1
|
Zemniaçak ÂB, Ribeiro RT, das Neves GM, Cunha SDA, Tavares TQ, Carvalho AVS, Netto CA, Castilho RF, Wajner M, Amaral AU. α-Ketoisocaproic Acid Disrupts Mitochondrial Bioenergetics in the Brain of Neonate Rats: Molecular Modeling Studies of α-ketoglutarate Dehydrogenase Subunits Inhibition. Neurochem Res 2025; 50:76. [PMID: 39786600 DOI: 10.1007/s11064-024-04328-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/21/2024] [Accepted: 12/23/2024] [Indexed: 01/12/2025]
Abstract
Brain accumulation of the branched-chain α-keto acids α-ketoisocaproic acid (KIC), α-keto-β-methylvaleric acid (KMV), and α-ketoisovaleric acid (KIV) occurs in maple syrup urine disease (MSUD), an inherited intoxicating metabolic disorder caused by defects of the branched-chain α-keto acid dehydrogenase complex. Patients commonly suffer life-threatening acute encephalopathy in the newborn period and develop chronic neurological sequelae of still undefined pathogenesis. Therefore, this work investigated the in vitro influence of pathological concentrations of KIC (5 mM), KMV (1 mM), and KIV (1 mM) on mitochondrial bioenergetics in the cerebral cortex of neonate (one-day-old) rats. KIC, but not KMV and KIV, decreased phosphorylating (stimulated by ADP) and uncoupled (induced by CCCP) mitochondrial respiration supported by pyruvate, malate, and glutamate, indicating metabolic inhibition. These effects were less evident after supplementing the medium with succinate. KIC also mildly increased non-phosphorylating respiration (in the presence of oligomycin) using pyruvate plus malate or glutamate plus malate as substrates, suggesting an uncoupling effect. Moreover, KIC markedly inhibited the activity of α-ketoglutarate dehydrogenase noncompetitively and decreased ATP synthesis. Finally, docking simulations demonstrated that KIC preferentially interacts with E2 and E3 subunits of α-ketoglutarate dehydrogenase at the dihydrolipoamide binding site and into an allosteric site of E1. The present data strongly indicate that KIC compromises mitochondrial bioenergetics in the neonatal rat brain, supporting the hypothesis that disruption of energy homeostasis caused by brain KIC accumulation in the first days of life may be implicated in the neuropathology of MSUD.
Collapse
Affiliation(s)
- Ângela Beatris Zemniaçak
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Rafael Teixeira Ribeiro
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Gustavo Machado das Neves
- Laboratório de Síntese Orgânica Medicinal (LaSOM), Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Sâmela de Azevedo Cunha
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Tailine Quevedo Tavares
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Andrey Vinícios Soares Carvalho
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Carlos Alexandre Netto
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Roger Frigério Castilho
- Departamento de Patologia, Faculdade de Ciências Médicas, Universidade Estadual de Campinas, Campinas, SP, Brazil
| | - Moacir Wajner
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Alexandre Umpierrez Amaral
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
- Programa de Pós-Graduação em Atenção Integral à Saúde (UNICRUZ/URI-Erechim/UNIJUÍ), Universidade Regional Integrada do Alto Uruguai e das Missões, Erechim, RS, Brazil.
| |
Collapse
|
2
|
Yan HH, He JJ, Fu C, Chen JH, Tang AH. ATAD1 Regulates Neuronal Development and Synapse Formation Through Tuning Mitochondrial Function. Int J Mol Sci 2024; 26:44. [PMID: 39795902 PMCID: PMC11719905 DOI: 10.3390/ijms26010044] [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: 11/04/2024] [Revised: 12/20/2024] [Accepted: 12/21/2024] [Indexed: 01/13/2025] Open
Abstract
Mitochondrial function is essential for synaptic function. ATAD1, an AAA+ protease involved in mitochondrial quality control, governs fission-fusion dynamics within the organelle. However, the distribution and functional role of ATAD1 in neurons remain poorly understood. In this study, we demonstrate that ATAD1 is primarily localized to mitochondria in dendrites and, to a lesser extent, in spines in cultured hippocampal neurons. We found that ATAD1 deficiency disrupts the mitochondrial fission-fusion balance, resulting in mitochondrial fragmentation. This deficiency also impairs dendritic branching, hinders dendritic spine maturation, and reduces glutamatergic synaptic transmission in hippocampal neuron. To further investigate the underlying mechanism, we employed an ATP hydrolysis-deficient mutant of ATAD1 to rescue the neuronal deficits associated with ATAD1 loss. We discovered that the synaptic deficits are independent of the mitochondrial morphology changes but rely on its ATP hydrolysis. Furthermore, we show that ATAD1 loss leads to impaired mitochondrial function, including decreased ATP production, impaired membrane potential, and elevated oxidative stress. In conclusion, our results provide evidence that ATAD1 is crucial for maintaining mitochondrial function and regulating neurodevelopment and synaptic function.
Collapse
Affiliation(s)
- Hao-Hao Yan
- Hefei National Laboratory for Physical Sciences at the Microscale, MOE Key Laboratory for Membrane-Less Organelles & Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (H.-H.Y.); (J.-J.H.); (C.F.)
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China
- Neurodegenerative Disorder Research Center and Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei 230027, China
| | - Jia-Jia He
- Hefei National Laboratory for Physical Sciences at the Microscale, MOE Key Laboratory for Membrane-Less Organelles & Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (H.-H.Y.); (J.-J.H.); (C.F.)
| | - Chuanhai Fu
- Hefei National Laboratory for Physical Sciences at the Microscale, MOE Key Laboratory for Membrane-Less Organelles & Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (H.-H.Y.); (J.-J.H.); (C.F.)
| | - Jia-Hui Chen
- Department of Anatomy, School of Basic Medicine, Anhui Medical University, Hefei 230032, China
| | - Ai-Hui Tang
- Hefei National Laboratory for Physical Sciences at the Microscale, MOE Key Laboratory for Membrane-Less Organelles & Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (H.-H.Y.); (J.-J.H.); (C.F.)
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China
- Neurodegenerative Disorder Research Center and Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei 230027, China
| |
Collapse
|
3
|
Vetrovoy OV, Potapova SS, Stratilov VA, Tyulkova EI. Prenatal Hypoxia Predisposes to Impaired Expression of the chrna4 and chrna7 Genes in Adult Rats without Affecting Acetylcholine Metabolism during Embryonic Development. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1950-1960. [PMID: 39647824 DOI: 10.1134/s0006297924110099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/28/2024] [Accepted: 06/03/2024] [Indexed: 12/10/2024]
Abstract
Previous studies have shown that the combined effect of fetal hypoxia and maternal stress hormones predetermines tendency to nicotine addiction in adulthood. This study in rats aimed to investigate the effect of prenatal severe hypoxia (PSH) on acetylcholine metabolism in the developing brain, as well as on expression of acetylcholine receptors chrna4 and chrna7 in both the developing brain and adult brain structures following nicotine consumption. In the developing brain of PSH rats, no changes were found in the activity of choline acetyltransferase (ChAT) and acetylcholinesterase (AChE) or disturbances in the acetylcholine levels. However, decreased chrna4 expression was detected on the day 15 of pregnancy, while elevation in the chrna7 expression was observed on the days 15 and 16 of embryogenesis. In adulthood, the consequences of PSH were manifested as decreased expression of chrna4 in the medial prefrontal cortex (PFC), nucleus accumbens (NAacc), and hypothalamus (HT), decreased expression of chrna7 in the PFC and hippocampus (HPC). Whereas, nicotine consumption did not decrease the expression levels of chrna4 and chrna7 compared to the control group in the adult PSH rats. Thus, prenatal hypoxia predisposes to impaired expression of the chrna4 and chrna7 genes in adult rats without affecting acetylcholine metabolism during embryonic development.
Collapse
Affiliation(s)
- Oleg V Vetrovoy
- Laboratory of Regulation of Brain Neuronal Functions, Pavlov Institute of Physiology, Russian Academy of Sciences, Saint Petersburg, 199034, Russia.
| | - Sofiia S Potapova
- Laboratory of Regulation of Brain Neuronal Functions, Pavlov Institute of Physiology, Russian Academy of Sciences, Saint Petersburg, 199034, Russia
| | - Viktor A Stratilov
- Laboratory of Regulation of Brain Neuronal Functions, Pavlov Institute of Physiology, Russian Academy of Sciences, Saint Petersburg, 199034, Russia
| | - Ekaterina I Tyulkova
- Laboratory of Regulation of Brain Neuronal Functions, Pavlov Institute of Physiology, Russian Academy of Sciences, Saint Petersburg, 199034, Russia
| |
Collapse
|
4
|
Hua J, Zhang T, Chen X, Zhu B, Zhao M, Fu K, Zhang Y, Tang H, Pang H, Guo Y, Han J, Yang L, Zhou B. Behavioral impairments and disrupted mitochondrial energy metabolism induced by polypropylene microplastics in zebrafish larvae. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 947:174541. [PMID: 38977091 DOI: 10.1016/j.scitotenv.2024.174541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 07/01/2024] [Accepted: 07/04/2024] [Indexed: 07/10/2024]
Abstract
Polypropylene microplastics (PP-MPs) are emerging pollutant commonly detected in various environmental matrices and organisms, while their adverse effects and mechanisms are not well known. Here, zebrafish embryos were exposed to environmentally relevant concentrations of PP-MPs (0.08-50 mg/L) from 2 h post-fertilization (hpf) until 120 hpf. The results showed that the body weight was increased at 2 mg/L, heart rate was reduced at 0.08 and 10 mg/L, and behaviors were impaired at 0.4, 10 or 50 mg/L. Subsequently, transcriptomic analysis in the 0.4 and 50 mg/L PP-MPs treatment groups indicated potential inhibition on the glycolysis/gluconeogenesis and oxidative phosphorylation pathways. These findings were validated through alterations in multiple biomarkers related to glucose metabolism. Moreover, abnormal mitochondrial ultrastructures were observed in the intestine and liver in 0.4 and 50 mg/L PP-MPs treatment groups, accompanied by significant decreases in the activities of four mitochondrial electron transport chain complexes and ATP contents. Oxidative stress was also induced, as indicated by significantly increased ROS levels and significant reduced activities of CAT and SOD and GSH contents. All the results suggested that environmentally relevant concentrations of PP-MPs could induce disrupted mitochondrial energy metabolism in zebrafish, which may be associated with the observed behavioral impairments. This study will provide novel insights into PP-MPs-induced adverse effects and highlight need for further research.
Collapse
Affiliation(s)
- Jianghuan Hua
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, China; Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; Hubei Shizhen Laboratory, Wuhan 430061, China.
| | - Taotao Zhang
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; Hubei Shizhen Laboratory, Wuhan 430061, China; School of Laboratory Medicine, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Xianglin Chen
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, China; Hubei Shizhen Laboratory, Wuhan 430061, China; School of Laboratory Medicine, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Biran Zhu
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, China; Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; Hubei Shizhen Laboratory, Wuhan 430061, China
| | - Min Zhao
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, China; Hubei Shizhen Laboratory, Wuhan 430061, China
| | - Kaiyu Fu
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Yindan Zhang
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Huijia Tang
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; School of Environmental Studies, China University of Geosciences, Wuhan 430074, China
| | - Hao Pang
- School of Chemistry and Environmental Engineering, Wuhan Institute of Technology, Wuhan 430205, China
| | - Yongyong Guo
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Jian Han
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Lihua Yang
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.
| | - Bingsheng Zhou
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| |
Collapse
|
5
|
Shimochi S, Ihalainen J, Parikka V, Kudomi N, Tolvanen T, Hietanen A, Kokkomäki E, Johansson S, Tsuji M, Kanaya S, Yatkin E, Grönroos TJ, Iida H. Small animal PET with spontaneous inhalation of 15O-labelled oxygen gases: Longitudinal assessment of cerebral oxygen metabolism in a rat model of neonatal hypoxic-ischaemic encephalopathy. J Cereb Blood Flow Metab 2024; 44:1024-1038. [PMID: 38112197 PMCID: PMC11318403 DOI: 10.1177/0271678x231220691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 10/05/2023] [Accepted: 11/12/2023] [Indexed: 12/21/2023]
Abstract
Perinatal hypoxic-ischaemic encephalopathy (HIE) is the leading cause of irreversible brain damage resulting in serious neurological dysfunction among neonates. We evaluated the feasibility of positron emission tomography (PET) methodology with 15O-labelled gases without intravenous or tracheal cannulation for assessing temporal changes in cerebral blood flow (CBF) and cerebral metabolic rate for oxygen (CMRO2) in a neonatal HIE rat model. Sequential PET scans with spontaneous inhalation of 15O-gases mixed with isoflurane were performed over 14 days after the hypoxic-ischaemic insult in HIE pups and age-matched controls. CBF and CMRO2 in the injured hemispheres of HIE pups remarkably decreased 2 days after the insult, gradually recovering over 14 days in line with their increase found in healthy controls according to their natural maturation process. The magnitude of hemispheric tissue loss histologically measured after the last PET scan was significantly correlated with the decreases in CBF and CMRO2.This fully non-invasive imaging strategy may be useful for monitoring damage progression in neonatal HIE and for evaluating potential therapeutic outcomes.
Collapse
Affiliation(s)
- Saeka Shimochi
- Turku PET Centre, University of Turku, Turku, Finland
- MediCity Research Laboratory, University of Turku, Turku, Finland
- Nara Institute of Science and Technology, Ikoma City, Japan
| | - Jukka Ihalainen
- Turku PET Centre, University of Turku, Turku, Finland
- Department of Medical Physics, Turku University Hospital, Turku, Finland
- Accelerator Laboratory, Turku PET Centre, Åbo Akademi University, Turku, Finland
| | - Vilhelmiina Parikka
- Turku PET Centre, University of Turku, Turku, Finland
- MediCity Research Laboratory, University of Turku, Turku, Finland
- Department of Pediatrics and Adolescent Medicine, Turku University Hospital, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Nobuyuki Kudomi
- Department of Medical Physics, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Tuula Tolvanen
- Turku PET Centre, University of Turku, Turku, Finland
- Department of Medical Physics, Turku University Hospital, Turku, Finland
| | - Ari Hietanen
- Turku PET Centre, University of Turku, Turku, Finland
| | - Esa Kokkomäki
- Turku PET Centre, University of Turku, Turku, Finland
| | - Stefan Johansson
- Accelerator Laboratory, Turku PET Centre, Åbo Akademi University, Turku, Finland
| | - Masahiro Tsuji
- Department of Food and Nutrition, Kyoto Women's University, Kyoto, Japan
| | | | - Emrah Yatkin
- Central Animal Laboratory, University of Turku, Turku, Finland
| | - Tove J Grönroos
- Turku PET Centre, University of Turku, Turku, Finland
- MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Hidehiro Iida
- Turku PET Centre, University of Turku, Turku, Finland
- Nara Institute of Science and Technology, Ikoma City, Japan
| |
Collapse
|
6
|
Rae CD, Baur JA, Borges K, Dienel G, Díaz-García CM, Douglass SR, Drew K, Duarte JMN, Duran J, Kann O, Kristian T, Lee-Liu D, Lindquist BE, McNay EC, Robinson MB, Rothman DL, Rowlands BD, Ryan TA, Scafidi J, Scafidi S, Shuttleworth CW, Swanson RA, Uruk G, Vardjan N, Zorec R, McKenna MC. Brain energy metabolism: A roadmap for future research. J Neurochem 2024; 168:910-954. [PMID: 38183680 PMCID: PMC11102343 DOI: 10.1111/jnc.16032] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 11/29/2023] [Accepted: 12/05/2023] [Indexed: 01/08/2024]
Abstract
Although we have learned much about how the brain fuels its functions over the last decades, there remains much still to discover in an organ that is so complex. This article lays out major gaps in our knowledge of interrelationships between brain metabolism and brain function, including biochemical, cellular, and subcellular aspects of functional metabolism and its imaging in adult brain, as well as during development, aging, and disease. The focus is on unknowns in metabolism of major brain substrates and associated transporters, the roles of insulin and of lipid droplets, the emerging role of metabolism in microglia, mysteries about the major brain cofactor and signaling molecule NAD+, as well as unsolved problems underlying brain metabolism in pathologies such as traumatic brain injury, epilepsy, and metabolic downregulation during hibernation. It describes our current level of understanding of these facets of brain energy metabolism as well as a roadmap for future research.
