1
|
Fang Y, Zhang Y, Shen X, Dou A, Xie H, Zhang Y, Xie K. Utilization of lactate trajectory models for predicting acute kidney injury and mortality in patients with hyperlactatemia: insights across three independent cohorts. Ren Fail 2025; 47:2474205. [PMID: 40074720 PMCID: PMC11905305 DOI: 10.1080/0886022x.2025.2474205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 02/08/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
This study aims to investigate the association between lactate trajectories and the risk of acute kidney injury (AKI) and hospital mortality in patients with hyperlactatemia. We conducted a multicenter retrospective study using data from three independent cohorts. By the lactate levels during the first 48 h of ICU admission, patients were classified into distinct lactate trajectories using group-based trajectory modeling (GBTM) method. The primary outcomes were AKI incidence and hospital mortality. Logistic regression analysis assessed the association between lactate trajectories and clinical outcomes, with adjusting potential confounders. Patients were divided into three trajectories: mild hyperlactatemia with rapid recovery (Traj-1), severe hyperlactatemia with gradual recovery (Traj-2), and severe hyperlactatemia with persistence (Traj-3). Traj-3 was an independent risk factor of both hospital mortality (all p < 0.001) and AKI development (all p < 0.001). Notably, Traj-2 was also associated with increased risk of mortality and AKI development (all p < 0.05) using Traj-1 as reference, except for the result in the Tianjin Medical University General Hospital (TMUGH) cohort for mortality in adjusted model (p = 0.123). Our finding was still robust in subgroup and sensitivity analysis. In the combination cohort, both Traj-2 and Traj-3 were considered as independent risk factor for hospital mortality and AKI development (all p < 0.001). When compared with the Traj-3, Traj-2 was only significantly associated with the decreased risk of hospital mortality (OR 0.17, 95% CI 0.14-0.20, p < 0.001), but no with the likelihood of AKI development (OR 0.90, 95% CI 0.77-1.05, p = 0.172). Lactate trajectories provide valuable information for predicting AKI and mortality in critically ill patients.
Collapse
Affiliation(s)
- Yipeng Fang
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Ying Zhang
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Xuejun Shen
- Department of Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong Province, China
| | - Aizhen Dou
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Hui Xie
- Firth Clinical College, XinXiang Medical University, Xinxiang, Henan, China
| | - Yunfei Zhang
- Editorial Department of Journal, Tianjin Hospital, Tianjin, China
| | - Keliang Xie
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
2
|
Wang J, He Y, Chu C, Wu Z, Zhang T, Peng H, Jin L. Covalent sortase A inhibitor, α, β-unsaturated aldehydes, prevents Staphylococcus aureus infection. Microb Pathog 2025; 204:107472. [PMID: 40122408 DOI: 10.1016/j.micpath.2025.107472] [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: 02/10/2025] [Accepted: 03/11/2025] [Indexed: 03/25/2025]
Abstract
Sortase A (SrtA) plays a crucial role in the attachment of virulence factors characterized by the LPXTG sequence to the peptidoglycans present on the cell wall surface, thereby rendering it a significant target for anti-virulence agents aimed at combating Staphylococcus aureus (S. aureus) infections. This study focuses on the compound 3, 3'- (1,4-phenylene) diacrylaldehyde (DCA), selected as an anti-anchoring agent for covalent inhibitors. To elucidate the targets of DCA in inhibiting S. aureus and to investigate the molecular mechanism underlying the covalent inhibition of SrtA, proteomics and high-resolution mass spectrometry were employed. The findings indicate that compounds containing α, β-unsaturated aldehydes can covalently modify the catalytic residue Cys184 of SrtA. This modification effectively inhibits the transpeptidation function of SrtA, which is responsible for cell wall anchoring, thereby obstructing the attachment of virulence factors such as IgG-SpA, FnbA, ClfA, Clfb, SdrC, Sas, and Can to the cell wall. As a result, this inhibition impedes immune evasion, biofilm formation, adhesion, and subsequent infection by S. aureus. Furthermore, it disrupts the NADH/NAD+ redox homeostasis and diminishes the virulence phenotype by interfering with the glycolytic pathway and the tricarboxylic acid (TCA) cycle of S. aureus. These findings confirm at the molecular level that compounds containing α, β-unsaturated aldehydes possess the potential to serve as anti-virulence drugs that covalently inhibit SrtA.
Collapse
Affiliation(s)
- Jianchao Wang
- Key Laboratory of Medicinal Resources Chemistry and Pharmacology in Wuling Mountainous of Hunan Province College, School of Pharmaceutical Sciences, Jishou University, Jishou, China
| | - Yuan He
- Key Laboratory of Medicinal Resources Chemistry and Pharmacology in Wuling Mountainous of Hunan Province College, School of Pharmaceutical Sciences, Jishou University, Jishou, China
| | - Chenliang Chu
- School of Food and Pharmaceutical Engineering, Zhaoqing University, Zhaoqing, China
| | - Ziqi Wu
- Key Laboratory of Medicinal Resources Chemistry and Pharmacology in Wuling Mountainous of Hunan Province College, School of Pharmaceutical Sciences, Jishou University, Jishou, China
| | - Ting Zhang
- Key Laboratory of Medicinal Resources Chemistry and Pharmacology in Wuling Mountainous of Hunan Province College, School of Pharmaceutical Sciences, Jishou University, Jishou, China
| | - Huayong Peng
- Key Laboratory of Medicinal Resources Chemistry and Pharmacology in Wuling Mountainous of Hunan Province College, School of Pharmaceutical Sciences, Jishou University, Jishou, China.
| | - Lu Jin
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong, China.
| |
Collapse
|
3
|
Jiang Q, Huang B, Chen S, Zhao L, Ban Z, Zhang B. Intestinal inflammation disrupts energy metabolism in layer pullets: insights into energy partitioning and intestinal metabolomic profiling. J Anim Sci Biotechnol 2025; 16:76. [PMID: 40420183 DOI: 10.1186/s40104-025-01204-x] [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: 12/19/2024] [Accepted: 03/31/2025] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND Intestinal inflammation is an energy-consuming process that may alter energy supply and demand in poultry. During inflammation, the intestinal energy metabolic profile and the patterns of energy partitioning remain unclear. This study investigated the effects of intestinal inflammation on energy intake, heat production (HP), retained energy (RE) and intestinal energy metabolites in layer pullets. METHODS After 7 d dietary adaption, 32 "Jing Tint 6" layer pullets with average body weight (1,123.50 ± 8.55 g) were selected from 96 birds, and randomly assigned to two groups (CON: Control group, INFL: Inflammation group) with 8 replicates per group. Indirect calorimetry analysis was conducted over 7 d to determine HP and fasting HP (FHP). During this period, pullets in INFL group received 4 mL/d of 0.6 g/mL dextran sulfate sodium (DSS) via oral gavage to induce intestinal inflammation. After the calorimetry, intestinal tissues were collected post-euthanasia from one bird per replicate for morphological and mucosal metabolomic analysis. RESULTS Birds exhibited significantly lower apparent metabolizable energy (AME) intake (P < 0.001) during intestinal inflammation, accompanied by compromised RE and RE as fat (P < 0.001), suggesting that birds consumed body energy to sustain energy demands. Targeted metabolomic studies identified 11 energy metabolites differentially expressed in ileal mucosa between CON and INFL groups. Specifically, DSS induction significantly increased (P < 0.05) adenosine triphosphate (ATP) level and reduced (P < 0.001) nicotinamide adenine dinucleotide (NAD+) level in ileal mucosa of pullets. In parallel, metabolic adaptations such as enhanced glycolytic intermediates, reduced amino acids, α-ketoglutarate (α-KG) accumulation and suppressed expression of genes encoding enzymes involved in tricarboxylic acid (TCA) cycle were observed in the inflamed ileum of pullets. CONCLUSION Immune stimulation by DSS induced a negative energy balance in layer pullets, characterized by reduced AME intake (-190.47 kJ/kg BW0.75) and compromised RE (-18.81% of AME intake). Disruption of intestinal energy profiling was observed in inflammation-challenged pullets, such as accumulation of α-KG and ATP, reduced NAD+ and amino acids, which could provide valuable insights for developing effective intervention strategies.
Collapse
Affiliation(s)
- Qiuyu Jiang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Bingjian Huang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Simiao Chen
- Institute of Animal Nutrition and Feed Sciences, Jilin Academy of Agricultural Sciences, Gongzhuling, Jilin, 136100, China
| | - Lihua Zhao
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Zhibin Ban
- Institute of Animal Nutrition and Feed Sciences, Jilin Academy of Agricultural Sciences, Gongzhuling, Jilin, 136100, China
| | - Bingkun Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
4
|
Fatani F, Rauf S, Banerjee A, Shamim A. Breaking the Specificity Barrier in Microwave Sensing: Highly Specific Lactate Microwave Biosensor for Fitness and Exercise Optimization. ACS Sens 2025; 10:3658-3668. [PMID: 40181501 DOI: 10.1021/acssensors.5c00470] [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: 04/05/2025]
Abstract
Noninvasive biomarker sensing plays a vital role in health monitoring and sports physiology, particularly for tracking sweat lactate in real time to gauge exercise intensity without disrupting activity. This work introduces a high-specificity microwave biosensor for lactate detection, addressing the challenge of specificity seen in current microwave biosensors, which limits their practical applications. Our approach leverages a cost-effective complementary split-ring resonator (CSRR) combined with lactate oxidase (LOx) immobilized on spherical glass beads that act as mini-reactors within a microfluidic reservoir, enabling highly specific lactate sensing. The sensor was tested in phosphate buffer saline (PBS) and artificial sweat, achieving a high linear sensitivity of 10.9 and 11.3 MHz/mM, respectively, across lactate concentrations up to 150 mM and limit-of-detection (LOD) of 8.76 mM, with validation using the gold-standard HPLC method. It demonstrated excellent specificity against common interferences, including glucose, uric acid, and several ions. Testing with a diverse group of adult volunteers confirmed the sensor's capability to detect dynamic lactate changes during exercise and reliably identify the lactate threshold (LT), underscoring its promise for applications in sports physiology. This innovative method not only offers a powerful tool for lactate monitoring but also paves the way for enzyme-specific microwave biosensors adaptable to detect a range of biomarkers by simply exchanging the target enzyme.
Collapse
Affiliation(s)
- Firas Fatani
- Electrical and Computer Engineering (CEMSE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Sakandar Rauf
- Electrical and Computer Engineering (CEMSE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Apala Banerjee
- Electrical and Computer Engineering (CEMSE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Atif Shamim
- Electrical and Computer Engineering (CEMSE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
| |
Collapse
|
5
|
Zhong D, Yang X, Yang J, Luo Z, Feng Z, Ma M, Liao Y, Tang Y, Wen Y, Liu J, Hu S. Oxygen vacancy-engineered bimetallic nanozymes for disrupting electron transport chain and synergistic multi-enzyme activity to reverse oxaliplatin resistance in colorectal cancer. J Nanobiotechnology 2025; 23:352. [PMID: 40380190 PMCID: PMC12082870 DOI: 10.1186/s12951-025-03417-8] [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: 01/24/2025] [Accepted: 04/26/2025] [Indexed: 05/19/2025] Open
Abstract
In colorectal cancer treatment, chemotherapeutic agents induce reactive oxygen species (ROS) production, which promotes NAD+ accumulation in tumor cells, reducing treatment sensitivity and worsening patient prognosis. Targeted depletion of NAD+ presents a promising strategy to overcome tumor resistance and improve patient prognosis. Here, we designed a dual-metallic nanozyme (CuMnOx-V@Oxa@SP) with defect engineering, modified by soy phospholipids (SP) and loaded with oxaliplatin (Oxa). This nanozyme uses its oxygen-deficient active sites to rapidly and irreversibly degrade NAD⁺ and NADH into nicotinamide and ADP-ribose derivatives, disrupting the electron transport chain (ETC) and compromising tumor antioxidant defenses. It also inhibits the glutathione S-transferase P1 (GSTP1) pathway, weakening tumor detoxification and enhancing chemotherapy sensitivity. Density functional theory calculations revealed that the synergistic effect among multi-enzyme active centers endows the CuMnOx-V nanozymes with excellent catalytic activity. In the tumor microenvironment (TME), CuMnOx-V nanozymes exhibit peroxidase, oxidase, and NAD+ oxidase-mimicking activities. CuMnOx-V generates multiple ROS and depletes NAD+ while preventing their regeneration thereby triggering a cascade amplification of oxidative stress. This, coupled with targeted chemotherapy drug delivery, restores chemosensitivity in refractory tumors and exposes the vulnerabilities of resistant colorectal cancer cells to ROS.
Collapse
Affiliation(s)
- Dong Zhong
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China
| | - Xiaoxin Yang
- Department of Radiology, Second Xiangya Hospital of Central South University, 139 Renming Middle Road Changsha, Changsha, Hunan, China.
| | - Jinhui Yang
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China
| | - Zhisheng Luo
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China
| | - Zhichao Feng
- SJTU-Ruijin-UIH Institute for Medical Imaging Technology Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengtian Ma
- Department of Radiology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yunjie Liao
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yongxiang Tang
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China
| | - Yu Wen
- Furong Laboratory, Central South University, Changsha, Hunan, 410008, China
| | - Jun Liu
- Department of Radiology, Second Xiangya Hospital of Central South University, 139 Renming Middle Road Changsha, Changsha, Hunan, China.
| | - Shuo Hu
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders (XIANGYA), Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Key Laboratory of Biological Nanotechnology of National Health Commission, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
6
|
Yosef O, Cohen-Daniel L, Shamriz O, Bar-On Z, Salaymeh W, Saragovi A, Abramovich I, Agranovich B, Lutz V, Tam J, Permyakova A, Gottlieb E, Huber M, Berger M. Metabolic reprogramming driven by Ant2 deficiency augments T Cell function and anti-tumor immunity in mice. Nat Commun 2025; 16:4292. [PMID: 40341170 PMCID: PMC12062294 DOI: 10.1038/s41467-025-59310-3] [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: 08/29/2024] [Accepted: 04/18/2025] [Indexed: 05/10/2025] Open
Abstract
T cell activation requires a substantial increase in NAD+ production, often exceeding the capacity of oxidative phosphorylation (OXPHOS). To investigate how T cells adapt to this metabolic challenge, we generate T cell-specific ADP/ATP translocase-2 knockout (Ant2-/-) mice. Loss of Ant2, a crucial protein mediating ADP/ATP exchange between mitochondria and cytoplasm, induces OXPHOS restriction by limiting ATP synthase activity, thereby impeding NAD+ regeneration. Interestingly, Ant2-/- naïve T cells exhibit enhanced activation, proliferation and effector functions compared to wild-type controls. Metabolic profiling reveals that these T cells adopt an activated-like metabolic program with increased mitobiogenesis and anabolism. Lastly, pharmacological inhibition of ANT in wild-type T cells recapitulates the Ant2-/- phenotype and improves adoptive T cell therapy of cancer in mouse models. Our findings thus suggest that Ant2-deficient T cells bypass the typical metabolic reprogramming required for activation, leading to enhanced T cell function and highlighting the therapeutic potential of targeting ANT for immune modulation.
Collapse
Affiliation(s)
- Omri Yosef
- The Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Leonor Cohen-Daniel
- The Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Oded Shamriz
- The Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Zahala Bar-On
- The Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Wajeeh Salaymeh
- The Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Amijai Saragovi
- The Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ifat Abramovich
- Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Bella Agranovich
- Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Veronika Lutz
- Institute of Systems Immunology, Philipps University of Marburg, Marburg, Germany
| | - Joseph Tam
- Obesity and Metabolism Laboratory, The Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Anna Permyakova
- Obesity and Metabolism Laboratory, The Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Eyal Gottlieb
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Magdalena Huber
- Institute of Systems Immunology, Philipps University of Marburg, Marburg, Germany
| | - Michael Berger
- The Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
7
|
Brisudova P, Stojanovic D, Novak J, Nahacka Z, Oliveira GL, Vanatko O, Dvorakova S, Endaya B, Truksa J, Kubiskova M, Foltynova A, Jirak D, Jirat-Ziolkowska N, Kucera L, Chalupsky K, Klima K, Prochazka J, Sedlacek R, Mengarelli F, Orlando P, Tiano L, Oliveira P, Grasso C, Berridge MV, Zobalova R, Anderova M, Neuzil J. Functional mitochondrial respiration is essential for glioblastoma tumour growth. Oncogene 2025:10.1038/s41388-025-03429-6. [PMID: 40325182 DOI: 10.1038/s41388-025-03429-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 04/14/2025] [Accepted: 04/24/2025] [Indexed: 05/07/2025]
Abstract
Horizontal transfer of mitochondria from the tumour microenvironment to cancer cells to support proliferation and enhance tumour progression has been shown for various types of cancer in recent years. Glioblastoma, the most aggressive adult brain tumour, has proven to be no exception when it comes to dynamic intercellular mitochondrial movement, as shown in this study using an orthotopic tumour model of respiration-deficient glioblastoma cells. Although confirmed mitochondrial transfer was shown to facilitate tumour progression in glioblastoma, we decided to investigate whether the related electron transport chain recovery is necessary for tumour formation in the brain. Based on experiments using time-resolved analysis of tumour formation by glioblastoma cells depleted of their mitochondrial DNA, we conclude that functional mitochondrial respiration is essential for glioblastoma growth in vivo, because it is needed to support coenzyme Q redox cycling for de novo pyrimidine biosynthesis controlled by respiration-linked dihydroorotate dehydrogenase enzyme activity. We also demonstrate here that astrocytes are key mitochondrial donors in this model.
