1
|
Orellana AMM, Mazucanti CH, Andreotti DZ, de Sá Lima L, Kawamoto EM, Scavone C. Effects of decrease in Klotho protein expression on insulin signaling and levels of proteins related to brain energy metabolism. Eur J Pharmacol 2025; 997:177587. [PMID: 40187598 DOI: 10.1016/j.ejphar.2025.177587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 03/31/2025] [Accepted: 04/02/2025] [Indexed: 04/07/2025]
Abstract
Mutations in Klotho have been associated with premature ageing and cognitive dysfunction. Although highly expressed in specific regions of the brain, the actions of Klotho in the central nervous system (CNS) remain largely unknown. Here, we show that animals with a mutated hypomorphic Klotho gene have altered glycaemic regulation, suggesting higher insulin sensitivity. In the CNS, pathways related to insulin intracellular signalling were found to be up-regulated in the hippocampus, with higher activation of protein kinase B and mammalian target of rapamycin and inactivation of the transcription factors forkhead box O (FOXO)-1 and FOXO-3a. In addition, the present study showed that in the hippocampi of wild-type aged mice, where Klotho is naturally downregulated, the levels of some proteins related to energy metabolism and metabolic coupling between neurones and astrocytes, such as monocarboxylate transporter 2 and 4, 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase isoform 3 and lactate dehydrogenase enzymes isoforms A and B were altered. These findings suggest that Klotho plays an essential role in regulating proteins and genes related to metabolic coupling in the brain.
Collapse
Affiliation(s)
- Ana Maria Marques Orellana
- Laboratory of Molecular Neuropharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil; Laboratory of Molecular and Functional Neurobiology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Caio Henrique Mazucanti
- Laboratory of Molecular Neuropharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil; Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, Maryland, USA
| | - Diana Zukas Andreotti
- Laboratory of Molecular and Functional Neurobiology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Larissa de Sá Lima
- Laboratory of Molecular Neuropharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil; Laboratory of Molecular and Functional Neurobiology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Elisa Mitiko Kawamoto
- Laboratory of Molecular and Functional Neurobiology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Cristoforo Scavone
- Laboratory of Molecular Neuropharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
2
|
Plata-Gómez AB, Ho PC. Age- and diet-instructed metabolic rewiring of the tumor-immune microenvironment. J Exp Med 2025; 222:e20241102. [PMID: 40214641 PMCID: PMC11987706 DOI: 10.1084/jem.20241102] [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: 12/20/2024] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 04/14/2025] Open
Abstract
The tumor-immune microenvironment (TIME) plays a critical role in tumor development and metastasis, as it influences the evolution of tumor cells and fosters an immunosuppressive state by intervening the metabolic reprogramming of infiltrating immune cells. Aging and diet significantly impact the metabolic reprogramming of the TIME, contributing to cancer progression and immune evasion. With aging, immune cell function declines, leading to a proinflammatory state and metabolic alterations such as increased oxidative stress and mitochondrial dysfunction, which compromise antitumor immunity. Similarly, dietary factors, particularly high-fat and high-sugar diets, promote metabolic shifts, creating a permissive TIME by fostering tumor-supportive immune cell phenotypes while impairing the tumoricidal activity of immune cells. In contrast, dietary restrictions have been shown to restore immune function by modulating metabolism and enhancing antitumor immune responses. Here, we discuss the intricate interplay between aging, diet, and metabolic reprogramming in shaping the TIME, with a particular focus on T cells, and highlight therapeutic strategies targeting these pathways to empower antitumor immunity.
Collapse
Affiliation(s)
- Ana Belén Plata-Gómez
- Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Ping-Chih Ho
- Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
3
|
Farina S, Cattabiani A, Mandge D, Shichkova P, Isbister JB, Jacquemier J, King JG, Markram H, Keller D. A multiscale electro-metabolic model of a rat neocortical circuit reveals the impact of ageing on central cortical layers. PLoS Comput Biol 2025; 21:e1013070. [PMID: 40393041 DOI: 10.1371/journal.pcbi.1013070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 04/19/2025] [Indexed: 05/22/2025] Open
Abstract
The high energetic demands of the brain arise primarily from neuronal activity. Neurons consume substantial energy to transmit information as electrical signals and maintain their resting membrane potential. These energetic requirements are met by the neuro-glial-vascular (NGV) ensemble, which generates energy in a coupled metabolic process. In ageing, metabolic function becomes impaired, producing less energy and, consequently, the system is unable to sustain the neuronal energetic needs. We propose a multiscale model of electro-metabolic coupling in a reconstructed rat neocortex. This combines an electro-morphologically reconstructed electrophysiological model with a detailed NGV metabolic model. Our results demonstrate that the large-scale model effectively captures electro-metabolic processes at the circuit level, highlighting the importance of heterogeneity within the circuit, where energetic demands vary according to neuronal characteristics. Finally, in metabolic ageing, our model indicates that the middle cortical layers are particularly vulnerable to energy impairment.
Collapse
Affiliation(s)
- Sofia Farina
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Alessandro Cattabiani
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Darshan Mandge
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Polina Shichkova
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Geneva, Switzerland
- Biognosys AG, Schlieren, Switzerland
| | - James B Isbister
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Jean Jacquemier
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - James G King
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Henry Markram
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Geneva, Switzerland
- Brain Mind Institute, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Daniel Keller
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Geneva, Switzerland
| |
Collapse
|
4
|
Varghese N, Szabo L, Cader MZ, Lejri I, Grimm A, Eckert A. Tracing mitochondrial marks of neuronal aging in iPSCs-derived neurons and directly converted neurons. Commun Biol 2025; 8:723. [PMID: 40346193 PMCID: PMC12064796 DOI: 10.1038/s42003-025-08152-2] [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: 09/02/2024] [Accepted: 05/01/2025] [Indexed: 05/11/2025] Open
Abstract
This study aims to determine if neurons derived from induced pluripotent stem cells (iPSCsNs) and directly converted neurons (iNs) from the same source cells exhibit changes in mitochondrial properties related to aging. This research addresses the uncertainty around whether aged iPSCsNs retain aging-associated mitochondrial impairments upon transitioning through pluripotency while direct conversion maintains these impairments. We observe that both aged models exhibit characteristics of aging, such as decreased ATP, mitochondrial membrane potential, respiration, NAD+/NADH ratio, and increased radicals and mitochondrial mass. In addition, both neuronal models show a fragmented mitochondrial network. However, aged iPSCsNs do not exhibit a metabolic shift towards glycolysis, unlike aged iNs. Furthermore, mRNA expression differed significantly between aged iPSCsNs and aged iNs. The study concludes that aged iPSCsNs may differ in transcriptomics and the aging-associated glycolytic shift but can be a valuable tool for studying specific feature of mitochondrial neuronal aging in vitro alongside aged iNs.
Collapse
Affiliation(s)
- Nimmy Varghese
- Research Cluster Molecular and Cognitive Neurosciences, University of Basel, Basel, Switzerland
- Neurobiology Lab for Brain Aging and Mental Health, University Psychiatric Clinics Basel, Basel, Switzerland
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Leonora Szabo
- Research Cluster Molecular and Cognitive Neurosciences, University of Basel, Basel, Switzerland
- Neurobiology Lab for Brain Aging and Mental Health, University Psychiatric Clinics Basel, Basel, Switzerland
| | - M Zameel Cader
- Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, UK
| | - Imane Lejri
- Research Cluster Molecular and Cognitive Neurosciences, University of Basel, Basel, Switzerland
- Neurobiology Lab for Brain Aging and Mental Health, University Psychiatric Clinics Basel, Basel, Switzerland
| | - Amandine Grimm
- Research Cluster Molecular and Cognitive Neurosciences, University of Basel, Basel, Switzerland
- Neurobiology Lab for Brain Aging and Mental Health, University Psychiatric Clinics Basel, Basel, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Anne Eckert
- Research Cluster Molecular and Cognitive Neurosciences, University of Basel, Basel, Switzerland.
- Neurobiology Lab for Brain Aging and Mental Health, University Psychiatric Clinics Basel, Basel, Switzerland.
| |
Collapse
|
5
|
Asadian E, Hekmat F, Hafezi Kahnamouei M, Mohammadpour R, Shahrokhian S, Sasanpour P. Supercapacitor-powered wearable biosensor for continuous lactate monitoring from sweat. Biosens Bioelectron 2025; 275:117226. [PMID: 39933404 DOI: 10.1016/j.bios.2025.117226] [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: 11/05/2024] [Revised: 01/29/2025] [Accepted: 02/02/2025] [Indexed: 02/13/2025]
Abstract
The development of wearable sensing platforms for continuous monitoring of sweat biomarkers has gained significant attention, particularly for lactate detection. This study presents the design and fabrication of a novel wearable lactate biosensor that integrates a flexible supercapacitor power supply with an advanced lactate sensing platform. The sensing platform features NiCo nanosheets electrodeposited onto nanocages of bimetallic CoFe Prussian Blue analogue (PBA), providing an optimal microenvironment for the immobilization of lactate oxidase (LOx) enzymes. The CoFe PBA nanocages act as efficient electrocatalysts for the reduction of hydrogen peroxide, enhancing the sensor's performance. The electrode exhibits a sensitivity of 262 μA mM-1cm-2 and demonstrates a short response time (<5 s), making it suitable for real-time monitoring applications. Additionally, the energy supply unit is constructed using a wearable conductive carbon textile (CCT) substrate modified with NiCoS through electrochemical deposition, achieving the necessary electrical conductivity. A flexible asymmetric supercapacitor (ASC) is then developed utilizing NiCoS@CCT and FeS@CNT@CCT as the positive and negative electrodes, respectively. This ASC exhibits remarkable electrochemical properties, including a high specific capacitance of 205 F g⁻1, notable energy density at elevated power densities, and excellent rate capability. Integrating these components with a custom-designed electronic circuit board results in a lightweight wearable sensor capable of continuous lactate monitoring in perspiration. This innovative approach demonstrates significant potential for advancing point-of-care health monitoring technologies.
Collapse
Affiliation(s)
- Elham Asadian
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, 19689-17313, Iran; Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 19689-17313, Iran.
| | - Farzaneh Hekmat
- Department of Chemistry, Shahid Beheshti University, Tehran, 1983969411, Iran.
| | - Mohammad Hafezi Kahnamouei
- Department of Conservation of Cultural-Historical Properties and Archaeometry, Art University of Isfahan, P. O. Box: 1744, Isfahan, Iran
| | - Raheleh Mohammadpour
- Center for Nanoscience and Nanotechnology, Institute for Convergence Science & Technology, Sharif University of Technology, Tehran, Iran
| | - Saeed Shahrokhian
- Department of Chemistry, Sharif University of Technology, Azadi Avenue, Tehran, 11155-9516, Iran
| | - Pezhman Sasanpour
- Department of Medical Physics & Biomedical Engineering, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; School of Nanoscience, Institute for Research in Fundamental Sciences (IPM), 19395-5531, Tehran, Iran.
| |
Collapse
|
6
|
Na D, Zhang Z, Meng M, Li M, Gao J, Kong J, Zhang G, Guo Y. Energy Metabolism and Brain Aging: Strategies to Delay Neuronal Degeneration. Cell Mol Neurobiol 2025; 45:38. [PMID: 40259102 PMCID: PMC12011708 DOI: 10.1007/s10571-025-01555-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 04/09/2025] [Indexed: 04/23/2025]
Abstract
Aging is characterized by a gradual decline in physiological functions, with brain aging being a major risk factor for numerous neurodegenerative diseases. Given the brain's high energy demands, maintaining an adequate ATP supply is crucial for its proper function. However, with advancing age, mitochondria dysfunction and a deteriorating energy metabolism lead to reduced overall energy production and impaired mitochondrial quality control (MQC). As a result, promoting healthy aging has become a key focus in contemporary research. This review examines the relationship between energy metabolism and brain aging, highlighting the connection between MQC and energy metabolism, and proposes strategies to delay brain aging by targeting energy metabolism.
Collapse
Affiliation(s)
- Donghui Na
- Department of Forensic Medicine, Hebei North University, Zhangjiakou, Hebei, China
| | - Zechen Zhang
- Mudi Meng Honors College, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Meng Meng
- Department of Forensic Medicine, Hebei North University, Zhangjiakou, Hebei, China
| | - Meiyu Li
- Department of Forensic Medicine, Hebei North University, Zhangjiakou, Hebei, China
- Department of Pathology, Hebei North University, Zhangjiakou, Hebei, China
| | - Junyan Gao
- Department of Forensic Medicine, Hebei North University, Zhangjiakou, Hebei, China
| | - Jiming Kong
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB, Canada.
| | - Guohui Zhang
- Department of Forensic Medicine, Hebei North University, Zhangjiakou, Hebei, China.
| | - Ying Guo
- Department of Forensic Medicine, Hebei North University, Zhangjiakou, Hebei, China.
- Department of Pathology, Hebei North University, Zhangjiakou, Hebei, China.
- Hebei Key Laboratory of Neuropharmacology, Hebei North University, Zhangjiakou, Hebei, China.
| |
Collapse
|
7
|
Altea-Manzano P, Decker-Farrell A, Janowitz T, Erez A. Metabolic interplays between the tumour and the host shape the tumour macroenvironment. Nat Rev Cancer 2025; 25:274-292. [PMID: 39833533 DOI: 10.1038/s41568-024-00786-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/10/2024] [Indexed: 01/22/2025]
Abstract
Metabolic reprogramming of cancer cells and the tumour microenvironment are pivotal characteristics of cancers, and studying these processes offer insights and avenues for cancer diagnostics and therapeutics. Recent advancements have underscored the impact of host systemic features, termed macroenvironment, on facilitating cancer progression. During tumorigenesis, these inherent features of the host, such as germline genetics, immune profile and the metabolic status, influence how the body responds to cancer. In parallel, as cancer grows, it induces systemic effects beyond the primary tumour site and affects the macroenvironment, for example, through inflammation, the metabolic end-stage syndrome of cachexia, and metabolic dysregulation. Therefore, understanding the intricate metabolic interplay between the tumour and the host is a growing frontier in advancing cancer diagnosis and therapy. In this Review, we explore the specific contribution of the metabolic fitness of the host to cancer initiation, progression and response to therapy. We then delineate the complex metabolic crosstalk between the tumour, the microenvironment and the host, which promotes disease progression to metastasis and cachexia. The metabolic relationships among the host, cancer pathogenesis and the consequent responsive systemic manifestations during cancer progression provide new perspectives for mechanistic cancer therapy and improved management of patients with cancer.