Collapse
Affiliation(s)
- Caroline D. Rae
- School of Psychology, The University of New South Wales, NSW 2052 & Neuroscience Research Australia, Randwick, New South Wales, Australia
| | - Joseph A. Baur
- Department of Physiology and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Karin Borges
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, QLD, Australia
| | - Gerald Dienel
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Department of Cell Biology and Physiology, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Carlos Manlio Díaz-García
- Department of Biochemistry and Molecular Biology, Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | | | - Kelly Drew
- Center for Transformative Research in Metabolism, Institute of Arctic Biology, University of Alaska Fairbanks, Fairbanks, Alaska, USA
| | - João M. N. Duarte
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, & Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Jordi Duran
- Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL), Barcelona, Spain
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Oliver Kann
- Institute of Physiology and Pathophysiology, University of Heidelberg, D-69120; Interdisciplinary Center for Neurosciences (IZN), University of Heidelberg, Heidelberg, Germany
| | - Tibor Kristian
- Veterans Affairs Maryland Health Center System, Baltimore, Maryland, USA
- Department of Anesthesiology and the Center for Shock, Trauma, and Anesthesiology Research (S.T.A.R.), University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Dasfne Lee-Liu
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Región Metropolitana, Chile
| | - Britta E. Lindquist
- Department of Neurology, Division of Neurocritical Care, Gladstone Institute of Neurological Disease, University of California at San Francisco, San Francisco, California, USA
| | - Ewan C. McNay
- Behavioral Neuroscience, University at Albany, Albany, New York, USA
| | - Michael B. Robinson
- Departments of Pediatrics and System Pharmacology & Translational Therapeutics, Children’s Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Douglas L. Rothman
- Magnetic Resonance Research Center and Departments of Radiology and Biomedical Engineering, Yale University, New Haven, Connecticut, USA
| | - Benjamin D. Rowlands
- School of Chemistry, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Timothy A. Ryan
- Department of Biochemistry, Weill Cornell Medicine, New York, New York, USA
| | - Joseph Scafidi
- Department of Neurology, Kennedy Krieger Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Susanna Scafidi
- Anesthesiology & Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - C. William Shuttleworth
- Department of Neurosciences, University of New Mexico School of Medicine Albuquerque, Albuquerque, New Mexico, USA
| | - Raymond A. Swanson
- Department of Neurology, University of California, San Francisco, and San Francisco Veterans Affairs Medical Center, San Francisco, California, USA
| | - Gökhan Uruk
- Department of Neurology, University of California, San Francisco, and San Francisco Veterans Affairs Medical Center, San Francisco, California, USA
| | - Nina Vardjan
- Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia
- Laboratory of Neuroendocrinology—Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Robert Zorec
- Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia
- Laboratory of Neuroendocrinology—Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Mary C. McKenna
- Department of Pediatrics and Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
7
|
Molloy JW, Barry D. The interplay between glucose and ketone bodies in neural stem cell metabolism. J Neurosci Res 2024; 102:e25342. [PMID: 38773878 DOI: 10.1002/jnr.25342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 04/29/2024] [Accepted: 05/05/2024] [Indexed: 05/24/2024]
Abstract
Glucose is the primary energy source for neural stem cells (NSCs), supporting their proliferation, differentiation, and quiescence. However, the high demand for glucose during brain development often exceeds its supply, leading to the utilization of alternative energy sources including ketone bodies. Ketone bodies, including β-hydroxybutyrate, are short-chain fatty acids produced through hepatic ketogenesis and play a crucial role in providing energy and the biosynthetic components for NSCs when required. The interplay between glucose and ketone metabolism influences NSC behavior and fate decisions, and disruptions in these metabolic pathways have been linked to neurodevelopmental, neuropsychiatric, and neurodegenerative disorders. Additionally, ketone bodies exert neuroprotective effects on NSCs and modulate cellular responses to oxidative stress, energy maintenance, deacetylation, and inflammation. As such, understanding the interdependence of glucose and ketone metabolism in NSCs is crucial to understanding their roles in NSC function and their implications for neurological conditions. This article reviews the mechanisms of glucose and ketone utilization in NSCs, their impact on NSC function, and the therapeutic potential of targeting these metabolic pathways in neurological disorders.
Collapse
Affiliation(s)
- Joseph W Molloy
- Discipline of Anatomy, School of Medicine, Trinity Biomedical Sciences Institute (TBSI), Trinity College Dublin, Dublin, Ireland
| | - Denis Barry
- Discipline of Anatomy, School of Medicine, Trinity Biomedical Sciences Institute (TBSI), Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
8
|
Rao RB. Biomarkers of Brain Dysfunction in Perinatal Iron Deficiency. Nutrients 2024; 16:1092. [PMID: 38613125 PMCID: PMC11013337 DOI: 10.3390/nu16071092] [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: 02/13/2024] [Revised: 03/28/2024] [Accepted: 04/04/2024] [Indexed: 04/14/2024] Open
Abstract
Iron deficiency in the fetal and neonatal period (perinatal iron deficiency) bodes poorly for neurodevelopment. Given its common occurrence and the negative impact on brain development, a screening and treatment strategy that is focused on optimizing brain development in perinatal iron deficiency is necessary. Pediatric societies currently recommend a universal iron supplementation strategy for full-term and preterm infants that does not consider individual variation in body iron status and thus could lead to undertreatment or overtreatment. Moreover, the focus is on hematological normalcy and not optimal brain development. Several serum iron indices and hematological parameters in the perinatal period are associated with a risk of abnormal neurodevelopment, suggesting their potential use as biomarkers for screening and monitoring treatment in infants at risk for perinatal iron deficiency. A biomarker-based screening and treatment strategy that is focused on optimizing brain development will likely improve outcomes in perinatal iron deficiency.
Collapse
Affiliation(s)
- Raghavendra B. Rao
- Division of Neonatology, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN 55455, USA;
- Masonic Institute for the Developing Brain, Minneapolis, MN 55414, USA
| |
Collapse
|
9
|
Uittenbogaard M, Gropman AL, Whitehead MT, Brantner CA, Gropman E, Chiaramello A. Dysfunctional Postnatal Mitochondrial Energy Metabolism in a Patient with Neurodevelopmental Defects Caused by Intrauterine Growth Restriction Due to Idiopathic Placental Insufficiency. Int J Mol Sci 2024; 25:1386. [PMID: 38338665 PMCID: PMC10855472 DOI: 10.3390/ijms25031386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/16/2024] [Accepted: 01/20/2024] [Indexed: 02/12/2024] Open
Abstract
We report the case of a four-year-old male patient with a complex medical history born prematurely as the result of intrauterine growth restriction due to placental insufficiency. His clinical manifestations included severe neurodevelopmental deficits, global developmental delay, Pierre-Robin sequence, and intractable epilepsy with both generalized and focal features. The proband's low levels of citrulline and lactic acidosis provoked by administration of Depakoke were evocative of a mitochondrial etiology. The proband's genotype-phenotype correlation remained undefined in the absence of nuclear and mitochondrial pathogenic variants detected by deep sequencing of both genomes. However, live-cell mitochondrial metabolic investigations provided evidence of a deficient oxidative-phosphorylation pathway responsible for adenosine triphosphate (ATP) synthesis, leading to chronic energy crisis in the proband. In addition, our metabolic analysis revealed metabolic plasticity in favor of glycolysis for ATP synthesis. Our mitochondrial morphometric analysis by transmission electron microscopy confirmed the suspected mitochondrial etiology, as the proband's mitochondria exhibited an immature morphology with poorly developed and rare cristae. Thus, our results support the concept that suboptimal levels of intrauterine oxygen and nutrients alter fetal mitochondrial metabolic reprogramming toward oxidative phosphorylation (OXPHOS) leading to a deficient postnatal mitochondrial energy metabolism. In conclusion, our collective studies shed light on the long-term postnatal mitochondrial pathophysiology caused by intrauterine growth restriction due to idiopathic placental insufficiency and its negative impact on the energy-demanding development of the fetal and postnatal brain.
Collapse
Affiliation(s)
- Martine Uittenbogaard
- Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, George Washington University, 2300 I Street N.W., Washington, DC 20037, USA; (M.U.); (E.G.)
| | - Andrea L. Gropman
- Children’s National Medical Center, Division of Neurogenetics and Neurodevelopmental Pediatrics, Washington, DC 20010, USA;
| | - Matthew T. Whitehead
- Division on Neuroradiology, Department of Radiology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
| | - Christine A. Brantner
- Electron Microscopy Core Imaging Facility, School of Dentistry and School of Medicine, University of Maryland Baltimore, Baltimore, MD 21201, USA;
| | - Eliana Gropman
- Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, George Washington University, 2300 I Street N.W., Washington, DC 20037, USA; (M.U.); (E.G.)
| | - Anne Chiaramello
- Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, George Washington University, 2300 I Street N.W., Washington, DC 20037, USA; (M.U.); (E.G.)
| |
Collapse
|
10
|
Zhou T, Zhong Y, Zhang Y, Zhou Y. Pyruvate Dehydrogenase Complex in Neonatal Hypoxic-Ischemic Brain Injury. ACS Pharmacol Transl Sci 2024; 7:42-47. [PMID: 38230287 PMCID: PMC10789137 DOI: 10.1021/acsptsci.3c00191] [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: 08/17/2023] [Revised: 12/02/2023] [Accepted: 12/05/2023] [Indexed: 01/18/2024]
Abstract
The disruption of cerebral energy metabolism in relation to brain damage has been the subject of extensive research. However, the pyruvate dehydrogenase complex (PDHC), which is primarily characterized by poor cerebral energy metabolism following brain trauma, has received relatively little study in comparison to newborn hypoxic-ischemic brain injury. Mitochondrial PDHC, a multienzyme complex that functions as a crucial hub in energy metabolism and acts as a central metabolic node to mediate pyruvate oxidation after glycolysis and fuel the Krebs cycle to meet energy demands, has been reported to be one cause of energy metabolism dysfunction according to recent studies. Here we assess the potential mechanisms of neonatal hypoxic-ischemic brain injury-related brain dysfunction mediated by PDHC and further discuss the neuroprotective effects of therapeutic medicines that target PDHC activation. We also provide a summary of recent research on medicines that target PDHC in neonates with hypoxic-ischemic brain damage. Through an understanding of the mechanisms by which it is modulated and an investigation of the neuroprotective techniques available to activate brain PDHC and improve neonatal hypoxic-ischemic impairment, our review emphasizes the significance of PDHC impairment in neonatal hypoxic-ischemic brain injury.
Collapse
Affiliation(s)
- Tao Zhou
- Department
of Pharmaceutical and Medical Equipment, Rongtong Bayi Orthopedic Hospital of China, Chengdu 610031, China
| | - Yuangao Zhong
- Department
of Pharmaceutical Preparation Rongtong Bayi Orthopedic Hospital Of
China, Chengdu 610031, China
| | - Yong Zhang
- Department
of Pharmaceutical Preparation Rongtong Bayi Orthopedic Hospital Of
China, Chengdu 610031, China
| | - Yue Zhou
- Department
of Pharmacy, Xindu District People’s
Hospital of Chengdu, Chengdu 610500, China
| |
Collapse
|
11
|
Monko TR, Tripp EH, Burr SE, Gunderson KN, Lanier LM, Georgieff MK, Bastian TW. Cellular Iron Deficiency Disrupts Thyroid Hormone Regulated Gene Expression in Developing Hippocampal Neurons. J Nutr 2024; 154:49-59. [PMID: 37984740 PMCID: PMC10808837 DOI: 10.1016/j.tjnut.2023.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/06/2023] [Accepted: 11/03/2023] [Indexed: 11/22/2023] Open
Abstract
BACKGROUND Developing neurons have high thyroid hormone and iron requirements to support their metabolically demanding growth. Early-life iron and thyroid-hormone deficiencies are prevalent and often coexist, and each independently increases risk of permanently impaired neurobehavioral function in children. Early-life dietary iron deficiency reduces thyroid-hormone concentrations and impairs thyroid hormone-responsive gene expression in the neonatal rat brain, but it is unclear whether the effect is cell-intrinsic. OBJECTIVES This study determined whether neuronal-specific iron deficiency alters thyroid hormone-regulated gene expression in developing neurons. METHODS Iron deficiency was induced in primary mouse embryonic hippocampal neuron cultures with the iron chelator deferoxamine (DFO) beginning at 3 d in vitro (DIV). At 11DIV and 18DIV, thyroid hormone-regulated gene messenger ribonucleic acid (mRNA)concentrations indexing thyroid hormone homeostasis (Hairless, mu-crystallin, Type II deiodinase, solute carrier family member 1c1, and solute carrier family member 16a2) and neurodevelopment (neurogranin, Parvalbumin, and Krüppel-like factor 9) were quantified. To assess the effect of iron repletion, DFO was removed at 14DIV from a subset of DFO-treated cultures, and gene expression and adenosine 5'-triphosphate (ATP) concentrations were quantified at 21DIV. RESULTS At 11DIV and 18DIV, neuronal iron deficiency decreased neurogranin, Parvalbumin, and mu-crystallin, and by 18DIV, solute carrier family member 16a2, solute carrier family member 1c1, Type II deiodinase, and Hairless were increased, suggesting cellular sensing of a functionally abnormal thyroid hormone state. Dimensionality reduction with Principal component analysis reveals that thyroid hormone homeostatic genes strongly correlate with and predict iron status. Iron repletion from 14-21DIV did not restore ATP concentration, and Principal component analysis suggests that, after iron repletion, cultures maintain a gene expression signature indicative of previous iron deficiency. CONCLUSIONS These novel findings suggest there is an intracellular mechanism coordinating cellular iron/thyroid hormone activities. We speculate this is a part of the homeostatic response to acutely match neuronal energy production and growth signaling. However, the adaptation to iron deficiency may cause permanent deficits in thyroid hormone-dependent neurodevelopmental processes even after recovery from iron deficiency.
Collapse
Affiliation(s)
- Timothy R Monko
- Department of Pediatrics, School of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Emma H Tripp
- Department of Pediatrics, School of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Sierra E Burr
- Department of Pediatrics, School of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Karina N Gunderson
- Department of Pediatrics, School of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Lorene M Lanier
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota
| | - Michael K Georgieff
- Department of Pediatrics, School of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Thomas W Bastian
- Department of Pediatrics, School of Medicine, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
12
|
Su Z, Zhang G, Li X, Zhang H. Inverse correlation between Alzheimer's disease and cancer from the perspective of hypoxia. Neurobiol Aging 2023; 131:59-73. [PMID: 37572528 DOI: 10.1016/j.neurobiolaging.2023.07.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 06/02/2023] [Accepted: 07/03/2023] [Indexed: 08/14/2023]
Abstract
Sporadic Alzheimer's disease and cancer remain epidemiologically inversely related, and exploring the reverse pathogenesis is important for our understanding of both. Cognitive dysfunctions in Alzheimer's disease (AD) might result from the depletion of adaptive reserves in the brain. Energy storage in the brain is limited and is dynamically regulated by neurovascular and neurometabolic coupling. The research on neurodegenerative diseases has been dominated by the neurocentric view that neuronal defects cause the diseases. However, the proposal of the 2-hit vascular hypothesis in AD led us to focus on alterations in the vasculature, especially hypoperfusion. Chronic hypoxia is a feature shared by AD and cancer. It is interesting how contradicting chronic hypoxia's effects on both cancer and AD are. In this article, we discuss the potential links between the 2 diseases' etiology, from comparable upstream circumstances to diametrically opposed downstream effects. We suggest opposing potential mechanisms, including upregulation and downregulation of hypoxia-inducible factor-1α, the Warburg and reverse-Warburg effects, lactate-mediated intracellular acidic and alkaline conditions, and VDAC1-mediated apoptosis and antiapoptosis, and search for regulators that may be identified as the crossroads between cancer and AD.