Collapse
Affiliation(s)
- Petra Brisudova
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Vestec, Czech Republic
- Faculty of Science, Charles University, 128 00, Prague 2, Czech Republic
| | - Dana Stojanovic
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Vestec, Czech Republic
- Faculty of Science, Charles University, 128 00, Prague 2, Czech Republic
| | - Jaromir Novak
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Vestec, Czech Republic
- Faculty of Science, Charles University, 128 00, Prague 2, Czech Republic
| | - Zuzana Nahacka
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Vestec, Czech Republic
| | - Gabriela Lopes Oliveira
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Vestec, Czech Republic
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, 3060-197, Cantanhede, Portugal
- CCIBB, Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3060-197, Cantanhede, Portugal
- PPDBEB, Institute for Interdisciplinary Research, Doctoral Programme in Experimental Biology and Biomedicine, University of Coimbra, 3060-197, Cantanhede, Portugal
| | - Ondrej Vanatko
- Institute of Experimental Medicine, Czech Academy of Sciences, 142 00, Prague 4, Czech Republic
- Second Faculty of Medicine, Charles University, 150 06, Prague 5, Czech Republic
| | - Sarka Dvorakova
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Vestec, Czech Republic
| | - Berwini Endaya
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Vestec, Czech Republic
| | - Jaroslav Truksa
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Vestec, Czech Republic
| | - Monika Kubiskova
- Institute of Experimental Medicine, Czech Academy of Sciences, 142 00, Prague 4, Czech Republic
- Second Faculty of Medicine, Charles University, 150 06, Prague 5, Czech Republic
| | - Alice Foltynova
- Institute of Experimental Medicine, Czech Academy of Sciences, 142 00, Prague 4, Czech Republic
- Second Faculty of Medicine, Charles University, 150 06, Prague 5, Czech Republic
| | - Daniel Jirak
- Institute of Clinical and Experimental Medicine, 140 21, Prague 4, Czech Republic
| | - Natalia Jirat-Ziolkowska
- Institute of Clinical and Experimental Medicine, 140 21, Prague 4, Czech Republic
- Institute of Biophysics and Informatics, First Faculty of Medicine, Charles University, 121 08, Prague 4, Czech Republic
| | - Lukas Kucera
- Czech Centre for Phenogenomics, Institute of Molecular Genetics, Czech Academy of Sciences, 142 20, Prague 4, Czech Republic
| | - Karel Chalupsky
- Czech Centre for Phenogenomics, Institute of Molecular Genetics, Czech Academy of Sciences, 142 20, Prague 4, Czech Republic
| | - Krystof Klima
- Czech Centre for Phenogenomics, Institute of Molecular Genetics, Czech Academy of Sciences, 142 20, Prague 4, Czech Republic
| | - Jan Prochazka
- Czech Centre for Phenogenomics, Institute of Molecular Genetics, Czech Academy of Sciences, 142 20, Prague 4, Czech Republic
| | - Radislav Sedlacek
- Czech Centre for Phenogenomics, Institute of Molecular Genetics, Czech Academy of Sciences, 142 20, Prague 4, Czech Republic
| | - Francesco Mengarelli
- Department of Life and Environmental Sciences, Polytechnic University of Marche, 60131, Ancona, Italy
| | - Patrick Orlando
- Department of Life and Environmental Sciences, Polytechnic University of Marche, 60131, Ancona, Italy
| | - Luca Tiano
- Department of Life and Environmental Sciences, Polytechnic University of Marche, 60131, Ancona, Italy
| | - Paulo Oliveira
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, 3060-197, Cantanhede, Portugal
- CCIBB, Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3060-197, Cantanhede, Portugal
| | - Carole Grasso
- Malaghan Institute of Medical Research, Wellington, 6242, New Zealand
| | | | - Renata Zobalova
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Vestec, Czech Republic.
| | - Miroslava Anderova
- Institute of Experimental Medicine, Czech Academy of Sciences, 142 00, Prague 4, Czech Republic.
| | - Jiri Neuzil
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Vestec, Czech Republic.
- Faculty of Science, Charles University, 128 00, Prague 2, Czech Republic.
- First Faculty of Medicine, Charles University, 121 08, Prague 2, Czech Republic.
- School of Pharmacy and Medical Science, Griffith University, Southport, QLD, 4222, Australia.
| |
Collapse
|
8
|
Li H, Hu Q, Zhu D, Wu D. The Role of NAD + Metabolism in Cardiovascular Diseases: Mechanisms and Prospects. Am J Cardiovasc Drugs 2025; 25:307-327. [PMID: 39707143 DOI: 10.1007/s40256-024-00711-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/03/2024] [Indexed: 12/23/2024]
Abstract
Nicotinamide adenine dinucleotide (NAD+) is a promising anti-aging molecule that plays a role in cellular energy metabolism and maintains redox homeostasis. Additionally, NAD+ is involved in regulating deacetylases, DNA repair enzymes, inflammation, and epigenetics, making it indispensable in maintaining the basic functions of cells. Research on NAD+ has become a hotspot, particularly regarding its potential in cardiovascular disease (CVD). Many studies have demonstrated that NAD+ plays a crucial role in the occurrence and development of CVD. This review summarizes the biosynthesis and consumption of NAD+, along with its precursors and their effects on raising NAD+ levels. We also discuss new mechanisms of NAD+ regulation in cardiovascular risk factors and its effects of NAD+ on atherosclerosis, aortic aneurysm, heart failure, hypertension, myocardial ischemia-reperfusion injury, diabetic cardiomyopathy, and dilated cardiomyopathy, elucidating different mechanisms and potential treatments. NAD+-centered therapy holds promising advantages and prospects in the field of CVD.
Collapse
Affiliation(s)
- Huimin Li
- Department of Pharmacy, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Qingxun Hu
- Department of Pharmacy, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Deqiu Zhu
- Department of Pharmacy, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China.
| | - Dan Wu
- Department of Pharmacy, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China.
| |
Collapse
|
9
|
MacDonald TL, Ryback B, Aparecida da Silva Pereira J, Wei S, Mendez B, Cai EP, Ishikawa Y, Arbeau M, Weir G, Bonner-Weir S, Kissler S, Yi P. Renalase inhibition defends against acute and chronic β cell stress by regulating cell metabolism. Mol Metab 2025; 95:102115. [PMID: 39988068 PMCID: PMC11981795 DOI: 10.1016/j.molmet.2025.102115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 02/18/2025] [Accepted: 02/18/2025] [Indexed: 02/25/2025] Open
Abstract
OBJECTIVE Renalase (Rnls) is annotated as an oxidase enzyme. It has been implicated in Type 1 diabetes (T1D) risk via genome-wide association studies (GWAS). We previously discovered through CRISPR screening and validation experiments that Rnls inhibition prevents or delays T1D in multiple mouse models of diabetes in vivo, and protects pancreatic β cells against autoimmune killing, ER and oxidative stress in vitro. The molecular biochemistry and functions of Rnls are largely uncharted. Here we studied the mechanisms of Rnls inhibition that underlie β cell protection during diabetogenic stress. METHODS Akita mice were treated with oral Pargyline (PG) in vivo to bind and inhibit Rnls, and pancreas or islets were harvested for β cell mass and β cell function analyses. Genetic and pharmacological tools were used to inhibit Rnls in β cell lines. RNA sequencing, metabolomics and metabolic function experiments were conducted in vitro in NIT-1 mouse β cell lines and human stem cell-derived β cells. RESULTS In vivo, PG improved glycemia and mildly preserved β cell mass and function in females. Genetic strategies to mutate (Rnlsmut) or knockout (Rnls KO) Rnls induced a robust metabolic shift towards glycolysis in both mouse and human β cell lines, in vitro. Stress protection was abolished when glycolysis was blocked with 2-deoxyglucose (2-DG). Pharmacological Rnls inhibition with PG did not strongly mimic these newly identified metabolic mechanisms. CONCLUSIONS Our work illustrates a role for Rnls in regulating cell metabolism. We show that inhibiting Rnls protects against chronic stress in vivo, and shields against acute stress in β cell lines in vitro by rewiring cell metabolism towards glycolysis.
Collapse
Affiliation(s)
- Tara L MacDonald
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, USA; Section for Immunobiology, Joslin Diabetes Center, Boston, USA; Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Department of Medicine, Harvard Medical School, Boston, MA, USA.
| | - Birgitta Ryback
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, USA
| | - Jéssica Aparecida da Silva Pereira
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Siying Wei
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Bryhan Mendez
- Section for Immunobiology, Joslin Diabetes Center, Boston, USA
| | - Erica P Cai
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Yuki Ishikawa
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Meagan Arbeau
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Gordon Weir
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA; Diabetes Program, Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Susan Bonner-Weir
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA; Diabetes Program, Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Stephan Kissler
- Section for Immunobiology, Joslin Diabetes Center, Boston, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA; Diabetes Program, Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Peng Yi
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA; Diabetes Program, Harvard Stem Cell Institute, Cambridge, MA, USA.
| |
Collapse
|
10
|
Kim D, Kesavan R, Ryu K, Dey T, Marckx A, Menezes C, Praharaj PP, Morley S, Ko B, Soflaee MH, Tom HJ, Brown H, Vu HS, Tso SC, Brautigam CA, Lemoff A, Mettlen M, Mishra P, Cai F, Allen DK, Hoxhaj G. Mitochondrial NADPH fuels mitochondrial fatty acid synthesis and lipoylation to power oxidative metabolism. Nat Cell Biol 2025; 27:790-800. [PMID: 40258949 DOI: 10.1038/s41556-025-01655-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 03/12/2025] [Indexed: 04/23/2025]
Abstract
Nicotinamide adenine dinucleotide phosphate (NADPH) is a vital electron donor essential for macromolecular biosynthesis and protection against oxidative stress. Although NADPH is compartmentalized within the cytosol and mitochondria, the specific functions of mitochondrial NADPH remain largely unexplored. Here we demonstrate that NAD+ kinase 2 (NADK2), the principal enzyme responsible for mitochondrial NADPH production, is critical for maintaining protein lipoylation, a conserved lipid modification necessary for the optimal activity of multiple mitochondrial enzyme complexes, including the pyruvate dehydrogenase complex. The mitochondrial fatty acid synthesis (mtFAS) pathway utilizes NADPH for generating protein-bound acyl groups, including lipoic acid. By developing a mass-spectrometry-based method to assess mammalian mtFAS, we reveal that NADK2 is crucial for mtFAS activity. NADK2 deficiency impairs mtFAS-associated processes, leading to reduced cellular respiration and mitochondrial translation. Our findings support a model in which mitochondrial NADPH fuels the mtFAS pathway, thereby sustaining protein lipoylation and mitochondrial oxidative metabolism.
Collapse
Affiliation(s)
- Dohun Kim
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Rushendhiran Kesavan
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kevin Ryu
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Trishna Dey
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Austin Marckx
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Cameron Menezes
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Prakash P Praharaj
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Stewart Morley
- Donald Danforth Plant Science Center, St. Louis, MO, USA
| | - Bookyung Ko
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Mona H Soflaee
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Harrison J Tom
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Harrison Brown
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Hieu S Vu
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Shih-Chia Tso
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Chad A Brautigam
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Andrew Lemoff
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Marcel Mettlen
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Prashant Mishra
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Feng Cai
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Doug K Allen
- Donald Danforth Plant Science Center, St. Louis, MO, USA
- United States Department of Agriculture, Agriculture Research Service, St. Louis, MO, USA
| | - Gerta Hoxhaj
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
11
|
Jackson BT, Montero AM, Chakraborty S, Brunner JS, Arnold PK, Bridgeman AE, Todorova PK, Paras KI, Finley LWS. Intracellular metabolic gradients dictate dependence on exogenous pyruvate. Nat Metab 2025:10.1038/s42255-025-01289-8. [PMID: 40295877 DOI: 10.1038/s42255-025-01289-8] [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: 06/24/2024] [Accepted: 03/25/2025] [Indexed: 04/30/2025]
Abstract
During developmental transitions, cells frequently remodel metabolic networks, including changing reliance on metabolites such as glucose and glutamine to fuel intracellular metabolic pathways. Here we used embryonic stem (ES) cells as a model system to understand how changes in intracellular metabolic networks that characterize cell state transitions affect reliance on exogenous nutrients. We find that ES cells in the naive ground state of pluripotency increase uptake and reliance on exogenous pyruvate through the monocarboxylate transporter MCT1. Naive ES cells, but not their more committed counterparts, rely on exogenous pyruvate even when other sources of pyruvate (glucose, lactate) are abundant. Pyruvate dependence in naive ES cells is a consequence of their elevated mitochondrial pyruvate consumption at the expense of cytosolic NAD+ regeneration. Indeed, across a range of cell types, increased mitochondrial pyruvate consumption is sufficient to drive demand for extracellular pyruvate. Accordingly, restoring cytosolic NAD+ regeneration allows naive ES cells to tolerate pyruvate depletion in diverse nutrient microenvironments. Together, these data demonstrate that intracellular metabolic gradients dictate uptake and reliance on exogenous pyruvate and highlight mitochondrial pyruvate metabolism as a metabolic vulnerability of naive ES cells.
Collapse
Affiliation(s)
- Benjamin T Jackson
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, New York, NY, USA
| | - Angela M Montero
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY, USA
| | - Sangita Chakraborty
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Julia S Brunner
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Paige K Arnold
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, New York, NY, USA
| | - Anna E Bridgeman
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Pavlina K Todorova
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Katrina I Paras
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY, USA
| | - Lydia W S Finley
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
12
|
Schmidt M, Binder H, Schneider MR. The metabolic underpinnings of sebaceous lipogenesis. Commun Biol 2025; 8:670. [PMID: 40289206 PMCID: PMC12034822 DOI: 10.1038/s42003-025-08105-9] [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: 02/24/2025] [Accepted: 04/17/2025] [Indexed: 04/30/2025] Open
Abstract
Sebaceous glands synthesize and secrete sebum, a mélange of lipids and other cellular products that safeguards the mammalian integument. Differentiating sebocytes delaminate from the basal membrane and dislodge towards the gland's middle, where they eventually undergo a poorly understood death mode in which the whole cell becomes a secretion product (holocrine secretion). Supported by recent transcriptomics data, this review examines the idea that peripheral sebocytes have a remarkable ability to draw nutrients from the blood and become committed to unrestrainedly invest all available resources into synthetic processes for accomplishing sebum synthesis, thereby exploiting core metabolic fluxes as glycogen turnover, glutamine-directed anaplerosis, the pentose phosphate pathway and de novo lipogenesis. Finally, we propose that metabolic-driven processes are an important mechanistic component of holocrine secretion. A deeper understanding of these metabolic adaptations could indicate novel strategies for modulating sebum synthesis, a key pathogenic factor in acne and other skin diseases.
Collapse
Affiliation(s)
- Maria Schmidt
- Interdisciplinary Institute for Bioinformatics (IZBI), University of Leipzig, Leipzig, Germany
| | - Hans Binder
- Interdisciplinary Institute for Bioinformatics (IZBI), University of Leipzig, Leipzig, Germany
- Armenian Bioinformatics Institute (ABI), Yerevan, Armenia
| | - Marlon R Schneider
- Institute of Veterinary Physiology, Veterinary Faculty, University of Leipzig, Leipzig, Germany.
| |
Collapse
|
13
|
Xing W, Li M, Wang B, Huo L, Tian W, Ge F, Shen M, Sun L, Liu J, Yu S. Effect of 1-DNJ on Oxidative Stress-Induced Apoptosis in Porcine Ovarian GCs Through Modulation of the PERK-ATF4/MFN2 Signaling Pathway. Antioxidants (Basel) 2025; 14:456. [PMID: 40298787 PMCID: PMC12024285 DOI: 10.3390/antiox14040456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/07/2025] [Accepted: 04/08/2025] [Indexed: 04/30/2025] Open
Abstract
Oxidative stress (OS) is regarded as a major contributor to granulosa cellapoptosis in ovarian disease. 1-Deoxynojirimycin (1-DNJ), a naturally occurring plant alkaloid, exhibits antioxidant, anti-inflammatory, and metabolism-modulating properties. Mitochondria and endoplasmic reticulum (ER), crucial organelles regulating oxidative balance, interact through mitochondria-associated endoplasmic reticulum membranes (MAMs) for signaling and molecular exchange. However, it remains unclear whether 1-DNJ attenuates oxidative damage in ovarian granulosa cells (GCs) via MAMs-mediated ER-mitochondria crosstalk, which needs further exploration. This study aimed to investigate the mechanisms by which 1-DNJ affects oxidative damage and apoptosis induced by OS in porcine follicular GCs by regulating mitochondrial function, MAMs, and ER interactions. Here, we found that GCs suffered from OS, accompanied by the up-regulation of ROS and MDA, alongside reduced activity of antioxidant enzymes (CAT and T-SOD). Further studies revealed that the up-regulation of MAMs proteins (MFN2, MCU, and VDAC1) and pro-apoptosis proteins (BAX and Cleaved-capase3), along with increased mitochondrial ROS and Ca2+ levels, led to the down-regulation of MMP and ATP content. These, in turn, triggered mitochondrial dysfunction, and MAMs destabilization, and subsequent apoptosis. Additionally, the up-regulation of the protein levels of P-PERK/PERK, GRP78, ATF4, and CHOP protein expression activated the PERK-ATF4 signaling pathway, which triggered endoplasmic reticulum stress (ERS). Conversely, 1-DNJ alleviated H2O2-induced mitochondrial and MAMs dysfunction and ERS, which in turn attenuated apoptosis. Further, ATF4 knockdown inhibited MFN2 protein expression, which attenuated H2O2-induced MMP inhibition, Ca2+ overload, ROS production, and mitochondrial damage. In summary, 1-DNJ mitigated OS-induced mitochondrial dysfunction in GCs and regulated ER-mitochondrial communication through MAMs, reducing OS-induced apoptosis. The present study demonstrates that 1-DNJ protects ovarian GCs from OS-induced damage by modulating ER and mitochondrial homeostasis through MAMs, offering new perspectives and a theoretical basis for the treatment of ovarian diseases.
