinical Medicine, Nanjing Medical University, Nanjing, China
- Department of Hematology, The Huaian Clinical College of Xuzhou Medical University, Huai’an, China
| |
Collapse
|
8
|
Chang KJ, Shiau LY, Lin SC, Cheong HP, Wang CY, Ma C, Liang YW, Yang YP, Ko PS, Hsu CH, Chiou SH. N 6-methyladenosine and its epitranscriptomic effects on hematopoietic stem cell regulation and leukemogenesis. Mol Med 2024; 30:196. [PMID: 39497033 PMCID: PMC11536562 DOI: 10.1186/s10020-024-00965-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 10/20/2024] [Indexed: 11/06/2024] Open
Abstract
N6-methyladenosine (m6A) RNA modification orchestrates cellular epitranscriptome through tuning the homeostasis of transcript stability, translation efficiency, and the transcript affinity toward RNA-binding proteins (RBPs). An aberrant m6A deposition on RNA can lead toward oncogenic expression profile (mRNA), impaired mitochondrial metabolism (mtRNA), and translational suppression (rRNA) of tumor suppressor genes. In addition, non-coding RNAs (ncRNAs), such as X-inactive specific transcript (XIST), miRNAs, and α-ketoglutarate-centric metabolic transcripts are also regulated by the m6A epitranscriptome. Notably, recent studies had uncovered a myriad of m6A-modified transcripts the center of hematopoietic stem cell (HSC) regulation, in which m6A modification act as a context dependent switch to the on and off of hematopoietic stem cell (HSC) maintenance, lineage commitment and terminal differentiation. In this review, we sequentially unfold the m6A mediated epithelial-to-hematopoietic transition in progenitor blood cell production, lymphocytic lineage expansion (T cells, B cells, NK cells, and non-NK ILCs), and the m6A crosstalk with the onco-metabolic prospects of leukemogenesis. Together, an encompassing body of evidence highlighted the emerging m6A significance in the regulation of HSC biology and leukemogenesis.
Collapse
Affiliation(s)
- Kao-Jung Chang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Li-Yang Shiau
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Shiuan-Chen Lin
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Han-Ping Cheong
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Pharmacology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ching-Yun Wang
- Department of Medical Education, Taichung Veterans General Hospital, Taipei, Taiwan
| | - Chun Ma
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yan-Wen Liang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Life Sciences and Institute of Genomic Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Yi-Ping Yang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Po-Shen Ko
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Hematology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chih-Hung Hsu
- The Fourth Affiliated Hospital, and Department of Environmental Medicine, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Genetics, International School of Medicine, Zhejiang University, Hangzhou, China
| | - Shih-Hwa Chiou
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan.
- Institute of Pharmacology, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
9
|
Trejo-Solís C, Serrano-García N, Castillo-Rodríguez RA, Robledo-Cadena DX, Jimenez-Farfan D, Marín-Hernández Á, Silva-Adaya D, Rodríguez-Pérez CE, Gallardo-Pérez JC. Metabolic dysregulation of tricarboxylic acid cycle and oxidative phosphorylation in glioblastoma. Rev Neurosci 2024; 35:813-838. [PMID: 38841811 DOI: 10.1515/revneuro-2024-0054] [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: 04/16/2024] [Accepted: 05/21/2024] [Indexed: 06/07/2024]
Abstract
Glioblastoma multiforme (GBM) exhibits genetic alterations that induce the deregulation of oncogenic pathways, thus promoting metabolic adaptation. The modulation of metabolic enzyme activities is necessary to generate nucleotides, amino acids, and fatty acids, which provide energy and metabolic intermediates essential for fulfilling the biosynthetic needs of glioma cells. Moreover, the TCA cycle produces intermediates that play important roles in the metabolism of glucose, fatty acids, or non-essential amino acids, and act as signaling molecules associated with the activation of oncogenic pathways, transcriptional changes, and epigenetic modifications. In this review, we aim to explore how dysregulated metabolic enzymes from the TCA cycle and oxidative phosphorylation, along with their metabolites, modulate both catabolic and anabolic metabolic pathways, as well as pro-oncogenic signaling pathways, transcriptional changes, and epigenetic modifications in GBM cells, contributing to the formation, survival, growth, and invasion of glioma cells. Additionally, we discuss promising therapeutic strategies targeting key players in metabolic regulation. Therefore, understanding metabolic reprogramming is necessary to fully comprehend the biology of malignant gliomas and significantly improve patient survival.
Collapse
Affiliation(s)
- Cristina Trejo-Solís
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Laboratorio de Neurobiología Molecular y Celular, Laboratorio de Neurofarmacología Molecular y Nanotecnología, Instituto Nacional de Neurología y Neurocirugía, Ciudad de Mexico 14269, Mexico
| | - Norma Serrano-García
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Laboratorio de Neurobiología Molecular y Celular, Laboratorio de Neurofarmacología Molecular y Nanotecnología, Instituto Nacional de Neurología y Neurocirugía, Ciudad de Mexico 14269, Mexico
| | - Rosa Angelica Castillo-Rodríguez
- CICATA Unidad Morelos, Instituto Politécnico Nacional, Boulevard de la Tecnología, 1036 Z-1, P 2/2, Atlacholoaya, Xochitepec 62790, Mexico
| | - Diana Xochiquetzal Robledo-Cadena
- Departamento de Fisiopatología Cardio-Renal, Departamento de Bioquímica, Instituto Nacional de Cardiología, Ciudad de México 14080, Mexico
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Coyoacán, 04510, Ciudad de México, Mexico
| | - Dolores Jimenez-Farfan
- Laboratorio de Inmunología, División de Estudios de Posgrado e Investigación, Facultad de Odontología, Universidad Nacional Autónoma de México, Ciudad de Mexico 04510, Mexico
| | - Álvaro Marín-Hernández
- Departamento de Fisiopatología Cardio-Renal, Departamento de Bioquímica, Instituto Nacional de Cardiología, Ciudad de México 14080, Mexico
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Coyoacán, 04510, Ciudad de México, Mexico
| | - Daniela Silva-Adaya
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Laboratorio de Neurobiología Molecular y Celular, Laboratorio de Neurofarmacología Molecular y Nanotecnología, Instituto Nacional de Neurología y Neurocirugía, Ciudad de Mexico 14269, Mexico
| | - Citlali Ekaterina Rodríguez-Pérez
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Laboratorio de Neurobiología Molecular y Celular, Laboratorio de Neurofarmacología Molecular y Nanotecnología, Instituto Nacional de Neurología y Neurocirugía, Ciudad de Mexico 14269, Mexico
| | - Juan Carlos Gallardo-Pérez
- Departamento de Fisiopatología Cardio-Renal, Departamento de Bioquímica, Instituto Nacional de Cardiología, Ciudad de México 14080, Mexico
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Coyoacán, 04510, Ciudad de México, Mexico
| |
Collapse
|
10
|
Liu S, Zhou Y, Chen Y, Qiao Y, Bai L, Zhang S, Men D, Zhang H, Pan F, Gao Y, Wang J, Wang Y. Isocitrate dehydrogenases 2-mediated dysfunctional metabolic reprogramming promotes intestinal cancer progression via regulating HIF-1A signaling pathway. Int Immunopharmacol 2024; 140:112828. [PMID: 39094359 DOI: 10.1016/j.intimp.2024.112828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 08/04/2024]
Abstract
Changes in isocitrate dehydrogenases (IDH) lead to the production of the cancer-causing metabolite 2-hydroxyglutarate, making them a cause of cancer. However, the specific role of IDH in the progression of colon cancer is still not well understood. Our current study provides evidence that IDH2 is significantly increased in colorectal cancer (CRC) cells and actively promotes cell growth in vitro and the development of tumors in vivo. Inhibiting the activity of IDH2, either through genetic silencing or pharmacological inhibition, results in a significant increase in α-ketoglutarate (α-KG), indicating a decrease in the reductive citric acid cycle. The excessive accumulation of α-KG caused by the inactivation of IDH2 obstructs the generation of ATP in mitochondria and promotes the downregulation of HIF-1A, eventually inhibiting glycolysis. This dual metabolic impact results in a reduction in ATP levels and the suppression of tumor growth. Our study reveals a metabolic trait of colorectal cancer cells, which involves the active utilization of glutamine through reductive citric acid cycle metabolism. The data suggests that IDH2 plays a crucial role in this metabolic process and has the potential to be a valuable target for the advancement of treatments for colorectal cancer.
Collapse
Affiliation(s)
- Shixiong Liu
- Department of Geriatrics, The First Hospital of Lanzhou University, Lanzhou 730000, China; Center of Hyperbaric Oxygen Therapy, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Yun Zhou
- Department of Geriatrics, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Yarong Chen
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
| | - Yuqin Qiao
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
| | - Lumucao Bai
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
| | - Shenhua Zhang
- Center of R&D for New Drug Discovery and Innovation, Nanjing BioMed Institute, Nanjing 25000, China
| | - Dongfang Men
- Center of R&D for New Drug Discovery and Innovation, Nanjing BioMed Institute, Nanjing 25000, China
| | - Haibu Zhang
- Center of R&D for New Drug Discovery and Innovation, Nanjing BioMed Institute, Nanjing 25000, China
| | - Fen Pan
- Center of R&D for New Drug Discovery and Innovation, Nanjing BioMed Institute, Nanjing 25000, China
| | - Yongshen Gao
- Center of R&D for New Drug Discovery and Innovation, Nanjing BioMed Institute, Nanjing 25000, China
| | - Jijing Wang
- Center of R&D for New Drug Discovery and Innovation, Nanjing BioMed Institute, Nanjing 25000, China
| | - Yuping Wang
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
11
|
Garcia I, Cornely K, Peterson CN, Berkmen MB. Roles of the oncometabolite enantiomers of 2-hydroxyglutarate and their metabolism by diverse dehydrogenases. Essays Biochem 2024; 68:161-171. [PMID: 38919140 DOI: 10.1042/ebc20230077] [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: 04/30/2024] [Revised: 06/09/2024] [Accepted: 06/11/2024] [Indexed: 06/27/2024]
Abstract
2-Hydroxyglutarate (2HG) is an oncometabolite that can contribute to tumor progression. Two enantiomer forms, L-2HG and D-2HG, arise from independent pathways starting from the precursor α-ketoglutarate (αKG). L-2HG production occurs through the promiscuous activities of malate dehydrogenase (MDH) and lactate dehydrogenase (LDH) under acidic and/or hypoxic conditions. D-2HG frequently accumulates by gain-of-function mutations in the genes encoding two isoforms of isocitrate dehydrogenase (IDH1 and IDH2). Cognate metabolite repair enzymes, L- and D-2-hydroxyglutarate dehydrogenases, oxidize the enantiomers and cause abnormally high 2HG accumulation and disease when mutated. Elevated levels of either oncometabolite affect redox homeostasis, metabolism, and immune system functioning. Moreover, the oncometabolites inhibit several α-ketoglutarate-dependent dioxygenases resulting in epigenetic changes such as DNA and histone hypermethylation as well as deficiencies in DNA repair. L-2HG, and D-2HG in some cases, inhibit degradation of hypoxia-inducible factor (HIF1α), a transcription factor that alters gene expression to adapt to hypoxic conditions, favoring tumorigenesis. Patients with the rare disease 2-hydroxyglutaric aciduria (2HGA) have exceedingly high levels of 2HG, which is neurotoxic, causing developmental delays and brain abnormalities. D-2HG also has specific effects on collagen production and NADPH pools. Recently, D-2HG has been targeted in new chemotherapies aimed at disrupting the gain-of-function IDH1 and IDH2 mutants, resulting in successful clinical trials for several cancers.
Collapse
Affiliation(s)
- Ivelitza Garcia
- Department of Chemistry, Allegheny College, Meadville, PA, U.S.A
| | - Kathleen Cornely
- Department of Chemistry and Biochemistry, Providence College, Providence, RI, U.S.A
| | | | - Melanie B Berkmen
- Department of Biochemistry, Chemistry, Environment, and Physics, Suffolk University, Boston, MA, U.S.A
| |
Collapse
|
12
|
Álvarez-González E, Sierra LM. Tricarboxylic Acid Cycle Relationships with Non-Metabolic Processes: A Short Story with DNA Repair and Its Consequences on Cancer Therapy Resistance. Int J Mol Sci 2024; 25:9054. [PMID: 39201738 PMCID: PMC11355010 DOI: 10.3390/ijms25169054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/08/2024] [Accepted: 08/20/2024] [Indexed: 09/03/2024] Open
Abstract
Metabolic changes involving the tricarboxylic acid (TCA) cycle have been linked to different non-metabolic cell processes. Among them, apart from cancer and immunity, emerges the DNA damage response (DDR) and specifically DNA damage repair. The oncometabolites succinate, fumarate and 2-hydroxyglutarate (2HG) increase reactive oxygen species levels and create pseudohypoxia conditions that induce DNA damage and/or inhibit DNA repair. Additionally, by influencing DDR modulation, they establish direct relationships with DNA repair on at least four different pathways. The AlkB pathway deals with the removal of N-alkylation DNA and RNA damage that is inhibited by fumarate and 2HG. The MGMT pathway acts in the removal of O-alkylation DNA damage, and it is inhibited by the silencing of the MGMT gene promoter by 2HG and succinate. The other two pathways deal with the repair of double-strand breaks (DSBs) but with opposite effects: the FH pathway, which uses fumarate to help with the repair of this damage, and the chromatin remodeling pathway, in which oncometabolites inhibit its repair by impairing the homologous recombination repair (HRR) system. Since oncometabolites inhibit DNA repair, their removal from tumor cells will not always generate a positive response in cancer therapy. In fact, their presence contributes to longer survival and/or sensitization against tumor therapy in some cancer patients.
