1
|
Yi Q, Xiong L. From sensory organs to internal pathways: A comprehensive review of amino acid sensing in Drosophila. Comp Biochem Physiol A Mol Integr Physiol 2025; 303:111828. [PMID: 39983896 DOI: 10.1016/j.cbpa.2025.111828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/18/2025] [Accepted: 02/18/2025] [Indexed: 02/23/2025]
Abstract
Organisms require various nutrients to provide energy, support growth, and maintain metabolic balance. Amino acid is among the most basic nutrients, serving as fundamental building blocks for protein synthesis while playing vital roles in growth, development, and reproduction. Understanding the mechanisms by which organisms perceive amino acids is key to unraveling how they select appropriate food sources and adapt to environmental challenges. The fruit fly, Drosophila melanogaster, serves as a powerful model for understanding fundamental genetic and physiological processes. This review focuses on recent advances in amino acid sensing mechanisms in Drosophila melanogaster and their relevance to feeding behavior, nutrient homeostasis, and adaptive responses, and integrates insights into peripheral sensory systems, such as the legs and proboscis, as well as internal regulatory mechanisms within the gut, fat body, and brain. It highlights key molecular players, including ionotropic receptors, gut-derived hormones, neuropeptides, and the microbiome-gut-brain axis. Additionally, the manuscript identifies knowledge gaps and proposes directions for future research, providing a comprehensive overview of this dynamic field.
Collapse
Affiliation(s)
- Quan Yi
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Liangyao Xiong
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
2
|
Li L, Kang Y, Cheng R, Liu F, Wu F, Liu Z, Kou J, Zhang Z, Li W, Zhao H, He X, Du W. The de novo synthesis of GABA and its gene regulatory function control hepatocellular carcinoma metastasis. Dev Cell 2025; 60:1053-1069.e6. [PMID: 39740661 DOI: 10.1016/j.devcel.2024.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 09/14/2024] [Accepted: 12/04/2024] [Indexed: 01/02/2025]
Abstract
The neurotransmitter gamma-aminobutyric acid (GABA) has been thought to be involved in the development of some types of cancer. Yet, the de novo synthesis of GABA and how it functions in hepatocellular carcinoma (HCC) remain unclear. Here, we report that SLC6A12 acts as a transporter of GABA, and that aldehyde dehydrogenase 9 family member A1 (ALDH9A1), not glutamate decarboxylase 1 (GAD1), generates GABA in human HCC. Interestingly, SLC6A12 and ALDH9A1 are upregulated during lung metastases of HCC, and depletion of either of them leads to impaired HCC metastasis. Mechanistically, GABA directly binds and stabilizes β-catenin, resulting in activated Wnt/β-catenin signaling, and thereby enhancing HCC metastasis. Reciprocally, β-catenin transcriptionally upregulates SLC6A12 to import more GABA to stabilize β-catenin. Thus, our findings identify ALDH9A1 as the major GABA synthetase in HCC, demonstrate a positive-feedback regulatory mechanism for sustaining Wnt/β-catenin signaling, and reveal a role for β-catenin in sensing GABA, which contributes to HCC metastasis.
Collapse
Affiliation(s)
- 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
| | - Youli Kang
- 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
| | - Running Cheng
- 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
| | - Fangming Liu
- 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
| | - Fujia Wu
- 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
| | - Zizhao Liu
- 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
| | - Junjie Kou
- 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
| | - Haitao Zhao
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing 100730, China.
| | - Xiaojing He
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, 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
|
3
|
Do LK, Lee HM, Ha YS, Lee CH, Kim J. Amino acids in cancer: Understanding metabolic plasticity and divergence for better therapeutic approaches. Cell Rep 2025; 44:115529. [PMID: 40193251 DOI: 10.1016/j.celrep.2025.115529] [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: 07/31/2024] [Revised: 02/24/2025] [Accepted: 03/17/2025] [Indexed: 04/09/2025] Open
Abstract
Metabolic reprogramming is a hallmark of malignant transformation. While initial studies in the field of cancer metabolism focused on central carbon metabolism, the field has expanded to metabolism beyond glucose and glutamine and uncovered the important role of amino acids in tumorigenesis and tumor immunity as energy sources, signaling molecules, and precursors for (epi)genetic modification. As a result of the development and application of new technologies, a multifaceted picture has emerged, showing that context-dependent heterogeneity in amino acid metabolism exists between tumors and even within distinct regions of solid tumors. Understanding the complexity and flexibility of amino acid metabolism in cancer is critical because it can influence therapeutic responses and predict clinical outcomes. This overview discusses the current findings on the heterogeneity in amino acid metabolism in cancer and how understanding the metabolic diversity of amino acids can be translated into more clinically relevant therapeutic interventions.
Collapse
Affiliation(s)
- Linda K Do
- Department of Urology, Yale School of Medicine, New Haven, CT 06519, USA
| | - Hyun Min Lee
- Department of Urology, Yale School of Medicine, New Haven, CT 06519, USA
| | - Yun-Sok Ha
- Department of Urology, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu 41404, Korea
| | - Chan-Hyeong Lee
- Department of Urology, Yale School of Medicine, New Haven, CT 06519, USA
| | - Jiyeon Kim
- Department of Urology, Yale School of Medicine, New Haven, CT 06519, USA; Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT 06519, USA.
| |
Collapse
|
4
|
Sutherland AE. The role of serendipity in our investigation of embryo implantation. Dev Biol 2025; 520:135-140. [PMID: 39826766 PMCID: PMC11830518 DOI: 10.1016/j.ydbio.2025.01.010] [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: 09/16/2024] [Revised: 01/07/2025] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
Serendipity plays a huge role in science, and having a prepared mind that can seize upon a chance observation or occurrence can drive a project forward. This happened in my lab with a project centered on the regulation of trophoblast cell behavior at implantation. We discovered that amino acids regulate the onset of trophoblast motility through the activation of the kinase complex mTORC1, and that this acts as a checkpoint to trophoblast differentiation. This finding not only broadened our understanding of the mechanisms underlying embryo implantation, but also provided new ways of thinking about the regulation of diapause, a state of suspended embryonic development that occurs in many species. I should say that we re-discovered the fact that amino acids regulate the onset of trophoblast motility, as reading the literature showed us that others had made this same observation some 30 years previously and we were fortuitously able to build upon those findings. This project confirmed to me how valuable it is to read the literature widely, both historical papers and those in fields outside one's area of research, and to go to seminars on topics outside one's area.
Collapse
Affiliation(s)
- Ann E Sutherland
- Department of Cell Biology, University of Virginia Health System, PO Box 800732, Charlottesville, VA, 22908-0732, USA.
| |
Collapse
|
5
|
Zhou J, Yu J, Ren J, Ren Y, Zeng Y, Wu Y, Zhang Q, Xiao X. Association of maternal blood metabolomics and gestational diabetes mellitus risk: a systematic review and meta-analysis. Rev Endocr Metab Disord 2025; 26:205-222. [PMID: 39602052 DOI: 10.1007/s11154-024-09934-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/13/2024] [Indexed: 11/29/2024]
Abstract
Gestational diabetes mellitus (GDM) is a common complication of pregnancy that has short- and long-term adverse effects. Therefore, further exploration of the pathophysiology of GDM and related biomarkers is important. In this study, we performed a systematic review and meta-analysis to investigate the associations between metabolites in blood detected via metabolomics techniques and the risk of GDM and to identify possible biomarkers for predicting the occurrence of GDM. We retrieved case‒control and cohort studies of metabolomics and GDM published in PubMed, Embase, and Web of Science through March 29, 2024; extracted metabolite concentrations, odds ratios (ORs), or relative risks (RRs); and evaluated the integrated results with metabolites per-SD risk estimates and 95% CIs for GDM. We estimated the results via the random effects model and the inverse variance method. Our study is registered in PROSPERO (CRD42024539435). We included a total of 28 case‒control and cohort studies, including 17,370 subjects (4,372 GDM patients and 12,998 non-GDM subjects), and meta-analyzed 67 metabolites. Twenty-five of these metabolites were associated with GDM risk. Some amino acids (isoleucine, leucine, valine, alanine, aspartate, etc.), lipids (C16:0, C18:1n-9, C18:1n-7, lysophosphatidylcholine (LPC) (16:0), LPC (18:0), and palmitoylcarnitine), and carbohydrates and energy metabolites (glucose, pyruvate, lactate, 2-hydroxybutyrate, 3-hydroxybutyrate) were discovered to be associated with increased GDM risk (hazard ratio 1.06-2.77). Glutamine, histidine, C14:0, and sphingomyelin (SM) (34:1) were associated with lower GDM risk (hazard ratio 0.75-0.84). These findings suggest that these metabolites may play essential roles in GDM progression, and serve as biomarkers, contributing to the early diagnosis and prediction of GDM.
Collapse
Grants
- 81870545, 81870579, 82170854, 81570715, 81170736 National Natural Science Foundation of China
- 7202163 the Beijing Natural Science Foundation
- Z201100005520011 the Beijing Municipal Science and Technology Commission
- 2017YFC1309603, 2021YFC2501700, 2016YFA0101002, 2018YFC2001100 the National Key Research and Development Program of China
- 2019DCT-M-05 the Scientific Activities Foundation for Selected Returned Overseas Professionals of Human Resources and Social Security Ministry, Beijing Dongcheng District Outstanding Talent Funding Project
- 2017PT31036, 2018PT31021 the Medical Epigenetics Research Center, Chinese Academy of Medical Sciences
- 2023PT32010, 2017PT32020, 2018PT32001 the Non-profit Central Research Institute Fund of Chinese Academy of Medical Sciences
- CIFMS2017-I2M-1-008, CIFMS2021-I2M-1-002 the Chinese Academy of Medical Sciences Innovation Fund for Medical Sciences
- 2022-PUMCH-C-019 National High Level Hospital Clinical Research Funding
Collapse
Affiliation(s)
- Jing Zhou
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Diabetes Research Center of Chinese Academy of Medical Sciences, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Jie Yu
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Diabetes Research Center of Chinese Academy of Medical Sciences, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Jing Ren
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Diabetes Research Center of Chinese Academy of Medical Sciences, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yaolin Ren
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Diabetes Research Center of Chinese Academy of Medical Sciences, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yuan Zeng
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Diabetes Research Center of Chinese Academy of Medical Sciences, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yifan Wu
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Diabetes Research Center of Chinese Academy of Medical Sciences, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Qian Zhang
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Diabetes Research Center of Chinese Academy of Medical Sciences, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Xinhua Xiao
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Diabetes Research Center of Chinese Academy of Medical Sciences, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| |
Collapse
|
6
|
Abstract
Tuberous sclerosis complex (TSC) is an autosomal dominant inherited disease characterized by systemic hamartomas, neuropsychiatric symptoms known as TAND (TSC-associated neuropsychiatric disorders), and vitiligo. These symptoms are attributed to the constant activation of mechanistic target of rapamycin complex 1 (mTORC1) caused by genetic mutations in the causative genes TSC1 or TSC2. The elucidation of the pathogenesis of this disease and advances in diagnostic technologies have led to dramatic changes in the diagnosis and treatment of TSC. Diagnostic criteria have been created at a global level, and mTORC1 inhibitors have emerged as therapeutic agents for this disease. Previously, the treatment strategy was limited to symptomatic treatments such as surgery. Inhibitors of mTORC1 are effective against all symptoms of TSC, but they also have systemic side effects. Therefore, the need for a cross-disciplinary, collaborative medical care system has increased, resulting in the establishment of a practice structure known as the "TSC Board." Furthermore, to reduce the side effects of systemic administration of mTORC1 inhibitors, a topical formulation of mTORC1 inhibitor was developed in Japan for the treatment of skin lesions caused by TSC. This report summarizes the pathogenesis and current status of TSC and the contribution of the Neurocutaneous Syndrome Policy Research Group to the policies of the Ministry of Health, Labor, and Welfare with respect to this rare, intractable disease.
Collapse
Affiliation(s)
- Mari Wataya-Kaneda
- Department of Neurocutaneous Medicine, Division of Health Sciences, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Dermatology, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
7
|
Duerre DJ, Hansen JK, John SV, Jen A, Carrillo ND, Bui H, Bao Y, Fabregat M, Catrow JL, Chen LY, Overmyer KA, Shishkova E, Pearce Q, Keller MP, Anderson RA, Cryns VL, Attie AD, Cox JE, Coon JJ, Fan J, Galmozzi A. Haem biosynthesis regulates BCAA catabolism and thermogenesis in brown adipose tissue. Nat Metab 2025:10.1038/s42255-025-01253-6. [PMID: 40133548 DOI: 10.1038/s42255-025-01253-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/19/2025] [Indexed: 03/27/2025]
Abstract
The distinctive colour of brown adipose tissue (BAT) is attributed to its high content of haem-rich mitochondria. However, the mechanisms by which BAT regulates intracellular haem levels remain largely unexplored. Here we demonstrate that haem biosynthesis is the primary source of haem in brown adipocytes. Inhibiting haem biosynthesis results in an accumulation of the branched-chain amino acids (BCAAs) valine and isoleucine, owing to a haem-associated metabolon that channels BCAA-derived carbons into haem biosynthesis. Haem synthesis-deficient brown adipocytes display reduced mitochondrial respiration and lower UCP1 levels than wild-type cells. Although exogenous haem supplementation can restore intracellular haem levels and mitochondrial function, UCP1 downregulation persists. This sustained UCP1 suppression is linked to epigenetic regulation induced by the accumulation of propionyl-CoA, a byproduct of disrupted haem synthesis. Finally, disruption of haem biosynthesis in BAT impairs thermogenic response and, in female but not male mice, hinders the cold-induced clearance of circulating BCAAs in a sex-hormone-dependent manner. These findings establish adipose haem biosynthesis as a key regulator of thermogenesis and sex-dependent BCAA homeostasis.