Collapse
Affiliation(s)
- Wenwen Xing
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (W.X.); (M.L.); (B.W.); (L.H.); (W.T.); (F.G.); (M.S.); (L.S.)
| | - Mengxuan Li
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (W.X.); (M.L.); (B.W.); (L.H.); (W.T.); (F.G.); (M.S.); (L.S.)
| | - Binbin Wang
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (W.X.); (M.L.); (B.W.); (L.H.); (W.T.); (F.G.); (M.S.); (L.S.)
| | - Lele Huo
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (W.X.); (M.L.); (B.W.); (L.H.); (W.T.); (F.G.); (M.S.); (L.S.)
| | - Wanru Tian
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (W.X.); (M.L.); (B.W.); (L.H.); (W.T.); (F.G.); (M.S.); (L.S.)
| | - Fangcai Ge
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (W.X.); (M.L.); (B.W.); (L.H.); (W.T.); (F.G.); (M.S.); (L.S.)
| | - Manman Shen
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (W.X.); (M.L.); (B.W.); (L.H.); (W.T.); (F.G.); (M.S.); (L.S.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Liumei Sun
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (W.X.); (M.L.); (B.W.); (L.H.); (W.T.); (F.G.); (M.S.); (L.S.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Jiying Liu
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (W.X.); (M.L.); (B.W.); (L.H.); (W.T.); (F.G.); (M.S.); (L.S.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Shali Yu
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| |
Collapse
|
14
|
Hou Y, Liu D, Guo Z, Wei C, Cao F, Xu Y, Feng Q, Liu F. Lactate and Lactylation in AKI-to-CKD: Epigenetic Regulation and Therapeutic Opportunities. Cell Prolif 2025:e70034. [PMID: 40207870 DOI: 10.1111/cpr.70034] [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: 12/27/2024] [Revised: 02/20/2025] [Accepted: 03/21/2025] [Indexed: 04/11/2025] Open
Abstract
Lactate is not only a byproduct of glycolysis, but is also considered an energy source, gluconeogenic precursor, signalling molecule and protein modifier during the process of cellular metabolism. The discovery of lactylation reveals the multifaceted functions of lactate in cellular metabolism and opens new avenues for lactate-related research. Both lactate and lactylation have been implicated in regulating numerous biological processes, including tumour progression, ischemic-hypoxic injury, neurodevelopment and immune-related inflammation. The kidney plays a crucial role in regulating lactate metabolism, influencing lactate levels while also being regulated by lactate. Previous studies have demonstrated the importance of lactate in the pathogenesis of acute kidney injury (AKI) and chronic kidney disease (CKD). This review explores the role of lactate and lactylation in these diseases, comparing the function and metabolic mechanisms of lactate in normal and diseased kidneys from the perspective of lactylation. The key regulatory roles of lactylation in different organs, multiple systems, various pathological states and underlying mechanisms in AKI-to-CKD progression are summarised. Moreover, potential therapeutic targets and future research directions for lactate and lactylation across multiple kidney diseases are identified.
Collapse
Affiliation(s)
- Yi Hou
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, Henan Province, China
- Henan Province Research Center for Kidney Disease, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Dongwei Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, Henan Province, China
- Henan Province Research Center for Kidney Disease, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
- Innovation Center of Basic Research for Metabolic-Associated Fatty Liver Disease, Ministry of Education of China, Zhengzhou, Henan Province, China
| | - Zuishuang Guo
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, Henan Province, China
- Henan Province Research Center for Kidney Disease, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
- Innovation Center of Basic Research for Metabolic-Associated Fatty Liver Disease, Ministry of Education of China, Zhengzhou, Henan Province, China
| | - Cien Wei
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, Henan Province, China
- Henan Province Research Center for Kidney Disease, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Fengyu Cao
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, Henan Province, China
- Henan Province Research Center for Kidney Disease, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yue Xu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, Henan Province, China
- Henan Province Research Center for Kidney Disease, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Qi Feng
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, Henan Province, China
- Henan Province Research Center for Kidney Disease, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
- Innovation Center of Basic Research for Metabolic-Associated Fatty Liver Disease, Ministry of Education of China, Zhengzhou, Henan Province, China
| | - Fengxun Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, Henan Province, China
- Henan Province Research Center for Kidney Disease, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
- Innovation Center of Basic Research for Metabolic-Associated Fatty Liver Disease, Ministry of Education of China, Zhengzhou, Henan Province, China
| |
Collapse
|
15
|
Seyfried TN, Lee DC, Duraj T, Ta NL, Mukherjee P, Kiebish M, Arismendi-Morillo G, Chinopoulos C. The Warburg hypothesis and the emergence of the mitochondrial metabolic theory of cancer. J Bioenerg Biomembr 2025:10.1007/s10863-025-10059-w. [PMID: 40199815 DOI: 10.1007/s10863-025-10059-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/20/2025] [Indexed: 04/10/2025]
Abstract
Otto Warburg originally proposed that cancer arose from a two-step process. The first step involved a chronic insufficiency of mitochondrial oxidative phosphorylation (OxPhos), while the second step involved a protracted compensatory energy synthesis through lactic acid fermentation. His extensive findings showed that oxygen consumption was lower while lactate production was higher in cancerous tissues than in non-cancerous tissues. Warburg considered both oxygen consumption and extracellular lactate as accurate markers for ATP production through OxPhos and glycolysis, respectively. Warburg's hypothesis was challenged from findings showing that oxygen consumption remained high in some cancer cells despite the elevated production of lactate suggesting that OxPhos was largely unimpaired. New information indicates that neither oxygen consumption nor lactate production are accurate surrogates for quantification of ATP production in cancer cells. Warburg also did not know that a significant amount of ATP could come from glutamine-driven mitochondrial substrate level phosphorylation in the glutaminolysis pathway with succinate produced as end product, thus confounding the linkage of oxygen consumption to the origin of ATP production within mitochondria. Moreover, new information shows that cytoplasmic lipid droplets and elevated aerobic lactic acid fermentation are both biomarkers for OxPhos insufficiency. Warburg's original hypothesis can now be linked to a more complete understanding of how OxPhos insufficiency underlies dysregulated cancer cell growth. These findings can also address several questionable assumptions regarding the origin of cancer thus allowing the field to advance with more effective therapeutic strategies for a less toxic metabolic management and prevention of cancer.
Collapse
Affiliation(s)
- Thomas N Seyfried
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA.
| | - Derek C Lee
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA
| | - Tomas Duraj
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA
| | - Nathan L Ta
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA
| | - Purna Mukherjee
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA
| | | | - Gabriel Arismendi-Morillo
- Facultad de Medicina, Instituto de Investigaciones Biológicas, Universidad del Zulia, Maracaibo, Venezuela
- Department of Medicine, Faculty of Health Sciences, University of Deusto, Bilbao (Bizkaia), Spain
| | - Christos Chinopoulos
- Department of Medical Biochemistry, Semmelweis University, Budapest, 1094, Hungary
| |
Collapse
|
16
|
Cai H, Chen X, Liu Y, Chen Y, Zhong G, Chen X, Rong S, Zeng H, Zhang L, Li Z, Liao A, Zeng X, Xiong W, Guo C, Zhu Y, Deng KQ, Ren H, Yan H, Cai Z, Xu K, Zhou L, Lu Z, Wang F, Liu S. Lactate activates trained immunity by fueling the tricarboxylic acid cycle and regulating histone lactylation. Nat Commun 2025; 16:3230. [PMID: 40185732 PMCID: PMC11971257 DOI: 10.1038/s41467-025-58563-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 03/24/2025] [Indexed: 04/07/2025] Open
Abstract
Trained immunity refers to the long-term memory of the innate immune cells. However, little is known about how environmental nutrient availability influences trained immunity. This study finds that physiologic carbon sources impact glucose contribution to the tricarboxylic acid (TCA) cycle and enhance cytokine production of trained monocytes. Our experiments demonstrate that trained monocytes preferentially employe lactate over glucose as a TCA cycle substrate, and lactate metabolism is required for trained immune cell responses to bacterial and fungal infection. Except for the contribution to the TCA cycle, endogenous lactate or exogenous lactate also supports trained immunity by regulating histone lactylation. Further transcriptome analysis, ATAC-seq, and CUT&Tag-seq demonstrate that lactate enhance chromatin accessibility in a manner dependent histone lactylation. Inhibiting lactate-dependent metabolism by silencing lactate dehydrogenase A (LDHA) impairs both lactate fueled the TCA cycle and histone lactylation. These findings suggest that lactate is the hub of immunometabolic and epigenetic programs in trained immunity.
Collapse
Affiliation(s)
- Huanhuan Cai
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, 430072, China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, 430072, China
| | - Xueyuan Chen
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Yan Liu
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Yingbo Chen
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, 430072, China
- Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430072, China
| | - Gechang Zhong
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, 430072, China
- Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430072, China
| | - Xiaoyu Chen
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, 430072, China
- Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430072, China
| | - Shuo Rong
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, 430072, China
- Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430072, China
| | - Hao Zeng
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430072, China
| | - Lin Zhang
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, 430072, China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, 430072, China
| | - Zelong Li
- Department of General Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
- Department of Infectious Diseases, Longnan First People's Hospital, Longnan, 341700, China
| | - Aihua Liao
- Department of General Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
- Department of Infectious Diseases, Longnan First People's Hospital, Longnan, 341700, China
| | - Xiangtai Zeng
- Department of General Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
- Department of Infectious Diseases, Longnan First People's Hospital, Longnan, 341700, China
| | - Wei Xiong
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, 430072, China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, 430072, China
| | - Cihang Guo
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, 430072, China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, 430072, China
| | - Yanfang Zhu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, 430072, China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, 430072, China
| | - Ke-Qiong Deng
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, 430072, China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, 430072, China
| | - Hong Ren
- Shanghai Children's Medical Center, Affiliated Hospital to Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, China
| | - Huan Yan
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, 430072, China
- Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430072, China
| | - Zeng Cai
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, 430072, China
- Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430072, China
| | - Ke Xu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, 430072, China
- Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430072, China
| | - Li Zhou
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, 430072, China
- Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430072, China
| | - Zhibing Lu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, 430072, China.
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, 430072, China.
| | - Fubing Wang
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, 430072, China.
| | - Shi Liu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, 430072, China.
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, 430072, China.
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, 430072, China.
- Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430072, China.
| |
Collapse
|
17
|
Chen J, Liao Y, Sheng Y, Yao H, Li T, He Z, Ye WWY, Yin M, Tang H, Zhao Y, Zhang P, Wang Y, Fu X, Ji Y. FSH exacerbates bone loss by promoting osteoclast energy metabolism through the CREB-MDH2-NAD + axis. Metabolism 2025; 165:156147. [PMID: 39880362 DOI: 10.1016/j.metabol.2025.156147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/07/2025] [Accepted: 01/25/2025] [Indexed: 01/31/2025]
Abstract
AIMS Osteoclast energy metabolism is a promising target for treating diseases characterized by high osteoclast activity, such as osteoporosis. However, the regulatory factors involved in osteoclast bioenergetic processes are still in the early stages of being fully understood. This study reveals the effects of follicle-stimulating hormone (FSH) on osteoclast energy metabolism. METHODS The Lyz2-Cre-Flox model selectively deletes FSH receptor (FSHR) from osteoclast precursor cells to generate Fshrf/f; Lyz2-Cre (Fshrf/f; Cre) mice. Bone quality was assessed using micro-computed tomography, histomorphometric analysis, and dual-fluorescence labeling. The in vitro assays measured oxygen consumption rate, extracellular acidification rate, pyruvate content, and mitochondrial membrane potential to determine metabolic flux. RNA-seq, LC-MS, dual-luciferase reporter assays, and chromatin immunoprecipitation (ChIP) assays were used to elucidate the underlying mechanisms. RESULTS FSHR deficiency in osteoclasts protected bone from resorption under normal and ovariectomized conditions. FSHR-deficient osteoclasts have reduced nicotinamide adenine dinucleotide (NAD+) levels, impairing osteoclast activity and energy metabolism. Mechanistically, FSH influenced NAD+ levels via the CREB/MDH2 axis. Treatment with FSH monoclonal antibodies rescued bone loss in OVX mice and reduced bone marrow NAD+ levels. CONCLUSIONS Targeting FSH may be a promising metabolic modulation strategy for treating osteoporosis and other diseases associated with high osteoclast activity.
Collapse
Affiliation(s)
- Jingqiu Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, PR China
| | - Yilin Liao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, PR China
| | - Yue Sheng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, PR China
| | - Hantao Yao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, PR China
| | - Ting Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, PR China
| | - Zhenru He
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, PR China
| | - Weng Wan Yue Ye
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, PR China
| | - Mengjie Yin
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, PR China
| | - Huilin Tang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, PR China
| | - Yaoyu Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, PR China
| | - Peiqi Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, PR China
| | - Yuting Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, PR China
| | - Xiazhou Fu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, PR China.
| | - Yaoting Ji
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, PR China.
| |
Collapse
|
18
|
Lien EC, Vu N, Westermark AM, Danai LV, Lau AN, Gültekin Y, Kukurugya MA, Bennett BD, Vander Heiden MG. Effects of Aging on Glucose and Lipid Metabolism in Mice. Aging Cell 2025; 24:e14462. [PMID: 39731205 PMCID: PMC11984682 DOI: 10.1111/acel.14462] [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] [Revised: 12/08/2024] [Accepted: 12/10/2024] [Indexed: 12/29/2024] Open
Abstract
Aging is accompanied by multiple molecular changes that contribute to aging associated pathologies, such as accumulation of cellular damage and mitochondrial dysfunction. Tissue metabolism can also change with age, in part, because mitochondria are central to cellular metabolism. Moreover, the cofactor NAD+, which is reported to decline across multiple tissues during aging, plays a central role in metabolic pathways such as glycolysis, the tricarboxylic acid cycle, and the oxidative synthesis of nucleotides, amino acids, and lipids. To further characterize how tissue metabolism changes with age, we intravenously infused [U-13C]-glucose into young and old C57BL/6J, WSB/EiJ, and diversity outbred mice to trace glucose fate into downstream metabolites within plasma, liver, gastrocnemius muscle, and brain tissues. We found that glucose incorporation into central carbon and amino acid metabolism was robust during healthy aging across these different strains of mice. We also observed that levels of NAD+, NADH, and the NAD+/NADH ratio were unchanged in these tissues with healthy aging. However, aging tissues, particularly brain, exhibited evidence of upregulated fatty acid and sphingolipid metabolism reactions that regenerate NAD+ from NADH. These data suggest that NAD+-generating lipid metabolism reactions may help to maintain the NAD+/NADH ratio during healthy aging.