Collapse
Affiliation(s)
- Enol Álvarez-González
- Departamento de Biología Funcional, Área de Genética, University of Oviedo, C/Julián Clavería s/n, 33006 Oviedo, Spain;
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias, Avda. HUCA s/n, 33011 Oviedo, Spain
| | - Luisa María Sierra
- Departamento de Biología Funcional, Área de Genética, University of Oviedo, C/Julián Clavería s/n, 33006 Oviedo, Spain;
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias, Avda. HUCA s/n, 33011 Oviedo, Spain
| |
Collapse
|
13
|
Zhang T, Pan Z, Gao J, Wu Q, Bai G, Li Y, Tong L, Feng F, Lai M, Liu Y, Song P, Ning Y, Tang H, Luo W, Chen Y, Fang Y, Zhang H, Liu Q, Zhang Y, Wang H, Chen Z, Chen Y, Geng M, Ji H, Zhao G, Zhou H, Ding J, Xie H. Branched-chain amino acid transaminase 1 confers EGFR-TKI resistance through epigenetic glycolytic activation. Signal Transduct Target Ther 2024; 9:216. [PMID: 39143065 PMCID: PMC11324870 DOI: 10.1038/s41392-024-01928-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 06/26/2024] [Accepted: 07/24/2024] [Indexed: 08/16/2024] Open
Abstract
Third-generation EGFR tyrosine kinase inhibitors (TKIs), exemplified by osimertinib, have demonstrated promising clinical efficacy in the treatment of non-small cell lung cancer (NSCLC). Our previous work has identified ASK120067 as a novel third-generation EGFR TKI with remarkable antitumor effects that has undergone New Drug Application (NDA) submission in China. Despite substantial progress, acquired resistance to EGFR-TKIs remains a significant challenge, impeding the long-term effectiveness of therapeutic approaches. In this study, we conducted a comprehensive investigation utilizing high-throughput proteomics analysis on established TKI-resistant tumor models, and found a notable upregulation of branched-chain amino acid transaminase 1 (BCAT1) expression in both osimertinib- and ASK120067-resistant tumors compared with the parental TKI-sensitive NSCLC tumors. Genetic depletion or pharmacological inhibition of BCAT1 impaired the growth of resistant cells and partially re-sensitized tumor cells to EGFR TKIs. Mechanistically, upregulated BCAT1 in resistant cells reprogrammed branched-chain amino acid (BCAA) metabolism and promoted alpha ketoglutarate (α-KG)-dependent demethylation of lysine 27 on histone H3 (H3K27) and subsequent transcriptional derepression of glycolysis-related genes, thereby enhancing glycolysis and promoting tumor progression. Moreover, we identified WQQ-345 as a novel BCAT1 inhibitor exhibiting antitumor activity both in vitro and in vivo against TKI-resistant lung cancer with high BCAT1 expression. In summary, our study highlighted the crucial role of BCAT1 in mediating resistance to third-generation EGFR-TKIs through epigenetic activation of glycolysis in NSCLC, thereby supporting BCAT1 as a promising therapeutic target for the treatment of TKI-resistant NSCLC.
Collapse
MESH Headings
- Humans
- ErbB Receptors/genetics
- ErbB Receptors/metabolism
- Drug Resistance, Neoplasm/genetics
- Drug Resistance, Neoplasm/drug effects
- Transaminases/genetics
- Transaminases/metabolism
- Protein Kinase Inhibitors/pharmacology
- Glycolysis/drug effects
- Glycolysis/genetics
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/metabolism
- Epigenesis, Genetic/drug effects
- Epigenesis, Genetic/genetics
- Mice
- Lung Neoplasms/genetics
- Lung Neoplasms/drug therapy
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Acrylamides/pharmacology
- Animals
- Aniline Compounds/pharmacology
- Cell Line, Tumor
- Gene Expression Regulation, Neoplastic/drug effects
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Indoles
- Pyrimidines
Collapse
Affiliation(s)
- Tao Zhang
- Division of Antitumor Pharmacology & Analytical Research Center for Organic and Biological Molecules & State Key Laboratory of Drug Research & Small-Molecule Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Zilu Pan
- Division of Antitumor Pharmacology & Analytical Research Center for Organic and Biological Molecules & State Key Laboratory of Drug Research & Small-Molecule Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jing Gao
- Division of Antitumor Pharmacology & Analytical Research Center for Organic and Biological Molecules & State Key Laboratory of Drug Research & Small-Molecule Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Qingqing Wu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, China
| | - Gang Bai
- Division of Antitumor Pharmacology & Analytical Research Center for Organic and Biological Molecules & State Key Laboratory of Drug Research & Small-Molecule Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yan Li
- Division of Antitumor Pharmacology & Analytical Research Center for Organic and Biological Molecules & State Key Laboratory of Drug Research & Small-Molecule Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Linjiang Tong
- Division of Antitumor Pharmacology & Analytical Research Center for Organic and Biological Molecules & State Key Laboratory of Drug Research & Small-Molecule Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Fang Feng
- Division of Antitumor Pharmacology & Analytical Research Center for Organic and Biological Molecules & State Key Laboratory of Drug Research & Small-Molecule Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Mengzhen Lai
- Division of Antitumor Pharmacology & Analytical Research Center for Organic and Biological Molecules & State Key Laboratory of Drug Research & Small-Molecule Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yingqiang Liu
- Division of Antitumor Pharmacology & Analytical Research Center for Organic and Biological Molecules & State Key Laboratory of Drug Research & Small-Molecule Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Peiran Song
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, China
| | - Yi Ning
- Division of Antitumor Pharmacology & Analytical Research Center for Organic and Biological Molecules & State Key Laboratory of Drug Research & Small-Molecule Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Haotian Tang
- Division of Antitumor Pharmacology & Analytical Research Center for Organic and Biological Molecules & State Key Laboratory of Drug Research & Small-Molecule Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Wen Luo
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, China
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Yi Chen
- Division of Antitumor Pharmacology & Analytical Research Center for Organic and Biological Molecules & State Key Laboratory of Drug Research & Small-Molecule Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yan Fang
- Division of Antitumor Pharmacology & Analytical Research Center for Organic and Biological Molecules & State Key Laboratory of Drug Research & Small-Molecule Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Hui Zhang
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiupei Liu
- Division of Antitumor Pharmacology & Analytical Research Center for Organic and Biological Molecules & State Key Laboratory of Drug Research & Small-Molecule Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- Department of Chemical and Environmental Engineering, University of Nottingham, Ningbo, China
| | - Yudi Zhang
- Division of Antitumor Pharmacology & Analytical Research Center for Organic and Biological Molecules & State Key Laboratory of Drug Research & Small-Molecule Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Hua Wang
- University of Chinese Academy of Sciences, Beijing, China
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Zhiwei Chen
- Division of Antitumor Pharmacology & Analytical Research Center for Organic and Biological Molecules & State Key Laboratory of Drug Research & Small-Molecule Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yi Chen
- Division of Antitumor Pharmacology & Analytical Research Center for Organic and Biological Molecules & State Key Laboratory of Drug Research & Small-Molecule Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Meiyu Geng
- Division of Antitumor Pharmacology & Analytical Research Center for Organic and Biological Molecules & State Key Laboratory of Drug Research & Small-Molecule Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Hongbin Ji
- University of Chinese Academy of Sciences, Beijing, China
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Guilong Zhao
- Division of Antitumor Pharmacology & Analytical Research Center for Organic and Biological Molecules & State Key Laboratory of Drug Research & Small-Molecule Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, China.
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.
| | - Hu Zhou
- Division of Antitumor Pharmacology & Analytical Research Center for Organic and Biological Molecules & State Key Laboratory of Drug Research & Small-Molecule Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
| | - Jian Ding
- Division of Antitumor Pharmacology & Analytical Research Center for Organic and Biological Molecules & State Key Laboratory of Drug Research & Small-Molecule Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
| | - Hua Xie
- Division of Antitumor Pharmacology & Analytical Research Center for Organic and Biological Molecules & State Key Laboratory of Drug Research & Small-Molecule Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, China.
| |
Collapse
|
14
|
Ippolito L, Duatti A, Iozzo M, Comito G, Pardella E, Lorito N, Bacci M, Pranzini E, Santi A, Sandrini G, Catapano CV, Serni S, Spatafora P, Morandi A, Giannoni E, Chiarugi P. Lactate supports cell-autonomous ECM production to sustain metastatic behavior in prostate cancer. EMBO Rep 2024; 25:3506-3531. [PMID: 38907027 PMCID: PMC11315984 DOI: 10.1038/s44319-024-00180-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 05/30/2024] [Accepted: 06/07/2024] [Indexed: 06/23/2024] Open
Abstract
Extracellular matrix (ECM) is a major component of the tumor environment, promoting the establishment of a pro-invasive behavior. Such environment is supported by both tumor- and stromal-derived metabolites, particularly lactate. In prostate cancer (PCa), cancer-associated fibroblasts (CAFs) are major contributors of secreted lactate, able to impact on metabolic and transcriptional regulation in cancer cells. Here, we describe a mechanism by which CAF-secreted lactate promotes in PCa cells the expression of genes coding for the collagen family. Lactate-exploiting PCa cells rely on increased α-ketoglutarate (α-KG) which activates the α-KG-dependent collagen prolyl-4-hydroxylase (P4HA1) to support collagen hydroxylation. De novo synthetized collagen plays a signaling role by activating discoidin domain receptor 1 (DDR1), supporting stem-like and invasive features of PCa cells. Inhibition of lactate-induced collagen hydroxylation and DDR1 activation reduces the metastatic colonization of PCa cells. Overall, these results provide a new understanding of the link between collagen remodeling/signaling and the nutrient environment exploited by PCa.
Collapse
Grants
- 19515 Fondazione AIRC per la ricerca sul cancro ETS (AIRC)
- 24731 Fondazione AIRC per la ricerca sul cancro ETS (AIRC)
- 22941 Fondazione AIRC per la ricerca sul cancro ETS (AIRC)
- 26599 Fondazione AIRC per la ricerca sul cancro ETS (AIRC)
- KLS-4899-08-2019 Swiss Cancer League
- CN00000041 European Union, National Recovery and Resilience Plan, Mission 4 Component 2 - Investment 1.4 - National Center for Gene Therapy and Drugs based on RNA Technology - NextGenerationEU
- ECS_00000017 European Union, National Recovery and Resilience Plan, Mission 4 Component 2, Creation and strengthening of "innovation ecosystems", construction of "territorial R&D leaders"
- Fondazione Ticinese Ricerca sul Cancro
- European Union, National Recovery and Resilience Plan, Mission 4 Component 2, Creation and strengthening of "innovation ecosystems", construction of "territorial R&D leaders"
- Fondazione Pezcoller (Pezcoller Foundation)
Collapse
Affiliation(s)
- Luigi Ippolito
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134, Florence, Italy.
| | - Assia Duatti
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134, Florence, Italy
| | - Marta Iozzo
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134, Florence, Italy
| | - Giuseppina Comito
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134, Florence, Italy
| | - Elisa Pardella
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134, Florence, Italy
| | - Nicla Lorito
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134, Florence, Italy
| | - Marina Bacci
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134, Florence, Italy
| | - Erica Pranzini
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134, Florence, Italy
| | - Alice Santi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134, Florence, Italy
| | - Giada Sandrini
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), 6500, Bellinzona, Switzerland
| | - Carlo V Catapano
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), 6500, Bellinzona, Switzerland
| | - Sergio Serni
- Department of Minimally Invasive and Robotic Urologic Surgery and Kidney Transplantation, University of Florence, 50134, Florence, Italy
| | - Pietro Spatafora
- Department of Minimally Invasive and Robotic Urologic Surgery and Kidney Transplantation, University of Florence, 50134, Florence, Italy
| | - Andrea Morandi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134, Florence, Italy
| | - Elisa Giannoni
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134, Florence, Italy
| | - Paola Chiarugi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134, Florence, Italy.
| |
Collapse
|
15
|
Qiu Y, Stamatatos OT, Hu Q, Ruiter Swain J, Russo S, Sann A, Costa ASH, Violante S, Spector DL, Cross JR, Lukey MJ. The unique catalytic properties of PSAT1 mediate metabolic adaptation to glutamine blockade. Nat Metab 2024; 6:1529-1548. [PMID: 39192144 PMCID: PMC11490312 DOI: 10.1038/s42255-024-01104-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 07/09/2024] [Indexed: 08/29/2024]
Abstract
Cultured cancer cells frequently rely on the consumption of glutamine and its subsequent hydrolysis by glutaminase (GLS). However, this metabolic addiction can be lost in the tumour microenvironment, rendering GLS inhibitors ineffective in the clinic. Here we show that glutamine-addicted breast cancer cells adapt to chronic glutamine starvation, or GLS inhibition, via AMPK-mediated upregulation of the serine synthesis pathway (SSP). In this context, the key product of the SSP is not serine, but α-ketoglutarate (α-KG). Mechanistically, we find that phosphoserine aminotransferase 1 (PSAT1) has a unique capacity for sustained α-KG production when glutamate is depleted. Breast cancer cells with resistance to glutamine starvation or GLS inhibition are highly dependent on SSP-supplied α-KG. Accordingly, inhibition of the SSP prevents adaptation to glutamine blockade, resulting in a potent drug synergism that suppresses breast tumour growth. These findings highlight how metabolic redundancy can be context dependent, with the catalytic properties of different metabolic enzymes that act on the same substrate determining which pathways can support tumour growth in a particular nutrient environment. This, in turn, has practical consequences for therapies targeting cancer metabolism.
Collapse
Affiliation(s)
- Yijian Qiu
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | | | - Qingting Hu
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
- Graduate Program in Genetics, Stony Brook University, Stony Brook, NY, USA
| | - Jed Ruiter Swain
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
- CSHL School of Biological Sciences, Cold Spring Harbor, NY, USA
| | - Suzanne Russo
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Ava Sann
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Ana S H Costa
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
- Matterworks Inc., Somerville, MA, USA
| | - Sara Violante
- Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Justin R Cross
- Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | |
Collapse
|
16
|
Murcia-Mejía M, Canela-Capdevila M, García-Pablo R, Jiménez-Franco A, Jiménez-Aguilar JM, Badía J, Benavides-Villarreal R, Acosta JC, Arguís M, Onoiu AI, Castañé H, Camps J, Arenas M, Joven J. Combining Metabolomics and Machine Learning to Identify Diagnostic and Prognostic Biomarkers in Patients with Non-Small Cell Lung Cancer Pre- and Post-Radiation Therapy. Biomolecules 2024; 14:898. [PMID: 39199286 PMCID: PMC11353221 DOI: 10.3390/biom14080898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 09/01/2024] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths globally, with non-small cell lung cancer (NSCLC) accounting for over 85% of cases and poor prognosis in advanced stages. This study explored shifts in circulating metabolite levels in NSCLC patients versus healthy controls and examined the effects of conventionally fractionated radiation therapy (CFRT) and stereotactic body radiation therapy (SBRT). We enrolled 91 NSCLC patients (38 CFRT and 53 SBRT) and 40 healthy controls. Plasma metabolite levels were assessed using semi-targeted metabolomics, revealing 32 elevated and 18 reduced metabolites in patients. Key discriminatory metabolites included ethylmalonic acid, maltose, 3-phosphoglyceric acid, taurine, glutamic acid, glycocolic acid, and d-arabinose, with a combined Receiver Operating Characteristics curve indicating perfect discrimination between patients and controls. CFRT and SBRT affected different metabolites, but both changes suggested a partial normalization of energy and amino acid metabolism pathways. In conclusion, metabolomics identified distinct metabolic signatures in NSCLC patients with potential as diagnostic biomarkers. The differing metabolic responses to CFRT and SBRT reflect their unique therapeutic impacts, underscoring the utility of this technique in enhancing NSCLC diagnosis and treatment monitoring.