Collapse
Affiliation(s)
- Dylan J Duerre
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Julia K Hansen
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Steven V John
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
- Morgridge Institute for Research, Madison, WI, USA
| | - Annie Jen
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomolecular Chemistry, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Noah D Carrillo
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Molecular and Environmental Toxicology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Hoang Bui
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Yutong Bao
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Matias Fabregat
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - J Leon Catrow
- Metabolomics Core Research Facility, University of Utah, Salt Lake City, UT, USA
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Li-Yu Chen
- Graduate Program in Chemistry, University of Wisconsin-Madison, Madison, WI, USA
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Katherine A Overmyer
- Morgridge Institute for Research, Madison, WI, USA
- Department of Biomolecular Chemistry, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- National Center for Quantitative Biology of Complex Systems, Madison, WI, USA
| | - Evgenia Shishkova
- Department of Biomolecular Chemistry, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- National Center for Quantitative Biology of Complex Systems, Madison, WI, USA
| | - Quentinn Pearce
- Metabolomics Core Research Facility, University of Utah, Salt Lake City, UT, USA
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Mark P Keller
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Richard A Anderson
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Vincent L Cryns
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Alan D Attie
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - James E Cox
- Metabolomics Core Research Facility, University of Utah, Salt Lake City, UT, USA
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Joshua J Coon
- Morgridge Institute for Research, Madison, WI, USA
- Department of Biomolecular Chemistry, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
- National Center for Quantitative Biology of Complex Systems, Madison, WI, USA
| | - Jing Fan
- Morgridge Institute for Research, Madison, WI, USA
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Department of Medical Microbiology & Immunology, University of Wisconsin-Madison, Madison, WI, USA
| | - Andrea Galmozzi
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA.
- Department of Biomolecular Chemistry, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA.
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA.
| |
Collapse
|
8
|
Koundouros N, Nagiec MJ, Bullen N, Noch EK, Burgos-Barragan G, Li Z, He L, Cho S, Parang B, Leone D, Andreopoulou E, Blenis J. Direct sensing of dietary ω-6 linoleic acid through FABP5-mTORC1 signaling. Science 2025; 387:eadm9805. [PMID: 40080571 DOI: 10.1126/science.adm9805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 10/09/2024] [Accepted: 01/14/2025] [Indexed: 03/15/2025]
Abstract
Diet influences macronutrient availability to cells, and although mechanisms of sensing dietary glucose and amino acids are well characterized, less is known about sensing lipids. We defined a nutrient signaling mechanism involving fatty acid-binding protein 5 (FABP5) and mechanistic target of rapamycin complex 1 (mTORC1) that is activated by the essential polyunsaturated fatty acid (PUFA) ω-6 linoleic acid (LA). FABP5 directly bound to the regulatory-associated protein of mTOR (Raptor) to enhance formation of functional mTORC1 and substrate binding, ultimately converging on increased mTOR signaling and proliferation. The amounts of FABP5 protein were increased in tumors and serum from triple-negative compared with those from receptor-positive breast cancer patients, which highlights its potential role as a biomarker that mediates cellular responses to ω-6 LA intake in this disease subtype.
Collapse
Affiliation(s)
- Nikos Koundouros
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Michal J Nagiec
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Nayah Bullen
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Evan K Noch
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Department of Neurology, Division of Neuro-oncology, Weill Cornell Medicine, New York, NY, USA
| | - Guillermo Burgos-Barragan
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Zhongchi Li
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Long He
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Sungyun Cho
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Bobak Parang
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Dominique Leone
- Cancer Clinical Trials Office - Breast, Weill Cornell Medicine, New York, NY, USA
| | - Eleni Andreopoulou
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York Presbyterian Hospital, New York, NY, USA
| | - John Blenis
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
9
|
Wang N, Lu S, Cao Z, Li H, Xu J, Zhou Q, Yin H, Qian Q, Zhang X, Tao M, Jiang Q, Zhou P, Zheng L, Han L, Li H, Yin L, Gu Y, Dou X, Sun H, Wang W, Piao HL, Li F, Xu Y, Yang W, Chen S, Liu J. Pyruvate metabolism enzyme DLAT promotes tumorigenesis by suppressing leucine catabolism. Cell Metab 2025:S1550-4131(25)00066-X. [PMID: 40112809 DOI: 10.1016/j.cmet.2025.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 11/24/2024] [Accepted: 02/22/2025] [Indexed: 03/22/2025]
Abstract
Pyruvate and branched-chain amino acid (BCAA) metabolism are pivotal pathways in tumor progression, yet the intricate interplay between them and its implications for tumor progression remain elusive. Our research reveals that dihydrolipoamide S-acetyltransferase (DLAT), a pyruvate metabolism enzyme, promotes leucine accumulation and sustains mammalian target of rapamycin (mTOR) complex activation in hepatocellular carcinoma (HCC). Mechanistically, DLAT directly acetylates the K109 residue of AU RNA-binding methylglutaconyl-coenzyme A (CoA) hydratase (AUH), a critical enzyme in leucine catabolism, inhibiting its activity and leading to leucine accumulation. Notably, DLAT upregulation correlates with poor prognosis in patients with HCC. Therefore, we developed an AUHK109R-mRNA lipid nanoparticles (LNPs) therapeutic strategy, which effectively inhibits tumor growth by restoring leucine catabolism and inhibiting mTOR activation in vivo. In summary, our findings uncover DLAT's unexpected role as an acetyltransferase for AUH, suppressing leucine catabolism. Restoring leucine catabolism with AUHK109R-mRNA LNP effectively inhibits HCC development, highlighting a novel direction for cancer research.
Collapse
Affiliation(s)
- Ning Wang
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Sijia Lu
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Ziyi Cao
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Huimin Li
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Junting Xu
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Qian Zhou
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Hanrui Yin
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Qiqi Qian
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Xianjing Zhang
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Mijia Tao
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Quanxin Jiang
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Peihui Zhou
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Liaoyuan Zheng
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Liu Han
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Hongtao Li
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Limin Yin
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yunqing Gu
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Xuefeng Dou
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Haipeng Sun
- Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Wei Wang
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Hai-Long Piao
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Fuming Li
- Institute of Metabolism and Integrative Biology, Fudan University, Shanghai 200438, China
| | - Yingjie Xu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Weiwei Yang
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Suzhen Chen
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| | - Junli Liu
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| |
Collapse
|
10
|
Xie Z, Lin M, Xing B, Wang H, Zhang H, Cai Z, Mei X, Zhu ZJ. Citrulline regulates macrophage metabolism and inflammation to counter aging in mice. SCIENCE ADVANCES 2025; 11:eads4957. [PMID: 40053596 PMCID: PMC11887811 DOI: 10.1126/sciadv.ads4957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 01/31/2025] [Indexed: 03/09/2025]
Abstract
Metabolic dysregulation and altered metabolite concentrations are widely recognized as key characteristics of aging. Comprehensive exploration of endogenous metabolites that drive aging remains insufficient. Here, we conducted an untargeted metabolomics analysis of aging mice, revealing citrulline as a consistently down-regulated metabolite associated with aging. Systematic investigations demonstrated that citrulline exhibited antiaging effects by reducing cellular senescence, protecting against DNA damage, preventing cell cycle arrest, modulating macrophage metabolism, and mitigating inflammaging. Long-term citrulline supplementation in aged mice yielded beneficial effects and ameliorated age-associated phenotypes. We further elucidated that citrulline acts as an endogenous metabolite antagonist to inflammation, suppressing proinflammatory responses in macrophages. Mechanistically, citrulline served as a potential inhibitor of mammalian target of rapamycin (mTOR) activation in macrophage and regulated the mTOR-hypoxia-inducible factor 1α-glycolysis signaling pathway to counter inflammation and aging. These findings underscore the significance of citrulline deficiency as a driver of aging, highlighting citrulline supplementation as a promising therapeutic intervention to counteract aging-related changes.
Collapse
Affiliation(s)
- Zhangdan Xie
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Moubin Lin
- Center for Clinical Research and Translational Medicine, Yangpu Hospital, Tongji University School of Medicine, Shanghai 200090, P.R. China
| | - Beizi Xing
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Hongmiao Wang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Haosong Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Zimu Cai
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, P. R. China
| | - Xinyu Mei
- Center for Clinical Research and Translational Medicine, Yangpu Hospital, Tongji University School of Medicine, Shanghai 200090, P.R. China
| | - Zheng-Jiang Zhu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
- Shanghai Key Laboratory of Aging Studies, Shanghai 201210, P. R. China
| |
Collapse
|
11
|
Sui Y, Hoshi N, Okamoto N, Inoue Y, Funatsu T, Ku Y, Ooi M, Watanabe D, Miyazaki H, Agawa M, Nakamura H, Ohgaki R, Kanai Y, Yang H, Kodama Y. The role of LAT1 in AOM/DSS-induced colorectal tumorigenesis. Biochem Biophys Res Commun 2025; 751:151446. [PMID: 39922055 DOI: 10.1016/j.bbrc.2025.151446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 01/25/2025] [Accepted: 02/03/2025] [Indexed: 02/10/2025]
Abstract
Amino acid transporters are essential for supplying nutrients to cells and are implicated in tumor progression. L-type amino acid transporter 1 (LAT1) is reported to be overexpressed in various cancers, affecting tumor development. However, the exact mechanisms by which LAT1 affects colorectal cancer (CRC) arising from a chronic inflammatory background are not yet fully understood. This study aimed to explore the role of LAT1 in CRC. Mice with intestinal epithelium-specific deletions of LAT1 (LAT1fl/fl; vil-cre) were treated with azoxymethane (AOM)/dextran sulfate sodium (DSS) in a colitis-associated cancer (CAC) model. Our results demonstrated that LAT1 was detected in normal colon crypts and highly expressed in AOM/DSS-induced tumor tissue. During the chronic colitis phase, weight loss was more prominent in LAT1fl/fl; vil-cre mice, compared with that in LAT1fl/fl mice. IL-1β and IL-6 expressions significantly increased in LAT1-deleted tumors; however, no overall difference in colon tumor number or size was observed between LAT1fl/fl and LAT1fl/fl; vil-cre mice. Accordingly, cell proliferation and apoptotic cell number were similar when comparing LAT1-deleted tumors with those with sufficient LAT1. Our findings indicated that LAT1 might not phenotypically affect overall colonic tumor development in this model; however, it affected the chronic colitis phase and inflammatory status within the tumors. These findings suggest that severe inflammation in tumors might have compensated for tumor growth in defects of amino acid supplementation through LAT1 deficiency, and provide insights into the potential of LAT1-targeted therapies for clinical CRC treatment.
Collapse
Affiliation(s)
- Yunlong Sui
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510260, China; Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, 650-0017, Japan
| | - Namiko Hoshi
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, 650-0017, Japan; Division of Integrated Analyses of Bioresource and Health Care, Kobe University Graduate School of Medicine, Hyogo, 650-0047, Japan.
| | - Norihiro Okamoto
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, 650-0017, Japan
| | - Yuta Inoue
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, 650-0017, Japan
| | - Takumi Funatsu
- Division of Integrated Analyses of Bioresource and Health Care, Kobe University Graduate School of Medicine, Hyogo, 650-0047, Japan
| | - Yuna Ku
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, 650-0017, Japan
| | - Makoto Ooi
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, 650-0017, Japan
| | - Daisuke Watanabe
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, 650-0017, Japan
| | - Haruka Miyazaki
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, 650-0017, Japan
| | - Misaki Agawa
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, 650-0017, Japan
| | - Hirotaka Nakamura
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, 650-0017, Japan
| | - Ryuichi Ohgaki
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka, 565-0871, Japan
| | - Yoshikatsu Kanai
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka, 565-0871, Japan; Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, Osaka, 565-0871, Japan
| | - Hui Yang
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510260, China
| | - Yuzo Kodama
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, 650-0017, Japan
| |
Collapse
|
12
|
Fung TS, Ryu KW, Thompson CB. Arginine: at the crossroads of nitrogen metabolism. EMBO J 2025; 44:1275-1293. [PMID: 39920310 DOI: 10.1038/s44318-025-00379-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 02/09/2025] Open
Abstract
L-arginine is the most nitrogen-rich amino acid, acting as a key precursor for the synthesis of nitrogen-containing metabolites and an essential intermediate in the clearance of excess nitrogen. Arginine's side chain possesses a guanidino group which has unique biochemical properties, and plays a primary role in nitrogen excretion (urea), cellular signaling (nitric oxide) and energy buffering (phosphocreatine). The post-translational modification of protein-incorporated arginine by guanidino-group methylation also contributes to epigenetic gene control. Most human cells do not synthesize sufficient arginine to meet demand and are dependent on exogenous arginine. Thus, dietary arginine plays an important role in maintaining health, particularly upon physiologic stress. How cells adapt to changes in extracellular arginine availability is unclear, mostly because nearly all tissue culture media are supplemented with supraphysiologic levels of arginine. Evidence is emerging that arginine-deficiency can influence disease progression. Here, we review new insights into the importance of arginine as a metabolite, emphasizing the central role of mitochondria in arginine synthesis/catabolism and the recent discovery that arginine can act as a signaling molecule regulating gene expression and organelle dynamics.
Collapse
Affiliation(s)
- Tak Shun Fung
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Keun Woo Ryu
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Craig B Thompson
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
| |
Collapse
|
13
|
Haidurov A, Zheltukhin AO, Snezhkina AV, Krasnov GS, Kudryavtseva AV, Budanov AV. p53-regulated SESN1 and SESN2 regulate cell proliferation and cell death through control of STAT3. Cell Commun Signal 2025; 23:105. [PMID: 39985075 PMCID: PMC11846189 DOI: 10.1186/s12964-025-02104-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 02/11/2025] [Indexed: 02/24/2025] Open
Abstract
Sestrin1 and Sestrin2 (SESN1&2) are evolutionarily conserved, stress-responsive proteins that regulate cell growth and viability. The primary target of Sestrins is the mTORC1 protein kinase, an activator of anabolic processes and an autophagy inhibitor. Our previous studies showed that inactivating SESN1&2 in lung adenocarcinoma A549 cells accelerates cell proliferation and confers resistance to cell death without affecting mTORC1 activity, suggesting that SESN1&2 modulate cellular processes via mTORC1-independent mechanisms. This work describes a new mechanism through which SESN1&2 regulate cell proliferation and death by suppressing the STAT3 transcription factor. Normally activated in response to stress and inflammation, STAT3 is frequently overactivated in human cancers. This overactivation promotes the expression of pro-proliferative and anti-apoptotic genes that drive carcinogenesis. We demonstrate that SESN1&2 inactivation stimulates STAT3 by downregulating the PTPRD phosphatase, a protein responsible for STAT3 dephosphorylation. Our study demonstrates that SESN1&2 deficiency may cause STAT3 activation and facilitate carcinogenesis and drug resistance, making SESN1&2 reactivation a potential cancer treatment strategy.