Collapse
Affiliation(s)
- Zhan Su
- Department of Neurology and Neuroscience Centre, The First Hospital of Jilin University, Changchun, China
| | - Guimei Zhang
- Department of Neurology and Neuroscience Centre, The First Hospital of Jilin University, Changchun, China
| | - Xiangting Li
- Department of Neurology and Neuroscience Centre, The First Hospital of Jilin University, Changchun, China
| | - Haining Zhang
- Department of Neurology and Neuroscience Centre, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
13
|
Simicic D, Rackayova V, Braissant O, Toso C, Oldani G, Sessa D, McLin VA, Cudalbu C. Neurometabolic changes in a rat pup model of type C hepatic encephalopathy depend on age at liver disease onset. Metab Brain Dis 2023; 38:1999-2012. [PMID: 37148431 PMCID: PMC10348928 DOI: 10.1007/s11011-023-01210-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 04/02/2023] [Indexed: 05/08/2023]
Abstract
Chronic liver disease (CLD) is a serious condition where various toxins present in the blood affect the brain leading to type C hepatic encephalopathy (HE). Both adults and children are impacted, while children may display unique vulnerabilities depending on the affected window of brain development.We aimed to use the advantages of high field proton Magnetic Resonance Spectroscopy (1H MRS) to study longitudinally the neurometabolic and behavioural effects of Bile Duct Ligation (animal model of CLD-induced type C HE) on rats at post-natal day 15 (p15) to get closer to neonatal onset liver disease. Furthermore, we compared two sets of animals (p15 and p21-previously published) to evaluate whether the brain responds differently to CLD according to age onset.We showed for the first time that when CLD was acquired at p15, the rats presented the typical signs of CLD, i.e. rise in plasma bilirubin and ammonium, and developed the characteristic brain metabolic changes associated with type C HE (e.g. glutamine increase and osmolytes decrease). When compared to rats that acquired CLD at p21, p15 rats did not show any significant difference in plasma biochemistry, but displayed a delayed increase in brain glutamine and decrease in total-choline. The changes in neurotransmitters were milder than in p21 rats. Moreover, p15 rats showed an earlier increase in brain lactate and a different antioxidant response. These findings offer tentative pointers as to which neurodevelopmental processes may be impacted and raise the question of whether similar changes might exist in humans but are missed owing to 1H MRS methodological limitations in field strength of clinical magnet.
Collapse
Affiliation(s)
- Dunja Simicic
- CIBM Center for Biomedical Imaging, Lausanne, Switzerland.
- Animal Imaging and Technology, Ecole Polytechnique Federale de Lausanne (EPFL), Lausanne, Switzerland.
| | - Veronika Rackayova
- CIBM Center for Biomedical Imaging, Lausanne, Switzerland
- Animal Imaging and Technology, Ecole Polytechnique Federale de Lausanne (EPFL), Lausanne, Switzerland
| | - Olivier Braissant
- Service of Clinical Chemistry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Christian Toso
- Division of Abdominal and Transplantation Surgery, Department of Surgery, Faculty of Medicine, Geneva University Hospitals, Geneva, Switzerland
- Faculty of Medicine, Hepato-pancreato-biliary Centre, Geneva University Hospitals, Geneva, Switzerland
| | - Graziano Oldani
- Division of Abdominal and Transplantation Surgery, Department of Surgery, Faculty of Medicine, Geneva University Hospitals, Geneva, Switzerland
- Faculty of Medicine, Hepato-pancreato-biliary Centre, Geneva University Hospitals, Geneva, Switzerland
| | - Dario Sessa
- Swiss Pediatric Liver Center, Department of Pediatrics, Gynecology and Obstetrics, University of Geneva, Geneva, Switzerland
| | - Valérie A McLin
- Swiss Pediatric Liver Center, Department of Pediatrics, Gynecology and Obstetrics, University of Geneva, Geneva, Switzerland
| | - Cristina Cudalbu
- CIBM Center for Biomedical Imaging, Lausanne, Switzerland.
- Animal Imaging and Technology, Ecole Polytechnique Federale de Lausanne (EPFL), Lausanne, Switzerland.
| |
Collapse
|
14
|
Al-Kafaji G, Jahrami HA, Alwehaidah MS, Alshammari Y, Husni M. Mitochondrial DNA copy number in autism spectrum disorder and attention deficit hyperactivity disorder: a systematic review and meta-analysis. Front Psychiatry 2023; 14:1196035. [PMID: 37484684 PMCID: PMC10361772 DOI: 10.3389/fpsyt.2023.1196035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 06/21/2023] [Indexed: 07/25/2023] Open
Abstract
Background Several reports suggest that altered mitochondrial DNA copy number (mtDNA-cn), a common biomarker for aberrant mitochondrial function, is implicated in autism spectrum disorder (ASD) and attention deficit hyperactivity disorder (ADHD), but the results are still elusive. Methods A meta-analysis was performed to summarize the current indication and to provide a more precise assessment of the mtDNA-cn in ASD and ADHD. A search in the MEDLINE-PubMed, Scopus, and EMBASE databases was done to identify related studies up to the end of February 2023. The meta-analysis was conducted according to recommendations of the Cochrane Handbook of Systematic Reviews. Results Fourteen studies involving 666 cases with ASD and ADHD and 585 controls were collected and judged relevant for the systematic review and meta-analysis. The pooled results by a random effects meta-analysis was reported as a geometric mean of the estimated average response ratio and 95% confidence interval. Overall analysis of studies reported differences in mtDNA-cn in blood samples (k = 10) and non-blood samples (brain tissues and oral samples; k = 4) suggested significantly higher mtDNA-cn in patients compared to controls (p = 0.0275). Sub-analysis by stratifying studies based on tissue type, showed no significant increase in mtDNA-cn in blood samples among patients and controls (p = 0.284). Conversely, higher mtDNA-cn was observed in non-blood samples in patients than in controls (p = 0.0122). Further stratified analysis based on blood-cell compositions as potential confounds showed no significant difference in mtDNA-cn in peripheral blood samples of patients comparted to controls (p = 0.074). In addition, stratified analysis of aged-matched ASD and ADHD patients and controls revealed no significant difference in mtDNA-cn in blood samples between patients and controls (p = 0.214), whereas a significant increase in mtDNA-cn was observed in non-blood samples between patients and controls (p < 0.001). Finally, when the mtDNA-cn was analyzed in blood samples of aged-matched patients with ASD (peripheral blood, leukocytes, and PBMCs) or ADHD (peripheral blood), no significant difference in mtDNA-cn was observed between ASD patients and controls (p = 0.385), while a significant increase in mtDNA-cn was found between ADHD patients and controls (p = 0.033). Conclusion In this first meta-analysis of the evaluation of mtDNA-cn in ASD/ADHD, our results show elevated mtDNA-cn in ASD and ADHD, further emphasizing the implication of mitochondrial dysfunction in neurodevelopmental disorders. However, our results indicate that the mtDNA-cn in blood is not reflected in other tissues in ASD/ADHD, and the true relationship between blood-derived mtDNA-cn and ASD/ADHD remains to be defined in future studies. The importance of blood-cell compositions as confounders of blood-based mtDNA-cn measurement and the advantages of salivary mtDNA-cn should be considered in future studies. Moreover, the potential of mtDNA-cn as a biomarker for mitochondrial malfunction in neurodevelopmental disorders deserves further investigations.
Collapse
Affiliation(s)
- Ghada Al-Kafaji
- Department of Molecular Medicine and Al-Jawhara Centre for Molecular Medicine, Genetics, and Inherited Disorders, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
| | - Haitham Ali Jahrami
- Department of Psychiatry, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
- Government Hospital, Manama, Bahrain
| | - Materah Salem Alwehaidah
- Department of Medical Laboratory, Faculty of Allied Health, Kuwait University, Kuwait City, Kuwait
| | | | - Mariwan Husni
- Department of Psychiatry, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
- Department of Psychiatry, Northern Ontarion School of Medicine University, Thunder Bay, ON, Canada
| |
Collapse
|
15
|
Monko TR, Tripp EH, Burr SE, Gunderson KN, Lanier LM, Georgieff MK, Bastian TW. Cellular Iron Deficiency Disrupts Thyroid Hormone Regulated Gene Expression in Developing Hippocampal Neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.17.545408. [PMID: 37398002 PMCID: PMC10312787 DOI: 10.1101/2023.06.17.545408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Background Developing neurons have high thyroid hormone and iron requirements to support their metabolism and growth. Early-life iron and thyroid hormone deficiencies are prevalent, often coexist, and increase the risk of permanently impaired neurobehavioral function in children. Early-life dietary iron deficiency reduces thyroid hormone levels and impairs thyroid hormone-responsive gene expression in the neonatal rat brain. Objective This study determined whether neuronal-specific iron deficiency alters thyroid hormone-regulated gene expression in developing neurons. Methods Iron deficiency was induced in primary mouse embryonic hippocampal neuron cultures with the iron chelator deferoxamine (DFO) beginning at 3 days in vitro (DIV). At 11DIV and 18DIV, mRNA levels for thyroid hormone-regulated genes indexing thyroid hormone homeostasis (Hr, Crym, Dio2, Slco1c1, Slc16a2) and neurodevelopment (Nrgn, Pvalb, Klf9) were quantified. To assess the effect of iron repletion, DFO was removed at 14DIV from a subset of DFO-treated cultures and gene expression and ATP levels were quantified at 21DIV. Results At 11DIV and 18DIV, neuronal iron deficiency decreased Nrgn, Pvalb, and Crym, and by 18DIV, Slc16a2, Slco1c1, Dio2, and Hr were increased; collectively suggesting cellular sensing of a functionally abnormal thyroid hormone state. Dimensionality reduction with Principal Component Analysis (PCA) reveals that thyroid hormone homeostatic genes strongly correlate with and predict iron status (Tfr1 mRNA). Iron repletion from 14-21DIV restored neurodevelopmental genes, but not all thyroid hormone homeostatic genes, and ATP concentrations remained significantly altered. PCA clustering suggests that cultures replete with iron maintain a gene expression signature indicative of previous iron deficiency. Conclusions These novel findings suggest there is an intracellular mechanism coordinating cellular iron/thyroid hormone activities. We speculate this is a part of homeostatic response to match neuronal energy production and growth signaling for these important metabolic regulators. However, iron deficiency may cause permanent deficits in thyroid hormone-dependent neurodevelopmental processes even after recovery from iron deficiency.
Collapse
Affiliation(s)
- Timothy R Monko
- University of Minnesota, School of Medicine, Department of Pediatrics
| | - Emma H Tripp
- University of Minnesota, School of Medicine, Department of Pediatrics
| | - Sierra E Burr
- University of Minnesota, School of Medicine, Department of Pediatrics
| | | | | | | | - Thomas W Bastian
- University of Minnesota, School of Medicine, Department of Pediatrics
| |
Collapse
|
16
|
Satrom KM, Rao RB, Tkáč I. Neonatal hyperbilirubinemia differentially alters the neurochemical profiles of the developing cerebellum and hippocampus in a preterm Gunn rat model. NMR IN BIOMEDICINE 2023; 36:e4946. [PMID: 37009906 PMCID: PMC11809466 DOI: 10.1002/nbm.4946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 03/29/2023] [Accepted: 03/30/2023] [Indexed: 06/19/2023]
Abstract
Neonatal hyperbilirubinemia (NHB) can lead to brain injury in newborn infants by affecting specific regions including the cerebellum and hippocampus. Extremely preterm infants are more vulnerable to bilirubin neurotoxicity, but the mechanism and extent of injury is not well understood. A preterm version of the Gunn rat model was utilized to investigate severe preterm NHB. Homozygous/jaundiced Gunn rat pups were injected (i.p.) on postnatal day (P) 5 with sulfadimethoxine, which increases serum free bilirubin capable of crossing the blood-brain barrier and causing brain injury. The neurochemical profiles of the cerebellum and hippocampus were determined using in vivo 1 H MRS at 9.4 T on P30 and compared with those of heterozygous/non-jaundiced control rats. Transcript expression of related genes was determined by real-time quantitative PCR. MRI revealed significant morphological changes in the cerebellum of jaundiced rats. The concentrations of myo-inositol (+54%), glucose (+51%), N-acetylaspartylglutamate (+21%), and the sum of glycerophosphocholine and phosphocholine (+17%) were significantly higher in the cerebellum of the jaundiced group compared with the control group. Despite the lack of morphologic changes in the hippocampus, the concentration of myo-inositol (+9%) was higher and the concentrations of creatine (-8%) and of total creatine (-3%) were lower in the jaundiced group. In the hippocampus, expression of calcium/calmodulin dependent protein kinase II alpha (Camk2a), glucose transporter 1 (Glut1), and Glut3 transcripts were downregulated in the jaundiced group. In the cerebellum, glial fibrillary acidic protein (Gfap), myelin basic protein (Mbp), and Glut1 transcript expression was upregulated in the jaundiced group. These results indicate osmotic imbalance, gliosis, and changes in energy utilization and myelination, and demonstrate that preterm NHB critically affects brain development in a region-specific manner, with the cerebellum more severely impacted than the hippocampus.
Collapse
Affiliation(s)
- Katherine M Satrom
- Department of Pediatrics, Division of Neonatology, University of Minnesota, Minneapolis, MN, USA
| | - Raghavendra B Rao
- Department of Pediatrics, Division of Neonatology, University of Minnesota, Minneapolis, MN, USA
| | - Ivan Tkáč
- Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
17
|
Song Q, Gao Q, Chen T, Wen T, Wu P, Luo X, Chen QY. FAM3A Ameliorates Brain Impairment Induced by Hypoxia-Ischemia in Neonatal Rat. Cell Mol Neurobiol 2023; 43:251-264. [PMID: 34853925 PMCID: PMC9813043 DOI: 10.1007/s10571-021-01172-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 11/14/2021] [Indexed: 01/12/2023]
Abstract
Hypoxia-ischemia (HI) during crucial periods of brain formation can lead to changes in brain morphology, propagation of neuronal stimuli, and permanent neurodevelopmental impairment, which can have profound effects on cognitive function later in life. FAM3A, a subgroup of family with sequence similarity 3 (FAM3) gene family, is ubiquitously expressed in almost all cells. Overexpression of FAM3A has been evidenced to reduce hyperglycemia via the PI3K/Akt signaling pathway and protect mitochondrial function in neuronal HT22 cells. This study aims to evaluate the protective role of FAM3A in HI-induced brain impairment. Experimentally, maternal rats underwent uterine artery bilateral ligation to induce neonatal HI on day 14 of gestation. At 6 weeks of age, cognitive development assessments including NSS, wire grip, and water maze were carried out. The animals were then sacrificed to assess cerebral mitochondrial function as well as levels of FAM3A, TNF-α and IFN-γ. Results suggest that HI significantly reduced FAM3A expression in rat brain tissues, and that overexpression of FAM3A through lentiviral transduction effectively improved cognitive and motor functions in HI rats as reflected by improved NSS evaluation, cerebral water content, limb strength, as well as spatial learning and memory. At the molecular level, overexpression of FAM3A was able to promote ATP production, balance mitochondrial membrane potential, and reduce levels of pro-inflammatory cytokines TNF-α and IFN-γ. We conclude that FAM3A overexpression may have a protective effect on neuron morphology, cerebral mitochondrial as well as cognitive function. Created with Biorender.com.
Collapse
Affiliation(s)
- Qing Song
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China.
| | - Qingying Gao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China
- The Third Affiliated Hospital of Xi'an Medical University, Xi'an, 710049, Shaanxi, China
| | - Taotao Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China
| | - Ting Wen
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China
| | - Peng Wu
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China
| | - Xiao Luo
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, China.
- Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, China.
| | - Qiao Yi Chen
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China.
| |
Collapse
|
18
|
Kurki SN, Uvarov P, Pospelov AS, Trontti K, Hübner AK, Srinivasan R, Watanabe M, Hovatta I, Hübner CA, Kaila K, Virtanen MA. Expression patterns of NKCC1 in neurons and non-neuronal cells during cortico-hippocampal development. Cereb Cortex 2022; 33:5906-5923. [PMID: 36573432 PMCID: PMC10183754 DOI: 10.1093/cercor/bhac470] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/05/2022] [Accepted: 11/06/2022] [Indexed: 12/28/2022] Open
Abstract
Abstract
The Na-K-2Cl cotransporter NKCC1 is widely expressed in cells within and outside the brain. However, our understanding of its roles in brain functions throughout development, as well as in neuropsychiatric and neurological disorders, has been severely hindered by the lack of reliable data on its developmental and (sub)cellular expression patterns. We provide here the first properly controlled analysis of NKCC1 protein expression in various cell types of the mouse brain using custom-made antibodies and an NKCC1 knock-out validated immunohistochemical procedure, with parallel data based on advanced mRNA approaches. NKCC1 protein and mRNA are expressed at remarkably high levels in oligodendrocytes. In immature neurons, NKCC1 protein was located in the somata, whereas in adult neurons, only NKCC1 mRNA could be clearly detected. NKCC1 immunoreactivity is also seen in microglia, astrocytes, developing pericytes, and in progenitor cells of the dentate gyrus. Finally, a differential expression of NKCC1 splice variants was observed, with NKCC1a predominating in non-neuronal cells and NKCC1b in neurons. Taken together, our data provide a cellular basis for understanding NKCC1 functions in the brain and enable the identification of major limitations and promises in the development of neuron-targeting NKCC1-blockers.