Collapse
Affiliation(s)
| | | | | | - Ayelet Erez
- Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
8
|
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
|
9
|
Yu Z, Lozupone M, Chen J, Bao Z, Ruan Q, Panza F. The Biological Rationale for Integrating Intrinsic Capacity Into Frailty Models. Clin Interv Aging 2025; 20:273-286. [PMID: 40060276 PMCID: PMC11890019 DOI: 10.2147/cia.s509990] [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: 12/11/2024] [Accepted: 02/26/2025] [Indexed: 05/13/2025] Open
Abstract
The assessment and management of two function-centered clinical care models, frailty and intrinsic capacity decline have been proposed to achieve healthy aging. To implement these two care models, several different guidelines have been advocated by different health organizations, which has resulted in confusion and cost-ineffective results in healthcare practice. Although there are various operational definitions and screening tools of frailty, the most accepted operational definitions are based on the recognition of frailty phenotypes or deficit accumulation-based frailty indexes. Intrinsic capacity, referred to as the total physical and mental capacities for individual to undertake daily tasks in everyday life, is another care model, including five domains. Similar or identical instruments have been used to assess frailty and intrinsic capacity. In the present narrative review, we outlined the biological rationale for integrating intrinsic capacity into frailty models and highlighted the hierarchical and energy-dependent order of the intrinsic capacity domains. The vitality domain or energy metabolism-related capacity, is the highest order dimension and the basis of other intrinsic capacity domains. Vitality vulnerability manifests as a pre-frailty status in function-centered healthy aging. We provided a conceptual framework of frailty phenotypes and frailty indexes based on the hierarchical and energy-dependent order of the intrinsic capacity domains, particularly vitality capacity. To facilitate the clinical translation of the framework, some potential energy metabolism-related biomarkers have also been proposed as critical components for assessing and screening vitality capacity in older age. The integrating framework not only provides testable theoretical hypotheses, particularly about vitality as a foundational element in aging, but could serve as a starting point for further research to unravel the mechanisms of frailty. It also improves cost-effectiveness for optimizing aging interventions in clinical healthcare and public health policies of healthy aging.
Collapse
Affiliation(s)
- Zhuowei Yu
- Laboratory of Aging, Anti-Aging & Cognitive Performance, Shanghai Institute of Geriatrics and Gerontology, Huadong Hospital Affiliated to Fudan University, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of Clinical Geriatrics, Shanghai Institute of Geriatrics and Gerontology, Huadong hospital affiliated to Shanghai Medical college, Fudan University, Shanghai, People’s Republic of China
- Department of Geriatrics, Huadong Hospital affiliated to Fudan University, Shanghai, People’s Republic of China
| | - Madia Lozupone
- Department of Translational Biomedicine and Neuroscience “DiBraiN”, University of Bari Aldo Moro, Bari, Italy
| | - Jie Chen
- Shanghai Key Laboratory of Clinical Geriatrics, Shanghai Institute of Geriatrics and Gerontology, Huadong hospital affiliated to Shanghai Medical college, Fudan University, Shanghai, People’s Republic of China
- Department of Geriatrics, Huadong Hospital affiliated to Fudan University, Shanghai, People’s Republic of China
| | - Zhijun Bao
- Laboratory of Aging, Anti-Aging & Cognitive Performance, Shanghai Institute of Geriatrics and Gerontology, Huadong Hospital Affiliated to Fudan University, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of Clinical Geriatrics, Shanghai Institute of Geriatrics and Gerontology, Huadong hospital affiliated to Shanghai Medical college, Fudan University, Shanghai, People’s Republic of China
- Department of Geriatrics, Huadong Hospital affiliated to Fudan University, Shanghai, People’s Republic of China
| | - Qingwei Ruan
- Laboratory of Aging, Anti-Aging & Cognitive Performance, Shanghai Institute of Geriatrics and Gerontology, Huadong Hospital Affiliated to Fudan University, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of Clinical Geriatrics, Shanghai Institute of Geriatrics and Gerontology, Huadong hospital affiliated to Shanghai Medical college, Fudan University, Shanghai, People’s Republic of China
| | - Francesco Panza
- Department of Interdisciplinary Medicine, Medical Clinic, and Geriatrics “Cesare Frugoni”, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
10
|
Deery HA, Liang EX, Moran C, Egan GF, Jamadar SD. Metabolic connectivity has greater predictive utility for age and cognition than functional connectivity. Brain Commun 2025; 7:fcaf075. [PMID: 40008331 PMCID: PMC11851278 DOI: 10.1093/braincomms/fcaf075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 01/04/2025] [Accepted: 02/16/2025] [Indexed: 02/27/2025] Open
Abstract
Recently developed high temporal resolution functional (18F)-fluorodeoxyglucose positron emission tomography (fPET) offers promise as a method for indexing the dynamic metabolic state of the brain in vivo by directly measuring a time series of metabolism at the post-synaptic neuron. This is distinct from functional magnetic resonance imaging (fMRI) that reflects a combination of metabolic, haemodynamic and vascular components of neuronal activity. The value of using fPET to understand healthy brain ageing and cognition over fMRI is currently unclear. Here, we use simultaneous fPET/fMRI to compare metabolic and functional connectivity and test their predictive ability for ageing and cognition. Whole-brain fPET connectomes showed moderate topological similarities to fMRI connectomes in a cross-sectional comparison of 40 younger (mean age 27.9 years; range 20-42) and 46 older (mean 75.8; 60-89) adults. There were more age-related within- and between-network connectivity and graph metric differences in fPET than fMRI. fPET was also associated with performance in more cognitive domains than fMRI. These results suggest that ageing is associated with a reconfiguration of metabolic connectivity that differs from haemodynamic alterations. We conclude that metabolic connectivity has greater predictive utility for age and cognition than functional connectivity and that measuring glucodynamic changes has promise as a biomarker for age-related cognitive decline.
Collapse
Affiliation(s)
- Hamish A Deery
- School of Psychological Sciences, Monash University, Melbourne 3800, Australia
- Monash Biomedical Imaging, Monash University, Melbourne 3800, Australia
| | - Emma X Liang
- School of Psychological Sciences, Monash University, Melbourne 3800, Australia
- Monash Biomedical Imaging, Monash University, Melbourne 3800, Australia
| | - Chris Moran
- School of Public Health and Preventive Medicine, Monash University, Melbourne 3004, Australia
| | - Gary F Egan
- Monash Biomedical Imaging, Monash University, Melbourne 3800, Australia
| | - Sharna D Jamadar
- School of Psychological Sciences, Monash University, Melbourne 3800, Australia
- Monash Biomedical Imaging, Monash University, Melbourne 3800, Australia
| |
Collapse
|
11
|
Li P, Hu Y, Tong L, Bi X. High-intensity training on CREB activation for improving brain health: a narrative review of possible molecular talks. Front Endocrinol (Lausanne) 2025; 15:1498495. [PMID: 39902166 PMCID: PMC11788139 DOI: 10.3389/fendo.2024.1498495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 12/27/2024] [Indexed: 02/05/2025] Open
Abstract
Although physical exercise has obvious benefits in brain physiology, the molecular biomarkers induced by exercise protocols are inconclusive. Evidence indicates that exercise interventions are effective in shaping brain physiology. However, the potential mediator for improving brain functions is uncertain. CREB is one of the potential targets of exercise that triggers various molecular cross-talk to improve neurogenesis, long-term potentiation, and synaptogenesis. Therefore, CREB may be situated on the causal path between maintaining brain health and exercising. To support this, studies have shown that exercise-mediated CREB phosphorylation improves cognitive functions and memory. In addition, among the protocols of exercise (types, duration, and frequency), the intensity has been reported to be the most effective in triggering CREB-mediated molecular signaling. For example, HIT increases the synthesis of CREB, which may not only induce brain physiology but also induce brain pathology by higher activation of its downstream targets, such as BDNF. Therefore, this review aims to understand the effects of HIT on CREB function and how HIT can mediate the CREB-induced molecular cross-talk for maintaining brain health.
Collapse
Affiliation(s)
- Ping Li
- Faculty of Sports Science, Ningbo University, Ningbo, China
| | - Yan Hu
- Ningbo High-tech Zone Playing Kindergarten, Ningbo, China
| | - Ligang Tong
- Xianjiang Honors School of Arts and Physical Education, Ningbo Childhood Education College, Ningbo, China
| | - Xuecui Bi
- Physical Education Institute, Capital University of Physical Education and Sports, Beijing, China
| |
Collapse
|
12
|
Škandík M, Friess L, Vázquez-Cabrera G, Keane L, Grabert K, Cruz De Los Santos M, Posada-Pérez M, Baleviciute A, Cheray M, Joseph B. Age-associated microglial transcriptome leads to diminished immunogenicity and dysregulation of MCT4 and P2RY12/P2RY13 related functions. Cell Death Discov 2025; 11:16. [PMID: 39828750 PMCID: PMC11743796 DOI: 10.1038/s41420-025-02295-1] [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: 11/20/2024] [Revised: 12/17/2024] [Accepted: 01/10/2025] [Indexed: 01/22/2025] Open
Abstract
The aging process is marked by a time-dependent deterioration in cellular functions, particularly the immune and neural systems. Understanding the phenotype acquisition of microglia, the sentinel immune cells of the brain, is crucial for understanding the nature of age-related neurological diseases. However, the specific phenotype adopted by microglia during aging remains a subject of debate and is contingent on the chosen experimental model. To address these unresolved questions, we employed a novel and highly controlled approach utilizing long-term cultivated BV-2 microglia, exempted from additional external stimuli. Our findings revealed that aged microglial cells, in comparison to their younger counterparts, acquire a distinct gene expression profile, primarily characterized by alterations in microglial immune response. Indeed, pro-inflammatory stimulated aged and young BV-2 microglia exhibited similar transcriptomic profiles, yet the response intensity to the stimulus was markedly muted in the aged microglia. Functional neurotoxic assays confirmed diminished neuronal death in coculture with aged, activated microglia, underscoring a compromised immune response. Furthermore, a subsequent comparative analysis of aged BV-2 microglia with established transcriptomic microglial datasets from aged mice and humans identified 13 overlapping genes, laying the foundation for identifying core microglial aging signature. Particularly noteworthy were SLC16A3 and P2RY13, which consistently exhibited upregulation and downregulation, respectively, across all datasets. Additionally, four other genes-CAPG, LGALS3BP, NRIP1, and P2RY12-were found to share regulatory patterns in response to both aging and extrinsic activation. An in-depth investigation focused on SLC16A3, encoding the high-affinity lactate transporter MCT4, revealed disruptions in extracellular acidification rate and lactate concentration with age. Microglial purine sensing and motility capacities, regulated by P2RY12/P2RY13, displayed age-related alterations. Remarkably, protein analysis in human brain tissue validated the observed upregulation of MCT4 and downregulation of P2RY12 in aged microglia. In conclusion, our study unveils a distinct phenotype in aged microglia characterized by compromised immune responsiveness. Through the integration of in vitro cultured BV-2 microglia with primary microglia datasets, we identify critical molecular determinants of microglial cellular aging confirmed in human-aged brain tissue. This comprehensive approach offers potential insights for understanding and potentially reprogramming aged microglia, with implications for combating age-related neurological disorders.
Collapse
Affiliation(s)
- Martin Škandík
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lara Friess
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Lily Keane
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Kathleen Grabert
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Mireia Cruz De Los Santos
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Mercedes Posada-Pérez
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Center for Neuromusculoskeletal Restorative Medicine, Hong-Kong, China
| | - Austeja Baleviciute
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Mathilde Cheray
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Bertrand Joseph
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
- Center for Neuromusculoskeletal Restorative Medicine, Hong-Kong, China.
| |
Collapse
|
13
|
Lana D, Ugolini F, Iovino L, Attorre S, Giovannini MG. Astrocytes phenomics as new druggable targets in healthy aging and Alzheimer's disease progression. Front Cell Neurosci 2025; 18:1512985. [PMID: 39835288 PMCID: PMC11743640 DOI: 10.3389/fncel.2024.1512985] [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: 10/17/2024] [Accepted: 12/13/2024] [Indexed: 01/22/2025] Open
Abstract
For over a century after their discovery astrocytes were regarded merely as cells located among other brain cells to hold and give support to neurons. Astrocytes activation, "astrocytosis" or A1 functional state, was considered a detrimental mechanism against neuronal survival. Recently, the scientific view on astrocytes has changed. Accumulating evidence indicate that astrocytes are not homogeneous, but rather encompass heterogeneous subpopulations of cells that differ from each other in terms of transcriptomics, molecular signature, function and response in physiological and pathological conditions. In this review, we report and discuss the recent literature on the phenomic differences of astrocytes in health and their modifications in disease conditions, focusing mainly on the hippocampus, a region involved in learning and memory encoding, in the age-related memory impairments, and in Alzheimer's disease (AD) dementia. The morphological and functional heterogeneity of astrocytes in different brain regions may be related to their different housekeeping functions. Astrocytes that express diverse transcriptomics and phenomics are present in strictly correlated brain regions and they are likely responsible for interactions essential for the formation of the specialized neural circuits that drive complex behaviors. In the contiguous and interconnected hippocampal areas CA1 and CA3, astrocytes show different, finely regulated, and region-specific heterogeneity. Heterogeneous astrocytes have specific activities in the healthy brain, and respond differently to physiological or pathological stimuli, such as inflammaging present in normal brain aging or beta-amyloid-dependent neuroinflammation typical of AD. To become reactive, astrocytes undergo transcriptional, functional, and morphological changes that transform them into cells with different properties and functions. Alterations of astrocytes affect the neurovascular unit, the blood-brain barrier and reverberate to other brain cell populations, favoring or dysregulating their activities. It will be of great interest to understand whether the differential phenomics of astrocytes in health and disease can explain the diverse vulnerability of the hippocampal areas to aging or to different damaging insults, in order to find new astrocyte-targeted therapies that might prevent or treat neurodegenerative disorders.
Collapse
Affiliation(s)
- Daniele Lana
- Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy
| | - Filippo Ugolini
- Section of Pathological Anatomy, Department of Health Sciences, University of Florence, Florence, Italy
| | - Ludovica Iovino
- Institute of Neuroscience, National Research Council (CNR), Pisa, Italy
| | - Selene Attorre
- Section of Pathological Anatomy, Department of Health Sciences, University of Florence, Florence, Italy
| | - Maria Grazia Giovannini
- Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy
| |
Collapse
|
14
|
Mohan M, Mannan A, Kakkar C, Singh TG. Nrf2 and Ferroptosis: Exploring Translational Avenues for Therapeutic Approaches to Neurological Diseases. Curr Drug Targets 2025; 26:33-58. [PMID: 39350404 DOI: 10.2174/0113894501320839240918110656] [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: 05/09/2024] [Revised: 08/28/2024] [Accepted: 09/02/2024] [Indexed: 02/19/2025]
Abstract
Nrf2, a crucial protein involved in defense mechanisms, particularly oxidative stress, plays a significant role in neurological diseases (NDs) by reducing oxidative stress and inflammation. NDs, including Alzheimer's, Parkinson's, Huntington's, amyotrophic lateral sclerosis, stroke, epilepsy, schizophrenia, depression, and autism, exhibit ferroptosis, iron-dependent regulated cell death resulting from lipid and iron-dependent reactive oxygen species (ROS) accumulation. Nrf2 has been shown to play a critical role in regulating ferroptosis in NDs. Age-related decline in Nrf2 expression and its target genes (HO-1, Nqo-1, and Trx) coincides with increased iron-mediated cell death, leading to ND onset. The modulation of iron-dependent cell death and ferroptosis by Nrf2 through various cellular and molecular mechanisms offers a potential therapeutic pathway for understanding the pathological processes underlying these NDs. This review emphasizes the mechanistic role of Nrf2 and ferroptosis in multiple NDs, providing valuable insights for future research and therapeutic approaches.