Collapse
Affiliation(s)
- Alexander Haidurov
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Pearse Street, Dublin 2, Ireland
| | - Andrei O Zheltukhin
- Engelhardt Institute of Molecular Biology, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Moscow, Russia
| | - Anastasiya V Snezhkina
- Engelhardt Institute of Molecular Biology, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Moscow, Russia
| | - George S Krasnov
- Engelhardt Institute of Molecular Biology, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Moscow, Russia
| | - Anna V Kudryavtseva
- Engelhardt Institute of Molecular Biology, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Moscow, Russia
| | - Andrei V Budanov
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Pearse Street, Dublin 2, Ireland.
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia.
| |
Collapse
|
14
|
Sun W, Lin R, Li Y, Yao Y, Lu B, Yu Y. Circulating branched-chain amino acids and the risk of major adverse cardiovascular events in the UK biobank. Front Endocrinol (Lausanne) 2025; 16:1510910. [PMID: 40052157 PMCID: PMC11882422 DOI: 10.3389/fendo.2025.1510910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 01/21/2025] [Indexed: 03/09/2025] Open
Abstract
Objective To investigate the relationship between circulating branched-chain amino acids (BCAAs) and the risk of major adverse cardiovascular events (MACE) in a national population-based cohort study. Methods UK Biobank, a prospective study involving 22 recruitment centers across the United Kingdom. For this analysis, we included 266,840 participants from the UK Biobank who had available BCAA data and no history of MACE at baseline. Cox regression analysis was conducted to evaluate these associations, adjusting for potential confounders. Results During a 13.80 ± 0.83-year follow-up, 52,598 participants experienced MACE, with the incidence of MACE increasing progressively across quintiles of circulating BCAAs, isoleucine, leucine, and valine. Overall, the fifth quintile exhibited a 7-12% higher MACE risk compared to the second quintile. In males, BCAAs were not associated with MACE risk. However, increased risks were observed for isoleucine (8-12% in higher quintiles), leucine (9% in the first quintile and 6% in the fifth quintile), and valine (8% in the first quintile). In females, higher quintiles of BCAAs, isoleucine, leucine, and valine were associated with increased MACE risk, ranging from 9% to 12%. Among participants under 65y, higher quintiles of BCAAs, isoleucine, and leucine were associated with increased MACE risk, while valine showed no significant association. No association was found in participants aged 65 and older. These analyses were adjusted for multiple potential confounders. Conclusion Generally, higher levels of BCAAs, isoleucine, leucine, and valine were associated with an increased risk of MACE, except in participants older than 65. Additionally, in males, the lowest quintiles of leucine and valine were also associated with an increased risk of MACE.
Collapse
Affiliation(s)
- Wanwan Sun
- Department of Endocrinology, Huashan Hospital Affiliated to Fudan University, Shanghai, China
| | - Ruilang Lin
- Department of Biostatistics, School of Public Health, The Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China
| | - Yiming Li
- Department of Endocrinology, Huashan Hospital Affiliated to Fudan University, Shanghai, China
| | - Ye Yao
- Department of Biostatistics, School of Public Health, The Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China
| | - Bin Lu
- Department of Endocrinology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Yongfu Yu
- Department of Biostatistics, School of Public Health, The Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China
| |
Collapse
|
15
|
Wang ZP, You W, Peng J, Xu B, Yang X, Tang W, He Y, Yang A, Yu C, Nian W. Synthesis and structural modification of the natural product Ivesinol to discover novel autophagy activators. Eur J Med Chem 2025; 284:117180. [PMID: 39724726 DOI: 10.1016/j.ejmech.2024.117180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/07/2024] [Accepted: 12/11/2024] [Indexed: 12/28/2024]
Abstract
Autophagy is a lysosome-dependent cellular degradation pathway that responds to a variety of environmental and cellular stresses, which is defective in aging and age-related diseases, therefore, targeting autophagy with small-molecule activators has potential therapeutic benefits. In this study, we successfully completed the first total synthesis of Ivesinol, an identified antibacterial natural product, and efficiently constructed a library of its analogs. To measure the effect of Ivesinol analogs on autophagic activity, we performed cell imaging-based screening approach, and observed that several Ivesinol analogs exhibited potent autophagy-regulating activity. Specifically, the derivative B2 significantly activated autophagy activity in concentration- and time-dependent manners, and even outperformed the commonly used activator Torin1 in activating autophagy in MCF-7 cells at 0.5 μM. Bioinformatics analysis showed that B2 treatment significantly impacted ubiquitin mediated proteolysis and AMPK signaling pathway, with functionally related gene sets displaying strong correlations. Based on these findings, we proposed that B2 activates autophagy by mechanisms involved in downregulation of key HSP70 family members, activation of the UPR, and ultimately leading to autophagy. In conclusion, we suggest that B2 could be a promising and valuable autophagy activator with significant potential for further development.
Collapse
Affiliation(s)
- Zhi-Peng Wang
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Wenxin You
- School of Medicine, Chongqing University, Chongqing, 400030, China
| | - Jie Peng
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Biao Xu
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Xiaohong Yang
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China
| | - Wanyan Tang
- Department of Oncology, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China
| | - Yun He
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, China
| | - Aimin Yang
- School of Life Sciences, Chongqing University, Chongqing, 401331, China.
| | - Chao Yu
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China.
| | - Weiqi Nian
- Department of Oncology, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China.
| |
Collapse
|
16
|
Jie H, Wei J, Li Z, Yi M, Qian X, Li Y, Liu C, Li C, Wang L, Deng P, Liu L, Cen X, Zhao Y. Serine starvation suppresses the progression of esophageal cancer by regulating the synthesis of purine nucleotides and NADPH. Cancer Metab 2025; 13:10. [PMID: 39948566 PMCID: PMC11827256 DOI: 10.1186/s40170-025-00376-4] [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: 03/25/2024] [Accepted: 01/24/2025] [Indexed: 02/16/2025] Open
Abstract
Serine metabolism provides important metabolic intermediates that support the rapid proliferation of tumor cells. However, the role of serine metabolism in esophageal squamous cell carcinoma (ESCC) and the underlying mechanism remains unclear. Here, we show that serine starvation predominantly inhibits ESCC cell proliferation by suppressing purine nucleotides and NADPH synthesis. Mechanistically, serine depletion led to the accumulation of aminoimidazole carboxamide ribonucleoside (AICAR), an intermediate metabolite of de novo purine synthesis, and AMP/ATP ratio. These increases activated 5'-AMP-activated kinase (AMPK), which subsequently inhibited the mTORC1 pathway by phosphorylating Raptor at Ser792. Moreover, serine depletion decreased NADPH level followed by elevated reactive oxygen species (ROS) production and DNA damage, which induced p53-p21 mediated G1 phase cell cycle arrest. Conversely, serine starvation activated transcription factor 4 (ATF4)-mediated robust expression of phosphoserine aminotransferase 1 (PSAT1) which in turn promoted compensatory endogenous serine synthesis, thus maintaining ESCC cell survival under serine-limited conditions. Accordingly, serine deprivation combined with PSAT1 inhibition significantly suppressed ESCC tumor growth both in vitro and in vivo. Taken together, our findings demonstrate that serine starvation suppresses the proliferation of ESCC cells by disturbing the synthesis of purine nucleotides and NADPH, and the combination of serine deprivation and PSAT1 inhibition significantly impairs ESCC tumor growth. Our study provides a theoretical basis for targeting serine metabolism as a potential therapeutic strategy for ESCC.
Collapse
Affiliation(s)
- Hui Jie
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jing Wei
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhuoling Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Min Yi
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xinying Qian
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yan Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chunqi Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chuan Li
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Liang Wang
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Pengchi Deng
- Analytical & Testing Center, Sichuan University, Chengdu, 610041, China
| | - Lunxu Liu
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaobo Cen
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yinglan Zhao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
17
|
Pan J, Lin Y, Liu X, Zhang X, Liang T, Bai X. Harnessing amino acid pathways to influence myeloid cell function in tumor immunity. Mol Med 2025; 31:44. [PMID: 39905317 PMCID: PMC11796060 DOI: 10.1186/s10020-025-01099-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 01/21/2025] [Indexed: 02/06/2025] Open
Abstract
Amino acids are pivotal regulators of immune cell metabolism, signaling pathways, and gene expression. In myeloid cells, these processes underlie their functional plasticity, enabling shifts between pro-inflammatory, anti-inflammatory, pro-tumor, and anti-tumor activities. Within the tumor microenvironment, amino acid metabolism plays a crucial role in mediating the immunosuppressive functions of myeloid cells, contributing to tumor progression. This review delves into the mechanisms by which specific amino acids-glutamine, serine, arginine, and tryptophan-regulate myeloid cell function and polarization. Furthermore, we explore the therapeutic potential of targeting amino acid metabolism to enhance anti-tumor immunity, offering insights into novel strategies for cancer treatment.
Collapse
Affiliation(s)
- Jiongli Pan
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yi Lin
- Health Science Center, Ningbo University, Ningbo, China
| | - Xinyuan Liu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaozhen Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xueli Bai
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
18
|
Shafer M, Low V, Li Z, Blenis J. The emerging role of dysregulated propionate metabolism and methylmalonic acid in metabolic disease, aging, and cancer. Cell Metab 2025; 37:316-329. [PMID: 39908986 PMCID: PMC11984558 DOI: 10.1016/j.cmet.2025.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 10/10/2024] [Accepted: 01/08/2025] [Indexed: 02/07/2025]
Abstract
Propionate metabolism dysregulation has emerged as a source of metabolic health alterations linked to aging, cardiovascular and renal diseases, obesity and diabetes, and cancer. This is supported by several large cohort population studies and recent work revealing its role in cancer progression. Mutations in several enzymes of this metabolic pathway are associated with devastating inborn errors of metabolism, resulting in severe methylmalonic and propionic acidemias. Beyond these rare diseases, however, the broader pathological significance of propionate metabolism and its metabolites has been largely overlooked. Here, we summarize earlier studies and new evidence that the alteration of this pathway and associated metabolites are involved in the development of various metabolic diseases and link aging to cancer progression and metastasis.
Collapse
Affiliation(s)
- Moniquetta Shafer
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA; Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Vivien Low
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zhongchi Li
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA; Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA.
| | - John Blenis
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA; Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
19
|
Gong Q, Wang J, Luo D, Xu Y, Zhang R, Li X, Yin Z, Fang J, Wang H. Accumulation of branched-chain amino acids deteriorates the neuroinflammatory response of Müller cells in diabetic retinopathy via leucine/Sestrin2-mediated sensing of mTOR signaling. Acta Diabetol 2025; 62:227-240. [PMID: 39150511 PMCID: PMC11861416 DOI: 10.1007/s00592-024-02349-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 07/22/2024] [Indexed: 08/17/2024]
Abstract
AIMS This study aimed to investigate branched-chain amino acid (BCAA) catabolism in diabetic retinopathy (DR). METHODS Wild-type and db/db mice were fed BCAAs (5 or 10 mg/kg/day) for 12 weeks, and hyperglycemia-exposed Müller cells were treated with BCAAs (2 or 5 mmol/L) for 24 and 48 h. BCAA levels were measured using MS/MS. Western blotting was performed to detect proteins. Flow cytometry, oxygen consumption rate, and Cell Counting Kit-8 assays were used to evaluate Müller cell viability. Each experiment was conducted at least thrice. RESULTS BCAAs and branched-chain α-keto acids (BCKAs) were increased in the retina and systemic tissues of diabetic mice, and these changes were further enhanced to approximately 2-fold by extra BCAAs compared to wild-type group. In vitro, BCAAs and BCKAs were induced in hyperglycemic Müller cells, and augmented by BCAA supplementation. The aberrant BCAA catabolism was accompanied by mTORC1 activation and subsequently induced TNF-ɑ, VEGFA, GS, and GFAP in retinas and Müller cells under diabetic conditions. The cell apoptosis rate increased by approximately 50%, and mitochondrial respiration was inhibited by hyperglycemia and BCAA in Müller cells. Additionally, mTORC1 signaling was activated by leucine in Müller cells. Knockdown of Sestrin2 or LeuRS significantly abolished the leucine-induced mTORC1 phosphorylation and protected Müller cell viability under diabetic conditions. CONCLUSIONS We found that BCAA catabolism is hindered in DR through mTORC1 activation. Leucine plays a key role in inducing mTORC1 by sensing Sestrin2 in Müller cells. Targeting Sestrin2 may ameliorate the toxic effects of BCAA accumulation on Müller cells in DR.
Collapse
Affiliation(s)
- Qiaoyun Gong
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
- National Clinical Research Center for Eye Diseases, No. 100 Haining Road, Hongkou District, Shanghai, 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Disease, Shanghai, China
| | - Jingyi Wang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
- National Clinical Research Center for Eye Diseases, No. 100 Haining Road, Hongkou District, Shanghai, 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Disease, Shanghai, China
| | - Dawei Luo
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
- National Clinical Research Center for Eye Diseases, No. 100 Haining Road, Hongkou District, Shanghai, 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Disease, Shanghai, China
| | - Yupeng Xu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
- National Clinical Research Center for Eye Diseases, No. 100 Haining Road, Hongkou District, Shanghai, 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Disease, Shanghai, China
| | - Rulin Zhang
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai, China
| | - Xin Li
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Zihan Yin
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
- National Clinical Research Center for Eye Diseases, No. 100 Haining Road, Hongkou District, Shanghai, 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Disease, Shanghai, China
| | - Junwei Fang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
- National Clinical Research Center for Eye Diseases, No. 100 Haining Road, Hongkou District, Shanghai, 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Disease, Shanghai, China
| | - Haiyan Wang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China.
- National Clinical Research Center for Eye Diseases, No. 100 Haining Road, Hongkou District, Shanghai, 200080, China.
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China.
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China.