Collapse
Affiliation(s)
- Mauricio Murcia-Mejía
- Department of Radiation Oncology, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain; (M.M.-M.); (M.C.-C.); (R.G.-P.); (R.B.-V.); (J.C.A.); (M.A.)
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain; (A.J.-F.); (J.M.J.-A.); (A.-I.O.); (H.C.); (J.J.)
| | - Marta Canela-Capdevila
- Department of Radiation Oncology, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain; (M.M.-M.); (M.C.-C.); (R.G.-P.); (R.B.-V.); (J.C.A.); (M.A.)
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain; (A.J.-F.); (J.M.J.-A.); (A.-I.O.); (H.C.); (J.J.)
| | - Raquel García-Pablo
- Department of Radiation Oncology, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain; (M.M.-M.); (M.C.-C.); (R.G.-P.); (R.B.-V.); (J.C.A.); (M.A.)
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain; (A.J.-F.); (J.M.J.-A.); (A.-I.O.); (H.C.); (J.J.)
| | - Andrea Jiménez-Franco
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain; (A.J.-F.); (J.M.J.-A.); (A.-I.O.); (H.C.); (J.J.)
| | - Juan Manuel Jiménez-Aguilar
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain; (A.J.-F.); (J.M.J.-A.); (A.-I.O.); (H.C.); (J.J.)
| | - Joan Badía
- Statistical Support Platform, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain;
| | - Rocío Benavides-Villarreal
- Department of Radiation Oncology, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain; (M.M.-M.); (M.C.-C.); (R.G.-P.); (R.B.-V.); (J.C.A.); (M.A.)
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain; (A.J.-F.); (J.M.J.-A.); (A.-I.O.); (H.C.); (J.J.)
| | - Johana C. Acosta
- Department of Radiation Oncology, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain; (M.M.-M.); (M.C.-C.); (R.G.-P.); (R.B.-V.); (J.C.A.); (M.A.)
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain; (A.J.-F.); (J.M.J.-A.); (A.-I.O.); (H.C.); (J.J.)
| | - Mónica Arguís
- Department of Radiation Oncology, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain; (M.M.-M.); (M.C.-C.); (R.G.-P.); (R.B.-V.); (J.C.A.); (M.A.)
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain; (A.J.-F.); (J.M.J.-A.); (A.-I.O.); (H.C.); (J.J.)
| | - Alina-Iuliana Onoiu
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain; (A.J.-F.); (J.M.J.-A.); (A.-I.O.); (H.C.); (J.J.)
| | - Helena Castañé
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain; (A.J.-F.); (J.M.J.-A.); (A.-I.O.); (H.C.); (J.J.)
| | - Jordi Camps
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain; (A.J.-F.); (J.M.J.-A.); (A.-I.O.); (H.C.); (J.J.)
| | - Meritxell Arenas
- Department of Radiation Oncology, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain; (M.M.-M.); (M.C.-C.); (R.G.-P.); (R.B.-V.); (J.C.A.); (M.A.)
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain; (A.J.-F.); (J.M.J.-A.); (A.-I.O.); (H.C.); (J.J.)
| | - Jorge Joven
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain; (A.J.-F.); (J.M.J.-A.); (A.-I.O.); (H.C.); (J.J.)
| |
Collapse
|
17
|
Marques E, Kramer R, Ryan DG. Multifaceted mitochondria in innate immunity. NPJ METABOLIC HEALTH AND DISEASE 2024; 2:6. [PMID: 38812744 PMCID: PMC11129950 DOI: 10.1038/s44324-024-00008-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 04/14/2024] [Indexed: 05/31/2024]
Abstract
The ability of mitochondria to transform the energy we obtain from food into cell phosphorylation potential has long been appreciated. However, recent decades have seen an evolution in our understanding of mitochondria, highlighting their significance as key signal-transducing organelles with essential roles in immunity that extend beyond their bioenergetic function. Importantly, mitochondria retain bacterial motifs as a remnant of their endosymbiotic origin that are recognised by innate immune cells to trigger inflammation and participate in anti-microbial defence. This review aims to explore how mitochondrial physiology, spanning from oxidative phosphorylation (OxPhos) to signalling of mitochondrial nucleic acids, metabolites, and lipids, influences the effector functions of phagocytes. These myriad effector functions include macrophage polarisation, efferocytosis, anti-bactericidal activity, antigen presentation, immune signalling, and cytokine regulation. Strict regulation of these processes is critical for organismal homeostasis that when disrupted may cause injury or contribute to disease. Thus, the expanding body of literature, which continues to highlight the central role of mitochondria in the innate immune system, may provide insights for the development of the next generation of therapies for inflammatory diseases.
Collapse
Affiliation(s)
- Eloïse Marques
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Robbin Kramer
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Dylan G. Ryan
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| |
Collapse
|
18
|
Chen Y, Hu H, Atashi H, Grelet C, Wijnrocx K, Lemal P, Gengler N. Genetic analysis of milk citrate predicted by milk mid-infrared spectra of Holstein cows in early lactation. J Dairy Sci 2024; 107:3047-3061. [PMID: 38056571 DOI: 10.3168/jds.2023-23903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 11/08/2023] [Indexed: 12/08/2023]
Abstract
Milk citrate is regarded as an early biomarker of negative energy balance in dairy cows during early lactation and serves as a suitable candidate phenotype for genomic selection due to its wide availability across a large number of cows through milk mid-infrared spectra prediction. However, its genetic background is not well known. Therefore, the objectives of this study were to (1) analyze the genetic parameters of milk citrate; (2) identify genomic regions associated with milk citrate; and (3) analyze the functional annotation of candidate genes and quantitative trait loci (QTL) related to milk citrate in Walloon Holstein cows. In total, 134,517 test-day milk-citrate phenotypes (mmol/L) collected within the first 50 d in milk on 52,198 Holstein cows were used. These milk-citrate phenotypes, predicted by milk mid-infrared spectra, were divided into 3 traits according to the first (citrate1), second (citrate2), and third to fifth parity (citrate3+). Genomic information for 566,170 SNPs was available for 4,479 animals. A multiple-trait repeatability model was used to estimate genetic parameters. A single-step GWAS was used to identify candidate genes for citrate and post-GWAS analysis was done to investigate the relationship and function of the identified candidate genes. The heritabilities estimated for citrate1, citrate2, and citrate3+ were 0.40, 0.37, and 0.35, respectively. The genetic correlations among the 3 traits ranged from 0.98 to 0.99. The genomic correlations among the 3 traits were also close to 1.00 across the genomic regions (1 Mb) in the whole genome, which means that citrate can be considered as a single trait in the first 5 parities. In total, 603 significant SNPs located on 3 genomic regions (chromosome 7, 68.569-68.575 Mb; chromosome 14, 0.15-1.90 Mb; and chromosome 20, 54.00-64.28 Mb), were identified to be associated with milk citrate. We identified 89 candidate genes including GPT, ANKH, PPP1R16A, and 32 QTL reported in the literature related to the identified significant SNPs. These identified QTL were mainly reported associated with milk fatty acids and metabolic diseases in dairy cows. This study suggests that milk citrate in Holstein cows is highly heritable and has the potential to be used as an early proxy for the negative energy balance of Holstein cows in a breeding objective.
Collapse
Affiliation(s)
- Yansen Chen
- TERRA Teaching and Research Center, University of Liège, Gembloux Agro-Bio Tech (ULiège-GxABT), 5030 Gembloux, Belgium.
| | - Hongqing Hu
- TERRA Teaching and Research Center, University of Liège, Gembloux Agro-Bio Tech (ULiège-GxABT), 5030 Gembloux, Belgium
| | - Hadi Atashi
- TERRA Teaching and Research Center, University of Liège, Gembloux Agro-Bio Tech (ULiège-GxABT), 5030 Gembloux, Belgium; Department of Animal Science, Shiraz University, 71441-13131 Shiraz, Iran
| | - Clément Grelet
- Walloon Agricultural Research Center (CRA-W), 5030 Gembloux, Belgium
| | - Katrien Wijnrocx
- TERRA Teaching and Research Center, University of Liège, Gembloux Agro-Bio Tech (ULiège-GxABT), 5030 Gembloux, Belgium
| | - Pauline Lemal
- TERRA Teaching and Research Center, University of Liège, Gembloux Agro-Bio Tech (ULiège-GxABT), 5030 Gembloux, Belgium
| | - Nicolas Gengler
- TERRA Teaching and Research Center, University of Liège, Gembloux Agro-Bio Tech (ULiège-GxABT), 5030 Gembloux, Belgium
| |
Collapse
|
19
|
Ren J, Ren B, Liu X, Cui M, Fang Y, Wang X, Zhou F, Gu M, Xiao R, Bai J, You L, Zhao Y. Crosstalk between metabolic remodeling and epigenetic reprogramming: A new perspective on pancreatic cancer. Cancer Lett 2024; 587:216649. [PMID: 38311052 DOI: 10.1016/j.canlet.2024.216649] [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: 07/18/2023] [Revised: 09/21/2023] [Accepted: 01/13/2024] [Indexed: 02/06/2024]
Abstract
Pancreatic cancer is a highly malignant solid tumor with a poor prognosis and a high mortality rate. Thus, exploring the mechanisms underlying the development and progression of pancreatic cancer is critical for identifying targets for diagnosis and treatment. Two important hallmarks of cancer-metabolic remodeling and epigenetic reprogramming-are interconnected and closely linked to regulate one another, creating a complex interaction landscape that is implicated in tumorigenesis, invasive metastasis, and immune escape. For example, metabolites can be involved in the regulation of epigenetic enzymes as substrates or cofactors, and alterations in epigenetic modifications can in turn regulate the expression of metabolic enzymes. The crosstalk between metabolic remodeling and epigenetic reprogramming in pancreatic cancer has gained considerable attention. Here, we review the emerging data with a focus on the reciprocal regulation of metabolic remodeling and epigenetic reprogramming. We aim to highlight how these mechanisms could be applied to develop better therapeutic strategies.
Collapse
Affiliation(s)
- Jie Ren
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China.
| | - Bo Ren
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China.
| | - Xiaohong Liu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China.
| | - Ming Cui
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China.
| | - Yuan Fang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China.
| | - Xing Wang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China.
| | - Feihan Zhou
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China.
| | - Minzhi Gu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China.
| | - Ruiling Xiao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China.
| | - Jialu Bai
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China.
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China.
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China.
| |
Collapse
|
20
|
Liu X, Wu L, Si Y, Zhai Y, Niu M, Han M, Su T. Regulating Effect of Exogenous α-Ketoglutarate on Ammonium Assimilation in Poplar. Molecules 2024; 29:1425. [PMID: 38611705 PMCID: PMC11012726 DOI: 10.3390/molecules29071425] [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: 02/28/2024] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 04/14/2024] Open
Abstract
Extensive industrial activities and anthropogenic agricultural practices have led to substantial ammonia release to the environment. Although croplands can act as ammonia sinks, reduced crop production under high concentrations of ammonium has been documented. Alpha-ketoglutarate (AKG) is a critical carbon source, displaying pleiotropic physiological functions. The objective of the present study is to disclose the potential of AKG to enhance ammonium assimilation in poplars. It showed that AKG application substantially boosted the height, biomass, and photosynthesis activity of poplars exposed to excessive ammonium. AKG also enhanced the activities of key enzymes involved in nitrogen assimilation: glutamine synthetase (GS) and glutamate synthase (GOGAT), elevating the content of amino acids, sucrose, and the tricarboxylic acid cycle (TCA) metabolites. Furthermore, AKG positively modulated key genes tied to glucose metabolism and ATP synthesis, while suppressing ATP-depleting genes. Correspondingly, both H+-ATPase activity and ATP content increased. These findings demonstrate that exogenously applying AKG improves poplar growth under a high level of ammonium treatment. AKG might function through sufficient carbon investment, which enhances the carbon-nitrogen balance and energy stability in poplars, promoting ammonium assimilation at high doses of ammonium. Our study provides novel insight into AKG's role in improving poplar growth in response to excess ammonia exposure.
Collapse
Affiliation(s)
- Xiaoning Liu
- Co-Innovation Center for Sustainable Forestry in Southern China, College of Life Sciences, Nanjing Forestry University, Nanjing 210037, China (Y.Z.)
| | - Liangdan Wu
- Co-Innovation Center for Sustainable Forestry in Southern China, College of Life Sciences, Nanjing Forestry University, Nanjing 210037, China (Y.Z.)
| | - Yujia Si
- Co-Innovation Center for Sustainable Forestry in Southern China, College of Life Sciences, Nanjing Forestry University, Nanjing 210037, China (Y.Z.)
| | - Yujie Zhai
- Co-Innovation Center for Sustainable Forestry in Southern China, College of Life Sciences, Nanjing Forestry University, Nanjing 210037, China (Y.Z.)
| | - Mingyi Niu
- Co-Innovation Center for Sustainable Forestry in Southern China, College of Life Sciences, Nanjing Forestry University, Nanjing 210037, China (Y.Z.)
| | - Mei Han
- Co-Innovation Center for Sustainable Forestry in Southern China, College of Life Sciences, Nanjing Forestry University, Nanjing 210037, China (Y.Z.)
| | - Tao Su
- Co-Innovation Center for Sustainable Forestry in Southern China, College of Life Sciences, Nanjing Forestry University, Nanjing 210037, China (Y.Z.)