Collapse
Affiliation(s)
- Samu N Kurki
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| | - Pavel Uvarov
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| | - Alexey S Pospelov
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| | - Kalevi Trontti
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
- University of Helsinki SleepWell Research Program, Faculty of Medicine, , 00014 Helsinki , Finland
- University of Helsinki Department of Psychology and Logopedics, , 00014 Helsinki , Finland
| | - Antje K Hübner
- Jena University Hospital, Friedrich Schiller Universität Institute of Human Genetics, , 07747 Jena , Germany
| | - Rakenduvadhana Srinivasan
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| | - Masahiko Watanabe
- Hokkaido University Department of Anatomy, Faculty of Medicine, , Sapporo 060–8638 , Japan
| | - Iiris Hovatta
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
- University of Helsinki SleepWell Research Program, Faculty of Medicine, , 00014 Helsinki , Finland
- University of Helsinki Department of Psychology and Logopedics, , 00014 Helsinki , Finland
| | - Christian A Hübner
- Jena University Hospital, Friedrich Schiller Universität Institute of Human Genetics, , 07747 Jena , Germany
| | - Kai Kaila
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| | - Mari A Virtanen
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| |
Collapse
|
19
|
Mohammadi A, Higazy R, Gauda EB. PGC-1α activity and mitochondrial dysfunction in preterm infants. Front Physiol 2022; 13:997619. [PMID: 36225305 PMCID: PMC9548560 DOI: 10.3389/fphys.2022.997619] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/09/2022] [Indexed: 11/26/2022] Open
Abstract
Extremely low gestational age neonates (ELGANs) are born in a relatively hyperoxic environment with weak antioxidant defenses, placing them at high risk for mitochondrial dysfunction affecting multiple organ systems including the nervous, respiratory, ocular, and gastrointestinal systems. The brain and lungs are highly affected by mitochondrial dysfunction and dysregulation in the neonate, causing white matter injury (WMI) and bronchopulmonary dysplasia (BPD), respectively. Adequate mitochondrial function is important in providing sufficient energy for organ development as it relates to alveolarization and axonal myelination and decreasing oxidative stress via reactive oxygen species (ROS) and reactive nitrogen species (RNS) detoxification. Peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) is a master regulator of mitochondrial biogenesis and function. Since mitochondrial dysfunction is at the root of WMI and BPD pathobiology, exploring therapies that can regulate PGC-1α activity may be beneficial. This review article describes several promising therapeutic agents that can mitigate mitochondrial dysfunction through direct and indirect activation and upregulation of the PGC-1α pathway. Metformin, resveratrol, omega 3 fatty acids, montelukast, L-citrulline, and adiponectin are promising candidates that require further pre-clinical and clinical studies to understand their efficacy in decreasing the burden of disease from WMI and BPD in preterm infants.
Collapse
Affiliation(s)
- Atefeh Mohammadi
- The Hospital for Sick Children, Division of Neonatology, Department of Pediatrics and Translational Medicine Program, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Randa Higazy
- The Hospital for Sick Children, Division of Neonatology, Department of Pediatrics and Translational Medicine Program, Toronto, ON, Canada
| | - Estelle B. Gauda
- The Hospital for Sick Children, Division of Neonatology, Department of Pediatrics and Translational Medicine Program, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
20
|
Rodriguez GG, Yu Z, O′Donnell LF, Calderon L, Cloos MA, Madelin G. Repeatability of simultaneous 3D 1H MRF/ 23Na MRI in brain at 7 T. Sci Rep 2022; 12:14156. [PMID: 35986071 PMCID: PMC9391473 DOI: 10.1038/s41598-022-18388-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 08/10/2022] [Indexed: 11/21/2022] Open
Abstract
Proton MRI can provide detailed morphological images, but it reveals little information about cell homeostasis. On the other hand, sodium MRI can provide metabolic information but cannot resolve fine structures. The complementary nature of proton and sodium MRI raises the prospect of their combined use in a single experiment. In this work, we assessed the repeatability of normalized proton density (PD), T1, T2, and normalized sodium density-weighted quantification measured with simultaneous 3D 1H MRF/23Na MRI in the brain at 7 T, from ten healthy volunteers who were scanned three times each. The coefficients of variation (CV) and the intra-class correlation (ICC) were calculated for the mean and standard deviation (SD) of these 4 parameters in grey matter, white matter, and cerebrospinal fluid. As result, the CVs were lower than 3.3% for the mean values and lower than 6.9% for the SD values. The ICCs were higher than 0.61 in all 24 measurements. We conclude that the measurements of normalized PD, T1, T2, and normalized sodium density-weighted from simultaneous 3D 1H MRF/23Na MRI in the brain at 7 T showed high repeatability. We estimate that changes > 6.6% (> 2 CVs) in mean values of both 1H and 23Na metrics could be detectable with this method.
Collapse
Affiliation(s)
- Gonzalo G. Rodriguez
- grid.137628.90000 0004 1936 8753Center for Biomedical Imaging, Department of Radiology, New York University Grossman School Medicine, 660 1st Avenue, 4th floor, New York, NY 10016 USA
| | - Zidan Yu
- grid.137628.90000 0004 1936 8753Center for Biomedical Imaging, Department of Radiology, New York University Grossman School Medicine, 660 1st Avenue, 4th floor, New York, NY 10016 USA ,grid.240324.30000 0001 2109 4251Vilcek Institute of Graduate Biomedical Sciences, NYU Langone Health, New York, NY 10016 USA
| | - Lauren F. O′Donnell
- grid.137628.90000 0004 1936 8753Center for Biomedical Imaging, Department of Radiology, New York University Grossman School Medicine, 660 1st Avenue, 4th floor, New York, NY 10016 USA
| | - Liz Calderon
- grid.137628.90000 0004 1936 8753Center for Biomedical Imaging, Department of Radiology, New York University Grossman School Medicine, 660 1st Avenue, 4th floor, New York, NY 10016 USA
| | - Martijn A. Cloos
- grid.1003.20000 0000 9320 7537Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD Australia ,grid.1003.20000 0000 9320 7537ARC Training Centre for Innovation in Biomedical Imaging Technology, The University of Queensland, Brisbane, QLD Australia
| | - Guillaume Madelin
- Center for Biomedical Imaging, Department of Radiology, New York University Grossman School Medicine, 660 1st Avenue, 4th floor, New York, NY, 10016, USA. .,Vilcek Institute of Graduate Biomedical Sciences, NYU Langone Health, New York, NY, 10016, USA.
| |
Collapse
|
21
|
Stanescu S, Bravo-Alonso I, Belanger-Quintana A, Pérez B, Medina-Diaz M, Ruiz-Sala P, Flores NP, Buenache R, Arrieta F, Rodríguez-Pombo P. Mitochondrial bioenergetic is impaired in Monocarboxylate transporter 1 deficiency: a new clinical case and review of the literature. Orphanet J Rare Dis 2022; 17:243. [PMID: 35729663 PMCID: PMC9215049 DOI: 10.1186/s13023-022-02389-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 06/06/2022] [Indexed: 12/03/2022] Open
Abstract
Background Monocarboxylate transporter 1 (MCT1) deficiency has recently been described as a rare cause of recurrent ketosis, the result of impaired ketone utilization in extrahepatic tissues. To date, only six patients with this condition have been identified, and clinical and biochemical details remain incomplete. Results The present work reports a patient suffering from severe, recurrent episodes of metabolic acidosis and psychomotor delay, showing a pathogenic loss-of-function variation c.747_750del in homozygosity in SLC16A1 (which codes for MCT1). Persistent ketotic and lactic acidosis was accompanied by an abnormal excretion of organic acids related to redox balance disturbances. Together with an altered bioenergetic profile detected in patient-derived fibroblasts, this suggests possible mitochondrial dysfunction. Brain MRI revealed extensive, diffuse bilateral, symmetric signal alterations for the subcortical white matter and basal ganglia, together with corpus callosum agenesia. Conclusions These findings suggest that the clinical spectrum of MCT1 deficiency not only involves recurrent atacks of ketoacidosis, but may also cause lactic acidosis and neuromotor delay with a distinctive neuroimaging pattern including agenesis of corpus callosum and other brain signal alterations. Supplementary Information The online version contains supplementary material available at 10.1186/s13023-022-02389-4.
Collapse
Affiliation(s)
- Sinziana Stanescu
- Unidad de Enfermedades Metabólicas, Hospital Universitario Ramón y Cajal, IRYCIS, Crta de Colmenar Viejo, km 9,100, 28034, Madrid, Spain.
| | - Irene Bravo-Alonso
- Centro de Diagnóstico de Enfermedades Moleculares, Centro de Biología Molecular Severo Ochoa, UAM-CSIC, CIBERER, IdiPAZ, C/Francisco Tomás y Valiente, 7, 28049, Madrid, Spain
| | - Amaya Belanger-Quintana
- Unidad de Enfermedades Metabólicas, Hospital Universitario Ramón y Cajal, IRYCIS, Crta de Colmenar Viejo, km 9,100, 28034, Madrid, Spain
| | - Belen Pérez
- Centro de Diagnóstico de Enfermedades Moleculares, Centro de Biología Molecular Severo Ochoa, UAM-CSIC, CIBERER, IdiPAZ, C/Francisco Tomás y Valiente, 7, 28049, Madrid, Spain
| | - Montserrat Medina-Diaz
- Department of Neuroradiology, Hospital Universitario Ramón y Cajal, IRYCIS, Crta de Colmenar Viejo, km 9,100, 28034, Madrid, Spain
| | - Pedro Ruiz-Sala
- Centro de Diagnóstico de Enfermedades Moleculares, Centro de Biología Molecular, Universidad Autónoma de Madrid, CIBERER, IdiPAZ, C/Francisco Tomás y Valiente, 7, 28049, Madrid, Spain
| | - Nathaly Paola Flores
- Paediatric Department, Hospital General La Mancha Centro, Av. Constitución, 3, 13600, Alcázar de San Juan, Ciudad Real, Spain
| | - Raquel Buenache
- Neuropediatric Department, Hospital Universitario Ramón y Cajal, IRYCIS, Crta de Colmenar Viejo, km 9,100, 28034, Madrid, Spain
| | - Francisco Arrieta
- Unidad de Enfermedades Metabólicas, Hospital Universitario Ramón y Cajal, IRYCIS, CIBER-OBN, Crta de Colmenar Viejo, km 9,100, 28034, Madrid, Spain
| | - Pilar Rodríguez-Pombo
- Centro de Diagnóstico de Enfermedades Moleculares, Centro de Biología Molecular Severo Ochoa, UAM-CSIC, CIBERER, IdiPAZ, C/Francisco Tomás y Valiente, 7, 28049, Madrid, Spain
| |
Collapse
|
22
|
Yu Z, Hodono S, Dergachyova O, Hilbert T, Wang B, Zhang B, Brown R, Sodickson DK, Madelin G, Cloos MA. Simultaneous 3D acquisition of 1 H MRF and 23 Na MRI. Magn Reson Med 2022; 87:2299-2312. [PMID: 34971454 PMCID: PMC8847332 DOI: 10.1002/mrm.29135] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 11/23/2021] [Accepted: 12/10/2021] [Indexed: 11/08/2022]
Abstract
PURPOSE To develop a 3D MR technique to simultaneously acquire proton multiparametric maps (T1 , T2 , and proton density) and sodium density weighted images over the whole brain. METHODS We implemented a 3D stack-of-stars MR pulse sequence which consists of interleaved proton (1 H) and sodium (23 Na) excitations, tailored slice encoding gradients that can encode the same slice for both nuclei, and simultaneous readout with different radial trajectories (1 H, full-radial; 23 Na, center-out radial). The receive chain of our 7T scanner was modified to enable simultaneous acquisition of 1 H and 23 Na signal. A heuristically optimized flip angle train was implemented for proton MR fingerprinting (MRF). The SNR and the accuracy of proton T1 and T2 were evaluated in phantoms. Finally, in vivo application of the method was demonstrated in five healthy subjects. RESULTS The SNR for the simultaneous measurement was almost identical to that for the single-nucleus measurements (<2% change). The proton T1 and T2 maps remained similar to the results from a reference 2D MRF technique (normalized RMS error in T1 ≈ 4.2% and T2 ≈ 11.3%). Measurements in healthy subjects corroborated these results and demonstrated the feasibility of our method for in vivo application. The in vivo T1 values measured using our method were lower than the results measured by other conventional techniques. CONCLUSIONS With the 3D simultaneous implementation, we were able to acquire sodium and proton density weighted images in addition to proton T1 , T2 , and B1+ from 1 H MRF that covers the whole brain volume within 21 min.
Collapse
Affiliation(s)
- Zidan Yu
- Center for Advanced Imaging Innovation and Research (CAI2R), Department of Radiology, New York University Grossman School of Medicine, New York, NY 10016, USA,The Vilcek Institute of Graduate Biomedical Sciences, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Shota Hodono
- Center for Advanced Imaging Innovation and Research (CAI2R), Department of Radiology, New York University Grossman School of Medicine, New York, NY 10016, USA,The Vilcek Institute of Graduate Biomedical Sciences, New York University Grossman School of Medicine, New York, NY 10016, USA,The Centre for Advanced Imaging, The University of Queensland, St Lucia, QLD, Australia
| | - Olga Dergachyova
- Center for Advanced Imaging Innovation and Research (CAI2R), Department of Radiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Tom Hilbert
- Advanced Clinical Imaging Technology, Siemens Healthcare AG, Lausanne, Switzerland,Department of Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland,LTS5, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Bili Wang
- Center for Advanced Imaging Innovation and Research (CAI2R), Department of Radiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Bei Zhang
- Center for Advanced Imaging Innovation and Research (CAI2R), Department of Radiology, New York University Grossman School of Medicine, New York, NY 10016, USA,Advanced Imaging Research Center, UT Southwestern Medical Center, Dallas, TX, USA
| | - Ryan Brown
- Center for Advanced Imaging Innovation and Research (CAI2R), Department of Radiology, New York University Grossman School of Medicine, New York, NY 10016, USA,The Vilcek Institute of Graduate Biomedical Sciences, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Daniel K. Sodickson
- Center for Advanced Imaging Innovation and Research (CAI2R), Department of Radiology, New York University Grossman School of Medicine, New York, NY 10016, USA,The Vilcek Institute of Graduate Biomedical Sciences, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Guillaume Madelin
- Center for Advanced Imaging Innovation and Research (CAI2R), Department of Radiology, New York University Grossman School of Medicine, New York, NY 10016, USA,The Vilcek Institute of Graduate Biomedical Sciences, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Martijn A. Cloos
- Center for Advanced Imaging Innovation and Research (CAI2R), Department of Radiology, New York University Grossman School of Medicine, New York, NY 10016, USA,The Vilcek Institute of Graduate Biomedical Sciences, New York University Grossman School of Medicine, New York, NY 10016, USA,The Centre for Advanced Imaging, The University of Queensland, St Lucia, QLD, Australia
| |
Collapse
|
23
|
Liu X, Shi L, Shi L, Wei M, Zhao Z, Min W. Towards Mapping Mouse Metabolic Tissue Atlas by Mid-Infrared Imaging with Heavy Water Labeling. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105437. [PMID: 35319171 PMCID: PMC9131428 DOI: 10.1002/advs.202105437] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/26/2022] [Indexed: 06/14/2023]
Abstract
Understanding metabolism is of great significance to decipher various physiological and pathogenic processes. While great progress has been made to profile gene expression, how to capture organ-, tissue-, and cell-type-specific metabolic profile (i.e., metabolic tissue atlas) in complex mammalian systems is lagging behind, largely owing to the lack of metabolic imaging tools with high resolution and high throughput. Here, the authors applied mid-infrared imaging coupled with heavy water (D2 O) metabolic labeling to a scope of mouse organs and tissues. The premise is that, as D2 O participates in the biosynthesis of various macromolecules, the resulting broad C-D vibrational spectrum should interrogate a wide range of metabolic pathways. Applying multivariate analysis to the C-D spectrum, the authors successfully identified both inter-organ and intra-tissue metabolic signatures of mice. A large-scale metabolic atlas map between different organs from the same mice is thus generated. Moreover, leveraging the power of unsupervised clustering methods, spatially-resolved metabolic signatures of brain tissues are discovered, revealing tissue and cell-type specific metabolic profile in situ. As a demonstration of this technique, the authors captured metabolic changes during brain development and characterized intratumoral metabolic heterogeneity of glioblastoma. Altogether, the integrated platform paves a way to map the metabolic tissue atlas for complex mammalian systems.