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Disease, Shanghai, China.
| |
Collapse
|
20
|
Mansoori S, Ho MY, Ng KK, Cheng KK. Branched-chain amino acid metabolism: Pathophysiological mechanism and therapeutic intervention in metabolic diseases. Obes Rev 2025; 26:e13856. [PMID: 39455059 PMCID: PMC11711082 DOI: 10.1111/obr.13856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 09/02/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024]
Abstract
Branched-chain amino acids (BCAAs), including leucine, isoleucine, and valine, are essential for maintaining physiological functions and metabolic homeostasis. However, chronic elevation of BCAAs causes metabolic diseases such as obesity, type 2 diabetes (T2D), and metabolic-associated fatty liver disease (MAFLD). Adipose tissue, skeletal muscle, and the liver are the three major metabolic tissues not only responsible for controlling glucose, lipid, and energy balance but also for maintaining BCAA homeostasis. Under obese and diabetic conditions, different pathogenic factors like pro-inflammatory cytokines, lipotoxicity, and reduction of adiponectin and peroxisome proliferator-activated receptors γ (PPARγ) disrupt BCAA metabolism, leading to excessive accumulation of BCAAs and their downstream metabolites in metabolic tissues and circulation. Mechanistically, BCAAs and/or their downstream metabolites, such as branched-chain ketoacids (BCKAs) and 3-hydroxyisobutyrate (3-HIB), impair insulin signaling, inhibit adipogenesis, induce inflammatory responses, and cause lipotoxicity in the metabolic tissues, resulting in multiple metabolic disorders. In this review, we summarize the latest studies on the metabolic regulation of BCAA homeostasis by the three major metabolic tissues-adipose tissue, skeletal muscle, and liver-and how dysregulated BCAA metabolism affects glucose, lipid, and energy balance in these active metabolic tissues. We also summarize therapeutic approaches to restore normal BCAA metabolism as a treatment for metabolic diseases.
Collapse
Affiliation(s)
- Shama Mansoori
- Department of Health Technology and InformaticsThe Hong Kong Polytechnic UniversityHong Kong, China
| | - Melody Yuen‐man Ho
- Department of Health Technology and InformaticsThe Hong Kong Polytechnic UniversityHong Kong, China
| | - Kelvin Kwun‐wang Ng
- Department of Health Technology and InformaticsThe Hong Kong Polytechnic UniversityHong Kong, China
| | - Kenneth King‐yip Cheng
- Department of Health Technology and InformaticsThe Hong Kong Polytechnic UniversityHong Kong, China
- Hong Kong Polytechnic University Shenzhen Research InstituteShenzhenChina
| |
Collapse
|
21
|
Yan Z, Luan Y, Wang Y, Ren Y, Li Z, Zhao L, Shen L, Yang X, Liu T, Gao Y, Sun W. Constructing a Novel Amino Acid Metabolism Signature: A New Perspective on Pheochromocytoma Diagnosis, Immune Landscape, and Immunotherapy. Biochem Genet 2025; 63:850-874. [PMID: 38526709 PMCID: PMC11832799 DOI: 10.1007/s10528-024-10733-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 02/05/2024] [Indexed: 03/27/2024]
Abstract
Pheochromocytoma/paraganglioma (PGPG) is a rare neuroendocrine tumor. Amino acid metabolism is crucial for energy production, redox balance, and metabolic pathways in tumor cell proliferation. This study aimed to build a risk model using amino acid metabolism-related genes, enhancing PGPG diagnosis and treatment decisions. We analyzed RNA-sequencing data from the PCPG cohort in the GEO dataset as our training set and validated our findings using the TCGA dataset and an additional clinical cohort. WGCNA and LASSO were utilized to identify hub genes and develop risk prediction models. The single-sample gene set enrichment analysis, MCPCOUNTER, and ESTIMATE algorithm calculated the relationship between amino acid metabolism and immune cell infiltration in PCPG. The TIDE algorithm predicted the immunotherapy efficacy for PCPG patients. The analysis identified 292 genes with differential expression, which are involved in amino acid metabolism and immune pathways. Six genes (DDC, SYT11, GCLM, PSMB7, TYRO3, AGMAT) were identified as crucial for the risk prediction model. Patients with a high-risk profile demonstrated reduced immune infiltration but potentially higher benefits from immunotherapy. Notably, DDC and SYT11 showed strong diagnostic and prognostic potential. Validation through quantitative Real-Time Polymerase Chain Reaction and immunohistochemistry confirmed their differential expression, underscoring their significance in PCPG diagnosis and in predicting immunotherapy response. This study's integration of amino acid metabolism-related genes into a risk prediction model offers critical clinical insights for PCPG risk stratification, potential immunotherapy responses, drug development, and treatment planning, marking a significant step forward in the management of this complex condition.
Collapse
Affiliation(s)
- Zechen Yan
- BGI College and Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
- Department of Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
- Henan Engineering Research Center of Tumor Molecular Diagnosis and Treatment, Zhengzhou, 450001, Henan, People's Republic of China
- Institute of Molecular Cancer Surgery, Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
| | - Yongkun Luan
- BGI College and Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
- Department of Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
- Henan Engineering Research Center of Tumor Molecular Diagnosis and Treatment, Zhengzhou, 450001, Henan, People's Republic of China
- Institute of Molecular Cancer Surgery, Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
| | - Yu Wang
- BGI College and Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
- Department of Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
- Institute of Molecular Cancer Surgery, Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
| | - Yilin Ren
- Department of Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
- Henan Engineering Research Center of Tumor Molecular Diagnosis and Treatment, Zhengzhou, 450001, Henan, People's Republic of China
| | - Zhiyuan Li
- Department of Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
- Henan Engineering Research Center of Tumor Molecular Diagnosis and Treatment, Zhengzhou, 450001, Henan, People's Republic of China
| | - Luyang Zhao
- BGI College and Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
- Department of Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
- Henan Engineering Research Center of Tumor Molecular Diagnosis and Treatment, Zhengzhou, 450001, Henan, People's Republic of China
- Institute of Molecular Cancer Surgery, Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
| | - Linnuo Shen
- Department of Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
- Henan Engineering Research Center of Tumor Molecular Diagnosis and Treatment, Zhengzhou, 450001, Henan, People's Republic of China
- Institute of Molecular Cancer Surgery, Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
| | - Xiaojie Yang
- Department of Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
- Henan Engineering Research Center of Tumor Molecular Diagnosis and Treatment, Zhengzhou, 450001, Henan, People's Republic of China
- Institute of Molecular Cancer Surgery, Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
| | - Tonghu Liu
- Department of Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China.
- Henan Engineering Research Center of Tumor Molecular Diagnosis and Treatment, Zhengzhou, 450001, Henan, People's Republic of China.
- Institute of Molecular Cancer Surgery, Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China.
| | - Yukui Gao
- Department of Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China.
- Institute of Molecular Cancer Surgery, Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China.
| | - Weibo Sun
- Department of Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China.
- Institute of Molecular Cancer Surgery, Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China.
- Department of Radiation Oncology and Oncology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, 450000, China.
| |
Collapse
|
22
|
Yao P, Cao S, Zhu Z, Wen Y, Guo Y, Liang W, Xie J. Cellular Signaling of Amino Acid Metabolism in Prostate Cancer. Int J Mol Sci 2025; 26:776. [PMID: 39859489 PMCID: PMC11765784 DOI: 10.3390/ijms26020776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 01/14/2025] [Accepted: 01/15/2025] [Indexed: 01/30/2025] Open
Abstract
Prostate cancer is one of the most common malignancies affecting men worldwide and a leading cause of cancer-related mortality, necessitating a deeper understanding of its underlying biochemical pathways. Similar to other cancer types, prostate cancer is also characterised by aberrantly activated metabolic pathways that support tumour development, such as amino acid metabolism, which is involved in modulating key physiological and pathological cellular processes during the progression of this disease. The metabolism of several amino acids, such as glutamine and methionine, crucial for tumorigenesis, is dysregulated and commonly discussed in prostate cancer. And the roles of some less studied amino acids, such as histidine and glycine, have also been covered in prostate cancer studies. Aberrant regulation of two major signalling pathways, mechanistic target of rapamycin (mTOR) and general amino acid control non-depressible 2 (GCN2), is a key driver of reshaping the amino acid metabolism landscape in prostate cancer. By summarising our current understanding of how amino acid metabolism is modulated in prostate cancer, here, we provide further insights into certain potential therapeutic targets for managing prostate cancer through metabolic interventions.
Collapse
Affiliation(s)
- Ping Yao
- School of Biology and Biological Engineering, South China University of Technology, University Town, Guangzhou 510006, China
| | - Shiqi Cao
- School of Biology and Biological Engineering, South China University of Technology, University Town, Guangzhou 510006, China
| | - Ziang Zhu
- School of Biology and Biological Engineering, South China University of Technology, University Town, Guangzhou 510006, China
| | - Yunru Wen
- School of Biology and Biological Engineering, South China University of Technology, University Town, Guangzhou 510006, China
| | - Yawen Guo
- School of Biology and Biological Engineering, South China University of Technology, University Town, Guangzhou 510006, China
| | - Wenken Liang
- School of Biology and Biological Engineering, South China University of Technology, University Town, Guangzhou 510006, China
| | - Jianling Xie
- School of Biology and Biological Engineering, South China University of Technology, University Town, Guangzhou 510006, China
- Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA 5042, Australia
| |
Collapse
|
23
|
He L, Cho S, Blenis J. mTORC1, the maestro of cell metabolism and growth. Genes Dev 2025; 39:109-131. [PMID: 39572234 PMCID: PMC11789495 DOI: 10.1101/gad.352084.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
The mechanistic target of rapamycin (mTOR) pathway senses and integrates various environmental and intracellular cues to regulate cell growth and proliferation. As a key conductor of the balance between anabolic and catabolic processes, mTOR complex 1 (mTORC1) orchestrates the symphonic regulation of glycolysis, nucleic acid and lipid metabolism, protein translation and degradation, and gene expression. Dysregulation of the mTOR pathway is linked to numerous human diseases, including cancer, neurodegenerative disorders, obesity, diabetes, and aging. This review provides an in-depth understanding of how nutrients and growth signals are coordinated to influence mTOR signaling and the extensive metabolic rewiring under its command. Additionally, we discuss the use of mTORC1 inhibitors in various aging-associated metabolic diseases and the current and future potential for targeting mTOR in clinical settings. By deciphering the complex landscape of mTORC1 signaling, this review aims to inform novel therapeutic strategies and provide a road map for future research endeavors in this dynamic and rapidly evolving field.
Collapse
Affiliation(s)
- Long He
- Meyer Cancer Center, Weill Cornell Medicine, New York, New York 10021, USA;
- Department of Pharmacology, Weill Cornell Medicine, New York, New York 10021, USA
| | - Sungyun Cho
- Meyer Cancer Center, Weill Cornell Medicine, New York, New York 10021, USA
- Department of Pharmacology, Weill Cornell Medicine, New York, New York 10021, USA
| | - John Blenis
- Meyer Cancer Center, Weill Cornell Medicine, New York, New York 10021, USA;
- Department of Pharmacology, Weill Cornell Medicine, New York, New York 10021, USA
| |
Collapse
|
24
|
Fang Z, Ren G, Ke S, Xu Q, Chen Y, Shi X, Guo C, Huang J. Serum metabolomic profiling for predicting therapeutic response and toxicity in breast cancer neoadjuvant chemotherapy: a retrospective longitudinal study. Breast Cancer Res 2025; 27:2. [PMID: 39762945 PMCID: PMC11706045 DOI: 10.1186/s13058-024-01956-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Neoadjuvant chemotherapy (NACT) is the standard-of-care treatment for patients with locally advanced breast cancer (LABC), providing crucial benefits in tumor downstaging. Clinical parameters, such as molecular subtypes, influence the therapeutic impact of NACT. Moreover, severe adverse events delay the treatment process and reduce the effectiveness of therapy. Although metabolic changes during cancer treatment are crucial determinant factors in therapeutic responses and toxicities, related clinical research remains limited. METHODS One hundred paired blood samples were collected from 50 patients with LABC before and after a complete NACT treatment cycle. Untargeted metabolomics was used by liquid chromatography-mass spectrometry (LC-MS) to investigate the relationship between dynamically changing metabolites in serum and the responses and toxicities of NACT. RESULTS Firstly, we observed significant alterations in serum metabolite levels pre- and post-NACT, with a predominant enrichment in the sphingolipid and amino acid metabolism pathways. Second, pre-treatment serum metabolites successfully predicted the therapeutic response and hematotoxicities during NACT. In particular, molecular subtype variations in favorable treatment responses are linked to acyl carnitine levels. Finally, we discovered that the therapeutic effects of NACT could be attributed to essential amino acid metabolism. CONCLUSION This study elucidated the dynamic changes in metabolism during NACT treatment, providing a possibility for developing responsive metabolic signatures for personalized NACT treatment.
Collapse
Affiliation(s)
- Zhihao Fang
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Jiefang Road, Hangzhou, Zhejiang, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Guohong Ren
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Jiefang Road, Hangzhou, Zhejiang, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shouyu Ke
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Jiefang Road, Hangzhou, Zhejiang, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qimin Xu
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Jiefang Road, Hangzhou, Zhejiang, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yuhua Chen
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiaoyuan Shi
- Analytical Instrument Trading Co., Ltd, SCIEX, Shanghai, China
| | - Cheng Guo
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Jian Huang
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Jiefang Road, Hangzhou, Zhejiang, China.