- Key Laboratory of State Forestry Administration on Subtropical Forest Biodiversity Conservation, Nanjing Forestry University, Nanjing 210037, China
| |
Collapse
|
21
|
Pan J, Wang Y, Huang S, Mao S, Ling Q, Li C, Li F, Yu M, Huang X, Huang J, Lv Y, Li X, Ye W, Wang H, Wang J, Jin J. High expression of BCAT1 sensitizes AML cells to PARP inhibitor by suppressing DNA damage response. J Mol Med (Berl) 2024; 102:415-433. [PMID: 38340163 DOI: 10.1007/s00109-023-02409-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 11/27/2023] [Accepted: 12/11/2023] [Indexed: 02/12/2024]
Abstract
Previous evidence has confirmed that branched-chain aminotransferase-1 (BCAT1), a key enzyme governing branched-chain amino acid (BCAA) metabolism, has a role in cancer aggression partly by restricting αKG levels and inhibiting the activities of the αKG-dependent enzyme family. The oncogenic role of BCAT1, however, was not fully elucidated in acute myeloid leukemia (AML). In this study, we investigated the clinical significance and biological insight of BCAT1 in AML. Using q-PCR, we analyzed BCAT1 mRNAs in bone marrow samples from 332 patients with newly diagnosed AML. High BCAT1 expression independently predicts poor prognosis in patients with AML. We also established BCAT1 knockout (KO)/over-expressing (OE) AML cell lines to explore the underlying mechanisms. We found that BCAT1 affects cell proliferation and modulates cell cycle, cell apoptosis, and DNA damage/repair process. Additionally, we demonstrated that BCAT1 regulates histone methylation by reducing intracellular αKG levels in AML cells. Moreover, high expression of BCAT1 enhances the sensitivity of AML cells to the Poly (ADP-ribose) polymerase (PARP) inhibitor both in vivo and in vitro. Our study has demonstrated that BCAT1 expression can serve as a reliable predictor for AML patients, and PARP inhibitor BMN673 can be used as an effective treatment strategy for patients with high BCAT1 expression. KEY MESSAGES: High expression of BCAT1 is an independent risk factor for poor prognosis in patients with CN-AML. High BCAT1 expression in AML limits intracellular αKG levels, impairs αKG-dependent histone demethylase activity, and upregulates H3K9me3 levels. H3K9me3 inhibits ATM expression and blocks cellular DNA damage repair process. Increased sensitivity of BCAT1 high expression AML to PARP inhibitors may be used as an effective treatment strategy in AML patients.
Collapse
Affiliation(s)
- Jiajia Pan
- Department of Hematology, The First Affiliated Hospital, Zhejiang University College of Medicine, No.79 Qingchun Road, Hangzhou, 310003, Zhejiang, People's Republic of China
- Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, Zhejiang, Hangzhou, China
| | - Yungui Wang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University College of Medicine, No.79 Qingchun Road, Hangzhou, 310003, Zhejiang, People's Republic of China
- Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, Zhejiang, Hangzhou, China
| | - Shujuan Huang
- Department of Hematology, the First Affiliated Hospital of University of Science and Technology of China, Anhui, Hefei, China
| | - Shihui Mao
- Department of Hematology, The First Affiliated Hospital, Zhejiang University College of Medicine, No.79 Qingchun Road, Hangzhou, 310003, Zhejiang, People's Republic of China
- Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, Zhejiang, Hangzhou, China
| | - Qing Ling
- Department of Hematology, The First Affiliated Hospital, Zhejiang University College of Medicine, No.79 Qingchun Road, Hangzhou, 310003, Zhejiang, People's Republic of China
- Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, Zhejiang, Hangzhou, China
| | - Chenying Li
- Department of Hematology, The First Affiliated Hospital, Zhejiang University College of Medicine, No.79 Qingchun Road, Hangzhou, 310003, Zhejiang, People's Republic of China
- Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, Zhejiang, Hangzhou, China
| | - Fenglin Li
- Department of Hematology, The First Affiliated Hospital, Zhejiang University College of Medicine, No.79 Qingchun Road, Hangzhou, 310003, Zhejiang, People's Republic of China
- Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, Zhejiang, Hangzhou, China
| | - Mengxia Yu
- Department of Hematology, Affiliated Hangzhou First People's Hospital, Zhejiang University College of Medicine, Hangzhou, China
| | - Xin Huang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University College of Medicine, No.79 Qingchun Road, Hangzhou, 310003, Zhejiang, People's Republic of China
- Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, Zhejiang, Hangzhou, China
| | - Jiansong Huang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University College of Medicine, No.79 Qingchun Road, Hangzhou, 310003, Zhejiang, People's Republic of China
- Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, Zhejiang, Hangzhou, China
| | - Yunfei Lv
- Department of Hematology, The First Affiliated Hospital, Zhejiang University College of Medicine, No.79 Qingchun Road, Hangzhou, 310003, Zhejiang, People's Republic of China
- Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, Zhejiang, Hangzhou, China
| | - Xia Li
- Department of Hematology, The First Affiliated Hospital, Zhejiang University College of Medicine, No.79 Qingchun Road, Hangzhou, 310003, Zhejiang, People's Republic of China
- Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, Zhejiang, Hangzhou, China
| | - Wenle Ye
- Department of Hematology, The First Affiliated Hospital, Zhejiang University College of Medicine, No.79 Qingchun Road, Hangzhou, 310003, Zhejiang, People's Republic of China
- Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, Zhejiang, Hangzhou, China
| | - Huafeng Wang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University College of Medicine, No.79 Qingchun Road, Hangzhou, 310003, Zhejiang, People's Republic of China
- Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, Zhejiang, Hangzhou, China
| | - Jinghan Wang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University College of Medicine, No.79 Qingchun Road, Hangzhou, 310003, Zhejiang, People's Republic of China.
- Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, Zhejiang, Hangzhou, China.
- Cancer Center, Zhejiang University, Hangzhou, China.
| | - Jie Jin
- Department of Hematology, The First Affiliated Hospital, Zhejiang University College of Medicine, No.79 Qingchun Road, Hangzhou, 310003, Zhejiang, People's Republic of China.
- Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, Zhejiang, Hangzhou, China.
- Cancer Center, Zhejiang University, Hangzhou, China.
| |
Collapse
|
22
|
Nakanishi T, Kuragano T. Growing concerns about using hypoxia-inducible factor prolyl hydroxylase inhibitors for the treatment of renal anemia. Clin Kidney J 2024; 17:sfae051. [PMID: 38516524 PMCID: PMC10956400 DOI: 10.1093/ckj/sfae051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Indexed: 03/23/2024] Open
Abstract
Hypoxia-inducible factor prolyl hydroxylase inhibitors (HIF-PHIs) have emerged as a novel therapeutic class for treating anemia in patients with chronic kidney disease. Small molecule analogs of α-ketoglutarate (AKG), an essential substrate for 2-oxoglutarate-dependent dioxygenases (2-OGDDs), including prolyl hydroxylase domain proteins (PHDs), inhibit PHDs pharmacologically and thereby prevent HIF degradation. HIF stabilization alleviates anemia through several stimulatory effects on erythropoiesis, but it also affects the expression of many anemia-unrelated genes whose protein products exert important functions in vivo. Therefore, the pleiotropic effects of HIF stabilization under normoxic conditions deserve to be examined in more detail. Specifically, we believe that particular attention should be given to epigenetic modifications among the various AKG-based metabolic systems that may be altered by HIF-PHIs. It is noteworthy that AKG has been reported to exert health-protective actions. AKG-based metabolic systems include enzymes associated with the tricarboxylic acid cycle and amino acid metabolism, as well as 2-OGDD-mediated processes, which play important roles in many biological reactions. In this review, we examine the multifaceted effects of HIF-PHIs, encompassing not only their on-target effect of HIF stabilization but also their off-target inhibitory effects on various AKG-based metabolic systems. Furthermore, we examine its potential relevance to cardiovascular complications, based on clinical and animal studies suggesting its involvement in vascular calcification, thrombogenesis and heart failure. In conclusion, although HIF-PHIs offer a promising avenue for anemia treatment in CKD patients, their broader impact on multiple biological systems raises substantial concerns. The intricate interplay between HIF stabilization, AKG competition and cardiovascular complications warrants extensive, long-term investigations to ensure the safety and usefulness of HIF-PHIs in clinical practice.
Collapse
Affiliation(s)
- Takeshi Nakanishi
- Division of Kidney, Dialysis and Cardiology, Department of Internal Medicine, Hyogo Medical University, Nishinomiya, Hyogo, Japan
- Department of Nephrology, Gojinkai Sumiyoshigawa Hospital, Kobe, Hyogo, Japan
| | - Takahiro Kuragano
- Division of Kidney, Dialysis and Cardiology, Department of Internal Medicine, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| |
Collapse
|
23
|
Sekiya M, Kainoh K, Saito K, Yamazaki D, Tsuyuzaki T, Chen W, Kobari Y, Nakata A, Babe H, Shimano H. C-Terminal Binding Protein 2 Emerges as a Critical Player Linking Metabolic Imbalance to the Pathogenesis of Obesity. J Atheroscler Thromb 2024; 31:109-116. [PMID: 37793810 PMCID: PMC10857841 DOI: 10.5551/jat.rv22014] [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] [Accepted: 08/21/2023] [Indexed: 10/06/2023] Open
Abstract
Metabolism is one of the vital functions of cells and living organisms, and the systems to sense and respond to the metabolic alterations play pivotal roles in a plethora of biological processes, including cell proliferative activities, immune cell functions, aging processes, and neuronal functions. Recently, we have reported that a transcriptional cofactor, C-terminal binding protein 2 (CtBP2), serves as a critical metabolite sensor in this context. CtBP2 has a structural pocket called Rossmann fold to accommodate metabolites, and it has been reported to be activated upon binding to NADH/NAD+. Owing to its preferential binding affinity for NADH compared with NAD+, increased glycolysis activates CtBP2 by regenerating NADH from NAD+. Furthermore, we recently reported that fatty acyl-CoAs, metabolites accumulated under the condition of lipid overload, as represented by obesity, can inactivate CtBP2. These observations suggest that CtBP2 monitors not only redox state but also energy substrate preference in the maintenance of metabolic homeostasis. In line with these metabolite-sensing capabilities, CtBP2 is activated in healthy subjects to protect against metabolic disturbances, whereas inactivation of CtBP2 in obesity contributes to the pathogeneses of obesity.This metabolic system orchestrated by CtBP2 can provide a novel framework for understanding how cells maintain their homeostasis through coordination of metabolism, and CtBP2 incapacitation can be a critical point of the obesogenic cascade.
Collapse
Affiliation(s)
- Motohiro Sekiya
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Kenta Kainoh
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Kenji Saito
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Daichi Yamazaki
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Tomomi Tsuyuzaki
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Wanpei Chen
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Yuto Kobari
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Ayumi Nakata
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Haruka Babe
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Hitoshi Shimano
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| |
Collapse
|
24
|
Nagaoka K, Bai X, Liu D, Cao K, Mulla J, Ji C, Chen H, Nisar MA, Bay A, Mueller W, Hildebrand G, Gao JS, Lu S, Setoyama H, Tanaka Y, Wands JR, Huang CK. Elevated 2-oxoglutarate antagonizes DNA damage responses in cholangiocarcinoma chemotherapy through regulating aspartate beta-hydroxylase. Cancer Lett 2024; 580:216493. [PMID: 37977350 DOI: 10.1016/j.canlet.2023.216493] [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: 07/03/2023] [Revised: 09/22/2023] [Accepted: 11/07/2023] [Indexed: 11/19/2023]
Abstract
Cholangiocarcinoma (CCA) is resistant to systemic chemotherapies that kill malignant cells mainly through DNA damage responses (DDRs). Recent studies suggest that the involvement of 2-oxoglutarate (2-OG) dependent dioxygenases in DDRs may be associated with chemoresistance in malignancy, but how 2-OG impacts DDRs in CCA chemotherapy remains elusive. We examined serum 2-OG levels in CCA patients before receiving chemotherapy. CCA patients are classified as progressive disease (PD), partial response (PR), and stable disease (SD) after receiving chemotherapy. CCA patients classified as PD showed significantly higher serum 2-OG levels than those defined as SD and PR. Treating CCA cells with 2-OG reduced DDRs. Overexpression of full-length aspartate beta-hydroxylase (ASPH) could mimic the effects of 2-OG on DDRs, suggesting the important role of ASPH in chemoresistance. Indeed, the knockdown of ASPH improved chemotherapy in CCA cells. Targeting ASPH with a specific small molecule inhibitor also enhanced the effects of chemotherapy. Mechanistically, ASPH modulates DDRs by affecting ATM and ATR, two of the major regulators finely controlling DDRs. More importantly, targeting ASPH improved the therapeutic potential of chemotherapy in two preclinical CCA models. Our data suggested the impacts of elevated 2-OG and ASPH on chemoresistance through antagonizing DDRs. Targeting ASPH may enhance DDRs, improving chemotherapy in CCA patients.
Collapse
Affiliation(s)
- Katsuya Nagaoka
- Liver Research Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA; Department of Gastroenterology & Hepatology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Xuewei Bai
- Liver Research Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| | - Dan Liu
- Liver Research Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| | - Kevin Cao
- Liver Research Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| | - Joud Mulla
- Liver Research Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| | - Chengcheng Ji
- Liver Research Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| | - Hongze Chen
- Department of Pathology and Laboratory Medicine, Tulane University, New Orleans, LA, USA
| | - Muhammad Azhar Nisar
- Department of Pathology and Laboratory Medicine, Tulane University, New Orleans, LA, USA
| | - Amalia Bay
- Liver Research Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| | - William Mueller
- Liver Research Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| | - Grace Hildebrand
- Liver Research Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| | - Jin-Song Gao
- Liver Research Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| | - Shaolei Lu
- Department of Pathology and Laboratory Medicine, Alpert Medical School of Brown University, Providence, RI, 02903, USA
| | - Hiroko Setoyama
- Department of Gastroenterology & Hepatology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yasuhito Tanaka
- Department of Gastroenterology & Hepatology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Jack R Wands
- Liver Research Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| | - Chiung-Kuei Huang
- Liver Research Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA; Department of Pathology and Laboratory Medicine, Tulane University, New Orleans, LA, USA.
| |
Collapse
|
25
|
Caetano-Pinto P, Stahl SH. Renal Organic Anion Transporters 1 and 3 In Vitro: Gone but Not Forgotten. Int J Mol Sci 2023; 24:15419. [PMID: 37895098 PMCID: PMC10607849 DOI: 10.3390/ijms242015419] [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: 07/26/2023] [Revised: 10/12/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Organic anion transporters 1 and 3 (OAT1 and OAT3) play a crucial role in kidney function by regulating the secretion of multiple renally cleared small molecules and toxic metabolic by-products. Assessing the activity of these transporters is essential for drug development purposes as they can significantly impact drug disposition and safety. OAT1 and OAT3 are amongst the most abundant drug transporters expressed in human renal proximal tubules. However, their expression is lost when cells are isolated and cultured in vitro, which is a persistent issue across all human and animal renal proximal tubule cell models, including primary cells and cell lines. Although it is well known that the overall expression of drug transporters is affected in vitro, the underlying reasons for the loss of OAT1 and OAT3 are still not fully understood. Nonetheless, research into the regulatory mechanisms of these transporters has provided insights into the molecular pathways underlying their expression and activity. In this review, we explore the regulatory mechanisms that govern the expression and activity of OAT1 and OAT3 and investigate the physiological changes that proximal tubule cells undergo and that potentially result in the loss of these transporters. A better understanding of the regulation of these transporters could aid in the development of strategies, such as introducing microfluidic conditions or epigenetic modification inhibitors, to improve their expression and activity in vitro and to create more physiologically relevant models. Consequently, this will enable more accurate assessment for drug development and safety applications.