Collapse
Affiliation(s)
- Xinwen Liu
- Department of ChemistryColumbia UniversityNew YorkNY10027USA
| | - Lixue Shi
- Department of ChemistryColumbia UniversityNew YorkNY10027USA
| | - Lingyan Shi
- Department of ChemistryColumbia UniversityNew YorkNY10027USA
| | - Mian Wei
- Department of ChemistryColumbia UniversityNew YorkNY10027USA
| | - Zhilun Zhao
- Department of ChemistryColumbia UniversityNew YorkNY10027USA
| | - Wei Min
- Department of ChemistryColumbia UniversityNew YorkNY10027USA
| |
Collapse
|
24
|
Amaral AU, Wajner M. Pathophysiology of maple syrup urine disease: Focus on the neurotoxic role of the accumulated branched-chain amino acids and branched-chain α-keto acids. Neurochem Int 2022; 157:105360. [DOI: 10.1016/j.neuint.2022.105360] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 05/07/2022] [Accepted: 05/09/2022] [Indexed: 12/21/2022]
|
25
|
Sharanek A, Jahani-Asl A. Monitoring Mitochondrial Respiration in Mouse Cerebellar Granule Neurons. Methods Mol Biol 2022; 2515:1-15. [PMID: 35776342 DOI: 10.1007/978-1-0716-2409-8_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Defects in mitochondrial oxidative phosphorylation have been observed in numerous neurodegenerative disorders and are linked to bioenergetic crises leading to neuronal death. The distinct metabolic profile of neurons is predominantly oxidative, which is characterized by the oxidation of glucose or its metabolites in the mitochondria to produce ATP. This process involves the tricarboxylic acid cycle, electron transfer in the respiratory chain, and oxygen consumption. Therefore, measurement of oxygen consumption rates (OCR) can be accurately applied to assess the rate of mitochondrial respiration. In this chapter, we describe our optimized protocol for the assessment of OCR specifically in primary mouse cerebellar granule neurons (CGN). The protocol includes isolation and manipulation of mouse CGNs followed by real-time assessment of mitochondrial OCR using a Seahorse XFe96 extracellular flux analyzer.
Collapse
Affiliation(s)
- Ahmad Sharanek
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
- University of Ottawa Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada
| | - Arezu Jahani-Asl
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.
- University of Ottawa Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada.
- Division of Experimental Medicine , McGill University, Montreal, QC, Canada.
| |
Collapse
|
26
|
Chen X, Zheng J, Teng M, Zhang J, Qian L, Duan M, Cheng Y, Zhao W, Wang Z, Wang C. Tralopyril affects locomotor activity of zebrafish (Danio rerio) by impairing tail muscle tissue, the nervous system, and energy metabolism. CHEMOSPHERE 2022; 286:131866. [PMID: 34391112 DOI: 10.1016/j.chemosphere.2021.131866] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 08/08/2021] [Accepted: 08/09/2021] [Indexed: 06/13/2023]
Abstract
Tralopyril (TP), an antifouling biocide, is widely used to prevent heavy biofouling, and can have potential risks to aquatic organisms. In this study, the effect of TP on locomotor activity and related mechanisms were evaluated in zebrafish (Danio rerio) larvae. TP significantly reduced locomotor activity after 168 -h exposure. Adverse modifications in tail muscle tissue, the nervous system, and energy metabolism were also observed in larvae. TP caused thinning of the muscle bundle in the tail of larvae. In conjunction with the metabolomics results, changes in dopamine (DA) and acetylcholine (ACh), acetylcholinesterase (AChE) activity, and the expression of genes involved in neurodevelopment, indicate that TP may disrupt the nervous system in zebrafish larvae. The change in metabolites (e.g., glucose 6-phosphate, cis-Aconitic acid, acetoacetyl-CoA, coenzyme-A and 3-Oxohexanoyl-CoA) involved in carbohydrate and lipid metabolism indicates that TP may disrupt energy metabolism. TP exposure may inhibit the locomotor activity of zebrafish larvae by impairing tail muscle tissue, the nervous system, and energy metabolism.
Collapse
Affiliation(s)
- Xiangguang Chen
- Innovation Center of Pesticide Research, Department of Applied Chemistry, College of Sciences, China Agricultural University, Beijing, 100193, China
| | - Junyue Zheng
- Innovation Center of Pesticide Research, Department of Applied Chemistry, College of Sciences, China Agricultural University, Beijing, 100193, China
| | - Miaomiao Teng
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, 100012, China
| | - Jie Zhang
- Innovation Center of Pesticide Research, Department of Applied Chemistry, College of Sciences, China Agricultural University, Beijing, 100193, China
| | - Le Qian
- Innovation Center of Pesticide Research, Department of Applied Chemistry, College of Sciences, China Agricultural University, Beijing, 100193, China
| | - Manman Duan
- Innovation Center of Pesticide Research, Department of Applied Chemistry, College of Sciences, China Agricultural University, Beijing, 100193, China
| | - Yi Cheng
- Innovation Center of Pesticide Research, Department of Applied Chemistry, College of Sciences, China Agricultural University, Beijing, 100193, China
| | - Wentian Zhao
- Innovation Center of Pesticide Research, Department of Applied Chemistry, College of Sciences, China Agricultural University, Beijing, 100193, China
| | - Zhao Wang
- The Institute of Plant Production, Jilin Academy of Agriculture Science, Changchun, 130033, China
| | - Chengju Wang
- Innovation Center of Pesticide Research, Department of Applied Chemistry, College of Sciences, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
27
|
Head B, Traber MG. Expanding role of vitamin E in protection against metabolic dysregulation: Insights gained from model systems, especially the developing nervous system of zebrafish embryos. Free Radic Biol Med 2021; 176:80-91. [PMID: 34555455 DOI: 10.1016/j.freeradbiomed.2021.09.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/27/2021] [Accepted: 09/07/2021] [Indexed: 12/20/2022]
Abstract
This review discusses why the embryo requires vitamin E (VitE) and shows that its lack causes metabolic dysregulation and impacts morphological changes at very early stages in development, which occur prior to when a woman knows she is pregnant. VitE halts the chain reactions of lipid peroxidation (LPO). Metabolomic analyses indicate that thiols become depleted in E- embryos because LPO generates products that require compensation using limited amino acids and methyl donors that are also developmentally relevant. Thus, VitE protects metabolic networks and the integrated gene expression networks that control development. VitE is critical especially for neurodevelopment, which is dependent on trafficking by the α-tocopherol transfer protein (TTPa). VitE-deficient (E-) zebrafish embryos initially appear normal, but by 12 and 24 h post-fertilization (hpf) E- embryos are developmentally abnormal with expression of pax2a and sox10 mis-localized in the midbrain-hindbrain boundary, neural crest cells and throughout the spinal neurons. These patterning defects indicate cells that are especially in need of VitE-protection. They precede obvious morphological abnormalities (cranial-facial malformation, pericardial edema, yolksac edema, skewed body-axis) and impaired behavioral responses to locomotor activity tests. The TTPA gene (ttpa) is expressed at the leading edges of the brain ventricle border. Ttpa knockdown using morpholinos is 100% lethal by 24 hpf, while E- embryo brains are often over- or under-inflated at 24 hpf. Further, E- embryos prior to 24 hpf have increased expression of genes involved in glycolysis and the pentose phosphate pathway, and decreased expression of genes involved in anabolic pathways and transcription. Combined data from both gene expression and the metabolome in E- embryos at 24 hpf suggest that the activity of the mechanistic Target of Rapamycin (mTOR) signaling pathway is decreased, which may impact both metabolism and neurodevelopment. Further evaluation of VitE deficiency in neurogenesis and its subsequent impact on learning and behavior is needed.
Collapse
Affiliation(s)
- Brian Head
- Linus Pauling Institute, Corvallis, OR, USA; Molecular and Cell Biology Program, Corvallis, OR, USA
| | - Maret G Traber
- Linus Pauling Institute, Corvallis, OR, USA; School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, OR, USA.
| |
Collapse
|
28
|
Singh T, Jiao Y, Ferrando LM, Yablonska S, Li F, Horoszko EC, Lacomis D, Friedlander RM, Carlisle DL. Neuronal mitochondrial dysfunction in sporadic amyotrophic lateral sclerosis is developmentally regulated. Sci Rep 2021; 11:18916. [PMID: 34556702 PMCID: PMC8460779 DOI: 10.1038/s41598-021-97928-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 09/01/2021] [Indexed: 02/08/2023] Open
Abstract
Amyotrophic lateral sclerosis is an adult-onset neurodegenerative disorder characterized by loss of motor neurons. Mitochondria are essential for neuronal survival but the developmental timing and mechanistic importance of mitochondrial dysfunction in sporadic ALS (sALS) neurons is not fully understood. We used human induced pluripotent stem cells and generated a developmental timeline by differentiating sALS iPSCs to neural progenitors and to motor neurons and comparing mitochondrial parameters with familial ALS (fALS) and control cells at each developmental stage. We report that sALS and fALS motor neurons have elevated reactive oxygen species levels, depolarized mitochondria, impaired oxidative phosphorylation, ATP loss and defective mitochondrial protein import compared with control motor neurons. This phenotype develops with differentiation into motor neurons, the affected cell type in ALS, and does not occur in the parental undifferentiated sALS cells or sALS neural progenitors. Our work demonstrates a developmentally regulated unifying mitochondrial phenotype between patient derived sALS and fALS motor neurons. The occurrence of a unifying mitochondrial phenotype suggests that mitochondrial etiology known to SOD1-fALS may applicable to sALS. Furthermore, our findings suggest that disease-modifying treatments focused on rescue of mitochondrial function may benefit both sALS and fALS patients.
Collapse
Affiliation(s)
- Tanisha Singh
- grid.21925.3d0000 0004 1936 9000Neuroapoptosis Laboratory, Department of Neurological Surgery, University of Pittsburgh, B400 Presbyterian Hospital, 200 Lothrop Street, Pittsburgh, PA 15213 USA
| | - Yuanyuan Jiao
- grid.21925.3d0000 0004 1936 9000Neuroapoptosis Laboratory, Department of Neurological Surgery, University of Pittsburgh, B400 Presbyterian Hospital, 200 Lothrop Street, Pittsburgh, PA 15213 USA
| | - Lisa M. Ferrando
- grid.21925.3d0000 0004 1936 9000Neuroapoptosis Laboratory, Department of Neurological Surgery, University of Pittsburgh, B400 Presbyterian Hospital, 200 Lothrop Street, Pittsburgh, PA 15213 USA
| | - Svitlana Yablonska
- grid.21925.3d0000 0004 1936 9000Neuroapoptosis Laboratory, Department of Neurological Surgery, University of Pittsburgh, B400 Presbyterian Hospital, 200 Lothrop Street, Pittsburgh, PA 15213 USA
| | - Fang Li
- grid.21925.3d0000 0004 1936 9000Neuroapoptosis Laboratory, Department of Neurological Surgery, University of Pittsburgh, B400 Presbyterian Hospital, 200 Lothrop Street, Pittsburgh, PA 15213 USA
| | - Emily C. Horoszko
- grid.21925.3d0000 0004 1936 9000Neuroapoptosis Laboratory, Department of Neurological Surgery, University of Pittsburgh, B400 Presbyterian Hospital, 200 Lothrop Street, Pittsburgh, PA 15213 USA
| | - David Lacomis
- grid.21925.3d0000 0004 1936 9000Departments of Neurology and Pathology, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | - Robert M. Friedlander
- grid.21925.3d0000 0004 1936 9000Neuroapoptosis Laboratory, Department of Neurological Surgery, University of Pittsburgh, B400 Presbyterian Hospital, 200 Lothrop Street, Pittsburgh, PA 15213 USA
| | - Diane L. Carlisle
- grid.21925.3d0000 0004 1936 9000Neuroapoptosis Laboratory, Department of Neurological Surgery, University of Pittsburgh, B400 Presbyterian Hospital, 200 Lothrop Street, Pittsburgh, PA 15213 USA
| |
Collapse
|
29
|
Gailus B, Naundorf H, Welzel L, Johne M, Römermann K, Kaila K, Löscher W. Long-term outcome in a noninvasive rat model of birth asphyxia with neonatal seizures: Cognitive impairment, anxiety, epilepsy, and structural brain alterations. Epilepsia 2021; 62:2826-2844. [PMID: 34458992 DOI: 10.1111/epi.17050] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 07/30/2021] [Accepted: 08/09/2021] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Birth asphyxia is a major cause of hypoxic-ischemic encephalopathy (HIE) in neonates and often associated with mortality, neonatal seizures, brain damage, and later life motor, cognitive, and behavioral impairments and epilepsy. Preclinical studies on rodent models are needed to develop more effective therapies for preventing HIE and its consequences. Thus far, the most popular rodent models have used either exposure of intact animals to hypoxia-only, or a combination of hypoxia and carotid occlusion, for the induction of neonatal seizures and adverse outcomes. However, such models lack systemic hypercapnia, which is a fundamental constituent of birth asphyxia with major effects on neuronal excitability. Here, we use a recently developed noninvasive rat model of birth asphyxia with subsequent neonatal seizures to study later life adverse outcome. METHODS Intermittent asphyxia was induced for 30 min by exposing male and female postnatal day 11 rat pups to three 7 + 3-min cycles of 9% and 5% O2 at constant 20% CO2 . All pups exhibited convulsive seizures after asphyxia. A set of behavioral tests were performed systematically over 14 months following asphyxia, that is, a large part of the rat's life span. Video-electroencephalographic (EEG) monitoring was used to determine whether asphyxia led to the development of epilepsy. Finally, structural brain alterations were examined. RESULTS The animals showed impaired spatial learning and memory and increased anxiety when tested at an age of 3-14 months. Video-EEG at ~10 months showed an abundance of spontaneous seizures, which was paralleled by neurodegeneration in the hippocampus and thalamus, and by aberrant mossy fiber sprouting. SIGNIFICANCE The present model of birth asphyxia recapitulates several of the later life consequences associated with human HIE. This model thus allows evaluation of the efficacy of novel therapies designed to prevent HIE and seizures following asphyxia, and of how such therapies might alleviate long-term adverse consequences.
Collapse
Affiliation(s)
- Björn Gailus
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| | - Hannah Naundorf
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| | - Lisa Welzel
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany
| | - Marie Johne
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| | - Kerstin Römermann
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany
| | - Kai Kaila
- Molecular and Integrative Biosciences, University of Helsinki, Helsinki, Finland.,Neuroscience Center (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| |
Collapse
|
30
|
Mohamed RA, Abdallah DM, El-brairy AI, Ahmed KA, El-Abhar HS. Palonosetron/Methyllycaconitine Deactivate Hippocampal Microglia 1, Inflammasome Assembly and Pyroptosis to Enhance Cognition in a Novel Model of Neuroinflammation. Molecules 2021; 26:5068. [PMID: 34443654 PMCID: PMC8401912 DOI: 10.3390/molecules26165068] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/10/2021] [Accepted: 08/17/2021] [Indexed: 12/30/2022] Open
Abstract
Since westernized diet-induced insulin resistance is a risk factor in Alzheimer's disease (AD) development, and lipopolysaccharide (LPS) coexists with amyloid β (Aβ)1-42 in these patients, our AD novel model was developed to resemble sporadic AD by injecting LPS into high fat/fructose diet (HFFD)-fed rats. The neuroprotective potential of palonosetron and/or methyllycaconitine, 5-HT3 receptor and α7 nAChR blockers, respectively, was evaluated after 8 days of daily administration in HFFD/LPS rats. All regimens improved histopathological findings and enhanced spatial memory (Morris Water Maze); however, palonosetron alone or with methyllycaconitine promoted animal performance during novel object recognition tests. In the hippocampus, all regimens reduced the expression of glial fibrillary acidic protein and skewed microglia M1 to M2 phenotype, indicated by the decreased M1 markers and the enhanced M2 related parameters. Additionally, palonosetron and its combination regimen downregulated the expression of ASC/TMS1, as well as levels of inflammasome downstream molecules and abated cleaved caspase-1, interleukin (IL)-1β, IL-18 and caspase-11. Furthermore, ACh and 5-HT were augmented after being hampered by the insult. Our study speculates that blocking 5-HT3 receptor using palonosetron overrides methyllycaconitine to combat AD-induced neuroinflammation and inflammasome cascade, as well as to restore microglial function in a HFFD/LPS novel model for sporadic AD.