Collapse
Affiliation(s)
- Evan C. Lien
- Department of Metabolism and Nutritional ProgrammingVan Andel InstituteGrand RapidsMichiganUSA
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Ngoc Vu
- Calico Life Sciences LLCSouth San FranciscoCaliforniaUSA
| | - Anna M. Westermark
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Laura V. Danai
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
- Department of Biochemistry and Molecular BiologyUniversity of Massachusetts AmherstAmherstMassachusettsUSA
| | - Allison N. Lau
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Yetiş Gültekin
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | | | | | - Matthew G. Vander Heiden
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
- Department of Medical OncologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
- Department of BiologyMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| |
Collapse
|
19
|
Wu Z, Nguyen PT, Sondhi V, Yao RW, Dai T, Chiang JC, Shang Z, Cai F, Cai L, Zhang J, Moore MD, Alshamleh I, Li X, Ogu T, Zacharias LG, Winston R, Patricio JS, Johnson X, Chen WM, Cong Q, Mathews TP, Zhang Y, DeBerardinis RJ. NUDT5 regulates purine metabolism and thiopurine sensitivity by interacting with PPAT. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.29.646096. [PMID: 40236075 PMCID: PMC11996305 DOI: 10.1101/2025.03.29.646096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Cells generate purine nucleotides through both de novo purine biosynthesis (DNPB) and purine salvage. Purine accumulation represses energetically costly DNPB through feedback inhibition of the enzymatic steps that produce the precursor phosphoribosylamine. Excessive DNPB is associated with human diseases including neurological dysfunction and hyperuricemia. However, the mechanisms explaining how cells balance DNPB and purine salvage are incompletely understood. Data from a genome-wide CRISPR loss-of-function screen and extensive stable isotope tracing identified Nudix hydrolase 5 (NUDT5) as a suppressor of DNPB during purine salvage. NUDT5 ablation allows DNPB to persist in the presence of either native purines or thiopurine drugs; this renders NUDT5-deficient cells insensitive to thiopurine treatment. Surprisingly, this regulation occurs independently of NUDT5's known function in hydrolyzing ADP-ribose to AMP and ribose-5-phosphate. Rather, NUDT5 interacts with phosphoribosyl pyrophosphate amidotransferase (PPAT), the rate-limiting enzyme in DNPB that generates phosphoribosylamine. Upon induction of purine salvage, the PPAT-NUDT5 interaction is required to trigger disassembly of the purinosome, a cytosolic metabolon involved in efficient DNPB. Mutations that disrupt NUDT5's interaction with PPAT but leave its catalytic activity intact permit excessive DNPB during purine salvage, inducing thiopurine resistance. Collectively, our findings identify NUDT5 as a regulator governing the balance between DNPB and purine salvage, underscoring its impact on nucleotide metabolism and efficacy of thiopurine treatment.
Collapse
|
20
|
Parkhitko AA, Cracan V. Xenotopic synthetic biology: Prospective tools for delaying aging and age-related diseases. SCIENCE ADVANCES 2025; 11:eadu1710. [PMID: 40153513 DOI: 10.1126/sciadv.adu1710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 02/24/2025] [Indexed: 03/30/2025]
Abstract
Metabolic dysregulation represents one of the major driving forces in aging. Although multiple genetic and pharmacological manipulations are known to extend longevity in model organisms, aging is a complex trait, and targeting one's own genes may be insufficient to prevent age-dependent deterioration. An alternative strategy could be to use enzymes from other species to reverse age-associated metabolic changes. In this review, we discuss a set of enzymes from lower organisms that have been shown to affect various metabolic parameters linked to age-related processes. These enzymes include modulators of steady-state levels of amino acids (METase, ASNase, and ADI), NADPH/NADP+ and/or reduced form of coenzyme Q (CoQH2)/CoQ redox potentials (NDI1, AOX, LbNOX, TPNOX, EcSTH, RquA, LOXCAT, Grubraw, and ScURA), GSH (StGshF), mitochondrial membrane potential (mtON and mito-dR), or reactive oxygen species (DAAO and KillerRed-SOD1). We propose that leveraging non-mammalian enzymes represents an untapped resource that can be used to delay aging and age-related diseases.
Collapse
Affiliation(s)
- Andrey A Parkhitko
- Aging Institute of UPMC and the University of Pittsburgh, Pittsburgh, PA, USA
| | - Valentin Cracan
- Laboratory of Redox Biology and Metabolism, Scintillon Institute, San Diego, CA, USA
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
21
|
de Moura Ferreira MA, de Almeida ELM, da Silveira WB, Nikoloski Z. Protein-constrained models pinpoints the role of underground metabolism in robustness of metabolic phenotypes. iScience 2025; 28:112126. [PMID: 40160425 PMCID: PMC11951047 DOI: 10.1016/j.isci.2025.112126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 01/26/2025] [Accepted: 02/25/2025] [Indexed: 04/02/2025] Open
Abstract
Integrating enzyme parameters into constraint-based models have significantly improved the prediction of physiological and molecular traits. To further improve these models, we integrated promiscuous enzyme activities that jointly comprise the so-called underground metabolism by developing the CORAL toolbox, which increases the resolution of modeled enzyme resource allocation. Applying CORAL to a protein-constrained model of Escherichia coli revealed that underground metabolism resulted in larger flexibility of metabolic fluxes and enzyme usage. Simulating metabolic defects where the main activity of a promiscuous enzyme was blocked but promiscuous activities remained functional showed a small enzyme redistribution to the side activities. Further, blocking pairs of main activities showed that non-promiscuous enzymes exhibited larger impact on growth than promiscuous enzymes. These simulations showed that promiscuous enzymes can compensate for these defects, in line with experimental evidence. Together, our results indicated that promiscuous enzyme activities are vital to maintain robust metabolic function and growth.
Collapse
Affiliation(s)
| | | | | | - Zoran Nikoloski
- Bioinformatics, Institute of Biochemistry and Biology, University of Potsdam, 14476 Potsdam, Germany
- Systems Biology and Mathematical Modelling, Max Planck Institute of Molecular Plant Physiology, 14476 Potsdam, Germany
| |
Collapse
|
22
|
Rondeau NC, Raup-Collado J, Kogan HV, Cho R, Lovinger N, Wague F, Lopatkin AJ, Texeira NG, Flores ME, Rovnyak D, Snow JW. Remodeling of Cellular Respiration and Insulin Signaling Are Part of a Shared Stress Response in Divergent Bee Species. INSECTS 2025; 16:300. [PMID: 40266798 PMCID: PMC11942726 DOI: 10.3390/insects16030300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 03/04/2025] [Accepted: 03/10/2025] [Indexed: 04/25/2025]
Abstract
The honey bee (Apis mellifera) is of paramount importance to human activities through the pollination services they provide in agricultural settings. Honey bee colonies in the United States have suffered from an increased rate of annual die-off in recent years, stemming from a complex set of interacting stressors that remain poorly described. Defining the cellular responses that are perturbed by divergent stressors represents a key step in understanding these synergies. We found that multiple model stressors induce upregulated expression of the lactate dehydrogenase (Ldh) gene in the midgut of the eusocial honey bee and that the Ldh gene family is expanded in diverse bee species. Alterations in Ldh expression were concomitant with changes in the expression of other genes involved in cellular respiration and genes encoding insulin/insulin-like growth factor signaling (IIS) pathway components. Additionally, changes in metabolites in the midgut after stress, including increased levels of lactate, linked metabolic changes with the observed changes in gene expression. Select transcriptional changes in response to stress were similarly observed in the solitary alfalfa leafcutting bee (Megachile rotundata). Thus, increased Ldh expression may be part of a core stress response remodeling cellular respiration and insulin signaling. These findings suggest that a conserved cellular response that regulates metabolic demands under diverse stressful conditions may play a protective role in bees regardless of life history.
Collapse
Affiliation(s)
- Nicole C. Rondeau
- Biology Department, Barnard College, New York, NY 10027, USA; (N.C.R.); (H.V.K.); (M.E.F.)
| | - Joanna Raup-Collado
- Department of Chemistry, Bucknell University, Lewisburg, PA 17837, USA; (J.R.-C.); (D.R.)
| | - Helen V. Kogan
- Biology Department, Barnard College, New York, NY 10027, USA; (N.C.R.); (H.V.K.); (M.E.F.)
| | - Rachel Cho
- Biology Department, Barnard College, New York, NY 10027, USA; (N.C.R.); (H.V.K.); (M.E.F.)
| | - Natalie Lovinger
- Biology Department, Barnard College, New York, NY 10027, USA; (N.C.R.); (H.V.K.); (M.E.F.)
| | - Fatoumata Wague
- Biology Department, Barnard College, New York, NY 10027, USA; (N.C.R.); (H.V.K.); (M.E.F.)
| | - Allison J. Lopatkin
- Department of Chemical Engineering, University of Rochester, Rochester, NY 14642, USA;
| | - Noelle G. Texeira
- Biology Department, Barnard College, New York, NY 10027, USA; (N.C.R.); (H.V.K.); (M.E.F.)
| | - Melissa E. Flores
- Biology Department, Barnard College, New York, NY 10027, USA; (N.C.R.); (H.V.K.); (M.E.F.)
| | - David Rovnyak
- Department of Chemistry, Bucknell University, Lewisburg, PA 17837, USA; (J.R.-C.); (D.R.)
| | - Jonathan W. Snow
- Biology Department, Barnard College, New York, NY 10027, USA; (N.C.R.); (H.V.K.); (M.E.F.)
| |
Collapse
|
23
|
Zhu P, Pfrender EM, Steffeck AWT, Reczek CR, Zhou Y, Thakkar AV, Gupta NR, Kupai A, Willbanks A, Lieber RL, Roy I, Chandel NS, Peek CB. Immunomodulatory role of the stem cell circadian clock in muscle repair. SCIENCE ADVANCES 2025; 11:eadq8538. [PMID: 40043110 PMCID: PMC11881903 DOI: 10.1126/sciadv.adq8538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 01/30/2025] [Indexed: 03/09/2025]
Abstract
Circadian rhythms orchestrate physiological processes such as metabolism, immune function, and tissue regeneration, aligning them with the optimal time of day (TOD). This study identifies an interplay between the circadian clock within muscle stem cells (SCs) and their capacity to modulate the immune microenvironment during muscle regeneration. We reveal that the SC clock triggers TOD-dependent inflammatory gene transcription after injury, particularly genes related to neutrophil activity and chemotaxis. These responses are driven by cytosolic regeneration of the signaling metabolite nicotinamide adenine dinucleotide (oxidized form) (NAD+), as enhancing cytosolic NAD+ regeneration in SCs is sufficient to induce inflammatory responses that influence muscle regeneration. Mononuclear single-cell sequencing of the regenerating muscle niche further implicates the cytokine CCL2 in mediating SC-neutrophil cross-talk in a TOD-dependent manner. Our findings highlight the intersection between SC metabolic shifts and immune responses within the muscle microenvironment, dictated by circadian rhythms, and underscore the potential for targeting circadian and metabolic pathways to enhance tissue regeneration.
Collapse
Affiliation(s)
- Pei Zhu
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Eric M. Pfrender
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Adam W. T. Steffeck
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Colleen R. Reczek
- Department of Medicine, Division of Pulmonary and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yalu Zhou
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Feinberg Cardiovascular and Renal Research Institute, Chicago, IL, USA
| | - Abhishek Vijay Thakkar
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Neha R. Gupta
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ariana Kupai
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Amber Willbanks
- Shirley Ryan AbilityLab (formerly known as Rehabilitation Institute of Chicago), Chicago, IL, USA
| | - Richard L. Lieber
- Shirley Ryan AbilityLab (formerly known as Rehabilitation Institute of Chicago), Chicago, IL, USA
- Department of Physical Medicine and Rehabilitation, Northwestern University, Chicago, IL, USA
- Hines VA Hospital, Maywood, IL, USA
| | - Ishan Roy
- Shirley Ryan AbilityLab (formerly known as Rehabilitation Institute of Chicago), Chicago, IL, USA
- Department of Physical Medicine and Rehabilitation, Northwestern University, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
| | - Navdeep S. Chandel
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Division of Pulmonary and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Clara B. Peek
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
24
|
Yadav AK, Kushwaha R, Mandal AA, Mandal A, Banerjee S. Intracellular Photocatalytic NADH/NAD(P)H Oxidation for Cancer Drug Development. J Am Chem Soc 2025; 147:7161-7181. [PMID: 39980079 DOI: 10.1021/jacs.4c18328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Photocatalytic cancer therapy (PCT) has emerged as a cutting-edge anticancer mechanism of action, harnessing light energy to mediate the catalytic oxidation of intracellular substrates. PCT is of significant current importance due to its potential to address the limitations of conventional chemotherapy, particularly drug resistance and side effects. This approach offers a noninvasive, targeted cancer treatment option by utilizing metal-based photocatalysts to induce redox and metabolic disorders within cancer cells. The photocatalysts disrupt the cancer cell metabolism by converting NADH/NAD(P)H to NAD+/NAD(P)+ via catalytic photoredox processes, altering intracellular NAD+/NADH or NAD(P)+/NAD(P)H ratios, which are crucial for cellular metabolism. Ir(III), Ru(II), Re(I), and Os(II) photocatalysts demonstrated promising PCT efficacy. Despite these developments, gaps remain in the literature for translating this new anticancer mechanism into clinical trials. This Perspective critically examines the developments in this research area and provides future directions for designing efficient photocatalysts for PCT.
Collapse
Affiliation(s)
- Ashish Kumar Yadav
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Rajesh Kushwaha
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Arif Ali Mandal
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Apurba Mandal
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Samya Banerjee
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| |
Collapse
|
25
|
Cho HB, Kim H, Lee S, Cho CW, Park J, Youn S, So G, Kang S, Kim HJ, Park K. Near Infrared-Mediated Intracellular NADH Delivery Strengthens Mitochondrial Function and Stability in Muscle Dysfunction Model. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2415303. [PMID: 39887582 PMCID: PMC11948086 DOI: 10.1002/advs.202415303] [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: 11/20/2024] [Revised: 01/15/2025] [Indexed: 02/01/2025]
Abstract
Mitochondrial transfer emerges as a promising therapy for the restoration of mitochondrial function in damaged cells, mainly due to its limited immunogenicity. However, isolated mitochondria rapidly lose function because they produce little energy outside cells. Therefore, this study investigates whether near infrared (NIR)-mediated nicotinamide adenine dinucleotide (NADH) pre-treatment enhances mitochondrial function and stability in mitochondria-donor cells prior to transplantation. Clinical applications of NADH, an essential electron donor in the oxidative phosphorylation process, are restricted due to the limited cellular uptake of NADH. To address this, a photo-mediated method optimizes direct NADH delivery into cells and increases NADH absorption. L6 cells treated with NADH and irradiated with NIR enhanced NADH uptake, significantly improving mitochondrial energy production and function. Importantly, the improved functional characteristics of the mitochondria are maintained even after isolation from cells. Primed mitochondria, i.e., those enhanced by NIR-mediated NADH uptake (P-MT), are encapsulated in fusogenic liposomes and delivered into muscle cells with mitochondrial dysfunction. Compared to conventional mitochondria, P-MT mitochondria promote greater mitochondrial recovery and muscle regeneration. These findings suggest that NIR-mediated NADH delivery is an effective strategy for improving mitochondrial function, and has the potential to lead to novel treatments for mitochondrial disorders and muscle degeneration.
Collapse
Affiliation(s)
- Hui Bang Cho
- Department of Nano‐regenerative Medical EngineeringCollege of Life ScienceCHA University6F, CHA Biocomplex, Sampyeong‐Dong, Bundang‐guSeongnam‐si13488Republic of Korea
| | - Hye‐Ryoung Kim
- Department of Nano‐regenerative Medical EngineeringCollege of Life ScienceCHA University6F, CHA Biocomplex, Sampyeong‐Dong, Bundang‐guSeongnam‐si13488Republic of Korea
| | - Sujeong Lee
- Department of Nano‐regenerative Medical EngineeringCollege of Life ScienceCHA University6F, CHA Biocomplex, Sampyeong‐Dong, Bundang‐guSeongnam‐si13488Republic of Korea
| | - Chae Won Cho
- Department of Nano‐regenerative Medical EngineeringCollege of Life ScienceCHA University6F, CHA Biocomplex, Sampyeong‐Dong, Bundang‐guSeongnam‐si13488Republic of Korea
| | - Ji‐In Park
- Department of Nano‐regenerative Medical EngineeringCollege of Life ScienceCHA University6F, CHA Biocomplex, Sampyeong‐Dong, Bundang‐guSeongnam‐si13488Republic of Korea
| | - Seulki Youn
- Department of Nano‐regenerative Medical EngineeringCollege of Life ScienceCHA University6F, CHA Biocomplex, Sampyeong‐Dong, Bundang‐guSeongnam‐si13488Republic of Korea
| | - Gyuwon So
- Department of Nano‐regenerative Medical EngineeringCollege of Life ScienceCHA University6F, CHA Biocomplex, Sampyeong‐Dong, Bundang‐guSeongnam‐si13488Republic of Korea
| | - Sumin Kang
- Department of Nano‐regenerative Medical EngineeringCollege of Life ScienceCHA University6F, CHA Biocomplex, Sampyeong‐Dong, Bundang‐guSeongnam‐si13488Republic of Korea
| | - Hye Jin Kim
- Department of Nano‐regenerative Medical EngineeringCollege of Life ScienceCHA University6F, CHA Biocomplex, Sampyeong‐Dong, Bundang‐guSeongnam‐si13488Republic of Korea
| | - Keun‐Hong Park
- Department of Nano‐regenerative Medical EngineeringCollege of Life ScienceCHA University6F, CHA Biocomplex, Sampyeong‐Dong, Bundang‐guSeongnam‐si13488Republic of Korea
| |
Collapse
|
26
|
Mouli K, Liopo AV, McHugh EA, Underwood E, Zhao J, Dash PK, Vo ATT, Malojirao VH, Hegde ML, Tour JM, Derry PJ, Kent TA. Oxidized Carbon Nanoparticles Enhance Cellular Energetics With Application to Injured Brain. Adv Healthc Mater 2025; 14:e2401629. [PMID: 39329414 PMCID: PMC11937864 DOI: 10.1002/adhm.202401629] [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: 05/02/2024] [Revised: 07/26/2024] [Indexed: 09/28/2024]
Abstract
Pro-energetic effects of functionalized, oxidized carbon nanozymes (OCNs) are reported. OCNs, derived from harsh acid oxidation of single-wall carbon nanotubes or activated charcoal are previously shown to possess multiple nanozymatic activities including mimicking superoxide dismutase and catalyzing the oxidation of reduced nicotinamide adenine dinucleotide (NADH) to NAD+. These actions are predicted to generate a glycolytic shift and enhance mitochondrial energetics under impaired conditions. Impaired mitochondrial energy metabolism is increasingly recognized as an important facet of traumatic brain injury (TBI) pathophysiology and decreases the efficiency of electron transport chain (ETC)-coupled adenosine triphosphate (ATP) and NAD+ regeneration. In vitro, OCNs promote a pro-aerobic shift in energy metabolism that persists through ETC inhibition and enhances glycolytic flux, glycolytic ATP production, and cellular generation of lactate, a crucial auxiliary substrate for energy metabolism. To address specific mechanisms of iron injury from hemorrhage, OCNs with the iron chelator, deferoxamine (DEF), covalently-linked were synthesized. DEF-linked OCNs induce a glycolytic shift in-vitro and in-vivo in tissue sections from a rat model of TBI complicated by hemorrhagic contusion. OCNs further reduced hemorrhage volumes 3 days following TBI. These results suggest OCNs are promising as pleiotropic mediators of cell and tissue resilience to injury.