Collapse
Affiliation(s)
- Maneesh Mohan
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Chirag Kakkar
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | | |
Collapse
|
15
|
Bogdańska-Chomczyk E, Wojtacha P, Tsai ML, Huang ACW, Kozłowska A. Alterations in Striatal Architecture and Biochemical Markers' Levels During Postnatal Development in the Rat Model of an Attention Deficit/Hyperactivity Disorder (ADHD). Int J Mol Sci 2024; 25:13652. [PMID: 39769412 PMCID: PMC11680085 DOI: 10.3390/ijms252413652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/04/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
Attention deficit/hyperactivity disorder (ADHD) is defined as a neurodevelopmental condition. The precise underlying mechanisms remain incompletely elucidated. A body of research suggests disruptions in both the cellular architecture and neuronal function within the brain regions of individuals with ADHD, coupled with disturbances in the biochemical parameters. This study seeks to evaluate the morphological characteristics with a volume measurement of the striatal regions and a neuron density assessment within the studied areas across different developmental stages in Spontaneously Hypertensive Rats (SHRs) and Wistar Kyoto Rats (WKYs). Furthermore, the investigation aims to scrutinize the levels and activities of specific markers related to immune function, oxidative stress, and metabolism within the striatum of juvenile and maturing SHRs compared to WKYs. The findings reveal that the most pronounced reductions in striatal volume occur during the juvenile stage in SHRs, alongside alterations in neuronal density within these brain regions compared to WKYs. Additionally, SHRs exhibit heightened levels and activities of various markers, including RAC-alpha serine/threonine-protein kinase (AKT-1), glucocorticoid receptor (GCsRβ), malondialdehyde (MDA), sulfhydryl groups (-SH), glucose (G), iron (Fe), lactate dehydrogenase (LDH). alanine transaminase (ALT), and aspartate transaminase (AST). In summary, notable changes in striatal morphology and elevated levels of inflammatory, oxidative, and metabolic markers within the striatum may be linked to the disrupted brain development and maturation observed in ADHD.
Collapse
Affiliation(s)
- Ewelina Bogdańska-Chomczyk
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, Warszawska 30, 10-082 Olsztyn, Poland;
| | - Paweł Wojtacha
- Department of Psychology and Sociology of Health and Public Health, University of Warmia and Mazury, Warszawska 30, 10-082 Olsztyn, Poland;
| | - Meng-Li Tsai
- Department of Biomechatronic Engineering, National Ilan University, Ylan 26047, Taiwan;
| | | | - Anna Kozłowska
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, Warszawska 30, 10-082 Olsztyn, Poland;
| |
Collapse
|
16
|
Yadav S, Graham A, Al Hammood F, Garbark C, Vasudevan D, Pandey U, Asara JM, Rajasundaram D, Parkhitko AA. Unique tau- and synuclein-dependent metabolic reprogramming in neurons distinct from normal aging. Aging Cell 2024; 23:e14277. [PMID: 39137949 PMCID: PMC11561663 DOI: 10.1111/acel.14277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 06/25/2024] [Accepted: 06/27/2024] [Indexed: 08/15/2024] Open
Abstract
Neuronal cells are highly specialized cells and have a specific metabolic profile to support their function. It has been demonstrated that the metabolic profiles of different cells/tissues undergo significant reprogramming with advancing age, which has often been considered a contributing factor towards aging-related diseases including Alzheimer's (AD) and Parkinson's (PD) diseases. However, it is unclear if the metabolic changes associated with normal aging predispose neurons to disease conditions or a distinct set of metabolic alterations happen in neurons in AD or PD which might contribute to disease pathologies. To decipher the changes in neuronal metabolism with age, in AD, or in PD, we performed high-throughput steady-state metabolite profiling on heads in wildtype Drosophila and in Drosophila models relevant to AD and PD. Intriguingly, we found that the spectrum of affected metabolic pathways is dramatically different between normal aging, Tau, or Synuclein overexpressing neurons. Genetic targeting of the purine and glutamate metabolism pathways, which were dysregulated in both old age and disease conditions partially rescued the neurodegenerative phenotype associated with the overexpression of wildtype and mutant tau. Our findings support a "two-hit model" to explain the pathological manifestations associated with AD where both aging- and Tau/Synuclein- driven metabolic reprogramming events cooperate with each other, and targeting both could be a potent therapeutic strategy.
Collapse
Affiliation(s)
- Shweta Yadav
- Aging Institute of UPMC and the University of PittsburghPittsburghPennsylvaniaUSA
| | - Aidan Graham
- Aging Institute of UPMC and the University of PittsburghPittsburghPennsylvaniaUSA
| | - Farazdaq Al Hammood
- Aging Institute of UPMC and the University of PittsburghPittsburghPennsylvaniaUSA
| | - Chris Garbark
- Department of Cell BiologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Deepika Vasudevan
- Department of Cell BiologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Udai Pandey
- Department of Pediatrics, Children's Hospital of PittsburghUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - John M. Asara
- Division of Signal Transduction, Beth Israel Deaconess Medical Center, and Department of MedicineHarvard Medical SchoolBostonMassachusettsUSA
| | - Dhivyaa Rajasundaram
- Department of Pediatrics, Children's Hospital of PittsburghUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Andrey A. Parkhitko
- Aging Institute of UPMC and the University of PittsburghPittsburghPennsylvaniaUSA
| |
Collapse
|
17
|
Wang MY, Zhou Y, Li WL, Zhu LQ, Liu D. Friend or foe: Lactate in neurodegenerative diseases. Ageing Res Rev 2024; 101:102452. [PMID: 39127445 DOI: 10.1016/j.arr.2024.102452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 08/07/2024] [Indexed: 08/12/2024]
Abstract
Lactate, a byproduct of glycolysis, was considered as a metabolic waste until identified by studies on the Warburg effect. Increasing evidence elucidates that lactate functions as energy fuel, signaling molecule, and donor for protein lactylation. Altered lactate utilization is a common metabolic feature of the onset and progression of neurodegenerative diseases, such as Alzheimer's disease, multiple sclerosis, amyotrophic lateral sclerosis, Parkinson's disease and Huntington's disease. This review offers an overview of lactate metabolism from the perspective of production, transportation and clearance, and the role of lactate in neurodegenerative progression, as well as a summary of protein lactylation and the signaling function of lactate in neurodegenerative diseases. Besides, this review delves into the dual roles of changed lactate metabolism during neurodegeneration and explores prospective therapeutic methods targeting lactate. We propose that elucidating the correlation between lactate and neurodegeneration is pivotal for exploring innovative therapeutic interventions for neurodegenerative diseases.
Collapse
Affiliation(s)
- Ming-Yu Wang
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yang Zhou
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Wen-Lian Li
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ling-Qiang Zhu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Dan Liu
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
18
|
Shichkova P, Coggan JS, Markram H, Keller D. Brain Metabolism in Health and Neurodegeneration: The Interplay Among Neurons and Astrocytes. Cells 2024; 13:1714. [PMID: 39451233 PMCID: PMC11506225 DOI: 10.3390/cells13201714] [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: 08/01/2024] [Revised: 08/31/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024] Open
Abstract
The regulation of energy in the brain has garnered substantial attention in recent years due to its significant implications in various disorders and aging. The brain's energy metabolism is a dynamic and tightly regulated network that balances energy demand and supply by engaging complementary molecular pathways. The crosstalk among these pathways enables the system to switch its preferred fuel source based on substrate availability, activity levels, and cell state-related factors such as redox balance. Brain energy production relies on multi-cellular cooperation and is continuously supplied by fuel from the blood due to limited internal energy stores. Astrocytes, which interface with neurons and blood vessels, play a crucial role in coordinating the brain's metabolic activity, and their dysfunction can have detrimental effects on brain health. This review characterizes the major energy substrates (glucose, lactate, glycogen, ketones and lipids) in astrocyte metabolism and their role in brain health, focusing on recent developments in the field.
Collapse
Affiliation(s)
- Polina Shichkova
- Blue Brain Project, École Polytechnique Fédérale de Lausanne, 1202 Geneva, Switzerland
| | - Jay S. Coggan
- Blue Brain Project, École Polytechnique Fédérale de Lausanne, 1202 Geneva, Switzerland
| | - Henry Markram
- Blue Brain Project, École Polytechnique Fédérale de Lausanne, 1202 Geneva, Switzerland
- Laboratory of Neural Microcircuitry, Brain Mind Institute, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Daniel Keller
- Blue Brain Project, École Polytechnique Fédérale de Lausanne, 1202 Geneva, Switzerland
| |
Collapse
|
19
|
De Stefano S, Tiberi M, Salvatori I, De Bardi M, Gimenez J, Pirshayan M, Greco V, Borsellino G, Ferri A, Valle C, Mercuri NB, Chiurchiù V, Spalloni A, Longone P. Hydrogen Sulfide Modulates Astrocytic Toxicity in Mouse Spinal Cord Cultures: Implications for Amyotrophic Lateral Sclerosis. Antioxidants (Basel) 2024; 13:1241. [PMID: 39456494 PMCID: PMC11504967 DOI: 10.3390/antiox13101241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/07/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
Hydrogen sulfide (H2S), a known inhibitor of the electron transport chain, is endogenously produced in the periphery as well as in the central nervous system, where is mainly generated by glial cells. It affects, as a cellular signaling molecule, many different biochemical processes. In the central nervous system, depending on its concentration, it can be protective or damaging to neurons. In the study, we have demonstrated, in a primary mouse spinal cord cultures, that it is particularly harmful to motor neurons, is produced by glial cells, and is stimulated by inflammation. However, its role on glial cells, especially astrocytes, is still under-investigated. The present study was designed to evaluate the impact of H2S on astrocytes and their phenotypic heterogeneity, together with the functionality and homeostasis of mitochondria in primary spinal cord cultures. We found that H2S modulates astrocytes' morphological changes and their phenotypic transformation, exerts toxic properties by decreasing ATP production and the mitochondrial respiration rate, disturbs mitochondrial depolarization, and alters the energetic metabolism. These results further support the hypothesis that H2S is a toxic mediator, mainly released by astrocytes, possibly acting as an autocrine factor toward astrocytes, and probably involved in the non-cell autonomous mechanisms leading to motor neuron death.
Collapse
Affiliation(s)
- Susanna De Stefano
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (S.D.S.); (M.T.); (N.B.M.)
- Laboratory of Molecular Neurobiology, IRCCS Santa Lucia Foundation, 00143 Rome, Italy; (J.G.); (M.P.); (A.S.)
| | - Marta Tiberi
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (S.D.S.); (M.T.); (N.B.M.)
- Laboratory of Resolution of Neuroinflammation, IRCCS Santa Lucia Foundation, 00143 Rome, Italy;
| | - Illari Salvatori
- Laboratory of Neurochemistry, IRCCS Santa Lucia Foundation, 00143 Rome, Italy; (I.S.); (A.F.); (C.V.)
| | - Marco De Bardi
- Neuroimmunology Unit, IRCCS Santa Lucia Foundation, 00143 Rome, Italy; (M.D.B.); (G.B.)
| | - Juliette Gimenez
- Laboratory of Molecular Neurobiology, IRCCS Santa Lucia Foundation, 00143 Rome, Italy; (J.G.); (M.P.); (A.S.)
| | - Mahsa Pirshayan
- Laboratory of Molecular Neurobiology, IRCCS Santa Lucia Foundation, 00143 Rome, Italy; (J.G.); (M.P.); (A.S.)
| | - Viviana Greco
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy;
- Institute of Biochemistry and Clinical Biochemistry, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Giovanna Borsellino
- Neuroimmunology Unit, IRCCS Santa Lucia Foundation, 00143 Rome, Italy; (M.D.B.); (G.B.)
| | - Alberto Ferri
- Laboratory of Neurochemistry, IRCCS Santa Lucia Foundation, 00143 Rome, Italy; (I.S.); (A.F.); (C.V.)
- Institute of Translational Pharmacology (IFT), National Research Council (CNR), 00133 Rome, Italy
| | - Cristiana Valle
- Laboratory of Neurochemistry, IRCCS Santa Lucia Foundation, 00143 Rome, Italy; (I.S.); (A.F.); (C.V.)
- Institute of Translational Pharmacology (IFT), National Research Council (CNR), 00133 Rome, Italy
| | - Nicola B. Mercuri
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (S.D.S.); (M.T.); (N.B.M.)
- Laboratory of Experimental Neurology, Santa Lucia Foundation IRCCS, 00143 Rome, Italy
| | - Valerio Chiurchiù
- Laboratory of Resolution of Neuroinflammation, IRCCS Santa Lucia Foundation, 00143 Rome, Italy;
- Institute of Translational Pharmacology (IFT), National Research Council (CNR), 00133 Rome, Italy
| | - Alida Spalloni
- Laboratory of Molecular Neurobiology, IRCCS Santa Lucia Foundation, 00143 Rome, Italy; (J.G.); (M.P.); (A.S.)
| | - Patrizia Longone
- Laboratory of Molecular Neurobiology, IRCCS Santa Lucia Foundation, 00143 Rome, Italy; (J.G.); (M.P.); (A.S.)
| |
Collapse
|
20
|
Li X, Chen M, Chen X, He X, Li X, Wei H, Tan Y, Min J, Azam T, Xue M, Zhang Y, Dong M, Yin Q, Zheng L, Jiang H, Huo D, Wang X, Chen S, Ji Y, Chen H. TRAP1 drives smooth muscle cell senescence and promotes atherosclerosis via HDAC3-primed histone H4 lysine 12 lactylation. Eur Heart J 2024; 45:4219-4235. [PMID: 39088352 PMCID: PMC11481199 DOI: 10.1093/eurheartj/ehae379] [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: 08/11/2023] [Revised: 12/12/2023] [Accepted: 06/03/2024] [Indexed: 08/03/2024] Open
Abstract
BACKGROUND AND AIMS Vascular smooth muscle cell (VSMC) senescence is crucial for the development of atherosclerosis, characterized by metabolic abnormalities. Tumour necrosis factor receptor-associated protein 1 (TRAP1), a metabolic regulator associated with ageing, might be implicated in atherosclerosis. As the role of TRAP1 in atherosclerosis remains elusive, this study aimed to examine the function of TRAP1 in VSMC senescence and atherosclerosis. METHODS TRAP1 expression was measured in the aortic tissues of patients and mice with atherosclerosis using western blot and RT-qPCR. Senescent VSMC models were established by oncogenic Ras, and cellular senescence was evaluated by measuring senescence-associated β-galactosidase expression and other senescence markers. Chromatin immunoprecipitation (ChIP) analysis was performed to explore the potential role of TRAP1 in atherosclerosis. RESULTS VSMC-specific TRAP1 deficiency mitigated VSMC senescence and atherosclerosis via metabolic reprogramming. Mechanistically, TRAP1 significantly increased aerobic glycolysis, leading to elevated lactate production. Accumulated lactate promoted histone H4 lysine 12 lactylation (H4K12la) by down-regulating the unique histone lysine delactylase HDAC3. H4K12la was enriched in the senescence-associated secretory phenotype (SASP) promoter, activating SASP transcription and exacerbating VSMC senescence. In VSMC-specific Trap1 knockout ApoeKO mice (ApoeKOTrap1SMCKO), the plaque area, senescence markers, H4K12la, and SASP were reduced. Additionally, pharmacological inhibition and proteolysis-targeting chimera (PROTAC)-mediated TRAP1 degradation effectively attenuated atherosclerosis in vivo. CONCLUSIONS This study reveals a novel mechanism by which mitonuclear communication orchestrates gene expression in VSMC senescence and atherosclerosis. TRAP1-mediated metabolic reprogramming increases lactate-dependent H4K12la via HDAC3, promoting SASP expression and offering a new therapeutic direction for VSMC senescence and atherosclerosis.