Collapse
Affiliation(s)
- Reem A. Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, October University for Modern Sciences and Arts, 26 July Mehwar Road Intersection with Wahat Road, 6th of October City, Giza 12451, Egypt; (R.A.M.); (A.I.E.-b.)
| | - Dalaal M. Abdallah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Κasr El-Aini Str., Cairo 11562, Egypt;
| | - Amany I. El-brairy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, October University for Modern Sciences and Arts, 26 July Mehwar Road Intersection with Wahat Road, 6th of October City, Giza 12451, Egypt; (R.A.M.); (A.I.E.-b.)
| | - Kawkab A. Ahmed
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt;
| | - Hanan S. El-Abhar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Κasr El-Aini Str., Cairo 11562, Egypt;
| |
Collapse
|
31
|
Tengeler AC, Emmerzaal TL, Geenen B, Verweij V, van Bodegom M, Morava E, Kiliaan AJ, Kozicz T. Early-adolescent antibiotic exposure results in mitochondrial and behavioral deficits in adult male mice. Sci Rep 2021; 11:12875. [PMID: 34145328 PMCID: PMC8213690 DOI: 10.1038/s41598-021-92203-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 05/21/2021] [Indexed: 11/21/2022] Open
Abstract
Exposure to antibiotic treatment has been associated with increased vulnerability to various psychiatric disorders. However, a research gap exists in understanding how adolescent antibiotic therapy affects behavior and cognition. Many antibiotics that target bacterial translation may also affect mitochondrial translation resulting in impaired mitochondrial function. The brain is one of the most metabolically active organs, and hence is the most vulnerable to impaired mitochondrial function. We hypothesized that exposure to antibiotics during early adolescence would directly affect brain mitochondrial function, and result in altered behavior and cognition. We administered amoxicillin, chloramphenicol, or gentamicin in the drinking water to young adolescent male wild-type mice. Next, we assayed mitochondrial oxidative phosphorylation complex activities in the cerebral cortex, performed behavioral screening and targeted mass spectrometry-based acylcarnitine profiling in the cerebral cortex. We found that mice exposed to chloramphenicol showed increased repetitive and compulsive-like behavior in the marble burying test, an accurate and sensitive assay of anxiety, concomitant with decreased mitochondrial complex IV activity. Our results suggest that only adolescent chloramphenicol exposure leads to impaired brain mitochondrial complex IV function, and could therefore be a candidate driver event for increased anxiety-like and repetitive, compulsive-like behaviors.
Collapse
Affiliation(s)
- Anouk C Tengeler
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behaviour, Centre for Medical Neuroscience, Preclinical Imaging Centre PRIME, Nijmegen, The Netherlands
| | - Tim L Emmerzaal
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behaviour, Centre for Medical Neuroscience, Preclinical Imaging Centre PRIME, Nijmegen, The Netherlands.,Department of Clinical Genomics, Mayo Clinic, 200 First St. SW, Rochester, MN, USA
| | - Bram Geenen
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behaviour, Centre for Medical Neuroscience, Preclinical Imaging Centre PRIME, Nijmegen, The Netherlands
| | - Vivienne Verweij
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behaviour, Centre for Medical Neuroscience, Preclinical Imaging Centre PRIME, Nijmegen, The Netherlands
| | - Miranda van Bodegom
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behaviour, Centre for Medical Neuroscience, Preclinical Imaging Centre PRIME, Nijmegen, The Netherlands
| | - Eva Morava
- Department of Clinical Genomics, Mayo Clinic, 200 First St. SW, Rochester, MN, USA
| | - Amanda J Kiliaan
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behaviour, Centre for Medical Neuroscience, Preclinical Imaging Centre PRIME, Nijmegen, The Netherlands
| | - Tamas Kozicz
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behaviour, Centre for Medical Neuroscience, Preclinical Imaging Centre PRIME, Nijmegen, The Netherlands. .,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
32
|
Valencia M, Kim SR, Jang Y, Lee SH. Neuronal Autophagy: Characteristic Features and Roles in Neuronal Pathophysiology. Biomol Ther (Seoul) 2021; 29:605-614. [PMID: 33875624 PMCID: PMC8551733 DOI: 10.4062/biomolther.2021.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/02/2021] [Accepted: 03/23/2021] [Indexed: 11/12/2022] Open
Abstract
Autophagy is an important degradative pathway that eliminates misfolded proteins and damaged organelles from cells. Autophagy is crucial for neuronal homeostasis and function. A lack of or deficiency in autophagy leads to the accumulation of protein aggregates, which are associated with several neurodegenerative diseases. Compared with non-neuronal cells, neurons exhibit rapid autophagic flux because damaged organelles or protein aggregates cannot be diluted in post-mitotic cells; because of this, these cells exhibit characteristic features of autophagy, such as compartment-specific autophagy, which depends on polarized structures and rapid autophagy flux. In addition, neurons exhibit compartment-specific autophagy, which depends on polarized structures. Neuronal autophagy may have additional physiological roles other than amino acid recycling. In this review, we focus on the characteristics and regulatory factors of neuronal autophagy. We also describe intracellular selective autophagy in neurons and its association with neurodegenerative diseases.
Collapse
Affiliation(s)
- McNeil Valencia
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Sung Rae Kim
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Yeseul Jang
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Sung Hoon Lee
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| |
Collapse
|
33
|
Löscher W, Kaila K. Reply to the commentary by Ben-Ari and Delpire: Bumetanide and neonatal seizures: Fiction versus reality. Epilepsia 2021; 62:941-946. [PMID: 33764535 DOI: 10.1111/epi.16866] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 02/16/2021] [Indexed: 12/18/2022]
Abstract
In this response to a commentary by Ben-Ari and Delpire on our recent study on the pharmacology of neonatal seizures in a novel, physiologically validated rat model of birth asphyxia, we wish to rectify their inaccurate descriptions of our model and data. Furthermore, because Ben-Ari and Delpire suggest that negative data on bumetanide from preclinical and clinical trials of neonatal seizures have few implications for (alleged) bumetanide actions on neurons in other brain disorders, we will discuss this topic as well. Based on the poor brain penetration of bumetanide, combined with the extremely wide cellular expression patterns of the target protein NKCC1, it is obvious that the numerous actions of systemically applied bumetanide described in the literature are not mediated by the drug's effects on central neurons.
Collapse
Affiliation(s)
- Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| | - Kai Kaila
- Molecular and Integrative Biosciences, University of Helsinki, Helsinki, Finland.,Neuroscience Center (HiLIFE), University of Helsinki, Helsinki, Finland
| |
Collapse
|
34
|
Mitochondrial Dysfunction and Permeability Transition in Neonatal Brain and Lung Injuries. Cells 2021; 10:cells10030569. [PMID: 33807810 PMCID: PMC7999701 DOI: 10.3390/cells10030569] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/27/2021] [Accepted: 03/01/2021] [Indexed: 01/16/2023] Open
Abstract
This review discusses the potential mechanistic role of abnormally elevated mitochondrial proton leak and mitochondrial bioenergetic dysfunction in the pathogenesis of neonatal brain and lung injuries associated with premature birth. Providing supporting evidence, we hypothesized that mitochondrial dysfunction contributes to postnatal alveolar developmental arrest in bronchopulmonary dysplasia (BPD) and cerebral myelination failure in diffuse white matter injury (WMI). This review also analyzes data on mitochondrial dysfunction triggered by activation of mitochondrial permeability transition pore(s) (mPTP) during the evolution of perinatal hypoxic-ischemic encephalopathy. While the still cryptic molecular identity of mPTP continues to be a subject for extensive basic science research efforts, the translational significance of mitochondrial proton leak received less scientific attention, especially in diseases of the developing organs. This review is focused on the potential mechanistic relevance of mPTP and mitochondrial dysfunction to neonatal diseases driven by developmental failure of organ maturation or by acute ischemia-reperfusion insult during development.
Collapse
|
35
|
Dupas T, Denis M, Dontaine J, Persello A, Bultot L, Erraud A, Vertommen D, Bouchard B, Tessier A, Rivière M, Lebreton J, Bigot‐Corbel E, Montnach J, De Waard M, Gauthier C, Burelle Y, Olson AK, Rozec B, Des Rosiers C, Bertrand L, Issad T, Lauzier B. Protein O-GlcNAcylation levels are regulated independently of dietary intake in a tissue and time-specific manner during rat postnatal development. Acta Physiol (Oxf) 2021; 231:e13566. [PMID: 33022862 PMCID: PMC7988603 DOI: 10.1111/apha.13566] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/28/2020] [Accepted: 09/29/2020] [Indexed: 12/11/2022]
Abstract
Aim Metabolic sources switch from carbohydrates in utero, to fatty acids after birth and then a mix once adults. O‐GlcNAcylation (O‐GlcNAc) is a post‐translational modification considered as a nutrient sensor. The purpose of this work was to assess changes in protein O‐GlcNAc levels, regulatory enzymes and metabolites during the first periods of life and decipher the impact of O‐GlcNAcylation on cardiac proteins. Methods Heart, brain and liver were harvested from rats before and after birth (D‐1 and D0), in suckling animals (D12), after weaning with a standard (D28) or a low‐carbohydrate diet (D28F), and adults (D84). O‐GlcNAc levels and regulatory enzymes were evaluated by western blots. Mass spectrometry (MS) approaches were performed to quantify levels of metabolites regulating O‐GlcNAc and identify putative cardiac O‐GlcNAcylated proteins. Results Protein O‐GlcNAc levels decrease drastically and progressively from D‐1 to D84 (13‐fold, P < .05) in the heart, whereas the changes were opposite in liver and brain. O‐GlcNAc levels were unaffected by weaning diet in any tissues. Changes in expression of enzymes and levels of metabolites regulating O‐GlcNAc were tissue‐dependent. MS analyses identified changes in putative cardiac O‐GlcNAcylated proteins, namely those involved in the stress response and energy metabolism, such as ACAT1, which is only O‐GlcNAcylated at D0. Conclusion Our results demonstrate that protein O‐GlcNAc levels are not linked to dietary intake and regulated in a time and tissue‐specific manner during postnatal development. We have identified by untargeted MS putative proteins with a particular O‐GlcNAc signature across the development process suggesting specific role of these proteins.
Collapse
Affiliation(s)
- Thomas Dupas
- Université de NantesCHU NantesCNRSINSERM, l’institut du thorax Nantes France
| | - Manon Denis
- Université de NantesCHU NantesCNRSINSERM, l’institut du thorax Nantes France
| | - Justine Dontaine
- Université catholique de LouvainInstitut de Recherche Expérimentale et CliniquePole of Cardiovascular Research Brussels Belgium
| | - Antoine Persello
- Université de NantesCHU NantesCNRSINSERM, l’institut du thorax Nantes France
- InFlectis BioScience Nantes France
| | - Laurent Bultot
- Université catholique de LouvainInstitut de Recherche Expérimentale et CliniquePole of Cardiovascular Research Brussels Belgium
| | - Angélique Erraud
- Université de NantesCHU NantesCNRSINSERM, l’institut du thorax Nantes France
| | - Didier Vertommen
- Université catholique de Louvainde Duve InstituteMass Spectrometry Platform Brussels Belgium
| | - Bertrand Bouchard
- Montreal Heart Institute Research Center and Department of Nutrition Université de Montréal Montreal Québec Canada
| | - Arnaud Tessier
- Faculté des Sciences et des Techniques Université de NantesCNRSChimie et Interdisciplinarité: Synthèse, Analyse, Modélisation (CEISAM)UMR CNRS 6230 Nantes France
| | - Matthieu Rivière
- Faculté des Sciences et des Techniques Université de NantesCNRSChimie et Interdisciplinarité: Synthèse, Analyse, Modélisation (CEISAM)UMR CNRS 6230 Nantes France
| | - Jacques Lebreton
- Faculté des Sciences et des Techniques Université de NantesCNRSChimie et Interdisciplinarité: Synthèse, Analyse, Modélisation (CEISAM)UMR CNRS 6230 Nantes France
| | | | - Jérôme Montnach
- Université de NantesCHU NantesCNRSINSERM, l’institut du thorax Nantes France
| | - Michel De Waard
- Université de NantesCHU NantesCNRSINSERM, l’institut du thorax Nantes France
| | - Chantal Gauthier
- Université de NantesCHU NantesCNRSINSERM, l’institut du thorax Nantes France
| | - Yan Burelle
- Interdisciplinary School of Health Sciences Faculty of Health Sciences and Department of Cellular and Molecular Medicine Faculty of Medicine University of Ottawa Ottawa ON Canada
| | - Aaron K. Olson
- Division of Cardiology Department of Pediatrics University of Washington Seattle WA98105USA
- Seattle Children’s Research Institute Seattle WA98101USA
| | - Bertrand Rozec
- Université de NantesCHU NantesCNRSINSERM, l’institut du thorax Nantes France
| | - Christine Des Rosiers
- Montreal Heart Institute Research Center and Department of Nutrition Université de Montréal Montreal Québec Canada
| | - Luc Bertrand
- Université catholique de LouvainInstitut de Recherche Expérimentale et CliniquePole of Cardiovascular Research Brussels Belgium
- WELBIO Brussels Belgium
| | - Tarik Issad
- Université de ParisINSERM U1016CNRS UMR 8104 Paris France
| | - Benjamin Lauzier
- Université de NantesCHU NantesCNRSINSERM, l’institut du thorax Nantes France
| |
Collapse
|
36
|
Račkayová V, Simicic D, Donati G, Braissant O, Gruetter R, McLin VA, Cudalbu C. Late post-natal neurometabolic development in healthy male rats using 1 H and 31 P magnetic resonance spectroscopy. J Neurochem 2021; 157:508-519. [PMID: 33421129 DOI: 10.1111/jnc.15294] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 01/01/2020] [Accepted: 12/25/2020] [Indexed: 11/28/2022]
Abstract
Brain metabolism evolves rapidly during early post-natal development in the rat. While changes in amino acids, energy metabolites, antioxidants or metabolites involved in phospholipid metabolism have been reported in the early stages, neurometabolic changes during the later post-natal period are less well characterized. Therefore, we aimed to assess the neurometabolic changes in male Wistar rats between post-natal days 29 and 77 (p29-p77) using longitudinal magnetic resonance spectroscopy (MRS) in vivo at 9.4 Tesla. 1 H MRS was performed in the hippocampus between p29 and p77 at 1-week intervals (n = 7) and in the cerebellum between p35 and p77 at 2-week intervals (n = 7) using the SPECIAL sequence at ultra-short echo-time. NOE enhanced and 1 H decoupled 31 P MR spectra were acquired at p35, p48 and p63 (n = 7) in a larger voxel covering cortex, hippocampus and part of the striatum. The hippocampus showed a decrease in taurine concentration and an increase in glutamate (with more pronounced changes until p49), seemingly a continuation of their well-described changes in the early post-natal period. A constant increase in myo-inositol and choline-containing compounds in the hippocampus (in particular glycero-phosphocholine as shown by 31 P MRS) was measured throughout the observation period, probably related to membrane metabolism and myelination. The cerebellum showed only a significant increase in myo-inositol between p35 and p77. In conclusion, this study showed important changes in brain metabolites in both the hippocampus and cerebellum in the later post-natal period (p29/p35-p77) of male rats, something previously unreported. Based on these novel data, changes in some neurometabolites beyond p28-35, conventionally accepted as the cut off for adulthood, should be taken into account in both experimental design and data interpretation in this animal model.