Collapse
Affiliation(s)
- Pedro Caetano-Pinto
- Department of Urology, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany
| | - Simone H. Stahl
- CVRM Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, 310 Darwin Building, Cambridge Science Park, Milton Road, Cambridge CB4 0WG, UK;
| |
Collapse
|
26
|
Lee SJ, Emery D, Vukmanic E, Wang Y, Lu X, Wang W, Fortuny E, James R, Kaplan HJ, Liu Y, Du J, Dean DC. Metabolic transcriptomics dictate responses of cone photoreceptors to retinitis pigmentosa. Cell Rep 2023; 42:113054. [PMID: 37656622 PMCID: PMC10591869 DOI: 10.1016/j.celrep.2023.113054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/21/2023] [Accepted: 08/15/2023] [Indexed: 09/03/2023] Open
Abstract
Most mutations in retinitis pigmentosa (RP) arise in rod photoreceptors, but cone photoreceptors, responsible for high-resolution daylight and color vision, are subsequently affected, causing the most debilitating features of the disease. We used mass spectroscopy to follow 13C metabolites delivered to the outer retina and single-cell RNA sequencing to assess photoreceptor transcriptomes. The S cone metabolic transcriptome suggests engagement of the TCA cycle and ongoing response to ROS characteristic of oxidative phosphorylation, which we link to their histone modification transcriptome. Tumor necrosis factor (TNF) and its downstream effector RIP3, which drive ROS generation via mitochondrial dysfunction, are induced and activated as S cones undergo early apoptosis in RP. The long/medium-wavelength (L/M) cone transcriptome shows enhanced glycolytic capacity, which maintains their function as RP progresses. Then, as extracellular glucose eventually diminishes, L/M cones are sustained in long-term dormancy by lactate metabolism.
Collapse
Affiliation(s)
- Sang Joon Lee
- Department of Medicine, Brown Cancer Center, University of Louisville Health Sciences Center, Louisville, KY 40202, USA; Department of Ophthalmology, Kosin University College of Medicine, #262 Gamcheon-ro, Seo-gu, Busan 49267, Korea
| | - Douglas Emery
- Department of Medicine, Brown Cancer Center, University of Louisville Health Sciences Center, Louisville, KY 40202, USA
| | - Eric Vukmanic
- Department of Medicine, Brown Cancer Center, University of Louisville Health Sciences Center, Louisville, KY 40202, USA
| | - Yekai Wang
- Departments of Ophthalmology and Visual Sciences and Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV 26506, USA
| | - Xiaoqin Lu
- Department of Medicine, Brown Cancer Center, University of Louisville Health Sciences Center, Louisville, KY 40202, USA
| | - Wei Wang
- Department of Ophthalmology and Visual Sciences, University of Louisville Health Sciences Center, Louisville, KY 40202, USA
| | - Enzo Fortuny
- Department of Neurosurgery, University of Louisville Health Sciences Center, Louisville, KY 40202, USA
| | - Robert James
- Department of Neurosurgery, University of Louisville Health Sciences Center, Louisville, KY 40202, USA
| | - Henry J Kaplan
- Department of Ophthalmology, St. Louis University School of Medicine, St. Louis MO 63110, USA
| | - Yongqing Liu
- Department of Medicine, Brown Cancer Center, University of Louisville Health Sciences Center, Louisville, KY 40202, USA
| | - Jianhai Du
- Departments of Ophthalmology and Visual Sciences and Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV 26506, USA.
| | - Douglas C Dean
- Department of Medicine, Brown Cancer Center, University of Louisville Health Sciences Center, Louisville, KY 40202, USA.
| |
Collapse
|
27
|
Tarazi D, Maynes JT. Impact of Opioids on Cellular Metabolism: Implications for Metabolic Pathways Involved in Cancer. Pharmaceutics 2023; 15:2225. [PMID: 37765194 PMCID: PMC10534826 DOI: 10.3390/pharmaceutics15092225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 09/29/2023] Open
Abstract
Opioid utilization for pain management is prevalent among cancer patients. There is significant evidence describing the many effects of opioids on cancer development. Despite the pivotal role of metabolic reprogramming in facilitating cancer growth and metastasis, the specific impact of opioids on crucial oncogenic metabolic pathways remains inadequately investigated. This review provides an understanding of the current research on opioid-mediated changes to cellular metabolic pathways crucial for oncogenesis, including glycolysis, the tricarboxylic acid cycle, glutaminolysis, and oxidative phosphorylation (OXPHOS). The existing literature suggests that opioids affect energy production pathways via increasing intracellular glucose levels, increasing the production of lactic acid, and reducing ATP levels through impediment of OXPHOS. Opioids modulate pathways involved in redox balance which may allow cancer cells to overcome ROS-mediated apoptotic signaling. The majority of studies have been conducted in healthy tissue with a predominant focus on neuronal cells. To comprehensively understand the impact of opioids on metabolic pathways critical to cancer progression, research must extend beyond healthy tissue and encompass patient-derived cancer tissue, allowing for a better understanding in the context of the metabolic reprogramming already undergone by cancer cells. The current literature is limited by a lack of direct experimentation exploring opioid-induced changes to cancer metabolism as they relate to tumor growth and patient outcome.
Collapse
Affiliation(s)
- Doorsa Tarazi
- Department of Biochemistry, University of Toronto, Toronto, ON M5G 1A8, Canada;
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Jason T. Maynes
- Department of Biochemistry, University of Toronto, Toronto, ON M5G 1A8, Canada;
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
- Department of Anesthesia and Pain Medicine, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
- Department of Anesthesiology and Pain Medicine, University of Toronto, Toronto, ON M5G 1E2, Canada
| |
Collapse
|
28
|
Martino F, Lupi M, Giraudo E, Lanzetti L. Breast cancers as ecosystems: a metabolic perspective. Cell Mol Life Sci 2023; 80:244. [PMID: 37561190 PMCID: PMC10415483 DOI: 10.1007/s00018-023-04902-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/18/2023] [Accepted: 07/28/2023] [Indexed: 08/11/2023]
Abstract
Breast cancer (BC) is the most frequently diagnosed cancer and one of the major causes of cancer death. Despite enormous progress in its management, both from the therapeutic and early diagnosis viewpoints, still around 700,000 patients succumb to the disease each year, worldwide. Late recurrency is the major problem in BC, with many patients developing distant metastases several years after the successful eradication of the primary tumor. This is linked to the phenomenon of metastatic dormancy, a still mysterious trait of the natural history of BC, and of several other types of cancer, by which metastatic cells remain dormant for long periods of time before becoming reactivated to initiate the clinical metastatic disease. In recent years, it has become clear that cancers are best understood if studied as ecosystems in which the impact of non-cancer-cell-autonomous events-dependent on complex interaction between the cancer and its environment, both local and systemic-plays a paramount role, probably as significant as the cell-autonomous alterations occurring in the cancer cell. In adopting this perspective, a metabolic vision of the cancer ecosystem is bound to improve our understanding of the natural history of cancer, across space and time. In BC, many metabolic pathways are coopted into the cancer ecosystem, to serve the anabolic and energy demands of the cancer. Their study is shedding new light on the most critical aspect of BC management, of metastatic dissemination, and that of the related phenomenon of dormancy and fostering the application of the knowledge to the development of metabolic therapies.
Collapse
Affiliation(s)
- Flavia Martino
- Department of Oncology, University of Torino Medical School, Turin, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
| | - Mariadomenica Lupi
- Department of Oncology, University of Torino Medical School, Turin, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
| | - Enrico Giraudo
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
- Department of Science and Drug Technology, University of Torino, Turin, Italy
| | - Letizia Lanzetti
- Department of Oncology, University of Torino Medical School, Turin, Italy.
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy.
| |
Collapse
|
29
|
Huang F, Luo X, Ou Y, Gao Z, Tang Q, Chu Z, Zhu X, He Y. Control of histone demethylation by nuclear-localized α-ketoglutarate dehydrogenase. Science 2023; 381:eadf8822. [PMID: 37440635 DOI: 10.1126/science.adf8822] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 05/19/2023] [Indexed: 07/15/2023]
Abstract
Methylations on nucleosomal histones play fundamental roles in regulating eukaryotic transcription. Jumonji C domain-containing histone demethylases (JMJs) dynamically control the level of histone methylations. However, how JMJ activity is generally regulated is unknown. We found that the tricarboxylic acid cycle-associated enzyme α-ketoglutarate (α-KG) dehydrogenase (KGDH) entered the nucleus, where it interacted with various JMJs to regulate α-KG-dependent histone demethylations by JMJs, and thus controlled genome-wide gene expression in plants. We show that nuclear targeting is regulated by environmental signals and that KGDH is enriched at thousands of loci in Arabidopsis thaliana. Chromatin-bound KGDH catalyzes α-KG decarboxylation and thus may limit its local availability to KGDH-coupled JMJs, inhibiting histone demethylation. Thus, our results uncover a regulatory mechanism for histone demethylations by JMJs.
Collapse
Affiliation(s)
- Fei Huang
- National Key Laboratory of Wheat Improvement, Peking-Tsinghua Center for Life Sciences, School of Advanced Agricultural Sciences, Peking University, Beijing 100871, China
- Shanghai Center for Plant Stress Biology, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences (CAS), Shanghai 201602, China
| | - Xiao Luo
- Shanghai Center for Plant Stress Biology, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences (CAS), Shanghai 201602, China
- Peking University Institute of Advanced Agricultural Sciences, Shandong Laboratory of Advanced Agricultural Sciences in Weifang, Shandong 261325, China
| | - Yang Ou
- National Key Laboratory of Wheat Improvement, Peking-Tsinghua Center for Life Sciences, School of Advanced Agricultural Sciences, Peking University, Beijing 100871, China
- Shanghai Center for Plant Stress Biology, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences (CAS), Shanghai 201602, China
| | - Zhaoxu Gao
- National Key Laboratory of Wheat Improvement, Peking-Tsinghua Center for Life Sciences, School of Advanced Agricultural Sciences, Peking University, Beijing 100871, China
| | - Qiming Tang
- National Key Laboratory for Plant Molecular Genetics, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, CAS, Shanghai, 200032, China
| | - Zhenzhen Chu
- Shanghai Center for Plant Stress Biology, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences (CAS), Shanghai 201602, China
| | - Xinguang Zhu
- National Key Laboratory for Plant Molecular Genetics, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, CAS, Shanghai, 200032, China
| | - Yuehui He
- National Key Laboratory of Wheat Improvement, Peking-Tsinghua Center for Life Sciences, School of Advanced Agricultural Sciences, Peking University, Beijing 100871, China
- Shanghai Center for Plant Stress Biology, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences (CAS), Shanghai 201602, China
- Peking University Institute of Advanced Agricultural Sciences, Shandong Laboratory of Advanced Agricultural Sciences in Weifang, Shandong 261325, China
| |
Collapse
|
30
|
Yang Y, Zhang Z, Li W, Si Y, Li L, Du W. αKG-driven RNA polymerase II transcription of cyclin D1 licenses malic enzyme 2 to promote cell-cycle progression. Cell Rep 2023; 42:112770. [PMID: 37422761 DOI: 10.1016/j.celrep.2023.112770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 04/28/2023] [Accepted: 06/22/2023] [Indexed: 07/11/2023] Open
Abstract
Increased metabolic activity usually provides energy and nutrients for biomass synthesis and is indispensable for the progression of the cell cycle. Here, we find a role for α-ketoglutarate (αKG) generation in regulating cell-cycle gene transcription. A reduction in cellular αKG levels triggered by malic enzyme 2 (ME2) or isocitrate dehydrogenase 1 (IDH1) depletion leads to a pronounced arrest in G1 phase, while αKG supplementation promotes cell-cycle progression. Mechanistically, αKG directly binds to RNA polymerase II (RNAPII) and increases the level of RNAPII binding to the cyclin D1 gene promoter via promoting pre-initiation complex (PIC) assembly, consequently enhancing cyclin D1 transcription. Notably, αKG addition is sufficient to restore cyclin D1 expression in ME2- or IDH1-depleted cells, facilitating cell-cycle progression and proliferation in these cells. Therefore, our findings indicate a function of αKG in gene transcriptional regulation and cell-cycle control.
Collapse
Affiliation(s)
- Yanting Yang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Cell Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Zhenxi Zhang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Cell Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Wei Li
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Cell Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Yufan Si
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Cell Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Li Li
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Cell Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Wenjing Du
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Cell Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China.
| |
Collapse
|
31
|
Davuluri GVN, Chan CH. Regulation of intrinsic and extrinsic metabolic pathways in tumour-associated macrophages. FEBS J 2023; 290:3040-3058. [PMID: 35486022 PMCID: PMC10711806 DOI: 10.1111/febs.16465] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 03/08/2022] [Accepted: 04/26/2022] [Indexed: 02/03/2023]
Abstract
Tumour-associated macrophages (TAMs) are highly plastic and are broadly grouped into two major functional states, namely the pro-inflammatory M1-type and the pro-tumoural M2-type. Conversion of the functional states of TAMs is regulated by various cytokines, chemokines growth factors and other secreted factors in the microenvironment. Dysregulated metabolism is a hallmark of cancer. Emerging evidence suggests that metabolism governs the TAM differentiation and functional conversation in support of tumour growth and metastasis. Aside from the altered metabolism reprogramming in TAMs, extracellular metabolites secreted by cancer, stromal and/or other cells within the tumour microenvironment have been found to regulate TAMs through passive competition for metabolite availability and direct regulation via receptor/transporter-mediated signalling reaction. In this review, we focus on the regulatory roles of different metabolites and metabolic pathways in TAM conversion and function. We also discuss if the dysregulated metabolism in TAMs can be exploited for the development of new therapeutic strategies against cancer.