Collapse
Affiliation(s)
- Karthik Mouli
- Center for Genomics and Precision MedicineDepartment of Translational MedicineInstitute of Biosciences and TechnologyTexas A&M Health Science CenterHoustonTX77030USA
| | - Anton V. Liopo
- Center for Genomics and Precision MedicineDepartment of Translational MedicineInstitute of Biosciences and TechnologyTexas A&M Health Science CenterHoustonTX77030USA
- Department of ChemistryRice UniversityHoustonTX77005USA
| | - Emily A. McHugh
- Department of ChemistryRice UniversityHoustonTX77005USA
- Smalley‐Curl InstituteRice UniversityHoustonTX77005USA
| | - Erica Underwood
- Department of Neurobiology and AnatomyThe University of TX McGovern Medical SchoolHoustonTX77030USA
| | - Jing Zhao
- Department of Neurobiology and AnatomyThe University of TX McGovern Medical SchoolHoustonTX77030USA
| | - Pramod K. Dash
- Department of Neurobiology and AnatomyThe University of TX McGovern Medical SchoolHoustonTX77030USA
| | - Anh T. T. Vo
- Center for Genomics and Precision MedicineDepartment of Translational MedicineInstitute of Biosciences and TechnologyTexas A&M Health Science CenterHoustonTX77030USA
| | - Vikas H. Malojirao
- Center for NeuroregenerationDepartment of NeurosurgeryDivision of DNA Repair ResearchHouston Methodist Research InstituteHoustonTX77030USA
| | - Muralidhar L. Hegde
- Center for NeuroregenerationDepartment of NeurosurgeryDivision of DNA Repair ResearchHouston Methodist Research InstituteHoustonTX77030USA
- Department of NeurosciencesWeill Cornell Medical CollegeNew YorkNYUSA
- EnMedSchool of Engineering MedicineTexas A&M UniversityHouston77030USA
| | - James M. Tour
- Department of ChemistryRice UniversityHoustonTX77005USA
- Smalley‐Curl InstituteRice UniversityHoustonTX77005USA
- Welch Institute for Advanced MaterialsRice UniversityHoustonTX77005USA
- The NanoCarbon CenterRice UniversityHoustonTX77005USA
| | - Paul J. Derry
- Center for Genomics and Precision MedicineDepartment of Translational MedicineInstitute of Biosciences and TechnologyTexas A&M Health Science CenterHoustonTX77030USA
- EnMedSchool of Engineering MedicineTexas A&M UniversityHouston77030USA
| | - Thomas A. Kent
- Center for Genomics and Precision MedicineDepartment of Translational MedicineInstitute of Biosciences and TechnologyTexas A&M Health Science CenterHoustonTX77030USA
- Department of ChemistryRice UniversityHoustonTX77005USA
- Stanley H. Appel Department of NeurologyHouston Methodist Hospital and Research InstituteHoustonTX77030USA
| |
Collapse
|
27
|
Peinado P, Stazi M, Ballabio C, Margineanu MB, Li Z, Colón CI, Hsieh MS, Pal Choudhuri S, Stastny V, Hamilton S, Le Marois A, Collingridge J, Conrad L, Chen Y, Ng SR, Magendantz M, Bhutkar A, Chen JS, Sahai E, Drapkin BJ, Jacks T, Vander Heiden MG, Kopanitsa MV, Robinson HPC, Li L. Intrinsic electrical activity drives small-cell lung cancer progression. Nature 2025; 639:765-775. [PMID: 39939778 PMCID: PMC11922742 DOI: 10.1038/s41586-024-08575-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 12/23/2024] [Indexed: 02/14/2025]
Abstract
Elevated or ectopic expression of neuronal receptors promotes tumour progression in many cancer types1,2; neuroendocrine (NE) transformation of adenocarcinomas has also been associated with increased aggressiveness3. Whether the defining neuronal feature, namely electrical excitability, exists in cancer cells and impacts cancer progression remains mostly unexplored. Small-cell lung cancer (SCLC) is an archetypal example of a highly aggressive NE cancer and comprises two major distinct subpopulations: NE cells and non-NE cells4,5. Here we show that NE cells, but not non-NE cells, are excitable, and their action potential firing directly promotes SCLC malignancy. However, the resultant high ATP demand leads to an unusual dependency on oxidative phosphorylation in NE cells. This finding contrasts with the properties of most cancer cells reported in the literature, which are non-excitable and rely heavily on aerobic glycolysis. Additionally, we found that non-NE cells metabolically support NE cells, a process akin to the astrocyte-neuron metabolite shuttle6. Finally, we observed drastic changes in the innervation landscape during SCLC progression, which coincided with increased intratumoural heterogeneity and elevated neuronal features in SCLC cells, suggesting an induction of a tumour-autonomous vicious cycle, driven by cancer cell-intrinsic electrical activity, which confers long-term tumorigenic capability and metastatic potential.
Collapse
Affiliation(s)
- Paola Peinado
- Cancer Neuroscience Laboratory, Francis Crick Institute, London, UK
| | - Marco Stazi
- Cancer Neuroscience Laboratory, Francis Crick Institute, London, UK
| | - Claudio Ballabio
- Cancer Neuroscience Laboratory, Francis Crick Institute, London, UK
| | | | - Zhaoqi Li
- Koch Institute of Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Caterina I Colón
- Koch Institute of Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Min-Shu Hsieh
- Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
| | - Shreoshi Pal Choudhuri
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Internal Medicine and Simmons Comprehensive Cancer Center, University of Texas, Southwestern Medical Center, Dallas, TX, USA
| | - Victor Stastny
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Internal Medicine and Simmons Comprehensive Cancer Center, University of Texas, Southwestern Medical Center, Dallas, TX, USA
| | - Seth Hamilton
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Internal Medicine and Simmons Comprehensive Cancer Center, University of Texas, Southwestern Medical Center, Dallas, TX, USA
| | - Alix Le Marois
- Tumour Cell Biology Laboratory, Francis Crick Institute, London, UK
| | - Jodie Collingridge
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Linus Conrad
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Yinxing Chen
- Koch Institute of Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sheng Rong Ng
- Koch Institute of Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Margaret Magendantz
- Koch Institute of Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Arjun Bhutkar
- Koch Institute of Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jin-Shing Chen
- Division of Thoracic Surgery, Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Erik Sahai
- Tumour Cell Biology Laboratory, Francis Crick Institute, London, UK
| | - Benjamin J Drapkin
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Internal Medicine and Simmons Comprehensive Cancer Center, University of Texas, Southwestern Medical Center, Dallas, TX, USA
| | - Tyler Jacks
- Koch Institute of Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Matthew G Vander Heiden
- Koch Institute of Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Maksym V Kopanitsa
- Cancer Neuroscience Laboratory, Francis Crick Institute, London, UK
- Charles River Discovery Services, Portishead, UK
| | - Hugh P C Robinson
- Cancer Neuroscience Laboratory, Francis Crick Institute, London, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Leanne Li
- Cancer Neuroscience Laboratory, Francis Crick Institute, London, UK.
- Koch Institute of Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
28
|
Liu L, Wang R, Pu X, Zha Y, Yang X, Fang X, Liu Y, Shao M, Zhu L, Ren X, Deng G, Yang K. D 4 -CHIP REVEALS IMPAIRED T-CELL FUNCTION IN SEPSIS: INSIGHTS FROM PLASMA MICROENVIRONMENT ANALYSIS AND MITOCHONDRIAL-TARGETED THERAPY. Shock 2025; 63:417-427. [PMID: 39178197 DOI: 10.1097/shk.0000000000002434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2024]
Abstract
ABSTRACT Background: Sepsis, a systemic inflammation syndrome initiated by infection, poses significant challenges due to its intricate pathophysiology. T cells play a crucial role in combating infections during sepsis. Despite previous observations indicating T-cell dysfunction in sepsis, reliable in vitro detection methods were lacking, and the factors influencing these impairments remained unclear. Methods: We developed a novel method using the D 4 -Chip to assess sepsis T-cell migration function. This microfluidic platform enabled precise analysis of migration function under controlled conditions. Additionally, We explored the impact of the plasma microenvironment on T-cell behavior, along with the redox environment in sepsis, and assessed the potential efficacy of Mitoquinone mesylate (MitoQ), a mitochondrial-targeted drug. Results: Our findings revealed impaired migration function in sepsis T cells compared to healthy controls. Interestingly, sepsis plasma enhanced the migration of healthy T cells, yet incubation with healthy plasma did not fully restore migration impairments in sepsis T cells. Subsequent investigations uncovered a significant increase in NADH/NAD+ levels in sepsis T cells, with healthy T cells exposed to various sepsis plasma conditions also showing elevated NADH/NAD+ levels. Importantly, MitoQ normalized abnormal intracellular NADH/NAD+ levels and enhanced the migration ability of T cells. Conclusions: Short-term incubation with sepsis plasma does not directly inhibit T-cell migration but instead affects T-cell function by disrupting the intracellular redox environment. Improving the intracellular redox environment of sepsis patients contributes to restoring impaired migration and proliferation, with MitoQ demonstrating therapeutic potential.
Collapse
Affiliation(s)
| | - Ruoyu Wang
- The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xuexue Pu
- The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yutao Zha
- The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | | | - Xiao Fang
- The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yong Liu
- Anhui Institute of Optics and Fine Mechanics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
| | - Min Shao
- The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ling Zhu
- Anhui Institute of Optics and Fine Mechanics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
| | - Xiaoou Ren
- Institute of Health Sciences and Technology, Institutes of Material Science and Information Technology, Anhui University, Hefei, China
| | - Guoqing Deng
- Anhui Institute of Optics and Fine Mechanics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
| | - Ke Yang
- Anhui Institute of Optics and Fine Mechanics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
| |
Collapse
|
29
|
Reddan B, Cummins EP. The regulation of cell metabolism by hypoxia and hypercapnia. J Biol Chem 2025; 301:108252. [PMID: 39914740 PMCID: PMC11923829 DOI: 10.1016/j.jbc.2025.108252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 01/13/2025] [Accepted: 01/25/2025] [Indexed: 03/06/2025] Open
Abstract
Every cell in the body is exposed to a certain level of CO2 and O2. Hypercapnia and hypoxia elicit stress signals to influence cellular metabolism and function. Both conditions exert profound yet distinct effects on metabolic pathways and mitochondrial dynamics, highlighting the need for cells to adapt to changes in the gaseous microenvironment. The interplay between hypercapnia and hypoxia signaling is the key for dictating cellular homeostasis as microenvironmental CO2 and O2 levels are inextricably linked. Hypercapnia, characterized by elevated pCO2, introduces metabolic adaptations within the aerobic metabolism pathways, affecting tricarboxylic acid cycle flux, lipid, and amino acid metabolism, oxidative phosphorylation and the electron transport chain. Hypoxia, defined by reduced oxygen availability, necessitates a shift from oxidative phosphorylation to anaerobic glycolysis to sustain ATP production, a process orchestrated by the stabilization of hypoxia-inducible factor-1α. Given that hypoxia and hypercapnia are present in both physiological and cancerous microenvironments, how might the coexistence of hypercapnia and hypoxia influence metabolic pathways and cellular function in physiological niches and the tumor microenvironment?
Collapse
Affiliation(s)
- Ben Reddan
- School of Medicine, University College Dublin, Dublin, Ireland; Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Eoin P Cummins
- School of Medicine, University College Dublin, Dublin, Ireland; Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland.
| |
Collapse
|
30
|
Zhang M, Kong X, Wu C, Li J, Yang H, Huang L. The role of lactate and lactylation in ischemic cardiomyopathy: Mechanisms and gene expression. Exp Mol Pathol 2025; 141:104957. [PMID: 40020527 DOI: 10.1016/j.yexmp.2025.104957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 02/07/2025] [Accepted: 02/12/2025] [Indexed: 03/03/2025]
Abstract
Ischemic cardiomyopathy (ICM) is a significant global public health issue, with its pathophysiology encompassing atherosclerotic plaque formation, thrombosis, hypoperfusion, ischemic cell death, and left ventricular remodeling. Lactate is not only regarded as an energy metabolite but also acts as a signaling molecule that influences various physiological processes, regulating metabolism and muscle contraction. Lactylation, an emerging epigenetic modification, affects protein functionality and gene expression through the P300 enzyme. In ICM, lactate accumulation leads to pH imbalance and myocardial cell dysfunction, impacting cellular signaling. This paper will analyze the role of lactylation in ICM, focusing on coronary artery disease (ASCVD) and myocardial infarction (MI). It will also explore the differential expression and immunological characteristics of lactylation-related genes in normal and ICM tissues, providing potential targets for future research.
Collapse
Affiliation(s)
- Mei Zhang
- Department of Clinical Nursing, The Second Xiangya Hospital of Central South University, Changsha, China; Xiangya School of Nursing, Central South University, Changsha, China
| | - Xue Kong
- Department of Clinical Nursing, The Second Xiangya Hospital of Central South University, Changsha, China; Xiangya School of Nursing, Central South University, Changsha, China
| | - Chenlu Wu
- Department of Cardiology, the Second Xiangya Hospital of Central South University, Changsha, China
| | - Jiuhong Li
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Hui Yang
- Department of Cardiology, the Second Xiangya Hospital of Central South University, Changsha, China.
| | - Lingzhi Huang
- Department of Clinical Nursing, The Second Xiangya Hospital of Central South University, Changsha, China; Xiangya School of Nursing, Central South University, Changsha, China.
| |
Collapse
|
31
|
Lee J, Roh JL. Ferroptosis: iron release mechanisms in the bioenergetic process. Cancer Metastasis Rev 2025; 44:36. [PMID: 40000477 DOI: 10.1007/s10555-025-10252-8] [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: 11/15/2024] [Accepted: 02/17/2025] [Indexed: 02/27/2025]
Abstract
Ferroptosis, an iron-dependent form of cell death, has been the focus of extensive research over the past decade, leading to the elucidation of key molecules and mechanisms involved in this process. While several studies have highlighted iron sources for the Fenton reaction, the predominant mechanism for iron release in ferroptosis has been identified as ferritinophagy, which occurs in response to iron starvation. However, much of the existing literature has concentrated on lipid peroxidation rather than on the mechanisms of iron release. This review proposes three distinct mechanisms of iron mobilization: ferritinophagy, reductive pathways with selective gating of ferritin pores, and quinone-mediated iron mobilization. Notably, the latter two mechanisms operate independently of iron starvation and rely primarily on reductants such as NADH and O2•-. The inhibition of the respiratory chain, particularly under the activation of α-ketoglutarate dehydrogenase, leads to the accumulation of these reductants, which in turn promotes iron release from ferritin and indirectly inhibits AMP-activated protein kinase through excessive iron levels. In this work, we delineate the intricate relationship between iron mobilization and bioenergetic processes under conditions of oxidative stress. Furthermore, this review aims to enhance the understanding of the connections between ferroptosis and these mechanisms.
Collapse
Affiliation(s)
- Jaewang Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University, Seongnam, Gyeonggi-Do, 13496, Republic of Korea
- Department of Biomedical Science, General Graduate School, CHA University, Pocheon, Republic of Korea
| | - Jong-Lyel Roh
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University, Seongnam, Gyeonggi-Do, 13496, Republic of Korea.