Collapse
Affiliation(s)
- Xuesong Li
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Minghong Chen
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Xiang Chen
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Xian He
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Xinyu Li
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Huiyuan Wei
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yongkang Tan
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Jiao Min
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Tayyiba Azam
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Mengdie Xue
- Department of Medicinal Chemistry, Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yunjia Zhang
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Mengdie Dong
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Quanwen Yin
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Longbin Zheng
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Hong Jiang
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Da Huo
- Department of Medicinal Chemistry, Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Xin Wang
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Shaoliang Chen
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yong Ji
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, State Key Laboratory of Reproductive Medicine, School of Pharmacy, the Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, Nanjing, Jiangsu, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Key Laboratory of Cardiovascular Medicine Research and Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, NHC Key Laboratory of Cell Transplantation, the Central Laboratory of the First Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| | - Hongshan Chen
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, China
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|
21
|
Nusinovici S, Rim TH, Li H, Yu M, Deshmukh M, Quek TC, Lee G, Chong CCY, Peng Q, Xue CC, Zhu Z, Chew EY, Sabanayagam C, Wong TY, Tham YC, Cheng CY. Application of a deep-learning marker for morbidity and mortality prediction derived from retinal photographs: a cohort development and validation study. THE LANCET. HEALTHY LONGEVITY 2024; 5:100593. [PMID: 39362226 PMCID: PMC11788570 DOI: 10.1016/s2666-7568(24)00089-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 05/03/2024] [Accepted: 05/03/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND Biological ageing markers are useful to risk stratify morbidity and mortality more precisely than chronological age. In this study, we aimed to develop a novel deep-learning-based biological ageing marker (referred to as RetiPhenoAge hereafter) using retinal images and PhenoAge, a composite biomarker of phenotypic age. METHODS We used retinal photographs from the UK Biobank dataset to train a deep-learning algorithm to predict the composite score of PhenoAge. We used a deep convolutional neural network architecture with multiple layers to develop our deep-learning-based biological ageing marker, as RetiPhenoAge, with the aim of identifying patterns and features in the retina associated with variations of blood biomarkers related to renal, immune, liver functions, inflammation, and energy metabolism, and chronological age. We determined the performance of this biological ageing marker for the prediction of morbidity (cardiovascular disease and cancer events) and mortality (all-cause, cardiovascular disease, and cancer) in three independent cohorts (UK Biobank, the Singapore Epidemiology of Eye Diseases [SEED], and the Age-Related Eye Disease Study [AREDS] from the USA). We also compared the performance of RetiPhenoAge with two other known ageing biomarkers (hand grip strength and adjusted leukocyte telomere length) and one lifestyle factor (physical activity) for risk stratification of mortality and morbidity. We explored the underlying biology of RetiPhenoAge by assessing its associations with different systemic characteristics (eg, diabetes or hypertension) and blood metabolite levels. We also did a genome-wide association study to identify genetic variants associated with RetiPhenoAge, followed by expression quantitative trait loci mapping, a gene-based analysis, and a gene-set analysis. Cox proportional hazards models were used to estimate the hazard ratios (HRs) and corresponding 95% CIs for the associations between RetiPhenoAge and the different morbidity and mortality outcomes. FINDINGS Retinal photographs for 34 061 UK Biobank participants were used to train the model, and data for 9429 participants from the SEED cohort and for 3986 participants from the AREDS cohort were included in the study. RetiPhenoAge was associated with all-cause mortality (HR 1·92 [95% CI 1·42-2·61]), cardiovascular disease mortality (1·97 [1·02-3·82]), cancer mortality (2·07 [1·29-3·33]), and cardiovascular disease events (1·70 [1·17-2·47]), independent of PhenoAge and other possible confounders. Similar findings were found in the two independent cohorts (HR 1·67 [1·21-2·31] for cardiovascular disease mortality in SEED and 2·07 [1·10-3·92] in AREDS). RetiPhenoAge had stronger associations with mortality and morbidity than did hand grip strength, telomere length, and physical activity. We identified two genetic variants that were significantly associated with RetiPhenoAge (single nucleotide polymorphisms rs3791224 and rs8001273), and were linked to expression quantitative trait locis in various tissues, including the heart, kidneys, and the brain. INTERPRETATION Our new deep-learning-derived biological ageing marker is a robust predictor of mortality and morbidity outcomes and could be used as a novel non-invasive method to measure ageing. FUNDING Singapore National Medical Research Council and Agency for Science, Technology and Research, Singapore.
Collapse
Affiliation(s)
- Simon Nusinovici
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Ophthalmology and Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore
| | - Tyler Hyungtaek Rim
- Ophthalmology and Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore; MediWhale, Seoul, South Korea
| | - Hengtong Li
- Department of Ophthalmology and Centre for Innovation and Precision Eye Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Marco Yu
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Ophthalmology and Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore
| | - Mihir Deshmukh
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Ten Cheer Quek
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | | | | | - Qingsheng Peng
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Ophthalmology and Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore
| | - Can Can Xue
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Zhuoting Zhu
- Department of Ophthalmology, Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China; Centre for Eye Research Australia, Ophthalmology, University of Melbourne, Melbourne, VIC, Australia
| | - Emily Y Chew
- National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Charumathi Sabanayagam
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Ophthalmology and Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore
| | - Tien-Yin Wong
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Tsinghua Medicine, Tsinghua University, Beijing, China
| | - Yih-Chung Tham
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Ophthalmology and Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore; Department of Ophthalmology and Centre for Innovation and Precision Eye Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Ching-Yu Cheng
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Ophthalmology and Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore; Department of Ophthalmology and Centre for Innovation and Precision Eye Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
22
|
Wang X, Zhang J, Xu X, Pan S, Cheng L, Dang K, Qi X, Li Y. Associations of daily eating frequency and nighttime fasting duration with biological aging in National Health and Nutrition Examination Survey (NHANES) 2003-2010 and 2015-2018. Int J Behav Nutr Phys Act 2024; 21:104. [PMID: 39300516 DOI: 10.1186/s12966-024-01654-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 09/11/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Information on the influences of daily eating frequency (DEF) and nighttime fasting duration (NFD) on biological aging is minimal. Our study investigated the potential associations of DEF and NFD with accelerated aging. METHODS Out of 24212 participants in NHANES 2003-2010 and 2015-2018, 4 predicted age metrics [homeostatic dysregulation (HD), Klemera-Doubal method (KDM), phenoAge (PA), and allostatic load (AL)] were computed based on 12 blood chemistry parameters. Utilizing 24-h dietary recall, DEF was measured by the frequency of eating occurrences, while NFD was determined by assessing the timing of the initial and final meals throughout the day. Weighted multivariate linear regression models and restricted cubic spline (RCS) were utilized to examine the associations. RESULTS Compared to DEF of ≤ 3.0 times, subjects with DEF ≥ 4.6 times demonstrated lower KDM residual [β: -0.57, 95% confidence-interval (CI): (-0.97, -0.17)] and PA residual [β: -0.47, 95% CI: (-0.69, -0.25)]. In comparison to NFD between 10.1 and 12.0 h, individuals with NFD ≤ 10.0 h were at higher HD [β: 0.03, 95% CI: (0.01, 0.04)], KDM residual [β: 0.34, 95% CI: (0.05, 0.63)], and PA residual [β: 0.38, 95% CI: (0.18, 0.57)]. Likewise, those with NFD ≥ 14.1 h also had higher HD [β: 0.02, 95% CI: (0.01, 0.04)] and KDM residual [β: 0.33, 95% CI: (0.03, 0.62)]. The results were confirmed by the dose-response relationships of DEF and NFD with predicted age metrics. Lactate dehydrogenase (LDH) and globulin (Glo) were acknowledged as implicated in and mediating the relationships. CONCLUSIONS DEF below 3.0 times and NFD less than 10.0 or more than 14.1 h were independently associated with higher predicted age metrics.
Collapse
Affiliation(s)
- Xuanyang Wang
- Department of Nutrition and Food Hygiene, School of Public Health, the National Key Discipline, Harbin Medical University, 157 Baojian Road, Harbin, 150081, P. R. China
| | - Jia Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, the National Key Discipline, Harbin Medical University, 157 Baojian Road, Harbin, 150081, P. R. China
| | - Xiaoqing Xu
- Department of Nutrition and Food Hygiene, School of Public Health, the National Key Discipline, Harbin Medical University, 157 Baojian Road, Harbin, 150081, P. R. China
| | - Sijia Pan
- Department of Nutrition and Food Hygiene, School of Public Health, the National Key Discipline, Harbin Medical University, 157 Baojian Road, Harbin, 150081, P. R. China
| | - Licheng Cheng
- Department of Nutrition and Food Hygiene, School of Public Health, the National Key Discipline, Harbin Medical University, 157 Baojian Road, Harbin, 150081, P. R. China
| | - Keke Dang
- Department of Nutrition and Food Hygiene, School of Public Health, the National Key Discipline, Harbin Medical University, 157 Baojian Road, Harbin, 150081, P. R. China
| | - Xiang Qi
- Department of Nutrition and Food Hygiene, School of Public Health, the National Key Discipline, Harbin Medical University, 157 Baojian Road, Harbin, 150081, P. R. China
| | - Ying Li
- Department of Nutrition and Food Hygiene, School of Public Health, the National Key Discipline, Harbin Medical University, 157 Baojian Road, Harbin, 150081, P. R. China.
| |
Collapse
|
23
|
Dong H, Hu F, Hao B, Jin X, Zheng Q, Su Y. Single-cell analysis reveals the disparities in immune profiles between younger and elder patients. Eur Geriatr Med 2024:10.1007/s41999-024-01032-8. [PMID: 39244673 DOI: 10.1007/s41999-024-01032-8] [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: 11/01/2023] [Accepted: 07/26/2024] [Indexed: 09/10/2024]
Abstract
PURPOSE The immune profiles of elder patients with non-small cell lung cancer (NSCLC) differ significantly from those of younger patients. The tumor microenvironment (TME) is a crucial factor in cancer progression and therapeutic responses. The present study aims to decipher the difference in TME between younger and elderly patients with lung cancers. METHODS We downloaded single-cell RNA data from public databases. The algorithm of uniform manifold approximation and projection (UMAP) was applied to cluster and visualize single-cell sequencing data. Gene set variation analysis (GSVA) and gene set enrichment analysis (GSEA) analysis were performed to evaluate the physiological functional characteristics in sub-group cells. CellPhoneDB was used to identify cell-cell interactions between immune cells within TME. RESULTS We conducted single-cell RNA sequencing on 96,491 cells from elderly patients and 169,207 cells from younger patients, respectively. We observed that epithelial cells were the predominant component of the TME in younger patients, whereas T/NK cells were the predominant cell type in the TME of elderly patients. We also found that there was a higher proportion of Tregs and a lower proportion of NK, effector CD8+T and γδT cells in elder patients compared with younger patients. In addition, a comparative GSEA analysis of NK cells between older and younger patients revealed that the pathways of Parkinson's disease, Alzheimer's disease, mismatch repair, and base excision repair were up-regulated in NK cells from elderly patients, while the pathways related to natural killer cell-mediated cytotoxicity and allograft rejection were downregulated. Furthermore, we identified tumor-associated neutrophils (TANs) in elder patients, and GSVA analysis demonstrated that the pathway of angiogenesis was upregulated, and the pathway of interferon_γ_response, inflammatory_response, TNFα_signaling_via_NFκB pathways were downregulated. Importantly, the pro-inflammatory response scores of complement C1q C chain positive (C1QC+) macrophages, tissue-resident macrophages (TRM), non-classical monocytes (NCM), secreted phosphoprotein 1 positive (SPP1+) macrophages, and classical monocytes (CM) in elder patients were significantly lower compared to those in younger patients. Finally, cell-to-cell communication analyses unveiled the disparities in regulatory patterns between elder and younger patients, namely the pairs of CXCL13-ACKR4 and CSF1-SIRPA in elder patients and the pairs of CTLA4-CD86 and TIGIT-NECTIN2 in younger patients. CONCLUSION This study reveals the distinct immune profiles between younger and elder NSCLC patients, and the elder patients were likely to exhibit a more immunosuppressive TME and attenuated tumor-killing capability compared with younger patients.
Collapse
Affiliation(s)
- Huixing Dong
- Department of Pulmonary and Critical Care Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 XianXia Road, Shanghai, 200336, China
| | - Feng Hu
- Department of Pulmonary and Critical Care Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 XianXia Road, Shanghai, 200336, China
| | - Bo Hao
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Xiaoyan Jin
- Department of Pulmonary and Critical Care Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 XianXia Road, Shanghai, 200336, China
| | - Qi Zheng
- Department of Respiratory and Critical Care Medicine, Huangpu Branch, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No.58, Pu Yu Dong Road, Shanghai, 200011, China.
| | - Yiliang Su
- Department of Pulmonary and Critical Care Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 XianXia Road, Shanghai, 200336, China.
| |
Collapse
|
24
|
Bagheri M, Habibzadeh S, Moeini M. Transient Changes in Cerebral Tissue Oxygen, Glucose, and Temperature by Microstrokes: A Computational Study. Microcirculation 2024; 31:e12872. [PMID: 38944839 DOI: 10.1111/micc.12872] [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: 02/17/2024] [Revised: 05/09/2024] [Accepted: 06/08/2024] [Indexed: 07/02/2024]
Abstract
OBJECTIVE This study focuses on evaluating the disruptions in key physiological parameters during microstroke events to assess their severity. METHODS A mathematical model was developed to simulate the changes in cerebral tissue pO2, glucose concentration, and temperature due to blood flow interruptions. The model considers variations in baseline cerebral blood flow (CBF), capillary density, and blood oxygen/glucose levels, as well as ambient temperature changes. RESULTS Simulations indicate that complete blood flow obstruction still allows for limited glucose availability, supporting nonoxidative metabolism and potentially exacerbating lactate buildup and acidosis. Partial obstructions decrease tissue pO2, with minimal impact on glucose level, which can remain almost unchanged or even slightly increase. Reduced CBF, capillary density, or blood oxygen due to aging or disease enhances hypoxia risk at lower obstruction levels, with capillary density having a significant effect on stroke severity by influencing both pO2 and glucose levels. Conditions could lead to co-occurrence of hypoxia/hypoglycemia or hypoxia/hyperglycemia, each worsening outcomes. Temperature effects were minimal in deep brain regions but varied near the skull by 0.2-0.8°C depending on ambient temperature. CONCLUSIONS The model provides insights into the conditions driving severe stroke outcomes based on estimated levels of hypoxia, hypoglycemia, hyperglycemia, and temperature changes.