Collapse
Affiliation(s)
| | - Chia-Hsin Chan
- Department of Molecular and Cellular Biology, Roswell Park Cancer Comprehensive Cancer Center, Buffalo, New York
| |
Collapse
|
32
|
Gao G, Shen S, Zhang T, Zhang J, Huang S, Sun Z, Zhang H. Lacticaseibacillus rhamnosus Probio-M9 enhanced the antitumor response to anti-PD-1 therapy by modulating intestinal metabolites. EBioMedicine 2023; 91:104533. [PMID: 37027929 PMCID: PMC10085781 DOI: 10.1016/j.ebiom.2023.104533] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 02/23/2023] [Accepted: 03/07/2023] [Indexed: 04/08/2023] Open
Abstract
BACKGROUND Probiotics have been increasingly proposed for enhancing immune checkpoint blockade (ICB) treatments against cancer. However, its causal relationship with immunotherapeutic efficacy remains unclear, which promoted us to explore if and how probiotic Lacticaseibacillus rhamnosus Probio-M9 manipulates gut microbiome for expected outcomes. METHODS We evaluated the effects of Probio-M9 on the anti-PD-1 treatment against colorectal cancer in mice via a multi-omics approach. We defined the mechanisms of Probio-M9-mediated antitumor immunity by comprehensive analyses of metagenome and metabolites of commensal gut microbes as well as the immunologic factors and serum metabolome of the host. FINDINGS The results indicated that Probio-M9 intervention strengthened the anti-PD-1-based tumor inhibition. Both prophylactic and therapeutic administration of Probio-M9 showed conspicuous performance in controlling tumor growth with ICB treatment. The supplement of Probio-M9 modulated enhanced immunotherapy response through promoting beneficial microbes (e.g., Lactobacillus and Bifidobacterium animalis), producing beneficial metabolites including butyric acids in the gut, and accumulating blood-derived α-ketoglutaric acid, N-acetyl-l-glutamic acid and pyridoxine in particular, which promoted the infiltration and activation of cytotoxic T lymphocytes (CTLs) and suppressing the function of regulatory T cells (Tregs) in the tumor microenvironment (TME). Subsequently, we found that enhanced immunotherapeutic response was transmissible by transplanting either post-probiotic-treatment gut microbes or intestinal metabolites to new tumor-bearing mice. INTERPRETATION This study offered valuable insight into the causal role of Probio-M9 in correcting the defects in gut microbiota that compromised anti-PD-1 therapeutic efficacy, which can be used as an alternative synergetic agent with ICB for clinical cancer treatment. FUNDING This research was supported by Research Fund for the National Key R&D Program of China (2022YFD2100702), Inner Mongolia Science and Technology Major Projects (2021ZD0014), and China Agriculture Research System of MOF and MARA.
Collapse
Affiliation(s)
- Guangqi Gao
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, 010018, China
| | - Siyuan Shen
- College of Food Science and Engineering, Hainan University, Haikou, China
| | - Tao Zhang
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, 010018, China
| | - Jiachao Zhang
- College of Food Science and Engineering, Hainan University, Haikou, China
| | - Shi Huang
- Faculty of Dentistry, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Zhihong Sun
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, 010018, China
| | - Heping Zhang
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, 010018, China.
| |
Collapse
|
33
|
Nadhan R, Kashyap S, Ha JH, Jayaraman M, Song YS, Isidoro C, Dhanasekaran DN. Targeting Oncometabolites in Peritoneal Cancers: Preclinical Insights and Therapeutic Strategies. Metabolites 2023; 13:618. [PMID: 37233659 PMCID: PMC10222714 DOI: 10.3390/metabo13050618] [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: 03/31/2023] [Revised: 04/27/2023] [Accepted: 04/28/2023] [Indexed: 05/27/2023] Open
Abstract
Peritoneal cancers present significant clinical challenges with poor prognosis. Understanding the role of cancer cell metabolism and cancer-promoting metabolites in peritoneal cancers can provide new insights into the mechanisms that drive tumor progression and can identify novel therapeutic targets and biomarkers for early detection, prognosis, and treatment response. Cancer cells dynamically reprogram their metabolism to facilitate tumor growth and overcome metabolic stress, with cancer-promoting metabolites such as kynurenines, lactate, and sphingosine-1-phosphate promoting cell proliferation, angiogenesis, and immune evasion. Targeting cancer-promoting metabolites could also lead to the development of effective combinatorial and adjuvant therapies involving metabolic inhibitors for the treatment of peritoneal cancers. With the observed metabolomic heterogeneity in cancer patients, defining peritoneal cancer metabolome and cancer-promoting metabolites holds great promise for improving outcomes for patients with peritoneal tumors and advancing the field of precision cancer medicine. This review provides an overview of the metabolic signatures of peritoneal cancer cells, explores the role of cancer-promoting metabolites as potential therapeutic targets, and discusses the implications for advancing precision cancer medicine in peritoneal cancers.
Collapse
Affiliation(s)
- Revathy Nadhan
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.N.); (S.K.); (J.H.H.); (M.J.)
| | - Srishti Kashyap
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.N.); (S.K.); (J.H.H.); (M.J.)
| | - Ji Hee Ha
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.N.); (S.K.); (J.H.H.); (M.J.)
- Department of Cell Biology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Muralidharan Jayaraman
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.N.); (S.K.); (J.H.H.); (M.J.)
- Department of Cell Biology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Yong Sang Song
- Department of Obstetrics and Gynecology, Cancer Research Institute, College of Medicine, Seoul National University, Seoul 151-921, Republic of Korea
| | - Ciro Isidoro
- Laboratory of Molecular Pathology and NanoBioImaging, Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy;
| | - Danny N. Dhanasekaran
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.N.); (S.K.); (J.H.H.); (M.J.)
- Department of Cell Biology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
34
|
Gkini V, Namba T. Glutaminolysis and the Control of Neural Progenitors in Neocortical Development and Evolution. Neuroscientist 2023; 29:177-189. [PMID: 35057642 PMCID: PMC10018057 DOI: 10.1177/10738584211069060] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Multiple types of neural progenitor cells (NPCs) contribute to the development of the neocortex, a brain region responsible for our higher cognitive abilities. Proliferative capacity of NPCs varies among NPC types, developmental stages, and species. The higher proliferative capacity of NPCs in the developing human neocortex is thought to be a major contributing factor why humans have the most expanded neocortex within primates. Recent studies have shed light on the importance of cell metabolism in the neocortical NPC proliferative capacity. Specifically, glutaminolysis, a metabolic pathway that converts glutamine to glutamate and then to α-ketoglutarate, has been shown to play a critical role in human NPCs, both in apical and basal progenitors. In this review, we summarize our current knowledge of NPC metabolism, focusing especially on glutaminolysis, and discuss the role of NPC metabolism in neocortical development, evolution, and neurodevelopmental disorders, providing a broader perspective on a newly emerging research field.
Collapse
Affiliation(s)
- Vasiliki Gkini
- Neuroscience Center, HiLIFE—Helsinki
Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Takashi Namba
- Neuroscience Center, HiLIFE—Helsinki
Institute of Life Science, University of Helsinki, Helsinki, Finland
- Takashi Namba, Neuroscience Center, HiLIFE
— Helsinki Institute of Life Science, University of Helsinki, PO 63,
Haartmaninkatu 8, Helsinki 00014, Finland.
| |
Collapse
|
35
|
Chen J, Feng D, Lu Y, Zhang Y, Jiang H, Yuan M, Xu Y, Zou J, Zhu Y, Zhang J, Ge C, Wang Y. A Novel Phenazine Analog, CPUL1, Suppresses Autophagic Flux and Proliferation in Hepatocellular Carcinoma: Insight from Integrated Transcriptomic and Metabolomic Analysis. Cancers (Basel) 2023; 15:cancers15051607. [PMID: 36900398 PMCID: PMC10001020 DOI: 10.3390/cancers15051607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 03/03/2023] [Indexed: 03/08/2023] Open
Abstract
BACKGROUND CPUL1, a phenazine analog, has demonstrated potent antitumor properties against hepatocellular carcinoma (HCC) and indicates a promising prospect in pharmaceutical development. However, the underlying mechanisms remain largely obscure. METHODS Multiple HCC cell lines were used to investigate the in vitro effects of CPUL1. The antineoplastic properties of CPUL1 were assessed in vivo by establishing a xenograft nude mice model. After that, metabolomics, transcriptomics, and bioinformatics were integrated to elucidate the mechanisms underlying the therapeutic efficacy of CPUL1, highlighting an unanticipated involvement of autophagy dysregulation. RESULTS CPUL1 suppressed HCC cell proliferation in vitro and in vivo, thereby endorsing the potential as a leading agent for HCC therapy. Integrative omics characterized a deteriorating scenario of metabolic debilitation with CPUL1, presenting an issue in the autophagy contribution of autophagy. Subsequent observations indicated that CPUL1 treatment could impede autophagic flow by suppressing autophagosome degradation rather than its formation, which supposedly exacerbated cellular damage triggered by metabolic impairment. Moreover, the observed late autophagosome degradation may be attributed to lysosome dysfunction, which is essential for the final stage of autophagy and cargo disposal. CONCLUSIONS Our study comprehensively profiled the anti-hepatoma characteristics and molecular mechanisms of CPUL1, highlighting the implications of progressive metabolic failure. This could partially be ascribed to autophagy blockage, which supposedly conveyed nutritional deprivation and intensified cellular vulnerability to stress.
Collapse
Affiliation(s)
- Jiaqin Chen
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Dong Feng
- Nanjing Southern Pharmaceutical Technology Co., Ltd., Nanjing 211100, China
| | - Yuanyuan Lu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Yanjun Zhang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Hanxiang Jiang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Man Yuan
- Department of Clinical Pharmacy, School of Basic Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yifan Xu
- Department of Clinical Pharmacy, School of Basic Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Jianjun Zou
- Department of Pharmacy, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
- Department of Clinical Pharmacology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Yubing Zhu
- Department of Pharmacy, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
- Department of Clinical Pharmacology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Jingjing Zhang
- Department of Pharmacy, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
- Department of Clinical Pharmacology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Chun Ge
- Department of Pharmacy, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
- Department of Clinical Pharmacology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
- Correspondence: (C.G.); (Y.W.)
| | - Ying Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
- Correspondence: (C.G.); (Y.W.)
| |
Collapse
|
36
|
Characterization of metabolic reprogramming by metabolomics in the oncocytic thyroid cancer cell line XTC.UC1. Sci Rep 2023; 13:149. [PMID: 36599897 PMCID: PMC9813134 DOI: 10.1038/s41598-023-27461-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 01/02/2023] [Indexed: 01/05/2023] Open
Abstract
Oncocytic thyroid cancer is characterized by the aberrant accumulation of abnormal mitochondria in the cytoplasm and a defect in oxidative phosphorylation. We performed metabolomics analysis to compare metabolic reprogramming among the oncocytic and non-oncocytic thyroid cancer cell lines XTC.UC1 and TPC1, respectively, and a normal thyroid cell line Nthy-ori 3-1. We found that although XTC.UC1 cells exhibit higher glucose uptake than TPC1 cells, the glycolytic intermediates are not only utilized to generate end-products of glycolysis, but also diverted to branching pathways such as lipid metabolism and the serine synthesis pathway. Glutamine is preferentially used to produce glutathione to reduce oxidative stress in XTC.UC1 cells, rather than to generate α-ketoglutarate for anaplerotic flux into the TCA cycle. Thus, growth, survival and redox homeostasis of XTC.UC1 cells rely more on both glucose and glutamine than do TPC1 cells. Furthermore, XTC.UC1 cells contained higher amounts of intracellular amino acids which is due to higher expression of the amino acid transporter ASCT2 and enhanced autophagy, thus providing the building blocks for macromolecules and energy production. These metabolic alterations are required for oncocytic cancer cells to compensate their defective mitochondrial function and to alleviate excess oxidative stress.
Collapse
|
37
|
Glutamine-dependent effects of nitric oxide on cancer cells subjected to hypoxia-reoxygenation. Nitric Oxide 2023; 130:22-35. [PMID: 36414197 DOI: 10.1016/j.niox.2022.11.003] [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: 07/07/2022] [Revised: 11/12/2022] [Accepted: 11/18/2022] [Indexed: 11/21/2022]
Abstract
Limited O2 availability can decrease essential processes in energy metabolism. However, cancers have developed distinct metabolic adaptations to these conditions. For example, glutaminolysis can maintain energy metabolism and hypoxia signaling. Additionally, it has been observed that nitric oxide (NO) possesses concentration-dependent, biphasic effects in cancer. NO has potent anti-tumor effects through modulating events such as angiogenesis and metastasis at low physiological concentrations and inducing cell death at higher concentrations. In this study, Ewing Sarcoma cells (A-673), MIA PaCa, and SKBR3 cells were treated with DetaNONOate (DetaNO) in a model of hypoxia (1% O2) and reoxygenation (21% O2). All 3 cell types showed NO-dependent inhibition of cellular O2 consumption which was enhanced as O2-tension decreased. L-Gln depletion suppressed the mitochondrial response to decreasing O2 tension in all 3 cell types and resulted in inhibition of Complex I activity. In A-673 cells the O2 tension dependent change in mitochondrial O2 consumption and increase in glycolysis was dependent on the presence of L-Gln. The response to hypoxia and Complex I activity were restored by α-ketoglutarate. NO exposure resulted in the A-673 cells showing greater sensitivity to decreasing O2 tension. Under conditions of L-Gln depletion, NO restored HIF-1α levels and the mitochondrial response to O2 tension possibly through the increase of 2-hydroxyglutarate. NO also resulted in suppression of cellular bioenergetics and further inhibition of Complex I which was not rescued by α-ketoglutarate. Taken together these data suggest that NO modulates the mitochondrial response to O2 differentially in the absence and presence of L-Gln. These data suggest a combination of metabolic strategies targeting glutaminolysis and Complex I in cancer cells.
Collapse
|
38
|
Ranjan P, Dubey VK. Krebs cycle enzymes for targeted therapeutics and immunotherapy for anti-leishmanial drug development using: Pathways, potential targets, and future perspectives. Life Sci 2022; 322:121314. [PMID: 36566880 DOI: 10.1016/j.lfs.2022.121314] [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: 10/08/2022] [Revised: 12/09/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022]
Abstract
Leishmaniasis is a parasitic and neglected tropical disease which majorly impacts poor and developing nations. One of the significant factors that impacts the severity of the pathological condition includes the socioeconomic background of the affected region. The rise of drug-resistant Leishmania is a serious concern for the effectiveness of the present treatment. As a result, the drug options need to be relooked immediately. Leishmania employs Krebs cycle intermediates for its needs after infection for establishing various defense mechanisms to escape the host immune responses. Nevertheless, a variety of immunological reactions are also seen during infection, which clear the parasites. One of the more promising strategies in this regard would involve combining targeted therapy and immunotherapy. The targeted treatments work by obstructing vital pathways that are required for Leishmania to grow and survive. The mechanism of action of immunotherapy is the control of the host immune response, which entails the blockage of molecular pathways essential for the growth and maintenance of the parasite. The Krebs cycle intermediates have important biochemical roles. Additionally, in macrophages and dendritic cells, they play roles as signalling molecules for controlling inflammatory responses. The review brings together the available literature about the importance of Krebs cycle metabolites as potential treatment targets for leishmaniasis.