- Department of Biomedical Science, General Graduate School, CHA University, Pocheon, Republic of Korea.
| |
Collapse
|
32
|
Valeros J, Jerome M, Tseyang T, Vo P, Do T, Fajardo Palomino D, Grotehans N, Kunala M, Jerrett AE, Hathiramani NR, Mireku M, Magesh RY, Yenilmez B, Rosen PC, Mann JL, Myers JW, Kunchok T, Manning TL, Boercker LN, Carr PE, Munim MB, Lewis CA, Sabatini DM, Kelly M, Xie J, Czech MP, Gao G, Shepherd JN, Walker AK, Kim H, Watson EV, Spinelli JB. Rhodoquinone carries electrons in the mammalian electron transport chain. Cell 2025; 188:1084-1099.e27. [PMID: 39909039 PMCID: PMC11845293 DOI: 10.1016/j.cell.2024.12.007] [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: 08/19/2024] [Accepted: 12/09/2024] [Indexed: 02/07/2025]
Abstract
Ubiquinone (UQ), the only known electron carrier in the mammalian electron transport chain (ETC), preferentially delivers electrons to the terminal electron acceptor oxygen (O2). In hypoxia, ubiquinol (UQH2) diverts these electrons onto fumarate instead. Here, we identify rhodoquinone (RQ), an electron carrier detected in mitochondria purified from certain mouse and human tissues that preferentially delivers electrons to fumarate through the reversal of succinate dehydrogenase, independent of environmental O2 levels. The RQ/fumarate ETC is strictly present in vivo and is undetectable in cultured mammalian cells. Using genetic and pharmacologic tools that reprogram the ETC from the UQ/O2 to the RQ/fumarate pathway, we establish that these distinct ETCs support unique programs of mitochondrial function and that RQ confers protection upon hypoxia exposure in vitro and in vivo. Thus, in discovering the presence of RQ in mammals, we unveil a tractable therapeutic strategy that exploits flexibility in the ETC to ameliorate hypoxia-related conditions.
Collapse
Affiliation(s)
- Jonathan Valeros
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Madison Jerome
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Tenzin Tseyang
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Paula Vo
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Thang Do
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Diana Fajardo Palomino
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA; Morningside Graduate School of Biomedical Sciences, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Nils Grotehans
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Manisha Kunala
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA; Morningside Graduate School of Biomedical Sciences, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Alexandra E Jerrett
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA; Morningside Graduate School of Biomedical Sciences, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Nicolai R Hathiramani
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA; Morningside Graduate School of Biomedical Sciences, UMass Chan Medical School, Worcester, MA 01605, USA; Diabetes Center of Excellence, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Michael Mireku
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Rayna Y Magesh
- Department of Systems Biology, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Batuhan Yenilmez
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Paul C Rosen
- Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Jessica L Mann
- Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Jacob W Myers
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | | | - Tanner L Manning
- Department of Chemistry and Biochemistry, Gonzaga University, Spokane, WA 99258, USA
| | - Lily N Boercker
- Department of Chemistry and Biochemistry, Gonzaga University, Spokane, WA 99258, USA
| | - Paige E Carr
- Department of Chemistry and Biochemistry, Gonzaga University, Spokane, WA 99258, USA
| | | | - Caroline A Lewis
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - David M Sabatini
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo n. 2, 166 10 Prague, Czech Republic; Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences Boston Branch, 840 Memorial Drive, Cambridge, MA 02139, USA
| | - Mark Kelly
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA; Division of Cardiovascular Medicine, Department of Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Jun Xie
- Horae Gene Therapy Center, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Michael P Czech
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA; Diabetes Center of Excellence, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Guangping Gao
- Horae Gene Therapy Center, UMass Chan Medical School, Worcester, MA 01605, USA; Li Weibo Institute for Rare Disease Research, UMass Chan Medical School, Worcester, MA 01655, USA; Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Jennifer N Shepherd
- Department of Chemistry and Biochemistry, Gonzaga University, Spokane, WA 99258, USA
| | - Amy K Walker
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Hahn Kim
- Small Molecule Screening Center, Princeton University, Princeton, NJ 08544, USA; Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
| | - Emma V Watson
- Diabetes Center of Excellence, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Jessica B Spinelli
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
33
|
Liu H, Wang S, Wang J, Guo X, Song Y, Fu K, Gao Z, Liu D, He W, Yang LL. Energy metabolism in health and diseases. Signal Transduct Target Ther 2025; 10:69. [PMID: 39966374 PMCID: PMC11836267 DOI: 10.1038/s41392-025-02141-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 11/08/2024] [Accepted: 12/25/2024] [Indexed: 02/20/2025] Open
Abstract
Energy metabolism is indispensable for sustaining physiological functions in living organisms and assumes a pivotal role across physiological and pathological conditions. This review provides an extensive overview of advancements in energy metabolism research, elucidating critical pathways such as glycolysis, oxidative phosphorylation, fatty acid metabolism, and amino acid metabolism, along with their intricate regulatory mechanisms. The homeostatic balance of these processes is crucial; however, in pathological states such as neurodegenerative diseases, autoimmune disorders, and cancer, extensive metabolic reprogramming occurs, resulting in impaired glucose metabolism and mitochondrial dysfunction, which accelerate disease progression. Recent investigations into key regulatory pathways, including mechanistic target of rapamycin, sirtuins, and adenosine monophosphate-activated protein kinase, have considerably deepened our understanding of metabolic dysregulation and opened new avenues for therapeutic innovation. Emerging technologies, such as fluorescent probes, nano-biomaterials, and metabolomic analyses, promise substantial improvements in diagnostic precision. This review critically examines recent advancements and ongoing challenges in metabolism research, emphasizing its potential for precision diagnostics and personalized therapeutic interventions. Future studies should prioritize unraveling the regulatory mechanisms of energy metabolism and the dynamics of intercellular energy interactions. Integrating cutting-edge gene-editing technologies and multi-omics approaches, the development of multi-target pharmaceuticals in synergy with existing therapies such as immunotherapy and dietary interventions could enhance therapeutic efficacy. Personalized metabolic analysis is indispensable for crafting tailored treatment protocols, ultimately providing more accurate medical solutions for patients. This review aims to deepen the understanding and improve the application of energy metabolism to drive innovative diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Hui Liu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuo Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianhua Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xin Guo
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yujing Song
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kun Fu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenjie Gao
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Danfeng Liu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Wei He
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Lei-Lei Yang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
34
|
Zhou Z, Yin X, Sun H, Lu J, Li Y, Fan Y, Lv P, Han M, Wu J, Li S, Liu Z, Zhao H, Sun H, Fan H, Wang S, Xin T. PTBP1 Lactylation Promotes Glioma Stem Cell Maintenance through PFKFB4-Driven Glycolysis. Cancer Res 2025; 85:739-757. [PMID: 39570804 DOI: 10.1158/0008-5472.can-24-1412] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 09/06/2024] [Accepted: 11/11/2024] [Indexed: 02/18/2025]
Abstract
Long-standing evidence implicates glioma stem cells (GSC) as the major driver for glioma propagation and recurrence. GSCs have a distinctive metabolic landscape characterized by elevated glycolysis. Lactate accumulation resulting from enhanced glycolytic activity can drive lysine lactylation to regulate protein functions, suggesting that elucidating the lactylation landscape in GSCs could provide insights into glioma biology. Herein, we have demonstrated that global lactylation was significantly elevated in GSCs compared with differentiated glioma cells. Polypyrimidine tract-binding protein 1 (PTBP1), a central regulator of RNA processing, was hyperlactylated in GSCs, and SIRT1 induced PTBP1 delactylation. PTBP1-K436 lactylation supported glioma progression and GSC maintenance. Mechanistically, K436 lactylation inhibited PTBP1 proteasomal degradation by attenuating the interaction with TRIM21. Moreover, PTBP1 lactylation enhanced RNA-binding capacity and facilitated PFKFB4 mRNA stabilization, which further increased glycolysis. Together, these findings uncovered a lactylation-mediated mechanism in GSCs driven by metabolic reprogramming that induces aberrant epigenetic modifications to further stimulate glycolysis, resulting in a vicious cycle to exacerbate tumorigenesis. Significance: Glycolysis-induced lactylation enhances the function of PTBP1 to promote glioma stem cell maintenance, indicating that PTBP1 lactylation could be a potential target for the development of innovative glioma therapies.
Collapse
Affiliation(s)
- Zijian Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Engineering Research Center of Precision Diagnosis and Treatment Technology for Neuro-Oncology, Jinan, China
| | - Xianyong Yin
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Engineering Research Center of Precision Diagnosis and Treatment Technology for Neuro-Oncology, Jinan, China
| | - Hao Sun
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Jiaze Lu
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Yuming Li
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Yang Fan
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Peiwen Lv
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Min Han
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Jing Wu
- Department of Pharmacy, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Shengjie Li
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Zihao Liu
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Hongbo Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Haohan Sun
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Hao Fan
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Shan Wang
- Shandong Key Laboratory of Reproductive Medicine, Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Tao Xin
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Engineering Research Center of Precision Diagnosis and Treatment Technology for Neuro-Oncology, Jinan, China
- Shandong Institute of Brain Science and Brain-Inspired Research, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
35
|
LeGrand EK. Beyond nutritional immunity: immune-stressing challenges basic paradigms of immunometabolism and immunology. Front Nutr 2025; 12:1508767. [PMID: 40013164 PMCID: PMC11860096 DOI: 10.3389/fnut.2025.1508767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/31/2025] [Indexed: 02/28/2025] Open
Abstract
Pathogens have the well-known advantage of rapid evolution due to short generation times and large populations. However, pathogens have the rarely noted disadvantage of the vulnerability to stress involved in proliferation as well as being localized. Presented here are numerous new paradigms in immunology, and especially immunometabolism, which are derived from examining how hosts capitalize on pathogen vulnerabilities to stress. Universally, proliferation requires both resources and synthesis, which are vulnerable to resource-limiting stress and damaging/noxious stress, respectively. Pathogens are particularly vulnerable to stress at the time when they are most threatening-when they are proliferating. Since immune cells actively controlling pathogens (effector cells) typically do not proliferate at infected sites, there is a "stress vulnerability gap" wherein proliferating pathogens are more vulnerable to any type of stress than are the attacking effector cells. Hosts actively stress vulnerable proliferating pathogens by restricting resources (resource-limiting stress) and generating noxious waste products (damaging/disruptive stress) in a fundamental defense here-in termed "immune-stressing." While nutritional immunity emphasizes denying pathogens micronutrients, immune-stressing extends the concept to restricting all resources, especially glucose and oxygen, coupled with the generation of noxious metabolic products such as lactic acid, reactive oxygen species (ROS), and heat to further harm or stress the pathogens. At present much of the field of immunometabolism centers on how nutrition and metabolism regulate immune function, a central feature being the inefficient use of glucose via aerobic glycolysis (with much lactate/lactic acid production) by effector immune cells. In contrast, immune-stressing emphasizes how the immune system uses nutrition and metabolism to control infections. Immune-stressing addresses effector cell glycolysis at the infected site by noting that the high uptake of glucose linked with high output of lactic acid is an ideal double-pronged stressor targeting proliferating pathogens. Once the basic vulnerability of pathogen proliferation is recognized, numerous other paradigms of immunometabolism, and immunology as a whole, are challenged.
Collapse
Affiliation(s)
- Edmund K. LeGrand
- Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, United States
| |
Collapse
|
36
|
Chen J, Huang Z, Chen Y, Tian H, Chai P, Shen Y, Yao Y, Xu S, Ge S, Jia R. Lactate and lactylation in cancer. Signal Transduct Target Ther 2025; 10:38. [PMID: 39934144 PMCID: PMC11814237 DOI: 10.1038/s41392-024-02082-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 10/07/2024] [Accepted: 11/18/2024] [Indexed: 02/13/2025] Open
Abstract
Accumulated evidence has implicated the diverse and substantial influence of lactate on cellular differentiation and fate regulation in physiological and pathological settings, particularly in intricate conditions such as cancer. Specifically, lactate has been demonstrated to be pivotal in molding the tumor microenvironment (TME) through its effects on different cell populations. Within tumor cells, lactate impacts cell signaling pathways, augments the lactate shuttle process, boosts resistance to oxidative stress, and contributes to lactylation. In various cellular populations, the interplay between lactate and immune cells governs processes such as cell differentiation, immune response, immune surveillance, and treatment effectiveness. Furthermore, communication between lactate and stromal/endothelial cells supports basal membrane (BM) remodeling, epithelial-mesenchymal transitions (EMT), metabolic reprogramming, angiogenesis, and drug resistance. Focusing on lactate production and transport, specifically through lactate dehydrogenase (LDH) and monocarboxylate transporters (MCT), has shown promise in the treatment of cancer. Inhibitors targeting LDH and MCT act as both tumor suppressors and enhancers of immunotherapy, leading to a synergistic therapeutic effect when combined with immunotherapy. The review underscores the importance of lactate in tumor progression and provides valuable perspectives on potential therapeutic approaches that target the vulnerability of lactate metabolism, highlighting the Heel of Achilles for cancer treatment.
Collapse
Affiliation(s)
- Jie Chen
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China
| | - Ziyue Huang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China
| | - Ya Chen
- Department of Radiology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
| | - Hao Tian
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China
| | - Peiwei Chai
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China
| | - Yongning Shen
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
| | - Yiran Yao
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China
| | - Shiqiong Xu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China.
| | - Shengfang Ge
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China.
| | - Renbing Jia
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China.
| |
Collapse
|
37
|
Huggler KS, Mellado Fritz CA, Flickinger KM, Chang GR, McGuire MF, Cantor JR. Hexokinase detachment from mitochondria drives the Warburg effect to support compartmentalized ATP production. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.07.637120. [PMID: 39975027 PMCID: PMC11839068 DOI: 10.1101/2025.02.07.637120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Hexokinase (HK) catalyzes the synthesis of glucose-6-phosphate, marking the first committed step of glucose metabolism. Most cancer cells express two homologous isoforms (HK1 and HK2) that can each bind to the outer mitochondrial membrane (OMM). CRISPR screens across hundreds of cancer cell lines indicate that both are dispensable for cell growth in traditional culture media. By contrast, HK2 deletion impairs cell growth in Human Plasma-Like Medium (HPLM). Here, we find that HK2 is required to maintain sufficient cytosolic (OMM-detached) HK activity under conditions that enhance HK1 binding to the OMM. Notably, OMM-detached rather than OMM-docked HK promotes "aerobic glycolysis" (Warburg effect), an enigmatic phenotype displayed by most proliferating cells. We show that several proposed theories for this phenotype cannot explain the HK2 dependence and instead find that HK2 deletion severely impairs glycolytic ATP production with little impact on total ATP yield for cells in HPLM. Our results reveal a basis for conditional HK2 essentiality and suggest that demand for compartmentalized ATP synthesis underlies the Warburg effect.
Collapse
|
38
|
Burtscher J, Denti V, Gostner JM, Weiss AK, Strasser B, Hüfner K, Burtscher M, Paglia G, Kopp M, Dünnwald T. The interplay of NAD and hypoxic stress and its relevance for ageing. Ageing Res Rev 2025; 104:102646. [PMID: 39710071 DOI: 10.1016/j.arr.2024.102646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 12/24/2024]
Abstract
Nicotinamide adenine dinucleotide (NAD) is an essential regulator of cellular metabolism and redox processes. NAD levels and the dynamics of NAD metabolism change with increasing age but can be modulated via the diet or medication. Because NAD metabolism is complex and its regulation still insufficiently understood, achieving specific outcomes without perturbing delicate balances through targeted pharmacological interventions remains challenging. NAD metabolism is also highly sensitive to environmental conditions and can be influenced behaviorally, e.g., by exercise. Changes in oxygen availability directly and indirectly affect NAD levels and may result from exposure to ambient hypoxia, increased oxygen demand during exercise, ageing or disease. Cellular responses to hypoxic stress involve rapid alterations in NAD metabolism and depend on many factors, including age, glucose status, the dose of the hypoxic stress and occurrence of reoxygenation phases, and exhibit complex time-courses. Here we summarize the known determinants of NAD-regulation by hypoxia and evaluate the role of NAD in hypoxic stress. We define the specific NAD responses to hypoxia and identify a great potential of the modulation of NAD metabolism regarding hypoxic injuries. In conclusion, NAD metabolism and cellular hypoxia responses are strongly intertwined and together mediate protective processes against hypoxic insults. Their interactions likely contribute to age-related changes and vulnerabilities. Targeting NAD homeostasis presents a promising avenue to prevent/treat hypoxic insults and - conversely - controlled hypoxia is a potential tool to regulate NAD homeostasis.