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
25
|
Soengas JL, Comesaña S, Blanco AM, Conde-Sieira M. Feed Intake Regulation in Fish: Implications for Aquaculture. REVIEWS IN FISHERIES SCIENCE & AQUACULTURE 2025; 33:8-60. [DOI: 10.1080/23308249.2024.2374259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Affiliation(s)
- José L. Soengas
- Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía, Laboratorio de Fisioloxía Animal, Centro de Investigación Mariña, Universidade de Vigo, Vigo, Spain
| | - Sara Comesaña
- Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía, Laboratorio de Fisioloxía Animal, Centro de Investigación Mariña, Universidade de Vigo, Vigo, Spain
| | - Ayelén M. Blanco
- Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía, Laboratorio de Fisioloxía Animal, Centro de Investigación Mariña, Universidade de Vigo, Vigo, Spain
| | - Marta Conde-Sieira
- Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía, Laboratorio de Fisioloxía Animal, Centro de Investigación Mariña, Universidade de Vigo, Vigo, Spain
| |
Collapse
|
26
|
Jin J, Meng T, Yu Y, Wu S, Jiao CC, Song S, Li YX, Zhang Y, Zhao YY, Li X, Wang Z, Liu YF, Huang R, Qin J, Chen Y, Cao H, Tan X, Ge X, Jiang C, Xue J, Yuan J, Wu D, Wu W, Jiang CZ, Wang P. Human HDAC6 senses valine abundancy to regulate DNA damage. Nature 2025; 637:215-223. [PMID: 39567688 DOI: 10.1038/s41586-024-08248-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 10/17/2024] [Indexed: 11/22/2024]
Abstract
As an essential branched amino acid, valine is pivotal for protein synthesis, neurological behaviour, haematopoiesis and leukaemia progression1-3. However, the mechanism by which cellular valine abundancy is sensed for subsequent cellular functions remains undefined. Here we identify that human histone deacetylase 6 (HDAC6) serves as a valine sensor by directly binding valine through a primate-specific SE14 repeat domain. The nucleus and cytoplasm shuttling of human, but not mouse, HDAC6 is tightly controlled by the intracellular levels of valine. Valine deprivation leads to HDAC6 retention in the nucleus and induces DNA damage. Mechanistically, nuclear-localized HDAC6 binds and deacetylates ten-eleven translocation 2 (TET2) to initiate active DNA demethylation, which promotes DNA damage through thymine DNA glycosylase-driven excision. Dietary valine restriction inhibits tumour growth in xenograft and patient-derived xenograft models, and enhances the therapeutic efficacy of PARP inhibitors. Collectively, our study identifies human HDAC6 as a valine sensor that mediates active DNA demethylation and DNA damage in response to valine deprivation, and highlights the potential of dietary valine restriction for cancer treatment.
Collapse
Affiliation(s)
- Jiali Jin
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Tong Meng
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yuanyuan Yu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Shuheng Wu
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Chen-Chen Jiao
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Sihui Song
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ya-Xu Li
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yu Zhang
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuan-Yuan Zhao
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xinran Li
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zixin Wang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Tongji Hospital affiliated to Tongji University, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yu-Fan Liu
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Runzhi Huang
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jieling Qin
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yihua Chen
- Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products and Yunnan College of Modern Biomedical Industry, Kunming Medical University, Kunming, China
| | - Hao Cao
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| | - Xiao Tan
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xin Ge
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Cong Jiang
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jianhuang Xue
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Tongji Hospital affiliated to Tongji University, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Jian Yuan
- Department of Biochemistry and Molecular Biology, Tongji University School of Medicine, Shanghai, China
| | - Dianqing Wu
- Department of Pharmacology, Yale School of Medicine, New Haven, CT, USA
| | - Wei Wu
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Ci-Zhong Jiang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Ping Wang
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
27
|
Gu X. Determination of Nutrient Ligand-Sensor Binding Affinity. Methods Mol Biol 2025; 2882:163-178. [PMID: 39992509 DOI: 10.1007/978-1-0716-4284-9_8] [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] [Indexed: 02/25/2025]
Abstract
Cells contain dedicated mechanisms to sense nutrient levels in the environment to regulate their growth by balancing anabolism and catabolism [1, 2]. The mechanistic Target of Rapamycin Complex 1 (mTORC1), a multi-protein kinase complex, serves as an essential growth regulator that integrates various upstream inputs including growth factors and nutrients like amino acids [1, 2] Nutrient sensors upstream of mTORC1 directly bind cognate nutrient ligands to convey their availability and thereby regulate mTORC1 signaling [1, 2]. A reliable method is needed to quantitatively determine the binding affinity (Kd) of the nutrient sensor to its ligand. In parallel, quantitative metabolomic analysis can reveal metabolite levels in fed and starved cells; which represent the physiological range of the nutrient of interest. Whether or not the binding affinity is within the physiological range serves as an indicator to determine the physiological relevance of the sensing mechanism. This chapter describes a generalizable protocol that allows reproducible determination of nutrient ligand-nutrient sensor binding affinity. Here, the S-adenosylmethionine (nutrient ligand)-SAMTOR (nutrient sensor) pair is used as an example [3]. Nutrient sensor purification, radioactive nutrient ligand incubation, and eventual scintillation counting are included, along with a description of the mathematical equation that is used to calculate the binding affinity.
Collapse
Affiliation(s)
- Xin Gu
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
28
|
DiMartino S, Revelo MP, Mallipattu SK, Piret SE. Activation of branched chain amino acid catabolism protects against nephrotoxic acute kidney injury. Am J Physiol Renal Physiol 2025; 328:F152-F163. [PMID: 39653371 PMCID: PMC11918290 DOI: 10.1152/ajprenal.00260.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 11/18/2024] [Accepted: 11/27/2024] [Indexed: 01/11/2025] Open
Abstract
Acute kidney injury (AKI) is a major risk factor for chronic kidney disease (CKD), and there are currently no therapies for AKI. Proximal tubules (PTs) are particularly susceptible to AKI, due to nephrotoxins such as aristolochic acid I (AAI). Normal PTs use fatty acid oxidation and branched chain amino acid (BCAA; valine, leucine, and isoleucine) catabolism to generate ATP; however, in AKI, these pathways are downregulated. Our aim was to investigate the utility of a pharmacological activator of BCAA catabolism, BT2, in preventing nephrotoxic AKI. Mice were administered two injections of AAI 3 days apart to induce AKI, with or without daily BT2 treatment. Mice treated with BT2 had significantly protected kidney function (reduced serum creatinine and urea nitrogen), reduced histological injury, preservation of PT (Lotus lectin staining), and less PT injury (cytokeratin-20 staining) and inflammatory gene expression compared with mice with AAI alone. Mice with AKI had increased circulating BCAA and accumulation of BCAA in the kidney cortex. Leucine is a potent activator of the mechanistic target of rapamycin complex 1 (mTORC1) signaling, and mTORC1 signaling was activated in mice treated with AAI. However, BT2 reduced kidney cortical BCAA accumulation and attenuated the mTORC1 signaling. In vitro, injured primary PT cells had compromised mitochondrial bioenergetics, but cells treated with AAI + BT2 had partially restored mitochondrial bioenergetics and improved injury markers compared with cells treated with AAI alone. Thus, pharmacological activation of BCAA catabolism using BT2 attenuated nephrotoxic AKI in mice.NEW & NOTEWORTHY This study explored the effects of pharmacological activation of branched chain amino acid (BCAA) catabolism using BT2 to prevent nephrotoxic acute kidney injury (AKI) in mice. Our results indicate that activation of BCAA catabolism protects against nephrotoxic AKI, in association with reduced BCAA accumulation, reduced mammalian target of rapamycin protein complex 1 signaling, and improved mitochondrial bioenergetics.
Collapse
Affiliation(s)
- Samaneh DiMartino
- Division of Nephrology and Hypertension, Department of Medicine, Stony Brook University, Stony Brook, New York, United States
| | - Monica P Revelo
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States
| | - Sandeep K Mallipattu
- Division of Nephrology and Hypertension, Department of Medicine, Stony Brook University, Stony Brook, New York, United States
- Renal Section, Northport VA Medical Center, Northport, New York, United States
| | - Sian E Piret
- Division of Nephrology and Hypertension, Department of Medicine, Stony Brook University, Stony Brook, New York, United States
| |
Collapse
|
29
|
Bo T, Fujii J. Primary Roles of Branched Chain Amino Acids (BCAAs) and Their Metabolism in Physiology and Metabolic Disorders. Molecules 2024; 30:56. [PMID: 39795113 PMCID: PMC11721030 DOI: 10.3390/molecules30010056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/25/2024] [Accepted: 12/25/2024] [Indexed: 01/13/2025] Open
Abstract
Leucine, isoleucine, and valine are collectively known as branched chain amino acids (BCAAs) and are often discussed in the same physiological and pathological situations. The two consecutive initial reactions of BCAA catabolism are catalyzed by the common enzymes referred to as branched chain aminotransferase (BCAT) and branched chain α-keto acid dehydrogenase (BCKDH). BCAT transfers the amino group of BCAAs to 2-ketoglutarate, which results in corresponding branched chain 2-keto acids (BCKAs) and glutamate. BCKDH performs an oxidative decarboxylation of BCKAs, which produces their coenzyme A-conjugates and NADH. BCAT2 in skeletal muscle dominantly catalyzes the transamination of BCAAs. Low BCAT activity in the liver reduces the metabolization of BCAAs, but the abundant presence of BCKDH promotes the metabolism of muscle-derived BCKAs, which leads to the production of glucose and ketone bodies. While mutations in the genes responsible for BCAA catabolism are involved in rare inherited disorders, an aberrant regulation of their enzymatic activities is associated with major metabolic disorders such as diabetes, cardiovascular disease, and cancer. Therefore, an understanding of the regulatory process of metabolic enzymes, as well as the functions of the BCAAs and their metabolites, make a significant contribution to our health.
Collapse
Affiliation(s)
- Tomoki Bo
- Laboratory Animal Center, Institute for Promotion of Medical Science Research, Faculty of Medicine, Yamagata University, Yamagata 990-9585, Japan
| | - Junichi Fujii
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, Yamagata 990-9585, Japan
| |
Collapse
|
30
|
Tian K, Yao Z, Pan D. Leveraging single-cell and multi-omics approaches to identify MTOR-centered deubiquitination signatures in esophageal cancer therapy. Front Immunol 2024; 15:1490623. [PMID: 39742278 PMCID: PMC11685190 DOI: 10.3389/fimmu.2024.1490623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 11/28/2024] [Indexed: 01/03/2025] Open
Abstract
Background Esophageal squamous cell carcinoma (ESCC) remains a significant challenge in oncology due to its aggressive nature and heterogeneity. As one of the deadliest malignancies, ESCC research lags behind other cancer types. The balance between ubiquitination and deubiquitination processes plays a crucial role in cellular functions, with its disruption linked to various diseases, including cancer. Methods Our study utilized diverse analytical approaches, encompassing Cox regression models, single-cell RNA sequencing, intercellular communication analysis, and Gene Ontology enrichment. We also conducted mutation profiling and explored potential immunotherapeutic agents. Furthermore, in vitro cellular experiments and in vivo mouse models were performed to validate findings. These methodologies aimed to establish deubiquitination-related gene signatures (DRGS) for predicting ESCC patient outcomes and response to immunotherapy. Results By integrating datasets from TCGA-ESCC and GSE53624, we developed a DRGS model based on 14 deubiquitination-related genes (DUBGs). This signature effectively forecasts ESCC prognosis, drug responsiveness, and immune cell infiltration patterns. It also influences the mutational landscape of patients. Those classified as high-risk exhibited reduced survival rates, increased genetic alterations, and more complex cellular interactions, potentially explaining their poor outcomes. Notably, in vitro and in vivo experiments identified MTOR, a key component of the signature, as a promising therapeutic target for ESCC treatment. Conclusion Our research highlights the significance of 14 DUBGs in ESCC progression. The risk score derived from this gene set enables clinical stratification of patients into distinct prognostic groups. Moreover, MTOR emerges as a potential target for personalized ESCC therapy, offering new avenues for treatment strategies.
Collapse
Affiliation(s)
- Kang Tian
- Department of Oncology, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, China
| | - Ziang Yao
- Department of Traditional Chinese Medicine, Peking University People’s Hospital, Beijing, China
| | - Da Pan
- Department of Gastroenterology, Wenzhou Central Hospital, Wenzhou, China
| |
Collapse
|
31
|
Hong J, Liu W, Xiao X, Gajendran B, Ben-David Y. Targeting pivotal amino acids metabolism for treatment of leukemia. Heliyon 2024; 10:e40492. [PMID: 39654725 PMCID: PMC11626780 DOI: 10.1016/j.heliyon.2024.e40492] [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: 06/27/2024] [Revised: 11/12/2024] [Accepted: 11/15/2024] [Indexed: 12/12/2024] Open
Abstract
Metabolic reprogramming is a crucial characteristic of cancer, allowing cancer cells to acquire metabolic properties that support their survival, immune evasion, and uncontrolled proliferation. Consequently, targeting cancer metabolism has become an essential therapeutic strategy. Abnormal amino acid metabolism is not only a key aspect of metabolic reprogramming but also plays a significant role in chemotherapy resistance and immune evasion, particularly in leukemia. Changes in amino acid metabolism in tumor cells are typically driven by a combination of signaling pathways and transcription factors. Current approaches to targeting amino acid metabolism in leukemia include inhibiting amino acid transporters, blocking amino acid biosynthesis, and depleting specific amino acids to induce apoptosis in leukemic cells. Different types of leukemic cells rely on the exogenous supply of specific amino acids, such as asparagine, glutamine, arginine, and tryptophan. Therefore, disrupting the supply of these amino acids may represent a vulnerability in leukemia. This review focuses on the pivotal role of amino acids in leukemia metabolism, their impact on leukemic stem cells, and their therapeutic potential.
Collapse
Affiliation(s)
- Jiankun Hong
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guian New Disctrict, 561113, Guizhou, PR China
- Natural Products Research Center of Guizhou. PR China
| | - Wuling Liu
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guian New Disctrict, 561113, Guizhou, PR China
- Natural Products Research Center of Guizhou. PR China
| | - Xiao Xiao
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guian New Disctrict, 561113, Guizhou, PR China
- Natural Products Research Center of Guizhou. PR China
| | - Babu Gajendran
- Institute of Pharmacology and Biological Activity, Natural Products Research Center of Guizhou Province, Guiyang, Guizhou, 550014, PR China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, 550025, Guizhou Province, PR China
| | - Yaacov Ben-David
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guian New Disctrict, 561113, Guizhou, PR China
- Natural Products Research Center of Guizhou. PR China
| |
Collapse
|
32
|
Kochavi A, Nagel R, Körner PR, Bleijerveld OB, Lin CP, Huinen Z, Malka Y, Proost N, van de Ven M, Feng X, Navarro JM, Pataskar A, Peeper DS, Champagne J, Agami R. Chemotherapeutic agents and leucine deprivation induce codon-biased aberrant protein production in cancer. Nucleic Acids Res 2024; 52:13964-13979. [PMID: 39588782 DOI: 10.1093/nar/gkae1110] [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: 07/29/2024] [Revised: 10/01/2024] [Accepted: 11/25/2024] [Indexed: 11/27/2024] Open
Abstract
Messenger RNA (mRNA) translation is a tightly controlled process frequently deregulated in cancer. Key to this deregulation are transfer RNAs (tRNAs), whose expression, processing and post-transcriptional modifications are often altered in cancer to support cellular transformation. In conditions of limiting levels of amino acids, this deregulated control of protein synthesis leads to aberrant protein production in the form of ribosomal frameshifting or misincorporation of non-cognate amino acids. Here, we studied leucine, an essential amino acid coded by six different codons. Surprisingly, we found that leucine deprivation leads to ribosomal stalling and aberrant protein production in various cancer cell types, predominantly at one codon, UUA. Similar effects were observed after treatment with chemotherapeutic agents, implying a shared mechanism controlling the downstream effects on mRNA translation. In both conditions, a limitation in the availability of tRNALeu(UAA) for protein production was shown to be the cause for this dominant effect on UUA codons. The induced aberrant proteins can be processed and immune-presented as neoepitopes and can direct T-cell killing. Altogether, we uncovered a novel mode of interplay between DNA damage, regulation of tRNA availability for mRNA translation and aberrant protein production in cancer that could be exploited for anti-cancer therapy.