Collapse
Affiliation(s)
- Preeti Ranjan
- School of Biochemical Engineering, Indian Institute of Technology BHU, Varanasi, UP 221005, India
| | - Vikash Kumar Dubey
- School of Biochemical Engineering, Indian Institute of Technology BHU, Varanasi, UP 221005, India.
| |
Collapse
|
39
|
Sollazzo M, De Luise M, Lemma S, Bressi L, Iorio M, Miglietta S, Milioni S, Kurelac I, Iommarini L, Gasparre G, Porcelli AM. Respiratory Complex I dysfunction in cancer: from a maze of cellular adaptive responses to potential therapeutic strategies. FEBS J 2022; 289:8003-8019. [PMID: 34606156 PMCID: PMC10078660 DOI: 10.1111/febs.16218] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 09/03/2021] [Accepted: 10/01/2021] [Indexed: 01/14/2023]
Abstract
Mitochondria act as key organelles in cellular bioenergetics and biosynthetic processes producing signals that regulate different molecular networks for proliferation and cell death. This ability is also preserved in pathologic contexts such as tumorigenesis, during which bioenergetic changes and metabolic reprogramming confer flexibility favoring cancer cell survival in a hostile microenvironment. Although different studies epitomize mitochondrial dysfunction as a protumorigenic hit, genetic ablation or pharmacological inhibition of respiratory complex I causing a severe impairment is associated with a low-proliferative phenotype. In this scenario, it must be considered that despite the initial delay in growth, cancer cells may become able to resume proliferation exploiting molecular mechanisms to overcome growth arrest. Here, we highlight the current knowledge on molecular responses activated by complex I-defective cancer cells to bypass physiological control systems and to re-adapt their fitness during microenvironment changes. Such adaptive mechanisms could reveal possible novel molecular players in synthetic lethality with complex I impairment, thus providing new synergistic strategies for mitochondrial-based anticancer therapy.
Collapse
Affiliation(s)
- Manuela Sollazzo
- Department of Pharmacy and Biotechnology (FABIT), Alma Mater Studiorum-University of Bologna, Bologna, Italy.,Center for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy
| | - Monica De Luise
- Center for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Silvia Lemma
- Center for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Licia Bressi
- Center for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Maria Iorio
- Center for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Stefano Miglietta
- Department of Pharmacy and Biotechnology (FABIT), Alma Mater Studiorum-University of Bologna, Bologna, Italy.,Center for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy
| | - Sara Milioni
- Department of Pharmacy and Biotechnology (FABIT), Alma Mater Studiorum-University of Bologna, Bologna, Italy.,Center for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy
| | - Ivana Kurelac
- Center for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum-University of Bologna, Bologna, Italy.,Centro di Studio e Ricerca sulle Neoplasie (CSR) Ginecologiche, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Luisa Iommarini
- Department of Pharmacy and Biotechnology (FABIT), Alma Mater Studiorum-University of Bologna, Bologna, Italy.,Center for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy.,Centro di Studio e Ricerca sulle Neoplasie (CSR) Ginecologiche, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Giuseppe Gasparre
- Center for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum-University of Bologna, Bologna, Italy.,Centro di Studio e Ricerca sulle Neoplasie (CSR) Ginecologiche, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Anna Maria Porcelli
- Department of Pharmacy and Biotechnology (FABIT), Alma Mater Studiorum-University of Bologna, Bologna, Italy.,Center for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy.,Centro di Studio e Ricerca sulle Neoplasie (CSR) Ginecologiche, Alma Mater Studiorum-University of Bologna, Bologna, Italy.,Interdepartmental Center for Industrial Research (CIRI) Life Sciences and Technologies for Health, Alma Mater Studiorum-University of Bologna, Ozzano dell'Emilia, Italy
| |
Collapse
|
40
|
Kant R, Manne RK, Anas M, Penugurti V, Chen T, Pan BS, Hsu CC, Lin HK. Deregulated transcription factors in cancer cell metabolisms and reprogramming. Semin Cancer Biol 2022; 86:1158-1174. [PMID: 36244530 PMCID: PMC11220368 DOI: 10.1016/j.semcancer.2022.10.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 09/10/2022] [Accepted: 10/11/2022] [Indexed: 01/27/2023]
Abstract
Metabolic reprogramming is an important cancer hallmark that plays a key role in cancer malignancies and therapy resistance. Cancer cells reprogram the metabolic pathways to generate not only energy and building blocks but also produce numerous key signaling metabolites to impact signaling and epigenetic/transcriptional regulation for cancer cell proliferation and survival. A deeper understanding of the mechanisms by which metabolic reprogramming is regulated in cancer may provide potential new strategies for cancer targeting. Recent studies suggest that deregulated transcription factors have been observed in various human cancers and significantly impact metabolism and signaling in cancer. In this review, we highlight the key transcription factors that are involved in metabolic control, dissect the crosstalk between signaling and transcription factors in metabolic reprogramming, and offer therapeutic strategies targeting deregulated transcription factors for cancer treatment.
Collapse
Affiliation(s)
- Rajni Kant
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston-Salem, NC 27101, USA
| | - Rajesh Kumar Manne
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston-Salem, NC 27101, USA
| | - Mohammad Anas
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston-Salem, NC 27101, USA
| | - Vasudevarao Penugurti
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston-Salem, NC 27101, USA
| | - Tingjin Chen
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston-Salem, NC 27101, USA
| | - Bo-Syong Pan
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston-Salem, NC 27101, USA
| | - Che-Chia Hsu
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston-Salem, NC 27101, USA
| | - Hui-Kuan Lin
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston-Salem, NC 27101, USA.
| |
Collapse
|
41
|
Bhardwaj V, He J, Jain A. Glutamine stabilizes myc via alpha-ketoglutarate and regulates paclitaxel sensitivity. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:227. [PMID: 36175776 DOI: 10.1007/s12032-022-01834-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 06/24/2022] [Indexed: 11/30/2022]
Abstract
Metabolic reprogramming wherein the cancer cells exhibit altered energetics is a hallmark of cancer. Although recent discoveries have enhanced our understanding of tumor metabolism, the therapeutic utility of targeting tumor metabolism is not yet realized. Glutamine, a non-essential amino acid, plays a critical role in regulating tumor metabolism and provides an alternative tumor energy source. In this study, we investigate the molecular mechanism regulated by glutamine and elucidate if targeting glutamine metabolism would enhance the efficacy of cancer chemotherapy. Using clonogenic and cell cycle analysis, we found that deprivation of glutamine suppress the growth of cancer cells. Mechanistically we demonstrate that glutamine stabilizes myc by preventing its ubiquitination through alpha-ketoglutarate. Inhibition of glutamine metabolism enhanced the sensitivity of tumor cells to chemotherapeutic agent paclitaxel. Our results delineate the mechanism behind glutamine-induced myc stabilization, and they provide a viable strategy to target cancer with a glutamine metabolism inhibitor in the clinic.
Collapse
Affiliation(s)
- Vikas Bhardwaj
- Department of Pharmaceutical Sciences, Jefferson College of Pharmacy, Thomas Jefferson University, Philadelphia, PA, USA.
| | - Jun He
- Department of Pathology, Anatomy & Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Aditi Jain
- The Jefferson Pancreas, Biliary and Related Cancer Center, Department of Surgery, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
42
|
Characterization of the Metabolome of Breast Tissues from Non-Hispanic Black and Non-Hispanic White Women Reveals Correlations between Microbial Dysbiosis and Enhanced Lipid Metabolism Pathways in Triple-Negative Breast Tumors. Cancers (Basel) 2022; 14:cancers14174075. [PMID: 36077608 PMCID: PMC9454857 DOI: 10.3390/cancers14174075] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 08/18/2022] [Accepted: 08/19/2022] [Indexed: 12/02/2022] Open
Abstract
Simple Summary We previously showed that breast tumor tissues from women display an imbalance in abundance and composition of microbiota compared to normal healthy breast tissues. It is unknown whether these differences in breast tumor microbiota may be driven by alterations in microbial metabolites, leading to potentially protective or pathogenic consequences. The aim of our study was to conduct global metabolic profiling on normal and breast tumor tissues to identify differences in metabolite profiles and to determine whether breast microbial dysbiosis may be associated with enrichment of microbial metabolites in triple-negative breast cancer (TNBC) which disproportionately affects women of African ancestry. We observed significant correlations between elevated lipid metabolism pathways and microbial dysbiosis in TNBC tissues from both non-Hispanic black and white women. This is the first study to report an association between breast microbial dysbiosis and alterations in host metabolic pathways in breast tumors, including TNBC, of non-Hispanic black and non-Hispanic white women. Abstract Triple-negative breast cancer (TNBC) is an aggressive form of breast cancer that is non-responsive to hormonal therapies and disproportionately impact women of African ancestry. We previously showed that TN breast tumors have a distinct microbial signature that differs from less aggressive breast tumor subtypes and normal breast tissues. However, it is unknown whether these differences in breast tumor microbiota may be driven by alterations in microbial metabolites, leading to potentially protective or pathogenic consequences. The goal of this global metabolomic profiling study was to investigate alterations in microbial metabolism pathways in normal and breast tumor tissues, including TNBC, of non-Hispanic black (NHB) and non-Hispanic white (NHW) women. In this study, we profiled the microbiome (16S rRNA) from breast tumor tissues and analyzed 984 metabolites from a total of 51 NHB and NHW women. Breast tumor tissues were collected from 15 patients with TNBC, 12 patients with less aggressive luminal A-type (Luminal) breast cancer, and 24 healthy controls for comparison using UHPLC-tandem mass spectrometry. Principal component analysis and hierarchical clustering of the global metabolomic profiling data revealed separation between metabolic signatures of normal and breast tumor tissues. Random forest analysis revealed a unique biochemical signature associated with elevated lipid metabolites and lower levels of microbial-derived metabolites important in controlling inflammation and immune responses in breast tumor tissues. Significant relationships between the breast microbiome and the metabolome, particularly lipid metabolism, were observed in TNBC tissues. Further investigations to determine whether alterations in sphingolipid, phospholipid, ceramide, amino acid, and energy metabolism pathways modulate Fusobacterium and Tenericutes abundance and composition to alter host metabolism in TNBC are necessary to help us understand the risk and underlying mechanisms and to identify potential microbial-based targets.
Collapse
|
43
|
Oliva CR, Ali MY, Flor S, Griguer CE. Effect of Expression of Nuclear-Encoded Cytochrome C Oxidase Subunit 4 Isoforms on Metabolic Profiles of Glioma Cells. Metabolites 2022; 12:metabo12080748. [PMID: 36005623 PMCID: PMC9415780 DOI: 10.3390/metabo12080748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/09/2022] [Accepted: 08/11/2022] [Indexed: 11/16/2022] Open
Abstract
Although often effective at treating newly diagnosed glioblastoma (GBM), increasing evidence suggests that chemo- and radiotherapy-induced alterations in tumor metabolism promote GBM recurrence and aggressiveness, as well as treatment resistance. Recent studies have demonstrated that alterations in glioma cell metabolism, induced by a switch in the isoform expression of cytochrome c oxidase subunit 4 (COX4), a key regulatory subunit of mammalian cytochrome c oxidase, could promote these effects. To understand how the two COX4 isoforms (COX4-1 and COX4-2) differentially affect glioma metabolism, glioma samples harvested from COX4-1- or COX4-2-overexpressing U251 cells were profiled using Gas chromatography–mass spectrometry GC-MS and Liquid Chromatography - Tandem Mass Spectrometry LC-MS/MS metabolomics platforms. The concentration of 362 metabolites differed significantly in the two cell types. The two most significantly upregulated pathways associated with COX4-1 overexpression were purine and glutathione metabolism; the two most significantly downregulated metabolic pathways associated with COX4-1 expression were glycolysis and fatty acid metabolism. Our study provides new insights into how Cytochrome c oxidase (CcO) regulatory subunits affect cellular metabolic networks in GBM and identifies potential targets that may be exploited for therapeutic benefit.
Collapse
Affiliation(s)
- Claudia R. Oliva
- Free Radical & Radiation Biology Program, Department of Radiation Oncology, The University of Iowa, Iowa City, IA 52242, USA
| | - Md Yousuf Ali
- Free Radical & Radiation Biology Program, Department of Radiation Oncology, The University of Iowa, Iowa City, IA 52242, USA
- Interdisciplinary Graduate Program in Human Toxicology, Department Radiation Oncology, The University of Iowa, Iowa City, IA 52242, USA
| | - Susanne Flor
- Free Radical & Radiation Biology Program, Department of Radiation Oncology, The University of Iowa, Iowa City, IA 52242, USA
| | - Corinne E. Griguer
- Free Radical & Radiation Biology Program, Department of Radiation Oncology, The University of Iowa, Iowa City, IA 52242, USA
- Correspondence:
| |
Collapse
|
44
|
Farooqi AA, Fayyaz S, Poltronieri P, Calin G, Mallardo M. Epigenetic deregulation in cancer: Enzyme players and non-coding RNAs. Semin Cancer Biol 2022; 83:197-207. [PMID: 32738290 DOI: 10.1016/j.semcancer.2020.07.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/26/2020] [Accepted: 07/27/2020] [Indexed: 02/08/2023]
Abstract
Data obtained from cutting-edge research have shown that deregulated epigenetic marks are critical hallmarks of cancer. Rapidly emerging scientific evidence has helped in developing a proper understanding of the mechanisms leading to control of cellular functions, from changes in chromatin accessibility, transcription and translation, and in post-translational modifications. Firstly, mechanisms of DNA methylation and demethylation are introduced, as well as modifications of DNA and RNA, with particular focus on N6-methyladenosine (m6A), discussing the effects of these modifications in normal cells and in malignancies. Then, chromatin modifying proteins and remodelling complexes are discussed. Many enzymes and accessory proteins in these complexes have been found mutated or have undergone differential splicing, leading to defective protein complexes. Epigenetic mechanisms acting on nucleosomes by polycomb repressive complexes and on chromatin by SWI/SNF complexes on nucleosome assembly/disassembly, as well as main mutated genes linked to cancers, are reviewed. Among enzymes acting on histones and other proteins erasing the reversible modifications are histone deacetylases (HDACs). Sirtuins are of interest since most of these enzymes not only deacylate histones and other proteins, but also post-translationally modify proteins adding a Mono-ADP-ribose (MAR) moiety. MAR can be read by MACRO-domain containing proteins such as histone MacroH2A1, with specific function in chromatin assembly. Finally, recent advances are presented on non-coding RNAs with a scaffold function, prospecting their role in assembly of chromatin modifying complexes, recruiting enzyme players to chromatin regions. Lastly, the imbalance in metabolites production due to mitochondrial dysfunction is presented, with the potential of these metabolites to inhibit enzymes, either writers, readers or erasers of epitranscriptome marks. In the perspectives, studies are overwied on drugs under development aiming to limit excessive enzyme activities and to reactivate chromatin modifying complexes, for therapeutic application. This knowledge may lead to novel drugs and personalised medicine for cancer patients.