Collapse
Affiliation(s)
- Johannes Burtscher
- Department of Sport Science, University of Innsbruck, Innsbruck, Austria.
| | - Vanna Denti
- School of Medicine and Surgery, University of Milano-Bicocca, Vedano al Lambro, MB, Italy
| | - Johanna M Gostner
- Medical University of Innsbruck, Biocenter, Institute of Medical Biochemistry, Innsbruck, Austria
| | - Alexander Kh Weiss
- Institute for Biomedical Aging Research, University of Innsbruck, Innsbruck, Austria
| | - Barbara Strasser
- Ludwig Boltzmann Institute for Rehabilitation Research, Vienna, Austria; Faculty of Medicine, Sigmund Freud Private University, Vienna, Austria
| | - Katharina Hüfner
- Department of Psychiatry, Psychotherapy, Psychosomatics and Medical Psychology, University Hospital for Psychiatry II, Medical University of Innsbruck, Innsbruck, Austria
| | - Martin Burtscher
- Department of Sport Science, University of Innsbruck, Innsbruck, Austria
| | - Giuseppe Paglia
- School of Medicine and Surgery, University of Milano-Bicocca, Vedano al Lambro, MB, Italy
| | - Martin Kopp
- Department of Sport Science, University of Innsbruck, Innsbruck, Austria
| | - Tobias Dünnwald
- Institute for Sports Medicine, Alpine Medicine and Health Tourism (ISAG), UMIT TIROL - Private University for Health Sciences and Health Technology, Hall in Tirol, Austria
| |
Collapse
|
39
|
Yang X, Zhou B. Unleashing metabolic power for axonal regeneration. Trends Endocrinol Metab 2025; 36:161-175. [PMID: 39069446 DOI: 10.1016/j.tem.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/13/2024] [Accepted: 07/03/2024] [Indexed: 07/30/2024]
Abstract
Axon regeneration requires the mobilization of intracellular resources, including proteins, lipids, and nucleotides. After injury, neurons need to adapt their metabolism to meet the biosynthetic demands needed to achieve axonal regeneration. However, the exact contribution of cellular metabolism to this process remains elusive. Insights into the metabolic characteristics of proliferative cells may illuminate similar mechanisms operating in axon regeneration; therefore, unraveling previously unappreciated roles of metabolic adaptation is critical to achieving neuron regrowth, which is connected to the therapeutic strategies for neurological conditions necessitating nerve repairs, such as spinal cord injury and stroke. Here, we outline the metabolic role in axon regeneration and discuss factors enhancing nerve regrowth, highlighting potential novel metabolic treatments for restoring nerve function.
Collapse
Affiliation(s)
- Xiaoyan Yang
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing 100191, China
| | - Bing Zhou
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing 100191, China; School of Engineering Medicine, Beihang University, Beijing 100191, China.
| |
Collapse
|
40
|
Dantzer R, Chelette B, Vichaya EG, West AP, Grossberg A. The metabolic basis of cancer-related fatigue. Neurosci Biobehav Rev 2025; 169:106035. [PMID: 39892436 PMCID: PMC11866516 DOI: 10.1016/j.neubiorev.2025.106035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 01/24/2025] [Accepted: 01/28/2025] [Indexed: 02/03/2025]
Abstract
Although we are all familiar with the sensation of fatigue, there are still profound divergences on what it represents and its mechanisms. Fatigue can take various forms depending on the condition in which it develops. Cancer-related fatigue is considered a symptom of exhaustion that is often present at the time of diagnosis, increases in intensity during cancer therapy, and does not always recede after completion of treatment. It is usually attributed to the inflammation induced by damage-associated molecular patterns released by tumor cells during cancer progression and in response to its treatment. In this review, we argue that it is necessary to go beyond the symptoms of fatigue to understand its nature and mechanisms. We propose to consider fatigue as a psychobiological process that regulates the behavioral activities an organism engages in to satisfy its needs, according to its physical ability to do so and to the capacity of its intermediary metabolism to exploit the resources procured by these activities. This last aspect is critical as it implies that these metabolic aspects need to be considered to understand fatigue. Based on the findings we have accumulated over several years of studying fatigue in diverse murine models of cancer, we show that energy metabolism plays a key role in the development and persistence of this condition. Cancer-related fatigue is dependent on the energy requirements of the tumor and the negative impact of cancer therapy on the mitochondrial function of the host. When inflammation is present, it adds to the organism's energy expenses. The organism needs to adjust its metabolism to the different forms of cellular stress it experiences thanks to specialized communication factors known as mitokines that act locally and at a distance from the cells in which they are produced. They induce the subjective, behavioral, and metabolic components of fatigue by acting in the brain. Therefore, the targeting of mitokines and their brain receptors offers a window of opportunity to treat fatigue when it is no longer adaptive but an obstacle to the quality of life of cancer survivors.
Collapse
Affiliation(s)
- Robert Dantzer
- Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Brandon Chelette
- Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Elisabeth G Vichaya
- Department of Psychology & Neuroscience, Baylor University, Waco, TX 76798, USA
| | | | - Aaron Grossberg
- Department of Radiation Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
41
|
Kern Coquillat N, Picq L, Hamond A, Megy P, Benezech S, Drouillard A, Lager-Lachaud N, Cahoreau E, Moreau M, Fallone L, Mathieu AL, Bellvert F, Nizard C, Bulteau AL, Walzer T, Marçais A. Pivotal role of exogenous pyruvate in human natural killer cell metabolism. Nat Metab 2025; 7:336-347. [PMID: 39753710 DOI: 10.1038/s42255-024-01188-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 11/20/2024] [Indexed: 02/27/2025]
Abstract
Resting natural killer (NK) cells display immediate effector functions after recognizing transformed or infected cells. The environmental nutrients and metabolic requirements to sustain these functions are not fully understood. Here, we show that NK cells rely on the use of extracellular pyruvate to support effector functions, signal transduction and cell viability. Glucose-derived carbons do not generate endogenous pyruvate. Consequently, NK cells import extracellular pyruvate that is reduced to lactate to regenerate glycolytic NAD+ and is oxidized in the tricarboxylic acid (TCA) cycle to produce ATP. This supports serine production through phosphoglycerate dehydrogenase, a pathway required for optimal proliferation following cytokine stimulation but dispensable for effector functions. In addition, like mouse NK cells, human NK cells rely on a citrate-malate configuration of the TCA cycle that is not fed by glutamine. Moreover, supraphysiologic pyruvate concentrations dose-dependently increase the effector functions of NK cells. Overall, this study highlights the role of exogenous pyruvate in NK cell biology, providing knowledge that could be exploited to boost NK cell potential in therapeutic settings.
Collapse
Affiliation(s)
- Nicolas Kern Coquillat
- CIRI, Centre International de Recherche en Infectiologie, (Team Lyacts), Univ Lyon, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
- LVMH Recherche, Saint Jean de Braye, France
| | - Louis Picq
- CIRI, Centre International de Recherche en Infectiologie, (Team Lyacts), Univ Lyon, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
- Institut Roche, Boulogne-Billancourt, France
| | - Ameline Hamond
- CIRI, Centre International de Recherche en Infectiologie, (Team Lyacts), Univ Lyon, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Pierre Megy
- CIRI, Centre International de Recherche en Infectiologie, (Team Lyacts), Univ Lyon, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Sarah Benezech
- CIRI, Centre International de Recherche en Infectiologie, (Team Lyacts), Univ Lyon, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Annabelle Drouillard
- CIRI, Centre International de Recherche en Infectiologie, (Team Lyacts), Univ Lyon, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Nina Lager-Lachaud
- Plateforme MetaToul FluxoMet (EAD13), Toulouse Biotechnology Institute, Bio & Chemical Engineering, Université de Toulouse, CNRS 5504, INRA 792, INSA TBI, INSA Toulouse, Toulouse, France
| | - Edern Cahoreau
- Plateforme MetaToul FluxoMet (EAD13), Toulouse Biotechnology Institute, Bio & Chemical Engineering, Université de Toulouse, CNRS 5504, INRA 792, INSA TBI, INSA Toulouse, Toulouse, France
| | | | - Lucie Fallone
- CIRI, Centre International de Recherche en Infectiologie, (Team Lyacts), Univ Lyon, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Anne-Laure Mathieu
- CIRI, Centre International de Recherche en Infectiologie, (Team Lyacts), Univ Lyon, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Floriant Bellvert
- Plateforme MetaToul FluxoMet (EAD13), Toulouse Biotechnology Institute, Bio & Chemical Engineering, Université de Toulouse, CNRS 5504, INRA 792, INSA TBI, INSA Toulouse, Toulouse, France
| | | | | | - Thierry Walzer
- CIRI, Centre International de Recherche en Infectiologie, (Team Lyacts), Univ Lyon, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France.
| | - Antoine Marçais
- CIRI, Centre International de Recherche en Infectiologie, (Team Lyacts), Univ Lyon, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France.
| |
Collapse
|
42
|
Liu T, Chen X, Sun Q, Li J, Wang Q, Wei P, Wang W, Li C, Wang Y. Valerenic acid attenuates pathological myocardial hypertrophy by promoting the utilization of multiple substrates in the mitochondrial energy metabolism. J Adv Res 2025; 68:241-256. [PMID: 38373650 PMCID: PMC11785575 DOI: 10.1016/j.jare.2024.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 01/31/2024] [Accepted: 02/15/2024] [Indexed: 02/21/2024] Open
Abstract
INTRODUCTION Valerenic acid (VA) is a unique and biologically active component in Valeriana officinalis L., which has been reported to have a regulatory effect on the cardiovascular system. However, its therapeutic effects on pathological myocardial hypertrophy (PMH) and the underlying mechanisms are undefined. OBJECTIVES Our study aims to elucidate how VA improves PMH, and preliminarily discuss its mechanism. METHODS The efficacy of VA on PMH was confirmed by in vivo and in vitro experiments and the underlying mechanism was investigated by molecular dynamics (MD) simulations and specific siRNA interference. RESULTS VA enhanced cardiomyocyte fatty acid oxidation (FAO), inhibited hyper-activated glycolysis, and improved the unbalanced pyruvate-lactate axis. VA could significantly improve impaired mitochondrial function and reduce the triglyceride (TG) in the hypertrophic myocardium while reducing the lactate (LD) content. Molecular mechanistic studies showed that VA up-regulated the expression of peroxisome proliferator-activated receptor-α (PPARα) and downstream FAO-related genes including CD36, CPT1A, EHHADH, and MCAD. VA reduced the expression of ENO1 and PDK4, the key enzymes in glycolysis. Meanwhile, VA improved the pyruvate-lactate axis and promoted the aerobic oxidation of pyruvate by inhibiting LDAH and MCT4. MD simulations confirmed that VA can bind with the F273 site of PPARα, which proposes VA as a potential activator of the PPARα. CONCLUSION Our results demonstrated that VA might be a potent activator for the PPARα-mediated pathway. VA directly targets the PPARα and subsequently promotes energy metabolism to attenuate PMH, which can be applied as a potentially effective drug for the treatment of HF.
Collapse
Affiliation(s)
- Tiantian Liu
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xu Chen
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Qianbin Sun
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Junjun Li
- School of Chinese Materia, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Qiyan Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Peng Wei
- Beijing Key Laboratory of TCM Syndrome and Formula, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Wei Wang
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; Beijing Key Laboratory of TCM Syndrome and Formula, Beijing University of Chinese Medicine, Beijing 100029, China; State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangdong 510006, China..
| | - Chun Li
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; Beijing Key Laboratory of TCM Syndrome and Formula, Beijing University of Chinese Medicine, Beijing 100029, China; State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangdong 510006, China..
| | - Yong Wang
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China; Yunnan University of Chinese Medicine, Yunnan 650500, China.
| |
Collapse
|
43
|
Zhang ZY, Wang F, Zhou G. Decrease of NAD + Inhibits the Apoptosis of OLP T Cells via Inducing Mitochondrial Fission. J Inflamm Res 2025; 18:1091-1106. [PMID: 39871961 PMCID: PMC11771177 DOI: 10.2147/jir.s502273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 01/12/2025] [Indexed: 01/29/2025] Open
Abstract
Purpose Oral lichen planus (OLP) is a chronic, immune-mediated inflammatory disease involving T cells. Mitochondrial fission plays a crucial role in T cell fate through structural remodeling. Nicotinamide adenine dinucleotide (NAD+) regulates mitochondrial remodeling and function. This study explored the role of NAD+ in modulating mitochondrial fission and apoptosis in T cells under the OLP immune-inflammatory environment. Patients and Methods T cells and plasma were isolated from peripheral blood. Mitochondrial morphology was characterized by transmission electron microscopy and Mito-Tracker staining. OLP plasma-exposed Jurkat T cells were infected with the Drp1 shRNA virus to investigate the role of mitochondrial fission in OLP T cell apoptosis. OLP T cells and OLP plasma-exposed Jurkat T cells were treated with either β-nicotinamide mononucleotide (an NAD+ synthesis precursor) or FK866 (an NAD+ synthesis inhibitor) to assess the effect of NAD+ regulation on mitochondrial remodeling and T cell apoptosis. Results OLP T cells exhibited fragmented mitochondria with elevated dynamin-related protein 1 (Drp1) and reduced mitofusin 2 (Mfn2) expression, accompanied by decreased apoptosis. Drp1 knockdown in OLP plasma-exposed Jurkat T cells increased apoptosis and reduced proliferation. NAD+ levels were reduced in both OLP T cells and OLP plasma-treated Jurkat T cells, leading to enhanced mitochondrial fission, decreased mitochondrial membrane potential (MMP) and respiration function, and reduced apoptosis rate. β-nicotinamide mononucleotide supplementation restored NAD+ levels, suppressed mitochondrial fission, improved MMP, and promoted apoptosis in these cells. Conclusion Reduced NAD+ levels in OLP T cells enhanced mitochondrial fission and contributed to decreased apoptosis. NAD+ supplementation mitigated these effects, suggesting a potential therapeutic strategy for restoring T cell homeostasis in OLP.
Collapse
Affiliation(s)
- Zhuo-Yu Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, People’s Republic of China
| | - Fang Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, People’s Republic of China
- Center for Cariology, Endodontics and Periodontics, Optical Valley Branch, School & Hospital of Stomatology, Wuhan University, Wuhan, People’s Republic of China
| | - Gang Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, People’s Republic of China
- Department of Oral Medicine, School and Hospital of Stomatology, Wuhan University, Wuhan, People’s Republic of China
| |
Collapse
|
44
|
Wang C, Ma C, Xu Y, Chang S, Wu H, Yan C, Chen J, Wu Y, An S, Xu J, Han Q, Jiang Y, Jiang Z, Chu X, Gao H, Zhang X, Chang Y. Dynamics of the mammalian pyruvate dehydrogenase complex revealed by in-situ structural analysis. Nat Commun 2025; 16:917. [PMID: 39843418 PMCID: PMC11754459 DOI: 10.1038/s41467-025-56171-8] [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: 02/29/2024] [Accepted: 01/10/2025] [Indexed: 01/24/2025] Open
Abstract
The multi-enzyme pyruvate dehydrogenase complex (PDHc) links glycolysis to the citric acid cycle and plays vital roles in metabolism, energy production, and cellular signaling. Although all components have been individually characterized, the intact PDHc structure remains unclear, hampering our understanding of its composition and dynamical catalytic mechanisms. Here, we report the in-situ architecture of intact mammalian PDHc by cryo-electron tomography. The organization of peripheral E1 and E3 components varies substantially among the observed PDHcs, with an average of 21 E1 surrounding each PDHc core, and up to 12 E3 locating primarily along the pentagonal openings. In addition, we observed dynamic interactions of the substrate translocating lipoyl domains (LDs) with both E1 and E2, and the interaction interfaces were further analyzed by molecular dynamics simulations. By revealing intrinsic dynamics of PDHc peripheral compositions, our findings indicate a distinctive activity regulation mechanism, through which the number of E1, E3 and functional LDs may be coordinated to meet constantly changing demands of metabolism.
Collapse
Affiliation(s)
- Chen Wang
- Department of Pathology of Sir Run Run Shaw Hospital and Department of Biophysics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Cheng Ma
- Protein Facility, Core Facilities, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yuanyou Xu
- Institute of Microbiology, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Shenghai Chang
- Department of Pathology of Sir Run Run Shaw Hospital and Department of Biophysics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Center of Cryo-Electron Microscopy, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Hangjun Wu
- Department of Pathology of Sir Run Run Shaw Hospital and Department of Biophysics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Center of Cryo-Electron Microscopy, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Chunlan Yan
- Department of Pathology of Sir Run Run Shaw Hospital and Department of Biophysics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jinghua Chen
- Institute of Microbiology, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yongping Wu
- College of Veterinary Medicine, College of Animal Science and Technology, Zhejiang A&F University, Hangzhou, Zhejiang, China
| | - Shaoya An
- Department of Pathology of Sir Run Run Shaw Hospital and Department of Biophysics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jiaqi Xu
- Department of Pathology of Sir Run Run Shaw Hospital and Department of Biophysics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qin Han
- Center of Cryo-Electron Microscopy, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yujie Jiang
- Department of Pathology of Sir Run Run Shaw Hospital and Department of Biophysics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhinong Jiang
- Department of Pathology of Sir Run Run Shaw Hospital and Department of Biophysics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiakun Chu
- Advanced Materials Thrust, The Hong Kong University of Science and Technology (Guangzhou), Guangzhou, Guangdong, China
| | - Haichun Gao
- Institute of Microbiology, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Xing Zhang
- Department of Pathology of Sir Run Run Shaw Hospital and Department of Biophysics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Center of Cryo-Electron Microscopy, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Yunjie Chang
- Center of Cryo-Electron Microscopy, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Department of Infectious Diseases of Sir Run Run Shaw Hospital and Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
45
|
Ni J, Huang S, Yang W, Chen Q, Lin Z. Electrochemiluminescence Detecting and Imaging of Yeast Metabolism Indicated by Endogenetic Co-reactant. Anal Chem 2025; 97:921-927. [PMID: 39700391 DOI: 10.1021/acs.analchem.4c05663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
Glycolysis, a pivotal step in yeast metabolism, plays an indispensable role as a carbohydrate utilization process crucial for cellular survival. Developing advanced technologies to elucidate this fundamental physiological process holds significant scientific implications. Electrochemiluminescence (ECL) imaging exhibits the advantage of negligible background interference and facilitates straightforward visualization, thereby conferring significant value in biomolecular observation. In this study, we present an ECL imaging method for investigating yeast metabolism by utilizing the endogenetic NADH as an efficient coreactant for ECL generation. The yeast glycolysis process drives the conversion of NAD+ to NADH, resulting in enhanced ECL response as well as the increased brightness of ECL images that can be used for quantification of yeast activity. There was a linear correlation between the reciprocal of both the gray value of ECL image and yeast concentration within the range of 6.25 × 106 - 6.25 × 108 CFU/mL. Due to the highly efficient coreactant behavior of NADH, our method demonstrated excellent selectivity with minimal interference. Furthermore, we employed this approach to investigate some toxic inhibitors on yeast metabolism, yielding reliable results. This ECL imaging method not only avoids the use of additional coreactants but also provides a sensitive and intuitive approach for monitoring yeast metabolism, demonstrating great potential in revealing various complex biological processes.