Collapse
Affiliation(s)
- Adva Kochavi
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, the Netherlands
| | - Remco Nagel
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, the Netherlands
| | - Pierre-Rene Körner
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, the Netherlands
| | - Onno B Bleijerveld
- NKI Proteomics Facility, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | - Chun-Pu Lin
- Division of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | - Zowi Huinen
- Division of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | - Yuval Malka
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, the Netherlands
| | - Natalie Proost
- Preclinical Intervention Unit and Pharmacology Unit of the Mouse Clinic for Cancer and Ageing (MCCA), The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | - Marieke van de Ven
- Preclinical Intervention Unit and Pharmacology Unit of the Mouse Clinic for Cancer and Ageing (MCCA), The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | - Xiaodong Feng
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, the Netherlands
| | - Jasmine Montenegro Navarro
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, the Netherlands
| | - Abhijeet Pataskar
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, the Netherlands
| | - Daniel S Peeper
- Division of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | - Julien Champagne
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, the Netherlands
| | - Reuven Agami
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, the Netherlands
- Erasmus MC, Rotterdam University, Dr. Molewaterplein 40, 3015GD, Rotterdam, the Netherlands
| |
Collapse
|
33
|
Galhuber M, Thedieck K. ODE-based models of signaling networks in autophagy. CURRENT OPINION IN SYSTEMS BIOLOGY 2024; 39:100519. [DOI: 10.1016/j.coisb.2024.100519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
34
|
Tucker SK, Eberhart JK. The convergence of mTOR signaling and ethanol teratogenesis. Reprod Toxicol 2024; 130:108720. [PMID: 39306261 DOI: 10.1016/j.reprotox.2024.108720] [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: 07/01/2024] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 10/04/2024]
Abstract
Ethanol is one of the most common teratogens and causes of human developmental disabilities. Fetal alcohol spectrum disorders (FASD), which describes the wide range of deficits due to prenatal ethanol exposure, are estimated to affect between 1.1 % and 5.0 % of births in the United States. Ethanol dysregulates numerous cellular mechanisms such as programmed cell death (apoptosis), protein synthesis, autophagy, and various aspects of cell signaling, all of which contribute to FASD. The mechanistic target of rapamycin (mTOR) regulates these cellular mechanisms via sensing of nutrients like amino acids and glucose, DNA damage, and growth factor signaling. Despite an extensive literature on ethanol teratogenesis and mTOR signaling, there has been less attention paid to their interaction. Here, we discuss the impact of ethanol teratogenesis on mTORC1's ability to coordinate growth factor and amino acid sensing with protein synthesis, autophagy, and apoptosis. Notably, the effect of ethanol exposure on mTOR signaling depends on the timing and dose of ethanol as well as the system studied. Overall, the overlap between the functions of mTORC1 and the phenotypes observed in FASD suggest a mechanistic interaction. However, more work is required to fully understand the impact of ethanol teratogenesis on mTOR signaling.
Collapse
Affiliation(s)
- Scott K Tucker
- Department of Molecular Biosciences, Waggoner Center for Alcohol and Addiction Research and Institute for Neuroscience, University of Texas, Austin, TX, USA
| | - Johann K Eberhart
- Department of Molecular Biosciences, Waggoner Center for Alcohol and Addiction Research and Institute for Neuroscience, University of Texas, Austin, TX, USA.
| |
Collapse
|
35
|
Arentson-Lantz EJ, Von Ruff Z, Connolly G, Albano F, Kilroe SP, Wacher A, Campbell WW, Paddon-Jones D. Meals Containing Equivalent Total Protein from Foods Providing Complete, Complementary, or Incomplete Essential Amino Acid Profiles do not Differentially Affect 24-h Skeletal Muscle Protein Synthesis in Healthy, Middle-Aged Women. J Nutr 2024; 154:3626-3638. [PMID: 39396760 DOI: 10.1016/j.tjnut.2024.10.010] [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: 04/18/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 10/15/2024] Open
Abstract
BACKGROUND Dietary protein quality can be assessed by skeletal muscle protein synthesis (MPS) stimulation. Limited knowledge exists on how consuming isonitrogenous meals with varied protein qualities affects postprandial and 24-h MPS. OBJECTIVES We assessed the effects of protein quality and complementary proteins on MPS. We hypothesized that meals containing a moderate amount of high-quality, complete protein would stimulate postprandial and 24-h MPS. Meals containing two complementary, plant-based incomplete proteins would stimulate MPS less, and meals containing plant-based incomplete proteins at each meal, but complementary over 24 h would not stimulate MPS. METHODS This quasi-experimental study included a randomized, crossover design to assess protein quality and a nonrandomized low-protein control. We measured postprandial and 24-h MPS responses of healthy middle-aged women (n = 9, age 56 ± 4 y), to 3 dietary conditions: isonitrogenous meals containing 23 g protein/meal from 1) complete protein (lean beef), 2) 2 incomplete, but complementary protein sources (navy/black beans and whole wheat bread), and 3) single incomplete protein sources (black beans or whole wheat bread at 1 meal), but providing a complete amino acid profile over 24 h. In the low-protein group women (n = 8, 54 ± 5 y) consumed a single breakfast meal containing 5 g of protein. Venous blood and vastus lateralis samples were obtained during primed, constant infusions of L-[ring-13C6]phenylalanine to measure mixed muscle fractional synthetic rates (FSR). RESULTS Meals containing complete, complementary, or incomplete proteins did not differentially influence FSR responses after breakfast (P = 0.90) or 24 h (P = 0.38). At breakfast, the complete (P = 0.030) and complementary (P = 0.031) protein meals, but not the incomplete protein meal (P = 0.38), had greater FSR responses compared with the low-protein control meal. CONCLUSIONS Isonitrogenous meals containing a moderate serving of total protein from foods providing complete, complementary, or incomplete essential amino acid profiles do not differentially stimulate muscle protein synthesis after a meal and daily. TRIAL REGISTRATION NUMBER This clinical trial was registered at clinicaltrials.gov as NCT03816579. URL: https://www. CLINICALTRIALS gov/ct2/show/NCT03816579?term=NCT03816579&draw=2&rank=1.
Collapse
Affiliation(s)
- Emily J Arentson-Lantz
- Department of Nutrition Sciences and Health Behavior, University of Texas Medical Branch, Galveston, TX, United States; Center for Health Promotion, Performance and Rehabilitation Science, University of Texas Medical Branch, Galveston, TX, United States.
| | - Zachary Von Ruff
- Center for Health Promotion, Performance and Rehabilitation Science, University of Texas Medical Branch, Galveston, TX, United States
| | - Gavin Connolly
- Department of Nutrition Science, Purdue University, West Lafayette, IN, United States
| | - Frank Albano
- Department of Nutrition Sciences and Health Behavior, University of Texas Medical Branch, Galveston, TX, United States
| | - Sean P Kilroe
- Center for Health Promotion, Performance and Rehabilitation Science, University of Texas Medical Branch, Galveston, TX, United States
| | - Adam Wacher
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, United States
| | - Wayne W Campbell
- Department of Nutrition Science, Purdue University, West Lafayette, IN, United States
| | - Douglas Paddon-Jones
- Department of Nutrition Sciences and Health Behavior, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
36
|
Liu Z, Wang H, Liang Y, Liu M, Huang Q, Wang M, Zhou J, Bu Q, Zhou H, Lu L. E2F2 Reprograms Macrophage Function By Modulating Material and Energy Metabolism in the Progression of Metabolic Dysfunction-Associated Steatohepatitis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2410880. [PMID: 39465673 PMCID: PMC11672278 DOI: 10.1002/advs.202410880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 09/30/2024] [Indexed: 10/29/2024]
Abstract
Macrophages are essential for the development of steatosis, hepatic inflammation, and fibrosis in metabolic dysfunction-associated steatohepatitis(MASH). However, the roles of macrophage E2F2 in the progression of MASH have not been elucidated. This study reveals that the expression of macrophage E2F2 is dramatically downregulated in MASH livers from mice and humans, and that this expression is adversely correlated with the severity of the disease. Myeloid-specific E2F2 depletion aggravates intrahepatic inflammation, hepatic stellate cell activation, and hepatocyte lipid accumulation during MASH progression. Mechanistically, E2F2 can inhibit the SLC7A5 transcription directly. E2F2 deficiency upregulates the expression of SLC7A5 to mediate amino acids flux, resulting in enhanced glycolysis, impaired mitochondrial function, and increased macrophages proinflammatory response in a Leu-mTORC1-dependent manner. Moreover, bioinformatics analysis and CUT &Tag assay identify the direct binding of Nrf2 to E2F2 promoter to promote its transcription and nuclear translocation. Genetic or pharmacological activation of Nrf2 effectively activates E2F2 to attenuate the MASH progression. Finally, patients treated with CDK4/6 inhibitors demonstrate reduced E2F2 activity but increased SLC7A5 activity in PBMCs. These findings indicated macrophage E2F2 suppresses MASH progression by reprogramming amino acid metabolism via SLC7A5- Leu-mTORC1 signaling pathway. Activating E2F2 holds promise as a therapeutic strategy for MASH.
Collapse
Affiliation(s)
- Zheng Liu
- Department of General Surgerythe First Affiliated Hospital of Nanjing Medical UniversityNanjing210029China
| | - Hao Wang
- Department of Liver SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100730China
| | - Yuan Liang
- Hepatobiliary CenterThe First Affiliated Hospital of Nanjing Medical University& Research Unit of Liver Transplantation and Transplant ImmunologyChinese Academy of Medical SciencesNanjing210029China
- School of Biological Science & Medical EngineeringSoutheast UniversityNanjing210096China
| | - Mu Liu
- Hepatobiliary CenterThe First Affiliated Hospital of Nanjing Medical University& Research Unit of Liver Transplantation and Transplant ImmunologyChinese Academy of Medical SciencesNanjing210029China
| | - Qiyuan Huang
- Hepatobiliary CenterThe First Affiliated Hospital of Nanjing Medical University& Research Unit of Liver Transplantation and Transplant ImmunologyChinese Academy of Medical SciencesNanjing210029China
| | - Mingming Wang
- Department of Liver SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100730China
| | - Jinren Zhou
- Hepatobiliary CenterThe First Affiliated Hospital of Nanjing Medical University& Research Unit of Liver Transplantation and Transplant ImmunologyChinese Academy of Medical SciencesNanjing210029China
| | - Qingfa Bu
- Hepatobiliary CenterThe First Affiliated Hospital of Nanjing Medical University& Research Unit of Liver Transplantation and Transplant ImmunologyChinese Academy of Medical SciencesNanjing210029China
| | - Haoming Zhou
- Hepatobiliary CenterThe First Affiliated Hospital of Nanjing Medical University& Research Unit of Liver Transplantation and Transplant ImmunologyChinese Academy of Medical SciencesNanjing210029China
| | - Ling Lu
- Affiliated Hospital of Xuzhou Medical UniversityXuzhou220005China
| |
Collapse
|
37
|
Kaspy MS, Hannaian SJ, Bell ZW, Churchward-Venne TA. The effects of branched-chain amino acids on muscle protein synthesis, muscle protein breakdown and associated molecular signalling responses in humans: an update. Nutr Res Rev 2024; 37:273-286. [PMID: 37681443 DOI: 10.1017/s0954422423000197] [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] [Indexed: 09/09/2023]
Abstract
Branched-chain amino acids (BCAA: leucine, isoleucine and valine) are three of the nine indispensable amino acids, and are frequently consumed as a dietary supplement by athletes and recreationally active individuals alike. The popularity of BCAA supplements is largely predicated on the notion that they can stimulate rates of muscle protein synthesis (MPS) and suppress rates of muscle protein breakdown (MPB), the combination of which promotes a net anabolic response in skeletal muscle. To date, several studies have shown that BCAA (particularly leucine) increase the phosphorylation status of key proteins within the mechanistic target of rapamycin (mTOR) signalling pathway involved in the regulation of translation initiation in human muscle. Early research in humans demonstrated that BCAA provision reduced indices of whole-body protein breakdown and MPB; however, there was no stimulatory effect of BCAA on MPS. In contrast, recent work has demonstrated that BCAA intake can stimulate postprandial MPS rates at rest and can further increase MPS rates during recovery after a bout of resistance exercise. The purpose of this evidence-based narrative review is to critically appraise the available research pertaining to studies examining the effects of BCAA on MPS, MPB and associated molecular signalling responses in humans. Overall, BCAA can activate molecular pathways that regulate translation initiation, reduce indices of whole-body and MPB, and transiently stimulate MPS rates. However, the stimulatory effect of BCAA on MPS rates is less than the response observed following ingestion of a complete protein source providing the full complement of indispensable amino acids.