Collapse
Affiliation(s)
- Veronika Račkayová
- CIBM Center for Biomedical Imaging, Lausanne, Switzerland.,Animal Imaging and Technology, Ecole Polytechnique Federale de Lausanne (EPFL), Lausanne, Switzerland.,Laboratory of Functional and Metabolic Imaging (LIFMET), Ecole Polytechnique Federale de Lausanne (EPFL), Lausanne, Switzerland
| | - Dunja Simicic
- CIBM Center for Biomedical Imaging, Lausanne, Switzerland.,Animal Imaging and Technology, Ecole Polytechnique Federale de Lausanne (EPFL), Lausanne, Switzerland.,Laboratory of Functional and Metabolic Imaging (LIFMET), Ecole Polytechnique Federale de Lausanne (EPFL), Lausanne, Switzerland
| | - Guillaume Donati
- Laboratory of Functional and Metabolic Imaging (LIFMET), Ecole Polytechnique Federale de Lausanne (EPFL), Lausanne, Switzerland
| | - Olivier Braissant
- Service of Clinical Chemistry, University of Lausanne and University Hospital of Lausanne, Lausanne, Switzerland
| | - Rolf Gruetter
- Laboratory of Functional and Metabolic Imaging (LIFMET), Ecole Polytechnique Federale de Lausanne (EPFL), Lausanne, Switzerland
| | - Valérie A McLin
- Swiss Pediatric Liver Center, University Hospitals Geneva, Geneva, Switzerland.,Department of Pediatrics, Gynecology, and Obstetrics, University of Geneva Medical School, Geneva, Switzerland
| | - Cristina Cudalbu
- CIBM Center for Biomedical Imaging, Lausanne, Switzerland.,Animal Imaging and Technology, Ecole Polytechnique Federale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
37
|
Davis JJ, Fournakis N, Ellison J. Ketogenic Diet for the Treatment and Prevention of Dementia: A Review. J Geriatr Psychiatry Neurol 2021; 34:3-10. [PMID: 31996078 DOI: 10.1177/0891988720901785] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Dementia (major neurocognitive disorder) is an increasingly common syndrome with a significant burden on patients, caregivers, the health-care system, and the society. The prevalence of dementia will certainly continue to grow as the US population ages. Current treatments for dementia, though, are limited. One proposed nonpharmacologic approach for the delay or prevention of dementia is the use of a ketogenic diet. The ketogenic diet was originally employed to treat refractory epilepsy and has shown promise in many neurologic diseases. It has also gained recent popularity for its weight loss effects. Several preclinical studies have confirmed a benefit of ketosis on cognition and systemic inflammation. Given the renewed emphasis on neuroinflammation as a pathogenic contributor to cognitive decline, and the decreased systemic inflammation observed with the ketogenic diet, it is plausible that this diet may delay, ameliorate, or prevent progression of cognitive decline. Several small human studies have shown benefit on cognition in dementia with a ketogenic diet intervention. Future, large controlled studies are needed to confirm this benefit; however, the ketogenic diet has shown promise in regard to delay or mitigation of symptoms of cognitive decline.
Collapse
Affiliation(s)
- Joshua J Davis
- Department of Emergency Medicine, 12311Penn State Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Nicole Fournakis
- Center for Community Health in the Office of Health Equity at 5973Christiana Care Health System, Newark, DE, USA
| | - James Ellison
- The Swank Foundation Endowed Chair in Memory Care and Geriatrics at 5973Christiana Care Health System, Newark, DE, USA
| |
Collapse
|
38
|
Giménez-Palomo A, Dodd S, Anmella G, Carvalho AF, Scaini G, Quevedo J, Pacchiarotti I, Vieta E, Berk M. The Role of Mitochondria in Mood Disorders: From Physiology to Pathophysiology and to Treatment. Front Psychiatry 2021; 12:546801. [PMID: 34295268 PMCID: PMC8291901 DOI: 10.3389/fpsyt.2021.546801] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 05/24/2021] [Indexed: 12/30/2022] Open
Abstract
Mitochondria are cellular organelles involved in several biological processes, especially in energy production. Several studies have found a relationship between mitochondrial dysfunction and mood disorders, such as major depressive disorder and bipolar disorder. Impairments in energy production are found in these disorders together with higher levels of oxidative stress. Recently, many agents capable of enhancing antioxidant defenses or mitochondrial functioning have been studied for the treatment of mood disorders as adjuvant therapy to current pharmacological treatments. A better knowledge of mitochondrial physiology and pathophysiology might allow the identification of new therapeutic targets and the development and study of novel effective therapies to treat these specific mitochondrial impairments. This could be especially beneficial for treatment-resistant patients. In this article, we provide a focused narrative review of the currently available evidence supporting the involvement of mitochondrial dysfunction in mood disorders, the effects of current therapies on mitochondrial functions, and novel targeted therapies acting on mitochondrial pathways that might be useful for the treatment of mood disorders.
Collapse
Affiliation(s)
- Anna Giménez-Palomo
- Bipolar and Depressives Disorders Unit, Hospital Clínic, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Mental Health Research Networking Center (CIBERSAM), Madrid, Spain
| | - Seetal Dodd
- Deakin University, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, VIC, Australia.,Department of Psychiatry, Centre for Youth Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Gerard Anmella
- Bipolar and Depressives Disorders Unit, Hospital Clínic, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Mental Health Research Networking Center (CIBERSAM), Madrid, Spain
| | - Andre F Carvalho
- Centre for Addiction and Mental Health, Toronto, ON, Canada.,Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Giselli Scaini
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Joao Quevedo
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States.,Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States.,Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciúma, Brazil.,Center of Excellence in Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Isabella Pacchiarotti
- Bipolar and Depressives Disorders Unit, Hospital Clínic, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Mental Health Research Networking Center (CIBERSAM), Madrid, Spain
| | - Eduard Vieta
- Bipolar and Depressives Disorders Unit, Hospital Clínic, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Mental Health Research Networking Center (CIBERSAM), Madrid, Spain
| | - Michael Berk
- School of Medicine, The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Barwon Health, Geelong, VIC, Australia.,Orygen, The National Centre of Excellence in Youth Mental Health, Parkville, VIC, Australia.,Centre for Youth Mental Health, Florey Institute for Neuroscience and Mental Health and the Department of Psychiatry, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
39
|
Espino De la Fuente-Muñoz C, Arias C. The therapeutic potential of mitochondrial transplantation for the treatment of neurodegenerative disorders. Rev Neurosci 2020; 32:203-217. [PMID: 33550783 DOI: 10.1515/revneuro-2020-0068] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 09/29/2020] [Indexed: 02/07/2023]
Abstract
Mitochondrial activity is essential to support neural functions, and changes in the integrity and activity of the mitochondria can contribute to synaptic damage and neuronal death, especially in degenerative diseases associated with age, such as Alzheimer's and Parkinson's disease. Currently, different approaches are used to treat these conditions, and one strategy under research is mitochondrial transplantation. For years, mitochondria have been shown to be transferred between cells of different tissues. This process has allowed several attempts to develop transplantation schemes by isolating functional mitochondria and introducing them into damaged tissue in particular to counteract the harmful effects of myocardial ischemia. Recently, mitochondrial transfer between brain cells has also been reported, and thus, mitochondrial transplantation for disorders of the nervous system has begun to be investigated. In this review, we focus on the relevance of mitochondria in the nervous system, as well as some mitochondrial alterations that occur in neurodegenerative diseases associated with age. In addition, we describe studies that have performed mitochondrial transplantation in various tissues, and we emphasize the advances in mitochondrial transplantation aimed at treating diseases of the nervous system.
Collapse
Affiliation(s)
- César Espino De la Fuente-Muñoz
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, AP 70-228, 04510, Ciudad de México, México
| | - Clorinda Arias
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, AP 70-228, 04510, Ciudad de México, México
| |
Collapse
|
40
|
Beiersdorf J, Hevesi Z, Calvigioni D, Pyszkowski J, Romanov R, Szodorai E, Lubec G, Shirran S, Botting CH, Kasper S, Guy GW, Gray R, Di Marzo V, Harkany T, Keimpema E. Adverse effects of Δ9-tetrahydrocannabinol on neuronal bioenergetics during postnatal development. JCI Insight 2020; 5:135418. [PMID: 33141759 PMCID: PMC7714410 DOI: 10.1172/jci.insight.135418] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 10/28/2020] [Indexed: 11/22/2022] Open
Abstract
Ongoing societal changes in views on the medical and recreational roles of cannabis increased the use of concentrated plant extracts with a Δ9-tetrahydrocannabinol (THC) content of more than 90%. Even though prenatal THC exposure is widely considered adverse for neuronal development, equivalent experimental data for young age cohorts are largely lacking. Here, we administered plant-derived THC (1 or 5 mg/kg) to mice daily during P5–P16 and P5–P35 and monitored its effects on hippocampal neuronal survival and specification by high-resolution imaging and iTRAQ proteomics, respectively. We found that THC indiscriminately affects pyramidal cells and both cannabinoid receptor 1+ (CB1R)+ and CB1R– interneurons by P16. THC particularly disrupted the expression of mitochondrial proteins (complexes I–IV), a change that had persisted even 4 months after the end of drug exposure. This was reflected by a THC-induced loss of membrane integrity occluding mitochondrial respiration and could be partially or completely rescued by pH stabilization, antioxidants, bypassed glycolysis, and targeting either mitochondrial soluble adenylyl cyclase or the mitochondrial voltage-dependent anion channel. Overall, THC exposure during infancy induces significant and long-lasting reorganization of neuronal circuits through mechanisms that, in large part, render cellular bioenergetics insufficient to sustain key developmental processes in otherwise healthy neurons. Repeated THC exposure in juvenile mice compromises the limbic circuitry, with life-long impairment to the respiration of neurons.
Collapse
Affiliation(s)
- Johannes Beiersdorf
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Zsofia Hevesi
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Daniela Calvigioni
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | | | - Roman Romanov
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Edit Szodorai
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Gert Lubec
- Paracelsus Private Medical University, Salzburg, Austria
| | - Sally Shirran
- School of Chemistry, University of St. Andrews, St. Andrews, United Kingdom
| | | | - Siegfried Kasper
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | | | - Roy Gray
- GW Phamaceuticals, Salisbury, Wiltshire, United Kingdom
| | - Vincenzo Di Marzo
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, Pozzuoli, Italy.,Canada Excellence Research Chair, Institut Universitaire de Cardiologie et de Pneumologie de Québec and Institut sur la Nutrition et les Aliments Fonctionnels, Université Laval, Québec, Québec, Canada
| | - Tibor Harkany
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria.,Department of Neuroscience, Biomedikum D7, Karolinska Institutet, Solna, Sweden
| | - Erik Keimpema
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
41
|
Decoding Aging: Understanding the Complex Relationship among Aging, Free Radicals, and GSH. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:3970860. [PMID: 33110472 PMCID: PMC7578726 DOI: 10.1155/2020/3970860] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/21/2020] [Accepted: 05/20/2020] [Indexed: 11/18/2022]
Abstract
N-aryl maleimides can undergo a 1,4-Michael-type addition reaction with reduced glutathione (GSH), leading to a decreased concentration of GSH and an increased concentration of free radicals (FRs) in cells. GSH is a critical scavenging molecule responsible for protecting cells from oxidation and for maintaining redox homeostasis. N-aryl maleimides disturb redox homeostasis in cells because they scavenge thiol-containing molecules, especially GSH. This study aimed at measuring the concentrations of GSH and FRs by electronic paramagnetic resonance (EPR), in the brain and liver tissue of male Wistar rats (ex vivo) at different ages and after treatment with 3,5-dimaleimylbenzoic acid (3,5-DMB). Our results showed a relationship between age and the concentrations of GSH and FRs in cells. In young rats, the concentration of GSH was higher than in old rats, while the concentration of FRs was higher in adult rats than in young rats, suggesting an inverse relationship between GSH and FRs. On the other hand, the reaction of 3,5-DMB (an electrophilic maleimide) with cellular GSH increased the FR content. The results of this study contribute to the awareness that the process of aging implies not only a loss of tissue function but also essential changes in the molecular contents of cells, especially the concentrations of FRs and GSH.
Collapse
|
42
|
Niatsetskaya Z, Sosunov S, Stepanova A, Goldman J, Galkin A, Neginskaya M, Pavlov E, Ten V. Cyclophilin D-dependent oligodendrocyte mitochondrial ion leak contributes to neonatal white matter injury. J Clin Invest 2020; 130:5536-5550. [PMID: 32925170 PMCID: PMC7524474 DOI: 10.1172/jci133082] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 07/07/2020] [Indexed: 12/26/2022] Open
Abstract
Postnatal failure of oligodendrocyte maturation has been proposed as a cellular mechanism of diffuse white matter injury (WMI) in premature infants. However, the molecular mechanisms for oligodendrocyte maturational failure remain unclear. In neonatal mice and cultured differentiating oligodendrocytes, sublethal intermittent hypoxic (IH) stress activated cyclophilin D-dependent mitochondrial proton leak and uncoupled mitochondrial respiration, leading to transient bioenergetic stress. This was associated with development of diffuse WMI: poor oligodendrocyte maturation, diffuse axonal hypomyelination, and permanent sensorimotor deficit. In normoxic mice and oligodendrocytes, exposure to a mitochondrial uncoupler recapitulated the phenotype of WMI, supporting the detrimental role of mitochondrial uncoupling in the pathogenesis of WMI. Compared with WT mice, cyclophilin D-knockout littermates did not develop bioenergetic stress in response to IH challenge and fully preserved oligodendrocyte maturation, axonal myelination, and neurofunction. Our study identified the cyclophilin D-dependent mitochondrial proton leak and uncoupling as a potentially novel subcellular mechanism for the maturational failure of oligodendrocytes and offers a potential therapeutic target for prevention of diffuse WMI in premature infants experiencing chronic IH stress.
Collapse
Affiliation(s)
| | | | | | - James Goldman
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | | | - Maria Neginskaya
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, New York, USA
| | - Evgeny Pavlov
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, New York, USA
| | | |
Collapse
|
43
|
Rodríguez M, Valez V, Cimarra C, Blasina F, Radi R. Hypoxic-Ischemic Encephalopathy and Mitochondrial Dysfunction: Facts, Unknowns, and Challenges. Antioxid Redox Signal 2020; 33:247-262. [PMID: 32295425 DOI: 10.1089/ars.2020.8093] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Significance: Hypoxic-ischemic events due to intrapartum complications represent the second cause of neonatal mortality and initiate an acute brain disorder known as hypoxic-ischemic encephalopathy (HIE). In HIE, the brain undergoes primary and secondary energy failure phases separated by a latent phase in which partial neuronal recovery is observed. A hypoxic-ischemic event leads to oxygen restriction causing ATP depletion, neuronal oxidative stress, and cell death. Mitochondrial dysfunction and enhanced oxidant formation in brain cells are characteristic phenomena associated with energy failure. Recent Advances: Mitochondrial sources of oxidants in neurons include complex I of the mitochondrial respiratory chain, as a key contributor to O2•- production via succinate by a reverse electron transport mechanism. The reaction of O2•- with nitric oxide (•NO) yields peroxynitrite, a mitochondrial and cellular toxin. Quantitation of the redox state of cytochrome c oxidase, through broadband near-infrared spectroscopy, represents a promising monitoring approach to evaluate mitochondrial dysfunction in vivo in humans, in conjunction with the determination of cerebral oxygenation and their correlation with the severity of brain injury. Critical Issues: The energetic failure being a key phenomenon in HIE connected with the severity of the encephalopathy, measurement of mitochondrial dysfunction in vivo provides an approach to assess evolution, prognosis, and adequate therapies. Restoration of mitochondrial redox homeostasis constitutes a key therapeutic goal. Future Directions: While hypothermia is the only currently accepted therapy in clinical management to preserve mitochondrial function, other mitochondria-targeted and/or redox-based treatments are likely to synergize to ensure further efficacy.
Collapse
Affiliation(s)
- Marianela Rodríguez
- Departamento de Bioquímica and Centro de Investigaciones Biomédicas (CEINBIO) and Facultad de Medicina, Hospital de Clínicas, Universidad de la República, Montevideo, Uruguay.,Departamento de Neonatología, Facultad de Medicina, Hospital de Clínicas, Universidad de la República, Montevideo, Uruguay
| | - Valeria Valez
- Departamento de Bioquímica and Centro de Investigaciones Biomédicas (CEINBIO) and Facultad de Medicina, Hospital de Clínicas, Universidad de la República, Montevideo, Uruguay
| | - Carolina Cimarra
- Departamento de Bioquímica and Centro de Investigaciones Biomédicas (CEINBIO) and Facultad de Medicina, Hospital de Clínicas, Universidad de la República, Montevideo, Uruguay
| | - Fernanda Blasina
- Departamento de Neonatología, Facultad de Medicina, Hospital de Clínicas, Universidad de la República, Montevideo, Uruguay
| | - Rafael Radi
- Departamento de Bioquímica and Centro de Investigaciones Biomédicas (CEINBIO) and Facultad de Medicina, Hospital de Clínicas, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
44
|
Pospelov AS, Puskarjov M, Kaila K, Voipio J. Endogenous brain-sparing responses in brain pH and PO 2 in a rodent model of birth asphyxia. Acta Physiol (Oxf) 2020; 229:e13467. [PMID: 32174009 DOI: 10.1111/apha.13467] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 03/11/2020] [Indexed: 12/12/2022]
Abstract
AIM To study brain-sparing physiological responses in a rodent model of birth asphyxia which reproduces the asphyxia-defining systemic hypoxia and hypercapnia. METHODS Steady or intermittent asphyxia was induced for 15-45 minutes in anaesthetized 6- and 11-days old rats and neonatal guinea pigs using gases containing 5% or 9% O2 plus 20% CO2 (in N2 ). Hypoxia and hypercapnia were induced with low O2 and high CO2 respectively. Oxygen partial pressure (PO2 ) and pH were measured with microsensors within the brain and subcutaneous ("body") tissue. Blood lactate was measured after asphyxia. RESULTS Brain and body PO2 fell to apparent zero with little recovery during 5% O2 asphyxia and 5% or 9% O2 hypoxia, and increased more than twofold during 20% CO2 hypercapnia. Unlike body PO2 , brain PO2 recovered rapidly to control after a transient fall (rat), or was slightly higher than control (guinea pig) during 9% O2 asphyxia. Asphyxia (5% O2 ) induced a respiratory acidosis paralleled by a progressive metabolic (lact)acidosis that was much smaller within than outside the brain. Hypoxia (5% O2 ) produced a brain-confined alkalosis. Hypercapnia outlasting asphyxia suppressed pH recovery and prolonged the post-asphyxia PO2 overshoot. All pH changes were accompanied by consistent shifts in the blood-brain barrier potential. CONCLUSION Regardless of brain maturation stage, hypercapnia can restore brain PO2 and protect the brain against metabolic acidosis despite compromised oxygen availability during asphyxia. This effect extends to the recovery phase if normocapnia is restored slowly, and it is absent during hypoxia, demonstrating that exposure to hypoxia does not mimic asphyxia.