Collapse
Affiliation(s)
- Marzieh Bagheri
- Department of Chemical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Sajjad Habibzadeh
- Department of Chemical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Mohammad Moeini
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| |
Collapse
|
25
|
Gomez-Pinilla F, Thapak P. Exercise epigenetics is fueled by cell bioenergetics: Supporting role on brain plasticity and cognition. Free Radic Biol Med 2024; 220:43-55. [PMID: 38677488 PMCID: PMC11144461 DOI: 10.1016/j.freeradbiomed.2024.04.237] [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: 02/29/2024] [Revised: 04/04/2024] [Accepted: 04/24/2024] [Indexed: 04/29/2024]
Abstract
Exercise has the unique aptitude to benefit overall health of body and brain. Evidence indicates that the effects of exercise can be saved in the epigenome for considerable time to elevate the threshold for various diseases. The action of exercise on epigenetic regulation seems central to building an "epigenetic memory" to influence long-term brain function and behavior. As an intrinsic bioenergetic process, exercise engages the function of the mitochondria and redox pathways to impinge upon molecular mechanisms that regulate synaptic plasticity and learning and memory. We discuss how the action of exercise uses mechanisms of bioenergetics to support a "epigenetic memory" with long-term implications for neural and behavioral plasticity. This information is crucial for directing the power of exercise to reduce the burden of neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Fernando Gomez-Pinilla
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA; Department of Neurosurgery, UCLA Brain Injury Research Center, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| | - Pavan Thapak
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| |
Collapse
|
26
|
Späte E, Zhou B, Sun T, Kusch K, Asadollahi E, Siems SB, Depp C, Werner HB, Saher G, Hirrlinger J, Möbius W, Nave KA, Goebbels S. Downregulated expression of lactate dehydrogenase in adult oligodendrocytes and its implication for the transfer of glycolysis products to axons. Glia 2024; 72:1374-1391. [PMID: 38587131 DOI: 10.1002/glia.24533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 03/25/2024] [Accepted: 03/26/2024] [Indexed: 04/09/2024]
Abstract
Oligodendrocytes and astrocytes are metabolically coupled to neuronal compartments. Pyruvate and lactate can shuttle between glial cells and axons via monocarboxylate transporters. However, lactate can only be synthesized or used in metabolic reactions with the help of lactate dehydrogenase (LDH), a tetramer of LDHA and LDHB subunits in varying compositions. Here we show that mice with a cell type-specific disruption of both Ldha and Ldhb genes in oligodendrocytes lack a pathological phenotype that would be indicative of oligodendroglial dysfunctions or lack of axonal metabolic support. Indeed, when combining immunohistochemical, electron microscopical, and in situ hybridization analyses in adult mice, we found that the vast majority of mature oligodendrocytes lack detectable expression of LDH. Even in neurodegenerative disease models and in mice under metabolic stress LDH was not increased. In contrast, at early development and in the remyelinating brain, LDHA was readily detectable in immature oligodendrocytes. Interestingly, by immunoelectron microscopy LDHA was particularly enriched at gap junctions formed between adjacent astrocytes and at junctions between astrocytes and oligodendrocytes. Our data suggest that oligodendrocytes metabolize lactate during development and remyelination. In contrast, for metabolic support of axons mature oligodendrocytes may export their own glycolysis products as pyruvate rather than lactate. Lacking LDH, these oligodendrocytes can also "funnel" lactate through their "myelinic" channels between gap junction-coupled astrocytes and axons without metabolizing it. We suggest a working model, in which the unequal cellular distribution of LDH in white matter tracts facilitates a rapid and efficient transport of glycolysis products among glial and axonal compartments.
Collapse
Affiliation(s)
- Erik Späte
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Baoyu Zhou
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Ting Sun
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Laboratory of Molecular Neurobiology, Department of Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Kathrin Kusch
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
| | - Ebrahim Asadollahi
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Sophie B Siems
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Constanze Depp
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Hauke B Werner
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Gesine Saher
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Johannes Hirrlinger
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Carl-Ludwig-Institute for Physiology, Faculty of Medicine, Leipzig University, Leipzig, Germany
| | - Wiebke Möbius
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Sandra Goebbels
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| |
Collapse
|
27
|
Yakovlev AV, Detterer AS, Yakovleva OV, Hermann A, Sitdikova GF. H 2S prevents the disruption of the blood-brain barrier in rats with prenatal hyperhomocysteinemia. J Pharmacol Sci 2024; 155:131-139. [PMID: 38880547 DOI: 10.1016/j.jphs.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/16/2024] [Accepted: 05/18/2024] [Indexed: 06/18/2024] Open
Abstract
Elevation of the homocysteine concentration in the plasma called hyperhomocysteinemia (hHCY) during pregnancy causes a number of pre- and postnatal developmental disorders. The aim of our study was to analyze the effects of H2S donors -NaHS and N-acetylcysteine (NAC) on blood-brain barrier (BBB) permeability in rats with prenatal hHCY. In rats with mild hHCY BBB permeability assessed by Evans Blue extravasation in brain increased markedly throughout life. Administration of NaHS or NAC during pregnancy attenuated hHCY-associated damage and increased endogenous concentrations of sulfides in brain tissues. Acute application of dl-homocysteine thiolactone induced BBB leakage, which was prevented by the NMDA receptor antagonist MK-801 or H2S donors. Rats with hHCY demonstrated high levels of NO metabolite - nitrites and proinflammatory cytokines (IL-1β, TNF-α, IL-6) in brain. Lactate dehydrogenase (LDH) activity in the serum was higher in rats with hHCY. Mitochondrial complex-I activity was lower in brain of hHCY rats. NaHS treatment during pregnancy restored levels of proinflammatory cytokines, nitrites and activity of the respiratory chain complex in brain as well as the LDH activity in serum. Our data suggest that H2S has neuroprotective effects against prenatal hHCY-associated BBB disturbance providing a potential strategy for the prevention of developmental impairments in newborns.
Collapse
Affiliation(s)
- A V Yakovlev
- Department of Human and Animal Physiology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kremlevskaya str 18, Kazan, 420008, Russia
| | - A S Detterer
- Department of Human and Animal Physiology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kremlevskaya str 18, Kazan, 420008, Russia
| | - O V Yakovleva
- Department of Human and Animal Physiology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kremlevskaya str 18, Kazan, 420008, Russia
| | - A Hermann
- Department of Cell Biology, Division of Cellular and Molecular Neurobiology, University of Salzburg, Department of Biosciences, Hellbrunnerstr. 34, Salzburg, 5020, Austria
| | - G F Sitdikova
- Department of Human and Animal Physiology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kremlevskaya str 18, Kazan, 420008, Russia.
| |
Collapse
|
28
|
Jamerson LE, Bradshaw PC. The Roles of White Adipose Tissue and Liver NADPH in Dietary Restriction-Induced Longevity. Antioxidants (Basel) 2024; 13:820. [PMID: 39061889 PMCID: PMC11273496 DOI: 10.3390/antiox13070820] [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: 05/29/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
Dietary restriction (DR) protocols frequently employ intermittent fasting. Following a period of fasting, meal consumption increases lipogenic gene expression, including that of NADPH-generating enzymes that fuel lipogenesis in white adipose tissue (WAT) through the induction of transcriptional regulators SREBP-1c and CHREBP. SREBP-1c knockout mice, unlike controls, did not show an extended lifespan on the DR diet. WAT cytoplasmic NADPH is generated by both malic enzyme 1 (ME1) and the pentose phosphate pathway (PPP), while liver cytoplasmic NADPH is primarily synthesized by folate cycle enzymes provided one-carbon units through serine catabolism. During the daily fasting period of the DR diet, fatty acids are released from WAT and are transported to peripheral tissues, where they are used for beta-oxidation and for phospholipid and lipid droplet synthesis, where monounsaturated fatty acids (MUFAs) may activate Nrf1 and inhibit ferroptosis to promote longevity. Decreased WAT NADPH from PPP gene knockout stimulated the browning of WAT and protected from a high-fat diet, while high levels of NADPH-generating enzymes in WAT and macrophages are linked to obesity. But oscillations in WAT [NADPH]/[NADP+] from feeding and fasting cycles may play an important role in maintaining metabolic plasticity to drive longevity. Studies measuring the WAT malate/pyruvate as a proxy for the cytoplasmic [NADPH]/[NADP+], as well as studies using fluorescent biosensors expressed in the WAT of animal models to monitor the changes in cytoplasmic [NADPH]/[NADP+], are needed during ad libitum and DR diets to determine the changes that are associated with longevity.
Collapse
Affiliation(s)
| | - Patrick C. Bradshaw
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| |
Collapse
|
29
|
Sun J, Jiang R, Hou L, Wang L, Li M, Dong H, Dong N, Lin Y, Zhu Z, Zhang G, Zhang Y. Identification of a combined hypoxia and lactate metabolism prognostic signature in lung adenocarcinoma. BMC Pulm Med 2024; 24:323. [PMID: 38965505 PMCID: PMC11225160 DOI: 10.1186/s12890-024-03132-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 06/25/2024] [Indexed: 07/06/2024] Open
Abstract
BACKGROUND In the tumor microenvironment (TME), a bidirectional relationship exists between hypoxia and lactate metabolism, with each component exerting a reciprocal influence on the other, forming an inextricable link. The aim of the present investigation was to develop a prognostic model by amalgamating genes associated with hypoxia and lactate metabolism. This model is intended to serve as a tool for predicting patient outcomes, including survival rates, the status of the immune microenvironment, and responsiveness to therapy in patients with lung adenocarcinoma (LUAD). METHODS Transcriptomic sequencing data and patient clinical information specific to LUAD were obtained from comprehensive repositories of The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO). A compendium of genes implicated in hypoxia and lactate metabolism was assembled from an array of accessible datasets. Univariate and multivariate Cox regression analyses were employed. Additional investigative procedures, including tumor mutational load (TMB), microsatellite instability (MSI), functional enrichment assessments and the ESTIMATE, CIBERSORT, and TIDE algorithms, were used to evaluate drug sensitivity and predict the efficacy of immune-based therapies. RESULTS A novel prognostic signature comprising five lactate and hypoxia-related genes (LHRGs), PKFP, SLC2A1, BCAN, CDKN3, and ANLN, was established. This model demonstrated that LUAD patients with elevated LHRG-related risk scores exhibited significantly reduced survival rates. Both univariate and multivariate Cox analyses confirmed that the risk score was a robust prognostic indicator of overall survival. Immunophenotyping revealed increased infiltration of memory CD4 + T cells, dendritic cells and NK cells in patients classified within the high-risk category compared to their low-risk counterparts. Higher probability of mutations in lung adenocarcinoma driver genes in high-risk groups, and the MSI was associated with the risk-score. Functional enrichment analyses indicated a predominance of cell cycle-related pathways in the high-risk group, whereas metabolic pathways were more prevalent in the low-risk group. Moreover, drug sensitivity analyses revealed increased sensitivity to a variety of drugs in the high-risk group, especially inhibitors of the PI3K-AKT, EGFR, and ELK pathways. CONCLUSIONS This prognostic model integrates lactate metabolism and hypoxia parameters, offering predictive insights regarding survival, immune cell infiltration and functionality, as well as therapeutic responsiveness in LUAD patients. This model may facilitate personalized treatment strategies, tailoring interventions to the unique molecular profile of each patient's disease.
Collapse
Affiliation(s)
- Jingyang Sun
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Enhanced Recovery After Surgery of Integrated Chinese and Western Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Biobank, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Rongxuan Jiang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Enhanced Recovery After Surgery of Integrated Chinese and Western Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Biobank, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Liren Hou
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Enhanced Recovery After Surgery of Integrated Chinese and Western Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Biobank, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Lei Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Enhanced Recovery After Surgery of Integrated Chinese and Western Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Biobank, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Meng Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China
| | - Huanhuan Dong
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Enhanced Recovery After Surgery of Integrated Chinese and Western Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Biobank, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Niuniu Dong
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Enhanced Recovery After Surgery of Integrated Chinese and Western Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Biobank, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Yihan Lin
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Enhanced Recovery After Surgery of Integrated Chinese and Western Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Biobank, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Zijiang Zhu
- Department of Thoracic Surgery, Gansu Province Central Hospital, Lanzhou, Gansu, 730070, China.
| | - Guangjian Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
- Key Laboratory of Enhanced Recovery After Surgery of Integrated Chinese and Western Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
- Biobank, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| | - Yanpeng Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
- Key Laboratory of Enhanced Recovery After Surgery of Integrated Chinese and Western Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
- Biobank, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| |
Collapse
|
30
|
Cai Y, Han Z, Cheng H, Li H, Wang K, Chen J, Liu ZX, Xie Y, Lin Y, Zhou S, Wang S, Zhou X, Jin S. The impact of ageing mechanisms on musculoskeletal system diseases in the elderly. Front Immunol 2024; 15:1405621. [PMID: 38774874 PMCID: PMC11106385 DOI: 10.3389/fimmu.2024.1405621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 04/22/2024] [Indexed: 05/24/2024] Open
Abstract
Ageing is an inevitable process that affects various tissues and organs of the human body, leading to a series of physiological and pathological changes. Mechanisms such as telomere depletion, stem cell depletion, macrophage dysfunction, and cellular senescence gradually manifest in the body, significantly increasing the incidence of diseases in elderly individuals. These mechanisms interact with each other, profoundly impacting the quality of life of older adults. As the ageing population continues to grow, the burden on the public health system is expected to intensify. Globally, the prevalence of musculoskeletal system diseases in elderly individuals is increasing, resulting in reduced limb mobility and prolonged suffering. This review aims to elucidate the mechanisms of ageing and their interplay while exploring their impact on diseases such as osteoarthritis, osteoporosis, and sarcopenia. By delving into the mechanisms of ageing, further research can be conducted to prevent and mitigate its effects, with the ultimate goal of alleviating the suffering of elderly patients in the future.
Collapse
Affiliation(s)
- Yijin Cai
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhongyu Han
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hong Cheng
- School of Automation Engineering, University of Electronic Science and Technology, Chengdu, China
| | - Hongpeng Li
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ke Wang
- Eye School of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jia Chen
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhi-Xiang Liu
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yulong Xie
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yumeng Lin
- Eye School of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shuwei Zhou
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Siyu Wang
- Department of Gastroenterology, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Xiao Zhou
- Second Clinical Medical College, Heilongjiang University of Chinese Medicine, Heilongjiang, China
| | - Song Jin
- Department of Rehabilitation, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
31
|
Filograna R, Gerlach J, Choi HN, Rigoni G, Barbaro M, Oscarson M, Lee S, Tiklova K, Ringnér M, Koolmeister C, Wibom R, Riggare S, Nennesmo I, Perlmann T, Wredenberg A, Wedell A, Motori E, Svenningsson P, Larsson NG. PARKIN is not required to sustain OXPHOS function in adult mammalian tissues. NPJ Parkinsons Dis 2024; 10:93. [PMID: 38684669 PMCID: PMC11058849 DOI: 10.1038/s41531-024-00707-0] [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: 09/12/2023] [Accepted: 04/11/2024] [Indexed: 05/02/2024] Open
Abstract
Loss-of-function variants in the PRKN gene encoding the ubiquitin E3 ligase PARKIN cause autosomal recessive early-onset Parkinson's disease (PD). Extensive in vitro and in vivo studies have reported that PARKIN is involved in multiple pathways of mitochondrial quality control, including mitochondrial degradation and biogenesis. However, these findings are surrounded by substantial controversy due to conflicting experimental data. In addition, the existing PARKIN-deficient mouse models have failed to faithfully recapitulate PD phenotypes. Therefore, we have investigated the mitochondrial role of PARKIN during ageing and in response to stress by employing a series of conditional Parkin knockout mice. We report that PARKIN loss does not affect oxidative phosphorylation (OXPHOS) capacity and mitochondrial DNA (mtDNA) levels in the brain, heart, and skeletal muscle of aged mice. We also demonstrate that PARKIN deficiency does not exacerbate the brain defects and the pro-inflammatory phenotype observed in mice carrying high levels of mtDNA mutations. To rule out compensatory mechanisms activated during embryonic development of Parkin-deficient mice, we generated a mouse model where loss of PARKIN was induced in adult dopaminergic (DA) neurons. Surprisingly, also these mice did not show motor impairment or neurodegeneration, and no major transcriptional changes were found in isolated midbrain DA neurons. Finally, we report a patient with compound heterozygous PRKN pathogenic variants that lacks PARKIN and has developed PD. The PARKIN deficiency did not impair OXPHOS activities or induce mitochondrial pathology in skeletal muscle from the patient. Altogether, our results argue that PARKIN is dispensable for OXPHOS function in adult mammalian tissues.