Collapse
Affiliation(s)
- Matthew S Kaspy
- Department of Kinesiology and Physical Education, McGill University, 475 Avenue Des Pins H2W 1S4, Montreal, QC, Canada
| | - Sarkis J Hannaian
- Department of Kinesiology and Physical Education, McGill University, 475 Avenue Des Pins H2W 1S4, Montreal, QC, Canada
- Research Institute of the McGill University Health Centre, Glen Site, 1001 Boul. Décarie, H4A 3J1 Montreal, QC, Canada
| | - Zachary W Bell
- Department of Kinesiology and Physical Education, McGill University, 475 Avenue Des Pins H2W 1S4, Montreal, QC, Canada
| | - Tyler A Churchward-Venne
- Department of Kinesiology and Physical Education, McGill University, 475 Avenue Des Pins H2W 1S4, Montreal, QC, Canada
- Division of Geriatric Medicine, McGill University, Montreal General Hospital, Room D6 237.F, 1650 Cedar Avenue, H3G 1A4, Montreal, QC, Canada
- Research Institute of the McGill University Health Centre, Glen Site, 1001 Boul. Décarie, H4A 3J1 Montreal, QC, Canada
| |
Collapse
|
38
|
Akkaya Fırat A, Özel A, Davutoğlu EA, Güngör ZB, Madazlı R. Maternal serum interleukin-1β, FoxO1 and Sestrin2 levels in predicting preterm delivery. J Matern Fetal Neonatal Med 2024; 37:2295807. [PMID: 38105533 DOI: 10.1080/14767058.2023.2295807] [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: 07/13/2023] [Accepted: 12/12/2023] [Indexed: 12/19/2023]
Abstract
The study aimed to investigate whether serum IL-1β, FoxO1and Sesn2 concentrations differed between threatened preterm labor (TPL) and uncomplicated pregnancies. This study was conducted on 54 women with TPL pregnancies and 26 healthy pregnant women. The TPL group was further divided into two subgroups according to the gestational age at delivery. Patients who gave birth within 48-72 hours after the hospitalization were referred to as preterm delivery (PD) and those who gave birth at ≥37 weeks were referred to as term delivery (TD). Maternal levels of serum IL-1β, FoxO1 and Sesn2 were measured with the use of enzyme-linked immunosorbent assay kits. The mean maternal serum IL-1β and FoxO1 of PD were significantly higher than TD (p<.000*) and the control group (p < .000*). The mean maternal serum IL-1β, FoxO1 level of TD was significantly higher than the control group (p<.000*). The mean maternal serum Sesn2 levels of TD and the control group were significantly higher than the preterm group (p<.000*). The mean maternal serum Sesn2 level of the control group was significantly higher than the TD group (p <.000*). A negative correlation was found between serum concentration of serum IL-1β, and FoxO1 with the gestational week of delivery (r= -0.722, p< .000*for, IL-1β; r = -0.625, p < .000* for FoxO1). A positive correlation was found between the serum concentration of serum Sesn2 with the gestational week of delivery (r = 0.507, p<.000* for sesn2). High serum IL-1β, FoxO1 levels, and low Sesn2 levels may have the potential to be used as biomarkers for the differentiation of PD and TD.
Collapse
Affiliation(s)
- Asuman Akkaya Fırat
- Department of Medical Biochemistry, Istanbul University Cerrahpasa Faculty of Medicine, Istanbul, Türkiye
| | - Aysegül Özel
- Department of Obstetrics and Gynecology, Perinatology Clinic, Istanbul University Cerrahpasa Faculty of Medicine, Istanbul, Türkiye
| | - Ebru Alıcı Davutoğlu
- Department of Obstetrics and Gynecology, Perinatology Clinic, Istanbul University Cerrahpasa Faculty of Medicine, Istanbul, Türkiye
| | - Zeynep Banu Güngör
- Department of Medical Biochemistry, Istanbul University Cerrahpasa Faculty of Medicine, Istanbul, Türkiye
| | - Rıza Madazlı
- Department of Obstetrics and Gynecology, Perinatology Clinic, Istanbul University Cerrahpasa Faculty of Medicine, Istanbul, Türkiye
| |
Collapse
|
39
|
Tang X, Li K, Wang Y, Rocchi S, Shen S, Cerezo M. Metabolism and mRNA translation: a nexus of cancer plasticity. Trends Cell Biol 2024:S0962-8924(24)00225-3. [PMID: 39603916 DOI: 10.1016/j.tcb.2024.10.009] [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: 05/24/2024] [Revised: 10/24/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024]
Abstract
Tumors often face energy deprivation due to mutations, hypoxia, and nutritional deficiencies within the harsh tumor microenvironment (TME), and as an effect of anticancer treatments. This metabolic stress triggers adaptive reprogramming of mRNA translation, which in turn adjusts metabolic plasticity and associated signaling pathways to ensure tumor cell survival. Emerging evidence is beginning to reveal the complex interplay between metabolism and mRNA translation, shedding light on the mechanisms that synchronize ribosome assembly and reconfigure translation programs under metabolic stress. This review explores recent advances in our understanding of the coordination between metabolism and mRNA translation, offering insights that could inform therapeutic strategies targeting both cancer metabolism and translation, with the aim of disrupting cancer cell plasticity and survival.
Collapse
Affiliation(s)
- Xinpu Tang
- Institute of Thoracic Oncology and Department of Thoracic Surgery, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Kaixiu Li
- Institute of Thoracic Oncology and Department of Thoracic Surgery, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yuqing Wang
- Institute of Thoracic Oncology and Department of Thoracic Surgery, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Stéphane Rocchi
- INSERM, U1065, Equipe 12, Centre Méditerranéen de Médecine Moléculaire (C3M), Bâtiment ARCHIMED, 151 route de saint Antoine de Ginestière, 06204, Nice cedex 3, France; Université Côte d'Azur, Nice, France
| | - Shensi Shen
- Institute of Thoracic Oncology and Department of Thoracic Surgery, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.
| | - Michael Cerezo
- INSERM, U1065, Equipe 12, Centre Méditerranéen de Médecine Moléculaire (C3M), Bâtiment ARCHIMED, 151 route de saint Antoine de Ginestière, 06204, Nice cedex 3, France; Université Côte d'Azur, Nice, France.
| |
Collapse
|
40
|
Li Z, Chen S, Wu X, Liu F, Zhu J, Chen J, Lu X, Chi R. Research advances in branched-chain amino acid metabolism in tumors. Mol Cell Biochem 2024. [DOI: 10.1007/s11010-024-05163-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 11/10/2024] [Indexed: 01/06/2025]
|
41
|
Wu T, Yu Z, Dai J, Li J, Ning F, Liu X, Zhu N, Zhang X. JPH203 alleviates peritoneal fibrosis via inhibition of amino acid-mediated mTORC1 signaling. Biochem Biophys Res Commun 2024; 734:150656. [PMID: 39362029 DOI: 10.1016/j.bbrc.2024.150656] [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: 07/11/2024] [Revised: 08/26/2024] [Accepted: 09/03/2024] [Indexed: 10/05/2024]
Abstract
BACKGROUND AND AIMS The mesothelial-mesenchymal transition (MMT) of mesothelial cells has been recognized as a critical process during progression of peritoneal fibrosis (PF). Despite its crucial role in amino acid transport and metabolism, the involvement of L-type amino acid transporter 1 (LAT1) and the potential therapeutic role of its inhibitor, JPH203, in fibrotic diseases remain unexplored. Considering the paucity of research on amino acid-mediated mTORC1 activation in PF, our study endeavors to elucidate the protective effects of JPH203 against PF and explore the involvement of amino acid-mediated mTORC1 signaling in this context. METHODS We established the transforming growth factor beta 1 (TGF-β1) induced MMT model in primary human mesothelial cells and the peritoneal dialysis fluid (PDF) induced PF model in mice. The therapeutic effects of JPH203 on PF were then examined on these two models by real-time quantitative polymerase chain reaction, western blotting, immunofluorescence staining, Masson's trichrome staining, H&E staining, picro-sirius red staining, and immunohistochemistry. The involvement of amino acid-mediated mTORC1 signaling was screened by RNA sequencing and further verified by western blotting in vitro. RESULTS LAT1 was significantly upregulated and JPH203 markedly attenuated fibrotic phenotype both in vitro and in vivo. RNA-seq unveiled a significant enrichment of mTOR signaling pathway in response to JPH203 treatment. Western blotting results indicated that JPH203 alleviates PF by inhibiting amino acid-mediated mTORC1 signaling, which differs from the direct inhibition observed with rapamycin. CONCLUSION JPH203 alleviates PF by inhibiting amino acid-mediated mTORC1 signaling.
Collapse
Affiliation(s)
- Tiangang Wu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Zanzhe Yu
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Junhao Dai
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Jiayang Li
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Fengling Ning
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Xin Liu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Nan Zhu
- Department of Nephrology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Xuemei Zhang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China; School of Pharmacy, East China Normal University, Shanghai, China.
| |
Collapse
|
42
|
Smiles WJ, Ovens AJ, Kemp BE, Galic S, Petersen J, Oakhill JS. New developments in AMPK and mTORC1 cross-talk. Essays Biochem 2024; 68:321-336. [PMID: 38994736 DOI: 10.1042/ebc20240007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/13/2024]
Abstract
Metabolic homeostasis and the ability to link energy supply to demand are essential requirements for all living cells to grow and proliferate. Key to metabolic homeostasis in all eukaryotes are AMPK and mTORC1, two kinases that sense nutrient levels and function as counteracting regulators of catabolism (AMPK) and anabolism (mTORC1) to control cell survival, growth and proliferation. Discoveries beginning in the early 2000s revealed that AMPK and mTORC1 communicate, or cross-talk, through direct and indirect phosphorylation events to regulate the activities of each other and their shared protein substrate ULK1, the master initiator of autophagy, thereby allowing cellular metabolism to rapidly adapt to energy and nutritional state. More recent reports describe divergent mechanisms of AMPK/mTORC1 cross-talk and the elaborate means by which AMPK and mTORC1 are activated at the lysosome. Here, we provide a comprehensive overview of current understanding in this exciting area and comment on new evidence showing mTORC1 feedback extends to the level of the AMPK isoform, which is particularly pertinent for some cancers where specific AMPK isoforms are implicated in disease pathogenesis.
Collapse
Affiliation(s)
- William J Smiles
- Metabolic Signalling Laboratory, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Research Program for Receptor Biochemistry and Tumour Metabolism, Department of Paediatrics, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Ashley J Ovens
- Protein Engineering in Immunity and Metabolism, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
| | - Bruce E Kemp
- Protein Chemistry and Metabolism, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Department of Medicine, University of Melbourne, Parkville, VIC 3010, Australia
- Mary Mackillop Institute for Health Research, Australian Catholic University, Fitzroy, Vic 3065, Vic. Australia
| | - Sandra Galic
- Department of Medicine, University of Melbourne, Parkville, VIC 3010, Australia
- Metabolic Physiology, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
| | - Janni Petersen
- Flinders Health and Medical Research Institute, Flinders Centre for Innovation in Cancer, Flinders University, Adelaide, SA 5042, Australia
- Nutrition and Metabolism, South Australia Health and Medical Research Institute, Adelaide, SA 5000, Australia
| | - Jonathan S Oakhill
- Metabolic Signalling Laboratory, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Department of Medicine, University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
43
|
Liu Y, Ohgaki R, Okanishi H, Xu M, Kanai Y. Amino acid transporter LAT1 is expressed on cancer cell-derived exosomes with potential as a diagnostic and prognostic biomarker. Sci Rep 2024; 14:28458. [PMID: 39557943 PMCID: PMC11574015 DOI: 10.1038/s41598-024-79425-9] [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: 07/12/2024] [Accepted: 11/08/2024] [Indexed: 11/20/2024] Open
Abstract
L-type amino acid transporter 1 (LAT1) is upregulated in various cancers and contributes to the growth and proliferation of cancer cells. Previous clinicopathological studies have revealed associations between the high expression of LAT1 and the poor prognosis in multiple cancer types. However, in those studies, the expression of LAT1 has been evaluated solely by immunohistochemistry on resected tumors or tissue biopsies. Cancer cell-derived exosomes are attracting increasing attention as a resource of diagnostic, prognostic, and therapeutic biomarkers. In this study, we revealed the expression of LAT1 on exosomes from pancreatic cancer T3M-4 cells by western blotting, immunoprecipitation, and immuno-transmission electron microscopy. LAT1 was generally detected by western blotting and ELISA on exosomes from several pancreatic, biliary tract, and ovarian cancer cells. Notably, similar trends existed between the abundance of exosomal LAT1 and the cellular LAT1 expression levels. The expression of LAT1 on exosomes in vivo was verified using the peritoneal wash from intraperitoneal T3M-4 tumor-bearing mice. These results indicate that exosomal LAT1 released from cancer cells holds significant potential as a novel biomarker for diagnosis and prognosis.
Collapse
Affiliation(s)
- Yumiao Liu
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, Japan
| | - Ryuichi Ohgaki
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, Japan.
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka, Japan.
| | - Hiroki Okanishi
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, Japan
| | - Minhui Xu
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, Japan
| | - Yoshikatsu Kanai
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, Japan.
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka, Japan.
- Department of Metabolic Reprogramming and Signal Regulation, Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
44
|
LaNoce E, Zhang DY, Garcia-Epelboim A, Su Y, Sun Y, Alepa G, Angelucci AR, Akay-Espinoza C, Jordan-Sciutto KL, Song H, Ming GL, Christian KM. Exposure to the antiretroviral drug dolutegravir impairs structure and neurogenesis in a forebrain organoid model of human embryonic cortical development. Front Mol Neurosci 2024; 17:1459877. [PMID: 39569018 PMCID: PMC11576471 DOI: 10.3389/fnmol.2024.1459877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 09/13/2024] [Indexed: 11/22/2024] Open
Abstract
Introduction For many therapeutic drugs, including antiretroviral drugs used to treat people living with HIV-1 (PLWH), we have little data on the potential effects on the developing human brain due to limited access to tissue and historical constraints on the inclusion of pregnant populations in clinical trials. Human induced pluripotent stem cells (iPSCs) offer a new avenue to gain insight on how drugs may impact human cell types representative of the developing central nervous system. To prevent vertical transmission of HIV and promote the health of pregnant PLWH, antiretroviral therapy must be initiated and/or maintained throughout pregnancy. However, many antiretroviral drugs are approved for widespread use following clinical testing only in non-pregnant populations and there may be limited information on potential teratogenicity until pregnancy outcomes are evaluated. The integrase strand transfer inhibitor dolutegravir (DTG) is a frontline antiretroviral drug that is effective in viral suppression of HIV but was previously reported to be associated with a slight increase in the risk for neural tube defects in one study, although this has not been replicated in other cohorts. Methods To directly investigate the potential impact of DTG on human cortical neurogenesis, we measured the effects of daily drug exposure on the early stages of corticogenesis in a human iPSC-based forebrain organoid model. We quantified organoid size and structure and analyzed gene and protein expression to evaluate the impact of several doses of DTG on organoid development. Results We observed deficits in organoid structure and impaired neurogenesis in DTG-treated organoids compared to vehicle-treated control organoids after 20 or 40 days in culture. Our highest dose of DTG (10 μM) resulted in significantly smaller organoids with a reduced density of neural rosette structures compared to vehicle-treated controls. Mechanistically, RNA-sequencing and immunohistological analysis suggests dysregulated amino acid transport and activation of the integrated stress response in the DTG-treated organoids, and functionally, a small molecule integrated stress response inhibitor (ISRIB) could partially rescue increased expression of proteins related to cell cycle regulation. Discussion Together, these results illustrate the potential for human iPSC-based strategies to reveal biological processes during neurogenesis that may be affected by therapeutic drugs and provide complementary data in relevant human cell types to augment preclinical investigations of drug safety during pregnancy.