Collapse
Affiliation(s)
- Alexey S. Pospelov
- Faculty of Biological and Environmental Sciences, Molecular and Integrative Biosciences University of Helsinki Helsinki Finland
| | - Martin Puskarjov
- Faculty of Biological and Environmental Sciences, Molecular and Integrative Biosciences University of Helsinki Helsinki Finland
| | - Kai Kaila
- Faculty of Biological and Environmental Sciences, Molecular and Integrative Biosciences University of Helsinki Helsinki Finland
- Neuroscience Center (HiLIFE) University of Helsinki Helsinki Finland
| | - Juha Voipio
- Faculty of Biological and Environmental Sciences, Molecular and Integrative Biosciences University of Helsinki Helsinki Finland
| |
Collapse
|
45
|
Bastian TW, Rao R, Tran PV, Georgieff MK. The Effects of Early-Life Iron Deficiency on Brain Energy Metabolism. Neurosci Insights 2020; 15:2633105520935104. [PMID: 32637938 PMCID: PMC7324901 DOI: 10.1177/2633105520935104] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 05/28/2020] [Indexed: 12/14/2022] Open
Abstract
Iron deficiency (ID) is one of the most prevalent nutritional deficiencies in the world. Iron deficiency in the late fetal and newborn period causes abnormal cognitive performance and emotional regulation, which can persist into adulthood despite iron repletion. Potential mechanisms contributing to these impairments include deficits in brain energy metabolism, neurotransmission, and myelination. Here, we comprehensively review the existing data that demonstrate diminished brain energetic capacity as a mechanistic driver of impaired neurobehavioral development due to early-life (fetal-neonatal) ID. We further discuss a novel hypothesis that permanent metabolic reprogramming, which occurs during the period of ID, leads to chronically impaired neuronal energetics and mitochondrial capacity in adulthood, thus limiting adult neuroplasticity and neurobehavioral function. We conclude that early-life ID impairs energy metabolism in a brain region- and age-dependent manner, with particularly strong evidence for hippocampal neurons. Additional studies, focusing on other brain regions and cell types, are needed.
Collapse
Affiliation(s)
- Thomas W Bastian
- Department of Pediatrics, Medical School, University of Minnesota, Minneapolis, MN, USA
| | - Raghavendra Rao
- Department of Pediatrics, Medical School, University of Minnesota, Minneapolis, MN, USA
| | - Phu V Tran
- Department of Pediatrics, Medical School, University of Minnesota, Minneapolis, MN, USA
| | - Michael K Georgieff
- Department of Pediatrics, Medical School, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
46
|
Steiner P. Brain Fuel Utilization in the Developing Brain. ANNALS OF NUTRITION AND METABOLISM 2020; 75 Suppl 1:8-18. [PMID: 32564020 DOI: 10.1159/000508054] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 03/16/2020] [Indexed: 11/19/2022]
Abstract
During pregnancy and infancy, the human brain is growing extremely fast; the brain volume increases significantly, reaching 36, 72, and 83% of the volume of adults at 2-4 weeks, 1 year, and 2 years of age, respectively, which is essential to establish the neuronal networks and capacity for the development of cognitive, motor, social, and emotional skills that will be continually refined throughout childhood and adulthood. Such dramatic changes in brain structure and function are associated with very large energetic demands exceeding by far those of other organs of the body. It has been estimated that during childhood the brain may account for up to 60% of the body basal energetic requirements. While the main source of energy for the adult brain is glucose, it appears that it is not sufficient to sustain the dramatic metabolic demands of the brain during its development. Recently, it has been proposed that this energetic challenge is solved by the ability of the brain to use ketone bodies (KBs), produced from fatty acid oxidation, as a complement source of energy. Here, we first describe the main cellular and physiological processes that drive brain development along time and how different brain metabolic pathways are engaged to support them. It has been assumed that the majority of energetic substrates are used to support neuronal activity and signal transmission. We discuss how glucose and KBs are metabolized to provide the carbon backbones used to synthesize lipids, nucleic acid, and cholesterol, which are indispensable building blocks of neuronal cell proliferation and are also used to establish and refine brain connectivity through synapse formation/elimination and myelination. We conclude that glucose and KBs are not only important to support the energy needs of the brain under development, but they are also essential substrates for the biosynthesis of macromolecules underlying structural brain growth and reorganization. We emphasize that glucose and fatty acids supporting the production of KBs are provided in complex food matrices, such as breast milk, and understanding how their availability impacts the brain will be key to promote adequate nutrition to support brain metabolism and, therefore, optimal brain development.
Collapse
Affiliation(s)
- Pascal Steiner
- Société des Produits Nestlé SA, Nestlé Research, Brain Health Department, Lausanne, Switzerland,
| |
Collapse
|
47
|
Rudnitskaya EA, Kozlova TA, Burnyasheva AO, Tarasova AE, Pankova TM, Starostina MV, Stefanova NA, Kolosova NG. Features of Postnatal Hippocampal Development in a Rat Model of Sporadic Alzheimer's Disease. Front Neurosci 2020; 14:533. [PMID: 32581685 PMCID: PMC7289999 DOI: 10.3389/fnins.2020.00533] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 04/29/2020] [Indexed: 12/26/2022] Open
Abstract
Aging is the major risk factor of the most common (∼95% of cases) sporadic Alzheimer’s disease (AD). Accumulating data indicate middle age as a critical period for the relevant pathological processes, however, the question of when AD starts to develop remains open. It has been reported only recently that in the early postnatal period—when brain development is completing—preconditions for a decrease in cognitive abilities and for accelerated aging can form. Here, we hypothesized that specific features of early postnatal brain development may be considered some of the prerequisites of AD development at an advanced age. To test this hypothesis, we used OXYS rats, which are a suitable model of sporadic AD. The duration of gestation, litter size, and weight at birth were lower in OXYS rats compared to control Wistar rats. The shortened duration of gestation may result in developmental retardation. Indeed, we noted decreased locomotor activity and increased anxiety in OXYS rats already at a young age: possible signs of altered brain development. We demonstrated retardation of the peak of postnatal neurogenesis in the hippocampal dentate gyrus of OXYS rats. Delayed neuronal maturation led to alterations of mossy-fiber formation: a shortened suprapyramidal bundle and longer infrapyramidal bundle, less pronounced fasciculation of granule cells’ axons, and smaller size and irregular shape of nuclei in the CA3 pyramidal layer. These changes were accompanied by altered astrocytic migration. The observed features of early development may be considered some of the risk factors of the AD-like pathology that manifests itself in OXYS rats late in life.
Collapse
Affiliation(s)
- Ekaterina A Rudnitskaya
- Laboratory of Molecular Mechanisms of Aging, The Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences (SB RAS), Novosibirsk, Russia
| | - Tatiana A Kozlova
- Laboratory of Molecular Mechanisms of Aging, The Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences (SB RAS), Novosibirsk, Russia
| | - Alena O Burnyasheva
- Laboratory of Molecular Mechanisms of Aging, The Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences (SB RAS), Novosibirsk, Russia
| | - Anna E Tarasova
- Laboratory of Central Mechanisms of Regulation, Institute of Molecular Biology and Biophysics, Federal Research Center of Fundamental and Translational Medicine, Novosibirsk, Russia
| | - Tatiana M Pankova
- Laboratory of Central Mechanisms of Regulation, Institute of Molecular Biology and Biophysics, Federal Research Center of Fundamental and Translational Medicine, Novosibirsk, Russia
| | - Marina V Starostina
- Laboratory of Central Mechanisms of Regulation, Institute of Molecular Biology and Biophysics, Federal Research Center of Fundamental and Translational Medicine, Novosibirsk, Russia
| | - Natalia A Stefanova
- Laboratory of Molecular Mechanisms of Aging, The Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences (SB RAS), Novosibirsk, Russia
| | - Nataliya G Kolosova
- Laboratory of Molecular Mechanisms of Aging, The Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences (SB RAS), Novosibirsk, Russia
| |
Collapse
|
48
|
Yu Z, Madelin G, Sodickson DK, Cloos MA. Simultaneous proton magnetic resonance fingerprinting and sodium MRI. Magn Reson Med 2020; 83:2232-2242. [PMID: 31746048 PMCID: PMC7047525 DOI: 10.1002/mrm.28073] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 10/07/2019] [Accepted: 10/21/2019] [Indexed: 12/15/2022]
Abstract
PURPOSE The goal of this work is to demonstrate a method for the simultaneous acquisition of proton multiparametric maps (T1 , T2 , and proton density) and sodium density images in 1 single scan. We hope that the development of such capabilities will help to ease the implementation of sodium MRI in clinical trials and provide more opportunities for researchers to investigate metabolism through sodium MRI. METHODS We developed a sequence based on magnetic resonance fingerprinting (MRF), which contains interleaved proton (1 H) and sodium (23 Na) excitations followed by a simultaneous center-out radial readout for both nuclei. The receive chain of a 7T scanner was modified to enable simultaneous acquisition of 1 H and 23 Na signal. The obtained signal-to-noise ratio (SNR) was evaluated, and the accuracy of both proton T1 , T2 , and B 1 + and sodium density maps were verified in phantoms. Finally, the method was demonstrated in 2 healthy subjects. RESULTS The SNR obtained using the simultaneous measurement was almost identical to single-nucleus measurements (<1% change). Similarly, the proton T1 and T2 maps remained stable (normalized root mean square error in T1 ≈ 2.2%, in T2 ≈ 1.4%, and B 1 + ≈ 5.4%), which indicates that the proposed sequence and hardware have no significant effects on the signal from either nucleus. In vivo measurements corroborated these results and demonstrated the feasibility of our method for in vivo application. CONCLUSIONS With the proposed approach, we were able to simultaneously acquire sodium density images in addition to proton T1 , T2 , and B 1 + maps as well as proton density images.
Collapse
Affiliation(s)
- Zidan Yu
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, NY, USA
- Center for Advanced Imaging Innovation and Research (CAIR), Department of Radiology, New York University School of Medicine, New York, NY, USA
- The Sackler Institute of Graduate Biomedical Sciences, New York University School of Medicine, New York, NY, USA
| | - Guillaume Madelin
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, NY, USA
- Center for Advanced Imaging Innovation and Research (CAIR), Department of Radiology, New York University School of Medicine, New York, NY, USA
- The Sackler Institute of Graduate Biomedical Sciences, New York University School of Medicine, New York, NY, USA
| | - Daniel K. Sodickson
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, NY, USA
- Center for Advanced Imaging Innovation and Research (CAIR), Department of Radiology, New York University School of Medicine, New York, NY, USA
- The Sackler Institute of Graduate Biomedical Sciences, New York University School of Medicine, New York, NY, USA
| | - Martijn A. Cloos
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, NY, USA
- Center for Advanced Imaging Innovation and Research (CAIR), Department of Radiology, New York University School of Medicine, New York, NY, USA
- The Sackler Institute of Graduate Biomedical Sciences, New York University School of Medicine, New York, NY, USA
| |
Collapse
|
49
|
Sandri BJ, Lubach GR, Lock EF, Georgieff MK, Kling PJ, Coe CL, Rao RB. Early-Life Iron Deficiency and Its Natural Resolution Are Associated with Altered Serum Metabolomic Profiles in Infant Rhesus Monkeys. J Nutr 2020; 150:685-693. [PMID: 31722400 PMCID: PMC7138653 DOI: 10.1093/jn/nxz274] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 07/23/2019] [Accepted: 10/11/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Iron deficiency is the most common nutrient deficiency in human infants aged 6 to 24 mo, and negatively affects many cellular metabolic processes, including energy production, electron transport, and oxidative degradation of toxins. There can be persistent influences on long-term metabolic health beyond its acute effects. OBJECTIVES The objective was to determine how iron deficiency in infancy alters the serum metabolomic profile and to test whether these effects persist after the resolution of iron deficiency in a nonhuman primate model of spontaneous iron deficiency. METHODS Blood was collected from naturally iron-sufficient (IS; n = 10) and iron-deficient (ID; n = 10) male and female infant rhesus monkeys (Macaca mulatta) at 6 mo of age. Iron deficiency resolved without intervention upon feeding of solid foods, and iron status was re-evaluated at 12 mo of age from the IS and formerly ID monkeys using hematological and other indices; sera were metabolically profiled using HPLC/MS and GC/MS with isobaric standards for identification and quantification at both time points. RESULTS A total of 413 metabolites were measured, with differences in 40 metabolites identified between IS and ID monkeys at 6 mo (P$\le $ 0.05). At 12 mo, iron-related hematological parameters had returned to normal, but the formerly ID infants remained metabolically distinct from the age-matched IS infants, with 48 metabolites differentially expressed between the groups. Metabolomic profiling indicated altered liver metabolites, differential fatty acid production, increased serum uridine release, and atypical bile acid production in the ID monkeys. CONCLUSIONS Pathway analyses of serum metabolites provided evidence of a hypometabolic state, altered liver function, differential essential fatty acid production, irregular uracil metabolism, and atypical bile acid production in ID infants. Many metabolites remained altered after the resolution of ID, suggesting long-term effects on metabolic health.
Collapse
Affiliation(s)
- Brian J Sandri
- Division of Neonatology, Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Gabriele R Lubach
- Harlow Center for Biological Psychology, University of Wisconsin, Madison, WI, USA
| | - Eric F Lock
- School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Michael K Georgieff
- Division of Neonatology, Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA,Center for Neurobehavioral Development, University of Minnesota, Minneapolis, MN, USA
| | - Pamela J Kling
- Division of Neonatology, Department of Pediatrics, University of Wisconsin, Madison, WI, USA
| | - Christopher L Coe
- Harlow Center for Biological Psychology, University of Wisconsin, Madison, WI, USA
| | - Raghavendra B Rao
- Division of Neonatology, Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA,Center for Neurobehavioral Development, University of Minnesota, Minneapolis, MN, USA,Address correspondence to RBR (e-mail: )
| |
Collapse
|
50
|
Pathological changes induced by phosphine poisoning: a study on 8 children. Int J Legal Med 2019; 134:217-228. [PMID: 31713064 DOI: 10.1007/s00414-019-02169-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 10/07/2019] [Indexed: 12/17/2022]
Abstract
Aluminum phosphide (ALP) has been extensively used as an economical and effective insecticide, rodenticide, and fumigant. The active ingredient of ALP is phosphine (PH3), the use of which can lead to accidental inhalation and mass poisoning with high mortality. Exposure to PH3 will give rise to global damage in the human body. This study reviewed 4 fatal accidents including 8 children with PH3 poisoning and aimed to determine the pathological changes that resulted from exposure to PH3 and, secondly, aimed to determine whether oxidative stress was involved in PH3-induced neurotoxicity using histopathological and immunohistochemistry (IHC) methods. After focusing on the pathological changes on the major organs, we found severe damage induced by PH3 in many systems, especially the neurological system, including neuronal, axonal, and vascular injuries as well as oxidative damage with increased expression of 4-hydroxy-2-trans-nonenal (4HNE), 8-hydroxy-2'-deoxyguanosine (8-OH-dG), and 3-nitrotyrosine (3-NT) in the brain, which indicated that oxidative stress was a crucial mechanism for neuronal death in PH3 toxicity. Moreover, we observed severe myocardial and hepatocellular fatty degeneration in the tissues of the heart and liver. We considered that these characteristic changes are a suggestive sign of PH3 poisoning and partly explained the toxic mechanism of PH3 (inhibition of mitochondrial oxidative phosphorylation). We hope that this research could improve the understanding of the toxicity of PH3 in both forensic and clinical practice.
Collapse
|