Collapse
Affiliation(s)
- Roberta Filograna
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden.
| | - Jule Gerlach
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Hae-Na Choi
- Institute for Biochemistry, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Giovanni Rigoni
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Michela Barbaro
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Mikael Oscarson
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Seungmin Lee
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Katarina Tiklova
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Markus Ringnér
- Department of Biology, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Lund University, Lund, Sweden
| | - Camilla Koolmeister
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Rolf Wibom
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Sara Riggare
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Inger Nennesmo
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Perlmann
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Anna Wredenberg
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Anna Wedell
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - Elisa Motori
- Institute for Biochemistry, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Per Svenningsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Nils-Göran Larsson
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
32
|
Delgado MG, Delgado R. Transient Synaptic Enhancement Triggered by Exogenously Supplied Monocarboxylate in Drosophila Motoneuron Synapse. Neuroscience 2024; 539:66-75. [PMID: 38220128 DOI: 10.1016/j.neuroscience.2024.01.003] [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: 05/26/2023] [Revised: 01/05/2024] [Accepted: 01/09/2024] [Indexed: 01/16/2024]
Abstract
Current evidence suggests that glial cells provide C3 carbon sources to fuel neuronal activity; however, this notion has become challenged by biosensor studies carried out in acute brain slices or in vivo, showing that neuronal activity does not rely on the import of astrocyte-produced L-lactate. Rather, stimulated neurons become net lactate exporters, as it was also shown in Drosophila neurons, in which astrocyte-provided lactate returns as lipid droplets to be stored in glial cells. In this view, we investigate whether exogenously supplied monocarboxylates can support Drosophila motoneuron neurotransmitter release (NTR). By assessing the excitatory post-synaptic current (EPSC) amplitude under voltage-clamp as NTR indicative, we found that both pyruvate and L-lactate, as the only carbon sources in the synapses bathing-solution, cause a large transient NTR enhancement, which declines to reach a synaptic depression state, from which the synapses do not recover. The FM1-43 pre-synaptic loading ability, however, is maintained under monocarboxylate, suggesting that SV cycling should not contribute to the synaptic depression state. The NTR recovery was reached by supplementing the monocarboxylate medium with sucrose. However, monocarboxylate addition to sucrose medium does not enhance NTR, but it does when the disaccharide concentration becomes too reduced. Thus, when pyruvate concentrations become too reduced, exogenously supplied L-lactate could be converted to pyruvate and metabolized by the neural mitochondria, triggering the NTR enhancement. SIGNIFICANCE STATEMENT: The question of whether monocarboxylic acids can fuel the Drosophila motoneuron NTR was challenged. Our findings show that exogenously supplied monocarboxylates trigger a large transient synaptic enhancement just under extreme glycolysis reduction but fail to maintain NTR under sustained synaptic demand, still at low frequency stimulation, driven to the synapses to a synaptic depression state. Glycolysis activation, by adding sucrose to the monocarboxylate bath solution, restores the motoneuron NTR ability, giving place to a hexoses role in SV recruitment. Moreover these results suggest exogenously supplied C3 carbon sources could have an additional role beyond providing energetic support for neural activity.
Collapse
Affiliation(s)
- María-Graciela Delgado
- Department of Biology, Faculty of Sciences, University of Chile, Las Palmeras 3425, 7800001 Santiago, Chile.
| | - Ricardo Delgado
- Department of Biology, Faculty of Sciences, University of Chile, Las Palmeras 3425, 7800001 Santiago, Chile.
| |
Collapse
|
33
|
Yin P, Chen M, Rao M, Lin Y, Zhang M, Xu R, Hu X, Chen R, Chai W, Huang X, Yu H, Yao Y, Zhao Y, Li Y, Zhang L, Tang P. Deciphering Immune Landscape Remodeling Unravels the Underlying Mechanism for Synchronized Muscle and Bone Aging. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304084. [PMID: 38088531 PMCID: PMC10837389 DOI: 10.1002/advs.202304084] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 10/30/2023] [Indexed: 02/04/2024]
Abstract
Evidence from numerous studies has revealed the synchronous progression of aging in bone and muscle; however, little is known about the underlying mechanisms. To this end, human muscles and bones are harvested and the aging-associated transcriptional dynamics of two tissues in parallel using single-cell RNA sequencing are surveyed. A subset of lipid-associated macrophages (triggering receptor expressed on myeloid cells 2, TREM2+ Macs) is identified in both aged muscle and bone. Genes responsible for muscle dystrophy and bone loss, such as secreted phosphoprotein 1 (SPP1), are also highly expressed in TREM2+ Macs, suggesting its conserved role in aging-related features. A common transition toward pro-inflammatory phenotypes in aged CD4+ T cells across tissues is also observed, activated by the nuclear factor kappa B subunit 1 (NFKB1). CD4+ T cells in aged muscle experience Th1-like differentiation, whereas, in bone, a skewing toward Th17 cells is observed. Furthermore, these results highlight that degenerated myocytes produce BAG6-containing exosomes that can communicate with Th17 cells in the bone through its receptor natural cytotoxicity triggering receptor 3 (NCR3). This communication upregulates CD6 expression in Th17 cells, which then interact with TREM2+ Macs through CD6-ALCAM signaling, ultimately stimulating the transcription of SPP1 in TREM2+ Macs. The negative correlation between serum exosomal BCL2-associated athanogene 6 (BAG6) levels and bone mineral density further supports its role in mediating muscle and bone synchronization with aging.
Collapse
Affiliation(s)
- Pengbin Yin
- Senior Department of OrthopedicsThe Fourth Medical Center of PLA General HospitalBeijing100048China
- National Clinical Research Center for OrthopedicsSports Medicine & RehabilitationBeijing100048China
| | - Ming Chen
- Senior Department of OrthopedicsThe Fourth Medical Center of PLA General HospitalBeijing100048China
- National Clinical Research Center for OrthopedicsSports Medicine & RehabilitationBeijing100048China
| | - Man Rao
- Senior Department of OrthopedicsThe Fourth Medical Center of PLA General HospitalBeijing100048China
- National Clinical Research Center for OrthopedicsSports Medicine & RehabilitationBeijing100048China
- Analytical Biosciences LimitedBeijing100191China
| | - Yuan Lin
- The Department of Orthopedic SurgerySecond Affiliated Hospital of Harbin Medical UniversityHarbin150086China
| | - Mingming Zhang
- Senior Department of OrthopedicsThe Fourth Medical Center of PLA General HospitalBeijing100048China
- National Clinical Research Center for OrthopedicsSports Medicine & RehabilitationBeijing100048China
| | - Ren Xu
- State Key Laboratory of Cellular Stress BiologySchool of MedicineFaculty of Medicine and Life SciencesXiamen UniversityXiamen361102China
| | - Xueda Hu
- Analytical Biosciences LimitedBeijing100191China
| | - Ruijing Chen
- Senior Department of OrthopedicsThe Fourth Medical Center of PLA General HospitalBeijing100048China
- National Clinical Research Center for OrthopedicsSports Medicine & RehabilitationBeijing100048China
| | - Wei Chai
- Senior Department of OrthopedicsThe Fourth Medical Center of PLA General HospitalBeijing100048China
- National Clinical Research Center for OrthopedicsSports Medicine & RehabilitationBeijing100048China
| | - Xiang Huang
- Senior Department of OrthopedicsThe Fourth Medical Center of PLA General HospitalBeijing100048China
- National Clinical Research Center for OrthopedicsSports Medicine & RehabilitationBeijing100048China
| | - Haikuan Yu
- Senior Department of OrthopedicsThe Fourth Medical Center of PLA General HospitalBeijing100048China
- National Clinical Research Center for OrthopedicsSports Medicine & RehabilitationBeijing100048China
| | - Yao Yao
- Center for Healthy Aging and Development StudiesNational School of DevelopmentPeking UniversityBeijing100871China
| | - Yali Zhao
- Central LaboratoryHainan Hospital of Chinese People's Liberation Army General HospitalSanya572013China
| | - Yi Li
- Senior Department of OrthopedicsThe Fourth Medical Center of PLA General HospitalBeijing100048China
- National Clinical Research Center for OrthopedicsSports Medicine & RehabilitationBeijing100048China
| | - Licheng Zhang
- Senior Department of OrthopedicsThe Fourth Medical Center of PLA General HospitalBeijing100048China
- National Clinical Research Center for OrthopedicsSports Medicine & RehabilitationBeijing100048China
| | - Peifu Tang
- Senior Department of OrthopedicsThe Fourth Medical Center of PLA General HospitalBeijing100048China
- National Clinical Research Center for OrthopedicsSports Medicine & RehabilitationBeijing100048China
| |
Collapse
|
34
|
Ross JM, Olson L, Coppotelli G. Mitochondrial Dysfunction and Protein Homeostasis in Aging: Insights from a Premature-Aging Mouse Model. Biomolecules 2024; 14:162. [PMID: 38397399 PMCID: PMC10886786 DOI: 10.3390/biom14020162] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/10/2024] [Accepted: 01/22/2024] [Indexed: 02/25/2024] Open
Abstract
Mitochondrial dysfunction has been implicated in aging and age-related disorders. Disturbed-protein homeostasis and clearance of damaged proteins have also been linked to aging, as well as to neurodegenerative diseases, cancers, and metabolic disorders. However, since mitochondrial oxidative phosphorylation, ubiquitin-proteasome, and autophagy-lysosome systems are tightly interdependent, it is not understood whether the facets observed in aging are the causes or consequences of one or all of these failed processes. We therefore used prematurely aging mtDNA-mutator mice and normally aging wild-type littermates to elucidate whether mitochondrial dysfunction per se is sufficient to impair cellular protein homeostasis similarly to that which is observed in aging. We found that both mitochondrial dysfunction and normal aging affect the ubiquitin-proteasome system in a tissue-dependent manner, whereas only normal aging markedly impairs the autophagy-lysosome system. Thus, our data show that the proteostasis network control in the prematurely aging mtDNA-mutator mouse differs in certain aspects from that found in normal aging. Taken together, our findings suggest that severe mitochondrial dysfunction drives an aging phenotype associated with the impairment of certain components of the protein homeostasis machinery, while others, such as the autophagy-lysosome system, are not affected or only minimally affected. Taken together, this shows that aging is a multifactorial process resulting from alterations of several integrated biological processes; thus, manipulating one process at the time might not be sufficient to fully recapitulate all changes associated with normal aging.
Collapse
Affiliation(s)
- Jaime M. Ross
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI 02881, USA
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| | - Lars Olson
- Department of Neuroscience, Karolinska Institutet, S-17177 Stockholm, Sweden;
| | - Giuseppe Coppotelli
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI 02881, USA
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| |
Collapse
|
35
|
Zhang Y, Liu L, Qi Y, Lou J, Chen Y, Liu C, Li H, Chang X, Hu Z, Li Y, Zhang Y, Feng C, Zhou Y, Zhai Y, Li C. Lactic acid promotes nucleus pulposus cell senescence and corresponding intervertebral disc degeneration via interacting with Akt. Cell Mol Life Sci 2024; 81:24. [PMID: 38212432 PMCID: PMC11071984 DOI: 10.1007/s00018-023-05094-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 12/12/2023] [Accepted: 12/14/2023] [Indexed: 01/13/2024]
Abstract
The accumulation of metabolites in the intervertebral disc is considered an important cause of intervertebral disc degeneration (IVDD). Lactic acid, which is a metabolite that is produced by cellular anaerobic glycolysis, has been proven to be closely associated with IVDD. However, little is known about the role of lactic acid in nucleus pulposus cells (NPCs) senescence and oxidative stress. The aim of this study was to investigate the effect of lactic acid on NPCs senescence and oxidative stress as well as the underlying mechanism. A puncture-induced disc degeneration (PIDD) model was established in rats. Metabolomics analysis revealed that lactic acid levels were significantly increased in degenerated intervertebral discs. Elimination of excessive lactic acid using a lactate oxidase (LOx)-overexpressing lentivirus alleviated the progression of IVDD. In vitro experiments showed that high concentrations of lactic acid could induce senescence and oxidative stress in NPCs. High-throughput RNA sequencing results and bioinformatic analysis demonstrated that the induction of NPCs senescence and oxidative stress by lactic acid may be related to the PI3K/Akt signaling pathway. Further study verified that high concentrations of lactic acid could induce NPCs senescence and oxidative stress by interacting with Akt and regulating its downstream Akt/p21/p27/cyclin D1 and Akt/Nrf2/HO-1 pathways. Utilizing molecular docking, site-directed mutation and microscale thermophoresis assays, we found that lactic acid could regulate Akt kinase activity by binding to the Lys39 and Leu52 residues in the PH domain of Akt. These results highlight the involvement of lactic acid in NPCs senescence and oxidative stress, and lactic acid may become a novel potential therapeutic target for the treatment of IVDD.
Collapse
Affiliation(s)
- Yuyao Zhang
- Department of Orthopedics, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Military Medical University, Chongqing, 400038, China
| | - Libangxi Liu
- Department of Orthopedics, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Military Medical University, Chongqing, 400038, China
- Department of Orthopedics, General Hospital of Central Theater Command of PLA, Wuhan, 430000, China
| | - Yuhan Qi
- Institute of Basic Theory of Traditional Chinese Medicine, China Academy of Chinese Medical Science, Beijing, 100000, China
| | - Jinhui Lou
- Department of Orthopedics, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Military Medical University, Chongqing, 400038, China
| | - Yuxuan Chen
- Department of Orthopedics, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Military Medical University, Chongqing, 400038, China
| | - Chao Liu
- Department of Orthopedics, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Military Medical University, Chongqing, 400038, China
| | - Haiyin Li
- Department of Orthopedics, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Military Medical University, Chongqing, 400038, China
| | - Xian Chang
- Department of Orthopedics, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Military Medical University, Chongqing, 400038, China
| | - Zhilei Hu
- Department of Orthopedics, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Military Medical University, Chongqing, 400038, China
| | - Yueyang Li
- Department of Orthopedics, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Military Medical University, Chongqing, 400038, China
| | - Yang Zhang
- Department of Orthopedics, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Military Medical University, Chongqing, 400038, China
| | - Chencheng Feng
- Department of Orthopedics, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Military Medical University, Chongqing, 400038, China
| | - Yue Zhou
- Department of Orthopedics, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Military Medical University, Chongqing, 400038, China
| | - Yu Zhai
- Department of Orthopedics, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China.
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Military Medical University, Chongqing, 400038, China.
| | - Changqing Li
- Department of Orthopedics, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China.
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Military Medical University, Chongqing, 400038, China.
| |
Collapse
|
36
|
Garcia-Segura ME, Pluchino S, Peruzzotti-Jametti L. Metabolic Control of Microglia. ADVANCES IN NEUROBIOLOGY 2024; 37:607-622. [PMID: 39207716 DOI: 10.1007/978-3-031-55529-9_34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Microglia, immune sentinels of the central nervous system (CNS), play a critical role in maintaining its health and integrity. This chapter delves into the concept of immunometabolism, exploring how microglial metabolism shapes their diverse immune functions. It examines the impact of cell metabolism on microglia during various CNS states, including homeostasis, development, aging, and inflammation. Particularly in CNS inflammation, the chapter discusses how metabolic rewiring in microglia can initiate, resolve, or perpetuate inflammatory responses. The potential of targeting microglial metabolism as a therapeutic strategy for chronic CNS disorders with prominent innate immune cell activation is also explored.