Collapse
Affiliation(s)
- Jiancong Ni
- Fujian Provincial Key Laboratory of Modern Analytical Science and Separation Technology, Fujian Provincial Key Laboratory of Pollution Monitoring and Control, College of Chemistry, Chemical Engineering and Environment, Minnan Normal University, Zhangzhou 363000, China
| | - Shengxiu Huang
- Fujian Provincial Key Laboratory of Modern Analytical Science and Separation Technology, Fujian Provincial Key Laboratory of Pollution Monitoring and Control, College of Chemistry, Chemical Engineering and Environment, Minnan Normal University, Zhangzhou 363000, China
| | - Weiqiang Yang
- Fujian Provincial Key Laboratory of Modern Analytical Science and Separation Technology, Fujian Provincial Key Laboratory of Pollution Monitoring and Control, College of Chemistry, Chemical Engineering and Environment, Minnan Normal University, Zhangzhou 363000, China
| | - Qiaoling Chen
- Fujian Provincial Key Laboratory of Modern Analytical Science and Separation Technology, Fujian Provincial Key Laboratory of Pollution Monitoring and Control, College of Chemistry, Chemical Engineering and Environment, Minnan Normal University, Zhangzhou 363000, China
| | - Zhenyu Lin
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, College of Chemistry, Fuzhou University, Fuzhou 350116, China
| |
Collapse
|
46
|
Shi Y, Zheng H, Wang T, Zhou S, Zhao S, Li M, Cao B. Targeting KRAS: from metabolic regulation to cancer treatment. Mol Cancer 2025; 24:9. [PMID: 39799325 PMCID: PMC11724471 DOI: 10.1186/s12943-024-02216-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 12/25/2024] [Indexed: 01/15/2025] Open
Abstract
The Kirsten rat sarcoma viral oncogene homolog (KRAS) protein plays a key pathogenic role in oncogenesis, cancer progression, and metastasis. Numerous studies have explored the role of metabolic alterations in KRAS-driven cancers, providing a scientific rationale for targeting metabolism in cancer treatment. The development of KRAS-specific inhibitors has also garnered considerable attention, partly due to the challenge of acquired treatment resistance. Here, we review the metabolic reprogramming of glucose, glutamine, and lipids regulated by oncogenic KRAS, with an emphasis on recent insights into the relationship between changes in metabolic mechanisms driven by KRAS mutant and related advances in targeted therapy. We also focus on advances in KRAS inhibitor discovery and related treatment strategies in colorectal, pancreatic, and non-small cell lung cancer, including current clinical trials. Therefore, this review provides an overview of the current understanding of metabolic mechanisms associated with KRAS mutation and related therapeutic strategies, aiming to facilitate the understanding of current challenges in KRAS-driven cancer and to support the investigation of therapeutic strategies.
Collapse
Affiliation(s)
- Yanyan Shi
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, 100191, China
| | - Huiling Zheng
- Department of Gastroenterology, Peking University Third Hospital, Beijing, 100191, China
| | - Tianzhen Wang
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology, Key Laboratory of Assisted Reproduction (Peking University), Peking University Third Hospital, Ministry of Education, Beijing, 100191, China
| | - Shengpu Zhou
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, 100191, China
| | - Shiqing Zhao
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, 100191, China
| | - Mo Li
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China.
- National Clinical Research Center for Obstetrics and Gynecology, Key Laboratory of Assisted Reproduction (Peking University), Peking University Third Hospital, Ministry of Education, Beijing, 100191, China.
| | - Baoshan Cao
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
47
|
Caturano A, Rocco M, Tagliaferri G, Piacevole A, Nilo D, Di Lorenzo G, Iadicicco I, Donnarumma M, Galiero R, Acierno C, Sardu C, Russo V, Vetrano E, Conte C, Marfella R, Rinaldi L, Sasso FC. Oxidative Stress and Cardiovascular Complications in Type 2 Diabetes: From Pathophysiology to Lifestyle Modifications. Antioxidants (Basel) 2025; 14:72. [PMID: 39857406 PMCID: PMC11759781 DOI: 10.3390/antiox14010072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 12/30/2024] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a chronic metabolic disorder that significantly increases the risk of cardiovascular disease, which is the leading cause of morbidity and mortality among diabetic patients. A central pathophysiological mechanism linking T2DM to cardiovascular complications is oxidative stress, defined as an imbalance between reactive oxygen species (ROS) production and the body's antioxidant defenses. Hyperglycemia in T2DM promotes oxidative stress through various pathways, including the formation of advanced glycation end products, the activation of protein kinase C, mitochondrial dysfunction, and the polyol pathway. These processes enhance ROS generation, leading to endothelial dysfunction, vascular inflammation, and the exacerbation of cardiovascular damage. Additionally, oxidative stress disrupts nitric oxide signaling, impairing vasodilation and promoting vasoconstriction, which contributes to vascular complications. This review explores the molecular mechanisms by which oxidative stress contributes to the pathogenesis of cardiovascular disease in T2DM. It also examines the potential of lifestyle modifications, such as dietary changes and physical activity, in reducing oxidative stress and mitigating cardiovascular risks in this high-risk population. Understanding these mechanisms is critical for developing targeted therapeutic strategies to improve cardiovascular outcomes in diabetic patients.
Collapse
Affiliation(s)
- Alfredo Caturano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.C.); (M.R.); (G.T.); (A.P.); (D.N.); (G.D.L.); (I.I.); (M.D.); (R.G.); (C.S.); (E.V.); (R.M.)
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy;
| | - Maria Rocco
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.C.); (M.R.); (G.T.); (A.P.); (D.N.); (G.D.L.); (I.I.); (M.D.); (R.G.); (C.S.); (E.V.); (R.M.)
| | - Giuseppina Tagliaferri
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.C.); (M.R.); (G.T.); (A.P.); (D.N.); (G.D.L.); (I.I.); (M.D.); (R.G.); (C.S.); (E.V.); (R.M.)
| | - Alessia Piacevole
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.C.); (M.R.); (G.T.); (A.P.); (D.N.); (G.D.L.); (I.I.); (M.D.); (R.G.); (C.S.); (E.V.); (R.M.)
| | - Davide Nilo
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.C.); (M.R.); (G.T.); (A.P.); (D.N.); (G.D.L.); (I.I.); (M.D.); (R.G.); (C.S.); (E.V.); (R.M.)
| | - Giovanni Di Lorenzo
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.C.); (M.R.); (G.T.); (A.P.); (D.N.); (G.D.L.); (I.I.); (M.D.); (R.G.); (C.S.); (E.V.); (R.M.)
| | - Ilaria Iadicicco
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.C.); (M.R.); (G.T.); (A.P.); (D.N.); (G.D.L.); (I.I.); (M.D.); (R.G.); (C.S.); (E.V.); (R.M.)
| | - Mariarosaria Donnarumma
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.C.); (M.R.); (G.T.); (A.P.); (D.N.); (G.D.L.); (I.I.); (M.D.); (R.G.); (C.S.); (E.V.); (R.M.)
| | - Raffaele Galiero
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.C.); (M.R.); (G.T.); (A.P.); (D.N.); (G.D.L.); (I.I.); (M.D.); (R.G.); (C.S.); (E.V.); (R.M.)
| | - Carlo Acierno
- Azienda Ospedaliera Regionale San Carlo, 85100 Potenza, Italy;
| | - Celestino Sardu
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.C.); (M.R.); (G.T.); (A.P.); (D.N.); (G.D.L.); (I.I.); (M.D.); (R.G.); (C.S.); (E.V.); (R.M.)
| | - Vincenzo Russo
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA;
- Division of Cardiology, Department of Medical Translational Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Erica Vetrano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.C.); (M.R.); (G.T.); (A.P.); (D.N.); (G.D.L.); (I.I.); (M.D.); (R.G.); (C.S.); (E.V.); (R.M.)
| | - Caterina Conte
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy;
- Department of Endocrinology, Nutrition and Metabolic Diseases, IRCCS MultiMedica, 20099 Milan, Italy
| | - Raffaele Marfella
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.C.); (M.R.); (G.T.); (A.P.); (D.N.); (G.D.L.); (I.I.); (M.D.); (R.G.); (C.S.); (E.V.); (R.M.)
| | - Luca Rinaldi
- Department of Medicine and Health Sciences “Vincenzo Tiberio”, Università degli Studi del Molise, 86100 Campobasso, Italy
| | - Ferdinando Carlo Sasso
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.C.); (M.R.); (G.T.); (A.P.); (D.N.); (G.D.L.); (I.I.); (M.D.); (R.G.); (C.S.); (E.V.); (R.M.)
| |
Collapse
|
48
|
Wang D, Zeng N, Li C, Li C, Wang Y, Chen B, Long J, Zhang N, Li B. Integrative analysis of transcriptome and metabolome profiling uncovers underlying mechanisms of the enhancement of the synthesis of biofilm in Sporobolomyces pararoseus NGR under acidic conditions. Microb Cell Fact 2025; 24:9. [PMID: 39773469 PMCID: PMC11706151 DOI: 10.1186/s12934-024-02636-2] [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: 03/27/2024] [Accepted: 12/24/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Sporobolomyces pararoseus is a well-studied oleaginous red yeast that can synthesize a variety of high value-added bioactive compounds. Biofilm is one of the important biological barriers for microbial cells to resist environmental stresses and maintain stable fermentation process. Here, the effect of acidic conditions on the biosynthesis of biofilms in S. pararoseus NGR was investigated through the combination of morphology, biochemistry, and multi-omics approaches. RESULTS The results showed that the acidic environment was the key factor to trigger the biofilm formation of S. pararoseus NGR. When S. pararoseus NGR was cultured under pH 4.7, the colony morphology was wrinkled, the cells were wrapped by a large amount of extracellular matrix, and the hydrophobicity and anti-oxidative stress ability were significantly improved, and the yield of intracellular carotenoids was significantly increased. Transcriptome and metabolome profiling indicated that carbohydrate metabolism, amino acid metabolism, lipid metabolism, and nucleic acid metabolism in S. pararoseus NGR cells were significantly enriched in biofilm cells under pH 4.7 culture conditions, including 56 differentially expressed genes and 341 differential metabolites. CONCLUSIONS These differential genes and metabolites may play an important role in the formation of biofilms by S. pararoseus NGR in response to acidic stress. The results will provide strategies for the development and utilization of beneficial microbial biofilms, and provide theoretical support for the industrial fermentation production of microorganisms to improve their resistance and maintain stable growth.
Collapse
Affiliation(s)
- Dandan Wang
- College of Land and Environment, Shenyang Agricultural University, Shenyang, 110866, People's Republic of China
| | - Nan Zeng
- College of Land and Environment, Shenyang Agricultural University, Shenyang, 110866, People's Republic of China
| | - Chunji Li
- College of Agriculture and Biology, Zhongkai University of Agriculture and Engineering, Guangzhou, 510225, People's Republic of China
- Innovative Institute for Plant Health, Zhongkai University of Agriculture and Engineering, Guangzhou, 510225, People's Republic of China
- Key Laboratory of Green Prevention and Control on Fruits and Vegetables in South China, Ministry of Agriculture and Rural Affairs, Guangzhou, 510225, People's Republic of China
| | - Chunwang Li
- College of Bioscience and Biotechnology, Shenyang Agricultural University, Shenyang, 110866, People's Republic of China
| | - Yunjiao Wang
- College of Bioscience and Biotechnology, Shenyang Agricultural University, Shenyang, 110866, People's Republic of China
| | - Bin Chen
- College of Bioscience and Biotechnology, Shenyang Agricultural University, Shenyang, 110866, People's Republic of China
| | - Jiajia Long
- College of Bioscience and Biotechnology, Shenyang Agricultural University, Shenyang, 110866, People's Republic of China
| | - Ning Zhang
- College of Bioscience and Biotechnology, Shenyang Agricultural University, Shenyang, 110866, People's Republic of China.
| | - Bingxue Li
- College of Land and Environment, Shenyang Agricultural University, Shenyang, 110866, People's Republic of China.
| |
Collapse
|
49
|
Kumar A, Das JK, Peng HY, Wang L, Ballard DJ, Ren Y, Xiong X, Ren X, Yang JM, de Figueiredo P, Song J. Metabolic fitness of NAC1-deficient Tregs in the tumor microenvironment fuels tumor growth. JCI Insight 2025; 10:e186000. [PMID: 39773913 PMCID: PMC11949012 DOI: 10.1172/jci.insight.186000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 01/02/2025] [Indexed: 01/11/2025] Open
Abstract
The nucleus accumbens-associated protein 1 (NAC1) has recently emerged as a pivotal factor in oncogenesis by promoting glycolysis. Deletion of NAC1 in regulatory T cells (Tregs) has been shown to enhance FoxP3 stability, a suppressor of glycolysis. This study delves into the intriguing dual role of NAC1, uncovering that Treg-specific deletion of NAC1 fosters metabolic fitness in Tregs, thereby promoting tumorigenesis. Our results unveil that NAC1-deficient Tregs exhibited prolonged survival and heightened function, particularly in acidic environments. Mechanistically, we find that NAC1-deficient Tregs adapted to adverse conditions by upregulating FoxP3 expression, engaging in CD36-mediated lipid metabolism, and enhancing peroxisome proliferator-activated receptor gamma coactivator 1-alpha-regulated mitochondrial function. In mouse tumor xenograft models, NAC1-deficient mice demonstrated increased susceptibility to tumor growth. Notably, Tregs lacking NAC1 not only displayed elevated lipid metabolism and mitochondrial fitness but also exhibited enhanced tumoral infiltration. Adoptive Treg transfer experiments further underscored the supportive role of NAC1-deficient Tregs in tumor growth. These findings suggest that modulating NAC1 expression in FoxP3+ Tregs could serve as a promising approach to augment antitumor immunity. Understanding the intricate interplay between NAC1 and Tregs opens avenues for potential therapeutic strategies targeting the tumor microenvironment.
Collapse
Affiliation(s)
- Anil Kumar
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Jugal Kishore Das
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Hao-Yun Peng
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, Texas, USA
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, USA
| | - Liqing Wang
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, Texas, USA
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, USA
| | - Darby Jane Ballard
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Yijie Ren
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Xiaofang Xiong
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Xingcong Ren
- Department of Toxicology and Cancer Biology, Department of Pharmacology and Nutritional Science, and Markey Cancer Center, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Jin-Ming Yang
- Department of Toxicology and Cancer Biology, Department of Pharmacology and Nutritional Science, and Markey Cancer Center, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Paul de Figueiredo
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, Missouri, USA
| | - Jianxun Song
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, Texas, USA
| |
Collapse
|
50
|
Alhumaidi R, Huang H, Saade MC, Clark AJ, Parikh SM. NAD + metabolism in acute kidney injury and chronic kidney disease transition. Trends Mol Med 2025:S1471-4914(24)00337-X. [PMID: 39757045 DOI: 10.1016/j.molmed.2024.12.004] [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: 09/12/2024] [Revised: 11/21/2024] [Accepted: 12/09/2024] [Indexed: 01/07/2025]
Abstract
Disturbances in kidney tubular cell metabolism are increasingly recognized as a feature of acute kidney injury (AKI). In AKI, tubular epithelial cells undergo abnormal metabolic shifts that notably disrupt NAD+ metabolism. Recent advancements have highlighted the critical role of NAD+ metabolism in AKI, revealing that acute disruptions may lead to lasting cellular changes, thereby promoting the transition to chronic kidney disease (CKD). This review explores the molecular mechanisms underlying metabolic dysfunction in AKI, with a focus on NAD+ metabolism, and proposes several cellular processes through which acute aberrations in NAD+ may contribute to long-term changes in the kidney.
Collapse
Affiliation(s)
- Rahil Alhumaidi
- Division of Nephrology, Department of Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Huihui Huang
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Marie Christelle Saade
- Division of Nephrology, Department of Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Amanda J Clark
- Division of Nephrology, Department of Pediatrics, University of Texas Southwestern and Children's Medical Center, Dallas, TX, USA
| | - Samir M Parikh
- Division of Nephrology, Department of Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|