Collapse
Affiliation(s)
- Ammad Ahmad Farooqi
- Department of Molecular Oncology, Institute of Biomedical and Genetic Engineering (IBGE), Islamabad 44000, Pakistan.
| | | | - Palmiro Poltronieri
- Institute of Sciences of Food Productions, National Research Council of Italy, via Monteroni Km 7, 73100 Lecce, Italy.
| | - George Calin
- Department of Experimental Therapeutics, and Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Massimo Mallardo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples, "Federico II" via Pansini 5, Napoli, Italy.
| |
Collapse
|
45
|
Wang Y, Shen N, Spurlin G, Korm S, Huang S, Anderson NM, Huiting LN, Liu H, Feng H. α-Ketoglutarate-Mediated DNA Demethylation Sustains T-Acute Lymphoblastic Leukemia upon TCA Cycle Targeting. Cancers (Basel) 2022; 14:2983. [PMID: 35740646 PMCID: PMC9221025 DOI: 10.3390/cancers14122983] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 06/07/2022] [Accepted: 06/14/2022] [Indexed: 02/06/2023] Open
Abstract
Despite the development of metabolism-based therapy for a variety of malignancies, resistance to single-agent treatment is common due to the metabolic plasticity of cancer cells. Improved understanding of how malignant cells rewire metabolic pathways can guide the rational selection of combination therapy to circumvent drug resistance. Here, we show that human T-ALL cells shift their metabolism from oxidative decarboxylation to reductive carboxylation when the TCA cycle is disrupted. The α-ketoglutarate dehydrogenase complex (KGDHC) in the TCA cycle regulates oxidative decarboxylation by converting α-ketoglutarate (α-KG) to succinyl-CoA, while isocitrate dehydrogenase (IDH) 1 and 2 govern reductive carboxylation. Metabolomics flux analysis of T-ALL reveals enhanced reductive carboxylation upon genetic depletion of the E2 subunit of KGDHC, dihydrolipoamide-succinyl transferase (DLST), mimicking pharmacological inhibition of the complex. Mechanistically, KGDHC dysfunction causes increased demethylation of nuclear DNA by α-KG-dependent dioxygenases (e.g., TET demethylases), leading to increased production of both IDH1 and 2. Consequently, dual pharmacologic inhibition of the TCA cycle and TET demethylases demonstrates additive efficacy in reducing the tumor burden in zebrafish xenografts. These findings provide mechanistic insights into how T-ALL develops resistance to drugs targeting the TCA cycle and therapeutic strategies to overcome this resistance.
Collapse
Affiliation(s)
- Yanwu Wang
- Taikang Medical School (School of Basic Medical Science), Wuhan University, Wuhan 430071, China;
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA; (N.S.); (G.S.); (S.K.); (S.H.); (N.M.A.); (L.N.H.)
| | - Ning Shen
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA; (N.S.); (G.S.); (S.K.); (S.H.); (N.M.A.); (L.N.H.)
- Department of Medicine, Section of Hematology and Medical Oncology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Gervase Spurlin
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA; (N.S.); (G.S.); (S.K.); (S.H.); (N.M.A.); (L.N.H.)
| | - Sovannarith Korm
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA; (N.S.); (G.S.); (S.K.); (S.H.); (N.M.A.); (L.N.H.)
| | - Sarah Huang
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA; (N.S.); (G.S.); (S.K.); (S.H.); (N.M.A.); (L.N.H.)
| | - Nicole M. Anderson
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA; (N.S.); (G.S.); (S.K.); (S.H.); (N.M.A.); (L.N.H.)
| | - Leah N. Huiting
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA; (N.S.); (G.S.); (S.K.); (S.H.); (N.M.A.); (L.N.H.)
| | - Hudan Liu
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China;
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan 430071, China
| | - Hui Feng
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA; (N.S.); (G.S.); (S.K.); (S.H.); (N.M.A.); (L.N.H.)
- Department of Medicine, Section of Hematology and Medical Oncology, Boston University School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
46
|
Jerves T, Blau N, Ferreira CR. Clinical and biochemical footprints of inherited metabolic diseases. VIII. Neoplasias. Mol Genet Metab 2022; 136:118-124. [PMID: 35422340 PMCID: PMC9189061 DOI: 10.1016/j.ymgme.2022.03.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/23/2022] [Accepted: 03/24/2022] [Indexed: 12/21/2022]
Abstract
Cancer, caused by multiple cumulative pathogenic variants in tumor suppressor genes and proto-oncogenes, is a leading cause of mortality worldwide. The uncontrolled and rapid cell growth of the tumors requires a reprogramming of the complex cellular metabolic network to favor anabolism. Adequate management and treatment of certain inherited metabolic diseases might prevent the development of certain neoplasias, such as hepatocellular carcinoma in tyrosinemia type 1 or hepatocellular adenomas in glycogen storage disorder type 1a. We reviewed and updated the list of known metabolic etiologies associated with various types of benign and malignant neoplasias, finding 64 relevant inborn errors of metabolism. This is the eighth article of the series attempting to create a comprehensive list of clinical and metabolic differential diagnosis by system involvement.
Collapse
Affiliation(s)
- Teodoro Jerves
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Nenad Blau
- Division of Metabolism, University Children's Hospital, Zürich, Switzerland.
| | - Carlos R Ferreira
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
47
|
Ding X, Jin F, Xu J, Zhang S, Chen D, Hu B, Hong Y. The impact of aquaculture system on the microbiome and gut metabolome of juvenile Chinese softshell turtle ( Pelodiscus sinensis). IMETA 2022; 1:e17. [PMID: 38868566 PMCID: PMC10989827 DOI: 10.1002/imt2.17] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 03/03/2022] [Accepted: 03/13/2022] [Indexed: 06/14/2024]
Abstract
The commercial aquatic animal microbiome may markedly affect the successful host's farming in various aquaculture systems. However, very little was known about it. Here, two different aquaculture systems, the rice-fish culture (RFC) and intensive pond culture (IPC) systems, were compared to deconstruct the skin, oral, and gut microbiome, as well as the gut metabolome of juvenile Chinese softshell turtle (Pelodiscus sinensis). Higher alpha-diversity and functional redundancy of P. sinensis microbial community were found in the RFC than those of the IPC. The aquaculture systems have the strongest influence on the gut microbiome, followed by the skin microbiome, and finally the oral microbiome. Source-tracking analysis showed that the RFC's microbial community originated from more unknown sources than that of the IPC across all body regions. Strikingly, the RFC's oral and skin microbiome exhibited a significantly higher proportion of generalists and broader habitat niche breadth than those of the IPC, but not the gut. Null model analysis revealed that the RFC's oral and skin microbial community assembly was governed by a significantly greater proportion of deterministic processes than that of the IPC, but not the gut. We further identified the key gene and microbial contribution to five significantly changed gut metabolites, 2-oxoglutarate, N-acetyl-d-mannosamine, cis-4-hydroxy-d-proline, nicotinamide, and l-alanine, which were significantly correlated with important categories of microbe-mediated processes, including the amino acid metabolism, GABAergic synapse, ABC transporters, biosynthesis of unsaturated fatty acids, as well as citrate cycle. Moreover, different aquaculture systems have a significant impact on the hepatic lipid metabolism and body shape of P. sinensis. Our results provide new insight into the influence of aquaculture systems on the microbial community structure feature and assembly mechanism in an aquatic animal, also highlighting the key microbiome and gene contributions to the metabolite variation in the gut microbiome-metabolome association.
Collapse
Affiliation(s)
- Xia Ding
- School of Life SciencesNanchang UniversityNanchangJiangxiChina
| | - Feng Jin
- School of Life SciencesNanchang UniversityNanchangJiangxiChina
| | - Jiawang Xu
- School of Life SciencesNanchang UniversityNanchangJiangxiChina
| | - Shulei Zhang
- School of Life SciencesNanchang UniversityNanchangJiangxiChina
| | - Dongxu Chen
- School of Life SciencesNanchang UniversityNanchangJiangxiChina
| | - Beijuan Hu
- School of Life SciencesNanchang UniversityNanchangJiangxiChina
| | - Yijiang Hong
- School of Life SciencesNanchang UniversityNanchangJiangxiChina
| |
Collapse
|
48
|
MYC, mitochondrial metabolism and O-GlcNAcylation converge to modulate the activity and subcellular localization of DNA and RNA demethylases. Leukemia 2022; 36:1150-1159. [PMID: 34997181 PMCID: PMC8983447 DOI: 10.1038/s41375-021-01489-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/17/2021] [Accepted: 11/25/2021] [Indexed: 12/31/2022]
Abstract
Mitochondria can function as signaling organelles, and part of this output leads to epigenetic remodeling. The full extent of this far-reaching interplay remains undefined. Here, we show that MYC transcriptionally activates IDH2 and increases alpha-ketoglutarate (αKG) levels. This regulatory step induces the activity of αKG-dependent DNA hydroxylases and RNA demethylases, thus reducing global DNA and RNA methylation. MYC, in a IDH2-dependent manner, also promotes the nuclear accumulation of TET1-TET2-TET3, FTO and ALKBH5. Notably, this subcellular movement correlated with the ability of MYC, in an IDH2-dependent manner, and, unexpectedly, of αKG to directly induce O-GlcNAcylation. Concordantly, modulation of the activity of OGT and OGA, enzymes that control the cycling of this non-canonical mono-glycosylation, largely recapitulated the effects of the MYC-IDH2-αKG axis on the subcellular movement of DNA and RNA demethylases. Together, we uncovered a hitherto unsuspected crosstalk between MYC, αKG and O-GlcNAcylation which could influence the epigenome and epitranscriptome homeostasis.
Collapse
|
49
|
Włodarczyk M, Nowicka G, Ciebiera M, Ali M, Yang Q, Al-Hendy A. Epigenetic Regulation in Uterine Fibroids-The Role of Ten-Eleven Translocation Enzymes and Their Potential Therapeutic Application. Int J Mol Sci 2022; 23:2720. [PMID: 35269864 PMCID: PMC8910916 DOI: 10.3390/ijms23052720] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/23/2022] [Accepted: 02/25/2022] [Indexed: 02/01/2023] Open
Abstract
Uterine fibroids (UFs) are monoclonal, benign tumors that contain abnormal smooth muscle cells and the accumulation of extracellular matrix (ECM). Although benign, UFs are a major source of gynecologic and reproductive dysfunction, ranging from menorrhagia and pelvic pain to infertility, recurrent miscarriage, and preterm labor. Many risk factors are involved in the pathogenesis of UFs via genetic and epigenetic mechanisms. The latter involving DNA methylation and demethylation reactions provide specific DNA methylation patterns that regulate gene expression. Active DNA demethylation reactions mediated by ten-eleven translocation proteins (TETs) and elevated levels of 5-hydroxymethylcytosine have been suggested to be involved in UF formation. This review paper summarizes the main findings regarding the function of TET enzymes and their activity dysregulation that may trigger the development of UFs. Understanding the role that epigenetics plays in the pathogenesis of UFs may possibly lead to a new type of pharmacological fertility-sparing treatment method.
Collapse
Affiliation(s)
- Marta Włodarczyk
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1B, 02-097 Warsaw, Poland;
- Centre for Preclinical Research, Medical University of Warsaw, Banacha 1B, 02-097 Warsaw, Poland
| | - Grażyna Nowicka
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1B, 02-097 Warsaw, Poland;
- Centre for Preclinical Research, Medical University of Warsaw, Banacha 1B, 02-097 Warsaw, Poland
| | - Michał Ciebiera
- The Center of Postgraduate Medical Education, Second Department of Obstetrics and Gynecology, 01-809 Warsaw, Poland;
| | - Mohamed Ali
- Clinical Pharmacy Department, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt;
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL 60637, USA; (Q.Y.); (A.A.-H.)
| | - Qiwei Yang
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL 60637, USA; (Q.Y.); (A.A.-H.)
| | - Ayman Al-Hendy
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL 60637, USA; (Q.Y.); (A.A.-H.)
| |
Collapse
|
50
|
Over-Reduced State of Mitochondria as a Trigger of "β-Oxidation Shuttle" in Cancer Cells. Cancers (Basel) 2022; 14:cancers14040871. [PMID: 35205619 PMCID: PMC8870273 DOI: 10.3390/cancers14040871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/27/2022] [Accepted: 02/07/2022] [Indexed: 11/17/2022] Open
Abstract
A considerable amount of data have accumulated in the last decade on the pronounced mitochondrial fatty acid oxidation (mFAO) in many types of cancer cells. As a result, mFAO was found to coexist with abnormally activated fatty acid synthesis (FAS) and the mevalonate pathway. Recent studies have demonstrated that overactivated mitochondrial β-oxidation may aggravate the impaired mitochondrial redox state and vice versa. Furthermore, the impaired redox state of cancerous mitochondria can ensure the continuous operation of β-oxidation by disconnecting it from the Krebs cycle and connecting it to the citrate-malate shuttle. This could create a new metabolic state/pathway in cancer cells, which we have called the "β-oxidation-citrate-malate shuttle", or "β-oxidation shuttle" for short, which forces them to proliferate. The calculation of the phosphate/oxygen ratio indicates that it is inefficient as an energy source and must consume significantly more oxygen per mole of ATP produced when combined with acetyl-CoA consuming pathways, such as the FAS and mevalonate pathways. The "β-oxidation shuttle" is an unconventional mFAO, a separate metabolic pathway that has not yet been explored as a source of energy, as well as a source of cataplerosis, leading to biomass accumulation, accelerated oxygen consumption, and, ultimately, a source of proliferation. The role of the "β-oxidation shuttle" and its contribution to redox-altered cancer metabolism provides a new direction for the development of future anticancer strategies. This may represent the metabolic "secret" of cancer underlying hypoxia and genomic instability.
Collapse
|