Collapse
Affiliation(s)
- Monica Emili Garcia-Segura
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, UK
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Stefano Pluchino
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Luca Peruzzotti-Jametti
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, UK.
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
| |
Collapse
|
37
|
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. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.17.572088. [PMID: 38187759 PMCID: PMC10769226 DOI: 10.1101/2023.12.17.572088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
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 co-factor NAD+, which is reported to decline across multiple tissue types 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 up-regulated fatty acid and sphingolipid metabolism reactions that regenerate NAD+ from NADH. Because mitochondrial respiration, a major source of NAD+ regeneration, is reported to decline with age, our data supports a model where NAD+-generating lipid metabolism reactions may buffer against changes in NAD+/NADH during healthy aging.
Collapse
Affiliation(s)
- Evan C. Lien
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI 49503, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ngoc Vu
- Calico Life Sciences LLC, South San Francisco, CA 94080, USA
| | - Anna M. Westermark
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Laura V. Danai
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Allison N. Lau
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Yetiş Gültekin
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | | | - Matthew G. Vander Heiden
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
38
|
Pilley SE, Esparza E, Mullen PJ. The aging tumor metabolic microenvironment. Curr Opin Biotechnol 2023; 84:102995. [PMID: 37783168 DOI: 10.1016/j.copbio.2023.102995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 08/24/2023] [Accepted: 08/31/2023] [Indexed: 10/04/2023]
Abstract
Despite the higher incidence of cancer with increasing age, few preclinical or clinical studies incorporate age. This, coupled with an aging world population, requires that we improve our understanding of how aging affects cancer development, progression, and treatment. One key area will be how the tumor microenvironment (TME) changes with age. Metabolite levels are an essential component of the TME, and they are affected by the metabolic requirements of the cells present and systemic metabolite availability. These factors are affected by aging, causing different TME metabolic states between young and older adults. In this review, we will summarize what is known about how aging impacts the TME metabolic state, and suggest how we can improve our understanding of it.
Collapse
Affiliation(s)
- Steven E Pilley
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Edgar Esparza
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Peter J Mullen
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA; Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
39
|
Wevers NR, De Vries HE. Microfluidic models of the neurovascular unit: a translational view. Fluids Barriers CNS 2023; 20:86. [PMID: 38008744 PMCID: PMC10680291 DOI: 10.1186/s12987-023-00490-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 11/15/2023] [Indexed: 11/28/2023] Open
Abstract
The vasculature of the brain consists of specialized endothelial cells that form a blood-brain barrier (BBB). This barrier, in conjunction with supporting cell types, forms the neurovascular unit (NVU). The NVU restricts the passage of certain substances from the bloodstream while selectively permitting essential nutrients and molecules to enter the brain. This protective role is crucial for optimal brain function, but presents a significant obstacle in treating neurological conditions, necessitating chemical modifications or advanced drug delivery methods for most drugs to cross the NVU. A deeper understanding of NVU in health and disease will aid in the identification of new therapeutic targets and drug delivery strategies for improved treatment of neurological disorders.To achieve this goal, we need models that reflect the human BBB and NVU in health and disease. Although animal models of the brain's vasculature have proven valuable, they are often of limited translational relevance due to interspecies differences or inability to faithfully mimic human disease conditions. For this reason, human in vitro models are essential to improve our understanding of the brain's vasculature under healthy and diseased conditions. This review delves into the advancements in in vitro modeling of the BBB and NVU, with a particular focus on microfluidic models. After providing a historical overview of the field, we shift our focus to recent developments, offering insights into the latest achievements and their associated constraints. We briefly examine the importance of chip materials and methods to facilitate fluid flow, emphasizing their critical roles in achieving the necessary throughput for the integration of microfluidic models into routine experimentation. Subsequently, we highlight the recent strides made in enhancing the biological complexity of microfluidic NVU models and propose recommendations for elevating the biological relevance of future iterations.Importantly, the NVU is an intricate structure and it is improbable that any model will fully encompass all its aspects. Fit-for-purpose models offer a valuable compromise between physiological relevance and ease-of-use and hold the future of NVU modeling: as simple as possible, as complex as needed.
Collapse
Affiliation(s)
- Nienke R Wevers
- MIMETAS BV, De Limes 7, Oegstgeest, 2342 DH, The Netherlands.
| | - Helga E De Vries
- Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam Neuroscience - Neuroinfection and Neuroinflammation, De Boelelaan 1117, Amsterdam, the Netherlands
| |
Collapse
|
40
|
Ikonen L, Pirnes-Karhu S, Pradhan S, Jacobs HT, Szibor M, Suomalainen A. Alternative oxidase causes cell type- and tissue-specific responses in mutator mice. Life Sci Alliance 2023; 6:e202302036. [PMID: 37657934 PMCID: PMC10474302 DOI: 10.26508/lsa.202302036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 08/17/2023] [Accepted: 08/18/2023] [Indexed: 09/03/2023] Open
Abstract
Energetic insufficiency, excess production of reactive oxygen species (ROS), and aberrant signaling partially account for the diverse pathology of mitochondrial diseases. Whether interventions affecting ROS, a regulator of stem cell pools, could modify somatic stem cell homeostasis remains unknown. Previous data from mitochondrial DNA mutator mice showed that increased ROS leads to oxidative damage in erythroid progenitors, causing lifespan-limiting anemia. Also unclear is how ROS-targeted interventions affect terminally differentiated tissues. Here, we set out to test in mitochondrial DNA mutator mice how ubiquitous expression of the Ciona intestinalis alternative oxidase (AOX), which attenuates ROS production, affects murine stem cell pools. We found that AOX does not affect neural stem cells but delays the progression of mutator-driven anemia. Furthermore, when combined with the mutator, AOX potentiates mitochondrial stress and inflammatory responses in skeletal muscle. These differential cell type-specific findings demonstrate that AOX expression is not a global panacea for curing mitochondrial dysfunction. ROS attenuation must be carefully studied regarding specific underlying defects before AOX can be safely used in therapy.
Collapse
Affiliation(s)
- Lilli Ikonen
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Sini Pirnes-Karhu
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Swagat Pradhan
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Howard T Jacobs
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Marten Szibor
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Cardiothoracic Surgery, Center for Sepsis Control and Care, Jena University Hospital, Friedrich-Schiller University of Jena, Jena, Germany
| | - Anu Suomalainen
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Helsinki University Hospital, HUSLAB, Helsinki, Finland
| |
Collapse
|
41
|
Chen W, Zhang Y, Liang J, Zhang Z, Zhang L, Huang E, Zhang G, Lu L, Han Y, Shi J. Disrupting astrocyte-neuron lactate transport prevents cocaine seeking after prolonged withdrawal. SCIENCE ADVANCES 2023; 9:eadi4462. [PMID: 37878699 PMCID: PMC10599624 DOI: 10.1126/sciadv.adi4462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 09/21/2023] [Indexed: 10/27/2023]
Abstract
Energy supply, especially the transfer of lactate from astrocytes to neurons, is critical for neuronal plasticity. However, its role in the incubation of cocaine craving remains largely unknown. Using an extended-access self-administration model and in vivo 1H-magnetic resonance spectroscopy, we found that lactate synthesis in the central amygdala (CeA) is required for the intensified cocaine craving after prolonged withdrawal. Furthermore, incubated cocaine seeking was associated with a selective increase in monocarboxylate transporter 2 (MCT2) and MCT4 expression levels. Down-regulation of astrocytic MCT4 or neuronal MCT2 using targeted antisense oligonucleotides or cell type-specific shRNA attenuated cocaine craving and reduced the expression of plasticity-related proteins and excitatory synaptic transmission. Meanwhile, lactate administration rescued MCT4 but not MCT2 disruption-induced behavioral changes due to the inability of lactate to be transported into neurons. Together, our study highlights the critical role of astrocyte-neuron lactate transport in the CeA in the incubation of cocaine craving and suggests a potential therapeutic target for drug addiction.
Collapse
Affiliation(s)
- Wenjun Chen
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Yan Zhang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
| | - Jie Liang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
| | - Zhongyu Zhang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
| | - Libo Zhang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
- Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Enze Huang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Guipeng Zhang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Lin Lu
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Ying Han
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
| | - Jie Shi
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
- The Key Laboratory for Neuroscience of the Ministry of Education and Health, Peking University, Beijing, 100191, China
- The State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, China
| |
Collapse
|
42
|
Chirumbolo S, Bertossi D, Magistretti P. Insights on the role of L-lactate as a signaling molecule in skin aging. Biogerontology 2023; 24:709-726. [PMID: 36708434 PMCID: PMC9883612 DOI: 10.1007/s10522-023-10018-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/17/2023] [Indexed: 01/29/2023]
Abstract
L-lactate is a catabolite from the anaerobic metabolism of glucose, which plays a paramount role as a signaling molecule in various steps of the cell survival. Its activity, as a master tuner of many mechanisms underlying the aging process, for example in the skin, is still presumptive, however its crucial position in the complex cross-talk between mitochondria and the process of cell survival, should suggest that L-lactate may be not a simple waste product but a fine regulator of the aging/survival machinery, probably via mito-hormesis. Actually, emerging evidence is highlighting that ROS are crucial in the signaling of skin health, including mechanisms underlying wound repair, renewal and aging. The ROS, including superoxide anion, hydrogen peroxide, and nitric oxide, play both beneficial and detrimental roles depending upon their levels and cellular microenvironment. Physiological ROS levels are essential for cutaneous health and the wound repair process. Aberrant redox signaling activity drives chronic skin disease in elderly. On the contrary, impaired redox modulation, due to enhanced ROS generation and/or reduced levels of antioxidant defense, suppresses wound healing via promoting lymphatic/vascular endothelial cell apoptosis and death. This review tries to elucidate this issue.
Collapse
Affiliation(s)
- Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, Unit of Human Anatomy, University of Verona, Strada Le Grazie 8, 37134, Verona, Italy.
| | - Dario Bertossi
- Department of Surgery, Dentistry, Paediatrics and Gynaecology-Unit of Maxillo-Facial Surgery, University of Verona, Verona, Italy
| | - Pierre Magistretti
- Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955, Saudi Arabia
| |
Collapse
|
43
|
Drulis‐Fajdasz D, Krzystyniak A, Puścian A, Pytyś A, Gostomska‐Pampuch K, Pudełko‐Malik N, Wiśniewski JŁ, Młynarz P, Miazek A, Wójtowicz T, Włodarczyk J, Duś‐Szachniewicz K, Gizak A, Wiśniewski JR, Rakus D. Glycogen phosphorylase inhibition improves cognitive function of aged mice. Aging Cell 2023; 22:e13928. [PMID: 37522798 PMCID: PMC10497847 DOI: 10.1111/acel.13928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 05/31/2023] [Accepted: 06/23/2023] [Indexed: 08/01/2023] Open
Abstract
Inhibition of glycogen breakdown blocks memory formation in young animals, but it stimulates the maintenance of the long-term potentiation, a cellular mechanism of memory formation, in hippocampal slices of old animals. Here, we report that a 2-week treatment with glycogen phosphorylase inhibitor BAY U6751 alleviated memory deficits and stimulated neuroplasticity in old mice. Using the 2-Novel Object Recognition and Novel Object Location tests, we discovered that the prolonged intraperitoneal administration of BAY U6751 improved memory formation in old mice. This was accompanied by changes in morphology of dendritic spines in hippocampal neurons, and by "rejuvenation" of hippocampal proteome. In contrast, in young animals, inhibition of glycogen degradation impaired memory formation; however, as in old mice, it did not alter significantly the morphology and density of cortical dendritic spines. Our findings provide evidence that prolonged inhibition of glycogen phosphorolysis improves memory formation of old animals. This could lead to the development of new strategies for treatment of age-related memory deficits.
Collapse
Affiliation(s)
| | - Adam Krzystyniak
- Laboratory of Cell BiophysicsNencki Institute of Experimental Biology, Polish Academy of SciencesWarsawPoland
| | - Alicja Puścian
- Nencki‐EMBL Partnership for Neural Plasticity and Brain Disorders – BRAINCITYNencki Institute of Experimental Biology, Polish Academy of SciencesWarsawPoland
| | - Agata Pytyś
- Laboratory of Cell BiophysicsNencki Institute of Experimental Biology, Polish Academy of SciencesWarsawPoland
| | - Kinga Gostomska‐Pampuch
- Department of Biochemistry and ImmunochemistryWroclaw Medical UniversityWroclawPoland
- Biochemical Proteomics Group, Department of Proteomics and Signal TransductionMax Planck Institute of BiochemistryMartinsriedGermany
| | - Natalia Pudełko‐Malik
- Department of Biochemistry, Molecular Biology and Biotechnology, Faculty of ChemistryWroclaw University of Science and TechnologyWroclawPoland
| | - Jerzy Ł. Wiśniewski
- Department of Biochemistry, Molecular Biology and Biotechnology, Faculty of ChemistryWroclaw University of Science and TechnologyWroclawPoland
| | - Piotr Młynarz
- Department of Biochemistry, Molecular Biology and Biotechnology, Faculty of ChemistryWroclaw University of Science and TechnologyWroclawPoland
| | - Arkadiusz Miazek
- Laboratory of Tumor ImmunologyHirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of SciencesWroclawPoland
| | - Tomasz Wójtowicz
- Laboratory of Cell BiophysicsNencki Institute of Experimental Biology, Polish Academy of SciencesWarsawPoland
| | - Jakub Włodarczyk
- Laboratory of Cell BiophysicsNencki Institute of Experimental Biology, Polish Academy of SciencesWarsawPoland
| | - Kamila Duś‐Szachniewicz
- Department of Clinical and Experimental PathologyInstitute of General and Experimental Pathology, Wroclaw Medical UniversityWroclawPoland
| | - Agnieszka Gizak
- Department of Molecular Physiology and NeurobiologyUniversity of WroclawWroclawPoland
| | - Jacek R. Wiśniewski
- Biochemical Proteomics Group, Department of Proteomics and Signal TransductionMax Planck Institute of BiochemistryMartinsriedGermany
| | - Dariusz Rakus
- Department of Molecular Physiology and NeurobiologyUniversity of WroclawWroclawPoland
| |
Collapse
|
44
|
Rojas-Valverde D, Bonilla DA, Gómez-Miranda LM, Calleja-Núñez JJ, Arias N, Martínez-Guardado I. Examining the Interaction between Exercise, Gut Microbiota, and Neurodegeneration: Future Research Directions. Biomedicines 2023; 11:2267. [PMID: 37626763 PMCID: PMC10452292 DOI: 10.3390/biomedicines11082267] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 08/08/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
Physical activity has been demonstrated to have a significant impact on gut microbial diversity and function. Emerging research has revealed certain aspects of the complex interactions between the gut, exercise, microbiota, and neurodegenerative diseases, suggesting that changes in gut microbial diversity and metabolic function may have an impact on the onset and progression of neurological conditions. This study aimed to review the current literature from several databases until 1 June 2023 (PubMed/MEDLINE, Web of Science, and Google Scholar) on the interplay between the gut, physical exercise, microbiota, and neurodegeneration. We summarized the roles of exercise and gut microbiota on neurodegeneration and identified the ways in which these are all connected. The gut-brain axis is a complex and multifaceted network that has gained considerable attention in recent years. Research indicates that gut microbiota plays vital roles in metabolic shifts during physiological or pathophysiological conditions in neurodegenerative diseases; therefore, they are closely related to maintaining overall health and well-being. Similarly, exercise has shown positive effects on brain health and cognitive function, which may reduce/delay the onset of severe neurological disorders. Exercise has been associated with various neurochemical changes, including alterations in cortisol levels, increased production of endorphins, endocannabinoids like anandamide, as well as higher levels of serotonin and dopamine. These changes have been linked to mood improvements, enhanced sleep quality, better motor control, and cognitive enhancements resulting from exercise-induced effects. However, further clinical research is necessary to evaluate changes in bacteria taxa along with age- and sex-based differences.