Collapse
Affiliation(s)
- Emma LaNoce
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Daniel Y. Zhang
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Alan Garcia-Epelboim
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Yijing Su
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Yusha Sun
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Giana Alepa
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Angelina R. Angelucci
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Cagla Akay-Espinoza
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Kelly L. Jordan-Sciutto
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- The Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Guo-li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Kimberly M. Christian
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
45
|
Akbay B, Omarova Z, Trofimov A, Sailike B, Karapina O, Molnár F, Tokay T. Double-Edge Effects of Leucine on Cancer Cells. Biomolecules 2024; 14:1401. [PMID: 39595578 PMCID: PMC11591885 DOI: 10.3390/biom14111401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/16/2024] [Accepted: 10/22/2024] [Indexed: 11/28/2024] Open
Abstract
Leucine is an essential amino acid that cannot be produced endogenously in the human body and therefore needs to be obtained from dietary sources. Leucine plays a pivotal role in stimulating muscle protein synthesis, along with isoleucine and valine, as the group of branched-chain amino acids, making them one of the most popular dietary supplements for athletes and gym-goers. The individual effects of leucine, however, have not been fully clarified, as most of the studies so far have focused on the grouped effects of branched-chain amino acids. In recent years, leucine and its metabolites have been shown to stimulate muscle protein synthesis mainly via the mammalian target of the rapamycin complex 1 signaling pathway, thereby improving muscle atrophy in cancer cachexia. Interestingly, cancer research suggests that leucine may have either anti-cancer or pro-tumorigenic effects. In the current manuscript, we aim to review leucine's roles in muscle protein synthesis, tumor suppression, and tumor progression, specifically summarizing the molecular mechanisms of leucine's action. The role of leucine is controversial in hepatocellular carcinoma, whereas its pro-tumorigenic effects have been demonstrated in breast and pancreatic cancers. In summary, leucine being used as nutritional supplement for athletes needs more attention, as its pro-oncogenic effects may have been identified by recent studies. Anti-cancer or pro-tumorigenic effects of leucine in various cancers should be further investigated to achieve clear conclusions.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Tursonjan Tokay
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Kabanbay Batyr 53, Astana 010000, Kazakhstan; (B.A.); (Z.O.); (A.T.); (B.S.); (O.K.); (F.M.)
| |
Collapse
|
46
|
Jiang C, Tan X, Liu N, Yan P, Hou T, Wei W. Nutrient sensing of mTORC1 signaling in cancer and aging. Semin Cancer Biol 2024; 106-107:1-12. [PMID: 39153724 DOI: 10.1016/j.semcancer.2024.08.001] [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: 06/27/2024] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 08/19/2024]
Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) is indispensable for preserving cellular and organismal homeostasis by balancing the anabolic and catabolic processes in response to various environmental cues, such as nutrients, growth factors, energy status, oxygen levels, and stress. Dysregulation of mTORC1 signaling is associated with the progression of many types of human disorders including cancer, age-related diseases, neurodegenerative disorders, and metabolic diseases. The way mTORC1 senses various upstream signals and converts them into specific downstream responses remains a crucial question with significant impacts for our perception of the related physiological and pathological process. In this review, we discuss the recent molecular and functional insights into the nutrient sensing of the mTORC1 signaling pathway, along with the emerging role of deregulating nutrient-mTORC1 signaling in cancer and age-related disorders.
Collapse
Affiliation(s)
- Cong Jiang
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China.
| | - Xiao Tan
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Ning Liu
- International Research Center for Food and Health, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Peiqiang Yan
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Tao Hou
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
47
|
Weston AH, Fernandes T, de Oliveira M, Gaskin S, Pilonero T, Hanigan MD. Effects of isoleucine, lysine, valine, and a group of nonessential amino acids on mammary amino acid metabolism in lactating dairy cows. J Dairy Sci 2024; 107:9155-9175. [PMID: 39067747 DOI: 10.3168/jds.2024-24774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 06/27/2024] [Indexed: 07/30/2024]
Abstract
Intracellular AA regulate milk protein synthesis within the mammary gland by modifying mammary plasma flow (MPF) and AA transporter activity. Amino acid transporters catalyze translocation using Na+ gradient, substrate gradient (uniporters), and exchange mechanisms; further, they exhibit specificity for individual AA or groups of AA with similar side-chain properties within each transport system. Nonessential AA are actively transported through Na+-dependent transporters and, thus, are often used as intracellular currencies for EAA transport through exchange transporters. Therefore, it was hypothesized that individual EAA supplementation would compete with other EAA for shared transporters, and supplementation with Ala, Gln, and Gly would stimulate EAA transport through exchange transporters. Ten primiparous lactating dairy cows were divided into 2 groups based on milk production and were randomly assigned to treatment sequences within 2 balanced 5 × 5 Latin squares by group. Period length was 14 d. Treatments were 9-d jugular infusions of (1) saline; (2) 34.5 g of Val per day; (3) a ratio of 32.7 g of Ala to 40 g of Gln to 26.7 g of Gly per day (AQG); (4) 43 g of Lys per day; or (5) 33.5 g of Ile per day. All cows were fed a common base diet formulated to contain 15.0% CP. Infusions of Ile, Lys, or AQG did not affect milk protein or milk production; however, Val infusion decreased both. The effects of Val infusion on milk protein production appeared to be partially driven by decreased DMI. The decline in milk protein percentage indicated that milk lactose production was also affected. Additionally, Val infusion increased MPF efficiency (MPF/milk; L/L) by approximately 44%. Infusion of Val tended to decrease or decreased mammary net uptakes of Lys, Leu, Met, and total AA. Infusion of Ile tended to increase its mammary net uptakes but did not affect any other AA. Infusions of Lys and AQG did not affect any mammary net uptakes. Infusion of Val tended to decrease Phe and total NEAA mammary clearance rates. Infusion with AQG stimulated Tyr clearance rates and tended to decline system N mammary clearance rates. Ratios of branched-chain AA mammary uptake to milk protein output (U:O) did not differ from 1 for Val-infused cows, which indicated that little intramammary catabolism was occurring. Additionally, the average NEAA U:O in response to all treatments except Val was 0.70, but Val-infused cows had NEAA U:O that averaged 0.09, indicating increased synthesis within the glands. The effects of Val on mammary net clearance rates of multiple EAA support the incorporation of AA limitations in ration optimizers to prevent AA imbalances. It is possible that oversupplementation of EAA other than Val may also decrease DMI and mammary activity. Identifying efficiency apexes for each of the EAA will allow more precise diet formulation and supplementation, leading to improved production efficiency.
Collapse
Affiliation(s)
- A H Weston
- School of Animal Sciences, Virginia Tech, Blacksburg, VA 24060
| | - T Fernandes
- School of Animal Sciences, Virginia Tech, Blacksburg, VA 24060
| | - M de Oliveira
- School of Animal Sciences, Virginia Tech, Blacksburg, VA 24060
| | - S Gaskin
- School of Animal Sciences, Virginia Tech, Blacksburg, VA 24060
| | - T Pilonero
- School of Animal Sciences, Virginia Tech, Blacksburg, VA 24060
| | - M D Hanigan
- School of Animal Sciences, Virginia Tech, Blacksburg, VA 24060.
| |
Collapse
|
48
|
Fernandes SA, Angelidaki DD, Nüchel J, Pan J, Gollwitzer P, Elkis Y, Artoni F, Wilhelm S, Kovacevic-Sarmiento M, Demetriades C. Spatial and functional separation of mTORC1 signalling in response to different amino acid sources. Nat Cell Biol 2024; 26:1918-1933. [PMID: 39385049 PMCID: PMC11567901 DOI: 10.1038/s41556-024-01523-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 09/09/2024] [Indexed: 10/11/2024]
Abstract
Amino acid (AA) availability is a robust determinant of cell growth through controlling mechanistic/mammalian target of rapamycin complex 1 (mTORC1) activity. According to the predominant model in the field, AA sufficiency drives the recruitment and activation of mTORC1 on the lysosomal surface by the heterodimeric Rag GTPases, from where it coordinates the majority of cellular processes. Importantly, however, the teleonomy of the proposed lysosomal regulation of mTORC1 and where mTORC1 acts on its effector proteins remain enigmatic. Here, by using multiple pharmacological and genetic means to perturb the lysosomal AA-sensing and protein recycling machineries, we describe the spatial separation of mTORC1 regulation and downstream functions in mammalian cells, with lysosomal and non-lysosomal mTORC1 phosphorylating distinct substrates in response to different AA sources. Moreover, we reveal that a fraction of mTOR localizes at lysosomes owing to basal lysosomal proteolysis that locally supplies new AAs, even in cells grown in the presence of extracellular nutrients, whereas cytoplasmic mTORC1 is regulated by exogenous AAs. Overall, our study substantially expands our knowledge about the topology of mTORC1 regulation by AAs and hints at the existence of distinct, Rag- and lysosome-independent mechanisms that control its activity at other subcellular locations. Given the importance of mTORC1 signalling and AA sensing for human ageing and disease, our findings will probably pave the way towards the identification of function-specific mTORC1 regulators and thus highlight more effective targets for drug discovery against conditions with dysregulated mTORC1 activity in the future.
Collapse
Affiliation(s)
- Stephanie A Fernandes
- Max Planck Institute for Biology of Ageing, Cologne, Germany
- Cologne Graduate School of Ageing Research, Cologne, Germany
| | | | - Julian Nüchel
- Max Planck Institute for Biology of Ageing, Cologne, Germany
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Jiyoung Pan
- Max Planck Institute for Biology of Ageing, Cologne, Germany
- Cologne Graduate School of Ageing Research, Cologne, Germany
| | | | - Yoav Elkis
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Filippo Artoni
- Max Planck Institute for Biology of Ageing, Cologne, Germany
- Cologne Graduate School of Ageing Research, Cologne, Germany
| | - Sabine Wilhelm
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | | | - Constantinos Demetriades
- Max Planck Institute for Biology of Ageing, Cologne, Germany.
- Cologne Graduate School of Ageing Research, Cologne, Germany.
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany.
| |
Collapse
|
49
|
Obata F, Miura M. Regulatory Mechanisms of Aging Through the Nutritional and Metabolic Control of Amino Acid Signaling in Model Organisms. Annu Rev Genet 2024; 58:19-41. [PMID: 38857535 DOI: 10.1146/annurev-genet-111523-102042] [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] [Indexed: 06/12/2024]
Abstract
Life activities are supported by the intricate metabolic network that is fueled by nutrients. Nutritional and genetic studies in model organisms have determined that dietary restriction and certain mutations in the insulin signaling pathway lead to lifespan extension. Subsequently, the detailed mechanisms of aging as well as various nutrient signaling pathways and their relationships have been investigated in a wide range of organisms, from yeast to mammals. This review summarizes the roles of nutritional and metabolic signaling in aging and lifespan with a focus on amino acids, the building blocks of organisms. We discuss how lifespan is affected by the sensing, transduction, and metabolism of specific amino acids and consider the influences of life stage, sex, and genetic background on the nutritional control of aging. Our goal is to enhance our understanding of how nutrients affect aging and thus contribute to the biology of aging and lifespan.
Collapse
Affiliation(s)
- Fumiaki Obata
- Laboratory of Molecular Cell Biology and Development, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
- Laboratory for Nutritional Biology, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan;
| | - Masayuki Miura
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan;
| |
Collapse
|
50
|
Seale B, Slotabec L, Nguyen JD, Wang H, Patterson C, Filho F, Rouhi N, Adenawoola MI, Li J. Sestrin2 serves as a scaffold protein to maintain cardiac energy and metabolic homeostasis during pathological stress. FASEB J 2024; 38:e70106. [PMID: 39404019 PMCID: PMC11698584 DOI: 10.1096/fj.202401404r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 09/23/2024] [Accepted: 09/30/2024] [Indexed: 10/25/2024]
Abstract
Cardiovascular diseases (CVDs) are a leading cause of morbidity and mortality worldwide. Metabolic imbalances and pathological stress often contribute to increased mortality. Sestrin2 (Sesn2) is a stress-inducible protein crucial in maintaining cardiac energy and metabolic homeostasis under pathological conditions. Sesn2 is upregulated in response to various stressors, including oxidative stress, hypoxia, and energy depletion, and mediates multiple cellular pathways to enhance antioxidant defenses, promote autophagy, and inhibit inflammation. This review explores the mechanisms through which Sesn2 regulates these pathways, focusing on the AMPK-mTORC1, Sesn2-Nrf2, and HIF1α-Sesn2 pathways, among others. We can identify the potential therapeutic targets for treating CVDs and related metabolic disorders by comprehending these complex mechanisms. Sesn2's unique ability to respond thoroughly to metabolic challenges, oxidative stress, and inflammation makes it a promising prospect for enhancing cardiac health and resilience against pathological stress.
Collapse
Affiliation(s)
- Blaise Seale
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Lily Slotabec
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Research, G.V. (Sonny) Montgomery VA Medical Center, Jackson, Mississippi, USA
| | - Jennie D. Nguyen
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Hao Wang
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Cory Patterson
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Fernanda Filho
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Nadiyeh Rouhi
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Michael I. Adenawoola
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Ji Li
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Research, G.V. (Sonny) Montgomery VA Medical Center, Jackson, Mississippi, USA
| |
Collapse
|