Collapse
Affiliation(s)
- Daniel Rojas-Valverde
- Nucleus of Studies for High Performance and Health (CIDISAD-NARS), School of Human Movement Sciences and Quality of Life (CIEMHCAVI), National University, Heredia 86-3000, Costa Rica
- Sports Injury Clinic (Rehab & Readapt), School of Human Movement Sciences and Quality of Life (CIEMHCAVI), National University, Heredia 86-3000, Costa Rica
| | - Diego A. Bonilla
- Research Division, Dynamical Business & Science Society—DBSS International SAS, Bogotá 110311, Colombia;
- Research Group in Biochemistry and Molecular Biology, Faculty of Sciences and Education, Universidad Distrital Francisco José de Caldas, Bogotá 110311, Colombia
- Research Group in Physical Activity, Sports and Health Sciences (GICAFS), Universidad de Córdoba, Montería 230002, Colombia
- Sport Genomics Research Group, Department of Genetics, Physical Anthropology and Animal Physiology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
| | - Luis M. Gómez-Miranda
- Sports Faculty, Autonomous University of Baja California, Tijuana 22615, Mexico; (L.M.G.-M.); (J.J.C.-N.)
| | - Juan J. Calleja-Núñez
- Sports Faculty, Autonomous University of Baja California, Tijuana 22615, Mexico; (L.M.G.-M.); (J.J.C.-N.)
| | - Natalia Arias
- BRABE Group, Department of Psychology, Faculty of Life and Natural Sciences, University of Nebrija, C/del Hostal, 28248 Madrid, Spain;
| | - Ismael Martínez-Guardado
- BRABE Group, Department of Psychology, Faculty of Life and Natural Sciences, University of Nebrija, C/del Hostal, 28248 Madrid, Spain;
| |
Collapse
|
45
|
Bednarik P, Goranovic D, Svatkova A, Niess F, Hingerl L, Strasser B, Deelchand DK, Spurny-Dworak B, Krssak M, Trattnig S, Hangel G, Scherer T, Lanzenberger R, Bogner W. 1H magnetic resonance spectroscopic imaging of deuterated glucose and of neurotransmitter metabolism at 7 T in the human brain. Nat Biomed Eng 2023; 7:1001-1013. [PMID: 37106154 PMCID: PMC10861140 DOI: 10.1038/s41551-023-01035-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 03/30/2023] [Indexed: 04/29/2023]
Abstract
Impaired glucose metabolism in the brain has been linked to several neurological disorders. Positron emission tomography and carbon-13 magnetic resonance spectroscopic imaging (MRSI) can be used to quantify the metabolism of glucose, but these methods involve exposure to radiation, cannot quantify downstream metabolism, or have poor spatial resolution. Deuterium MRSI (2H-MRSI) is a non-invasive and safe alternative for the quantification of the metabolism of 2H-labelled substrates such as glucose and their downstream metabolic products, yet it can only measure a limited number of deuterated compounds and requires specialized hardware. Here we show that proton MRSI (1H-MRSI) at 7 T has higher sensitivity, chemical specificity and spatiotemporal resolution than 2H-MRSI. We used 1H-MRSI in five volunteers to differentiate glutamate, glutamine, γ-aminobutyric acid and glucose deuterated at specific molecular positions, and to simultaneously map deuterated and non-deuterated metabolites. 1H-MRSI, which is amenable to clinically available magnetic-resonance hardware, may facilitate the study of glucose metabolism in the brain and its potential roles in neurological disorders.
Collapse
Affiliation(s)
- Petr Bednarik
- High-Field MR Centre, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria.
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Amager and Hvidovre, Copenhagen, Denmark.
- Department of Radiology, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Amager and Hvidovre, Copenhagen, Denmark.
| | - Dario Goranovic
- High-Field MR Centre, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Alena Svatkova
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Amager and Hvidovre, Copenhagen, Denmark
- Department of Radiology, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Amager and Hvidovre, Copenhagen, Denmark
- Department of Medicine III, Division of Endocrinology and Metabolism, Medical University of Vienna, Vienna, Austria
| | - Fabian Niess
- High-Field MR Centre, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Lukas Hingerl
- High-Field MR Centre, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Bernhard Strasser
- High-Field MR Centre, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Dinesh K Deelchand
- Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN, USA
| | - Benjamin Spurny-Dworak
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Martin Krssak
- Department of Medicine III, Division of Endocrinology and Metabolism, Medical University of Vienna, Vienna, Austria
| | - Siegfried Trattnig
- High-Field MR Centre, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Gilbert Hangel
- High-Field MR Centre, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
- Department of Neurosurgery, Medical University of Vienna, Vienna, Austria
| | - Thomas Scherer
- Department of Medicine III, Division of Endocrinology and Metabolism, Medical University of Vienna, Vienna, Austria
| | - Rupert Lanzenberger
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Bogner
- High-Field MR Centre, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
46
|
Kamalmaz N, Ben Bacha A, Alonazi M, Albasher G, Khayyat AIA, El-Ansary A. Unveiling sex-based differences in developing propionic acid-induced features in mice as a rodent model of ASD. PeerJ 2023; 11:e15488. [PMID: 37334116 PMCID: PMC10274690 DOI: 10.7717/peerj.15488] [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: 01/25/2023] [Accepted: 05/10/2023] [Indexed: 06/20/2023] Open
Abstract
Background Males are more likely to develop autism as a neurodevelopmental disorder than females are, although the mechanisms underlying male vulnerability are not fully understood. Therefore, studying the role of autism etiologies considering sex differences in the propionic acid (PPA) rodent model of autism would build greater understanding of how females are protected from autism spectrum disorder, which may be used as a treatment strategy for males with autism. Objectives This study aimed to investigate the sex differences in oxidative stress, glutamate excitotoxicity, neuroinflammation, and gut microbiota impairment as etiological mechanisms for many neurological diseases, with specific reference to autism. Method Forty albino mice were divided into four groups of 10 animals each with two control and two treated groups of both sexes received only phosphate-buffered saline or a neurotoxic dose of PPA (250 mg/kg body weight) for 3 days, respectively. Biochemical markers of energy metabolism, oxidative stress, neuroinflammation, and excitotoxicity were measured in mouse brain homogenates, whereas the presence of pathogenic bacteria was assessed in mouse stool samples. Furthermore, the repetitive behavior, cognitive ability, and physical-neural coordination of the animals were examined. Results Collectively, selected variables related to oxidative stress, glutamate excitotoxicity, neuroinflammation, and gut bacteria were impaired concomitantly with altered behavior in PPA-induced rodent model, with males being more susceptible than females. Conclusion This study explains the role of sex in the higher vulnerability of males to develop autistic biochemical and behavioral features compared with females. Female sex hormones and the higher detoxification capacity and higher glycolytic flux in females serve as neuroprotective contributors in a rodent model of autism.
Collapse
Affiliation(s)
- Nasreen Kamalmaz
- Biochemistry Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Abir Ben Bacha
- Biochemistry Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Mona Alonazi
- Biochemistry Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Gadah Albasher
- Zoology Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Arwa Ishaq A. Khayyat
- Biochemistry Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Afaf El-Ansary
- Central Research Laboratory, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
47
|
Latina V, Atlante A, Malerba F, La Regina F, Balzamino BO, Micera A, Pignataro A, Stigliano E, Cavallaro S, Calissano P, Amadoro G. The Cleavage-Specific Tau 12A12mAb Exerts an Anti-Amyloidogenic Action by Modulating the Endocytic and Bioenergetic Pathways in Alzheimer's Disease Mouse Model. Int J Mol Sci 2023; 24:ijms24119683. [PMID: 37298634 DOI: 10.3390/ijms24119683] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 05/27/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023] Open
Abstract
Beyond deficits in hippocampal-dependent episodic memory, Alzheimer's Disease (AD) features sensory impairment in visual cognition consistent with extensive neuropathology in the retina. 12A12 is a monoclonal cleavage specific antibody (mAb) that in vivo selectively neutralizes the AD-relevant, harmful N-terminal 20-22 kDa tau fragment(s) (i.e., NH2htau) without affecting the full-length normal protein. When systemically injected into the Tg2576 mouse model overexpressing a mutant form of Amyloid Precursor Protein (APP), APPK670/671L linked to early onset familial AD, this conformation-specific tau mAb successfully reduces the NH2htau accumulating both in their brain and retina and, thus, markedly alleviates the phenotype-associated signs. By means of a combined biochemical and metabolic experimental approach, we report that 12A12mAb downregulates the steady state expression levels of APP and Beta-Secretase 1 (BACE-1) and, thus, limits the Amyloid beta (Aβ) production both in the hippocampus and retina from this AD animal model. The local, antibody-mediated anti-amyloidogenic action is paralleled in vivo by coordinated modulation of the endocytic (BIN1, RIN3) and bioenergetic (glycolysis and L-Lactate) pathways. These findings indicate for the first time that similar molecular and metabolic retino-cerebral pathways are modulated in a coordinated fashion in response to 12A12mAb treatment to tackle the neurosensorial Aβ accumulation in AD neurodegeneration.
Collapse
Affiliation(s)
- Valentina Latina
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| | - Anna Atlante
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council (CNR), Via Amendola 122/O, 70126 Bari, Italy
| | - Francesca Malerba
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| | - Federico La Regina
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| | - Bijorn Omar Balzamino
- Research Laboratories in Ophthalmology, IRCCS-Fondazione Bietti, Via Santo Stefano Rotondo 6, 00184 Rome, Italy
| | - Alessandra Micera
- Research Laboratories in Ophthalmology, IRCCS-Fondazione Bietti, Via Santo Stefano Rotondo 6, 00184 Rome, Italy
| | - Annabella Pignataro
- Institute of Translational Pharmacology (IFT), National Research Council (CNR), Via Fosso del Cavaliere 100, 00133 Rome, Italy
| | - Egidio Stigliano
- Area of Pathology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Istituto di Anatomia Patologica, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Sebastiano Cavallaro
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), Via P. Gaifami 18, 95126 Catania, Italy
| | - Pietro Calissano
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| | - Giuseppina Amadoro
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
- Institute of Translational Pharmacology (IFT), National Research Council (CNR), Via Fosso del Cavaliere 100, 00133 Rome, Italy
| |
Collapse
|
48
|
Austin TT, Thomas CL, Lewis C, Blockley A, Warren B. Metabolic decline in an insect ear: correlative or causative for age-related auditory decline? Front Cell Dev Biol 2023; 11:1138392. [PMID: 37274746 PMCID: PMC10233746 DOI: 10.3389/fcell.2023.1138392] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 04/25/2023] [Indexed: 06/06/2023] Open
Abstract
One leading hypothesis for why we lose our hearing as we age is a decrease in ear metabolism. However, direct measurements of metabolism across a lifespan in any auditory system are lacking. Even if metabolism does decrease with age, a question remains: is a metabolic decrease a cause of age-related auditory decline or simply correlative? We use an insect, the desert locust Schistocerca gregaria, as a physiologically versatile model to understand how cellular metabolism correlates with age and impacts on age-related auditory decline. We found that auditory organ metabolism decreases with age as measured fluorometrically. Next, we measured the individual auditory organ's metabolic rate and its sound-evoked nerve activity and found no correlation. We found no age-related change in auditory nerve activity, using hook electrode recordings, and in the electrophysiological properties of auditory neurons, using patch-clamp electrophysiology, but transduction channel activity decreased. To further test for a causative role of the metabolic rate in auditory decline, we manipulated metabolism of the auditory organ through diet and cold-rearing but found no difference in sound-evoked nerve activity. We found that although metabolism correlates with age-related auditory decline, it is not causative. Finally, we performed RNA-Seq on the auditory organs of young and old locusts, and whilst we found enrichment for Gene Ontology terms associated with metabolism, we also found enrichment for a number of additional aging GO terms. We hypothesize that age-related hearing loss is dominated by accumulative damage in multiple cell types and multiple processes which outweighs its metabolic decline.
Collapse
|
49
|
Yang Z, Yu Z, Xiao B. Coordinated Regulation of Myelination by Growth Factor and Amino-acid Signaling Pathways. Neurosci Bull 2023; 39:453-465. [PMID: 36352321 PMCID: PMC10043148 DOI: 10.1007/s12264-022-00967-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 06/27/2022] [Indexed: 11/11/2022] Open
Abstract
Myelin-forming oligodendrocytes in the central nervous system (CNS) and Schwann cells in the peripheral nervous system (PNS) are essential for structural and functional homeostasis of nervous tissue. Albeit with certain similarities, the regulation of CNS and PNS myelination is executed differently. Recent advances highlight the coordinated regulation of oligodendrocyte myelination by amino-acid sensing and growth factor signaling pathways. In this review, we discuss novel insights into the understanding of differential regulation of oligodendrocyte and Schwann cell biology in CNS and PNS myelination, with particular focus on the roles of growth factor-stimulated RHEB-mTORC1 and GATOR2-mediated amino-acid sensing/signaling pathways. We also discuss recent progress on the metabolic regulation of oligodendrocytes and Schwann cells and the impact of their dysfunction on neuronal function and disease.
Collapse
Affiliation(s)
- Zhiwen Yang
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518000, China
| | - Zongyan Yu
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518000, China.
| | - Bo Xiao
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518000, China.
| |
Collapse
|
50
|
Stacpoole PW, McCall CE. The pyruvate dehydrogenase complex: Life's essential, vulnerable and druggable energy homeostat. Mitochondrion 2023; 70:59-102. [PMID: 36863425 DOI: 10.1016/j.mito.2023.02.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 01/30/2023] [Accepted: 02/13/2023] [Indexed: 03/04/2023]
Abstract
Found in all organisms, pyruvate dehydrogenase complexes (PDC) are the keystones of prokaryotic and eukaryotic energy metabolism. In eukaryotic organisms these multi-component megacomplexes provide a crucial mechanistic link between cytoplasmic glycolysis and the mitochondrial tricarboxylic acid (TCA) cycle. As a consequence, PDCs also influence the metabolism of branched chain amino acids, lipids and, ultimately, oxidative phosphorylation (OXPHOS). PDC activity is an essential determinant of the metabolic and bioenergetic flexibility of metazoan organisms in adapting to changes in development, nutrient availability and various stresses that challenge maintenance of homeostasis. This canonical role of the PDC has been extensively probed over the past decades by multidisciplinary investigations into its causal association with diverse physiological and pathological conditions, the latter making the PDC an increasingly viable therapeutic target. Here we review the biology of the remarkable PDC and its emerging importance in the pathobiology and treatment of diverse congenital and acquired disorders of metabolic integration.
Collapse
Affiliation(s)
- Peter W Stacpoole
- Department of Medicine (Division of Endocrinology, Metabolism and Diabetes), and Department of Biochemistry and Molecular Biology, University of Florida, College of Medicine, Gainesville, FL, United States.
| | - Charles E McCall
- Department of Internal Medicine and Translational Sciences, and Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|