1
|
Claeys W, Geerts A, Van Hoecke L, Van Steenkiste C, Vandenbroucke RE. Role of astrocytes and microglia in hepatic encephalopathy associated with advanced chronic liver disease: lessons from animal studies. Neural Regen Res 2025; 20:3461-3475. [PMID: 39688562 PMCID: PMC11974659 DOI: 10.4103/nrr.nrr-d-24-00600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 08/05/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Hepatic encephalopathy, defined as neuropsychiatric dysfunction secondary to liver disease, is a frequent decompensating event in cirrhosis. Its clinical impact is highlighted by a notable increase in patient mortality rates and a concomitant reduction in overall quality of life. Systemically, liver disease, liver function failure, portosystemic shunting, and associated multi-organ dysfunction result in the increase of disease-causing neurotoxins in the circulation, which impairs cerebral homeostasis. Key circulating neurotoxins are ammonia and inflammatory mediators. In the brain, pathophysiology is less well understood, but is thought to be driven by glial cell dysfunction. Astrocytes are the only brain resident cells that have ammonia-metabolizing machinery and are therefore putatively most susceptible to ammonia elevation. Based on a large body of mostly in vitro evidence, ammonia-induced cellular and molecular disturbances include astrocyte swelling and oxidative stress. Microglia, the brain resident macrophages, have been linked to the translation of systemic inflammation to the brain microenvironment. Recent evidence from animal studies has provided novel insights into old and new downstream effects of astrocyte and microglial dysfunction such as toxin clearance disruption and myeloid cell attraction to the central nervous system parenchyma. Furthermore, state of the art research increasingly implicates neuronal dysfunction and possibly even irreversible neuronal cell death. Cell-type specific investigation in animal models highlights the need for critical revision of the contribution of astrocytes and microglia to well-established and novel cellular and molecular alterations in hepatic encephalopathy. In this review, we therefore give a current and comprehensive overview of causes, features, and consequences of astrocyte and microglial dysfunction in hepatic encephalopathy, including areas of interest for future investigation.
Collapse
Affiliation(s)
- Wouter Claeys
- Department of Internal Medicine and Paediatrics, Hepatology Research Unit, Ghent University, Ghent, Belgium
- Liver Research Center Ghent, Ghent University, Ghent University Hospital, Ghent, Belgium
- Barriers in Inflammation, VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Anja Geerts
- Department of Internal Medicine and Paediatrics, Hepatology Research Unit, Ghent University, Ghent, Belgium
- Liver Research Center Ghent, Ghent University, Ghent University Hospital, Ghent, Belgium
- Department of Gastroenterology and Hepatology, Ghent University Hospital, Ghent, Belgium
| | - Lien Van Hoecke
- Barriers in Inflammation, VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Christophe Van Steenkiste
- Department of Gastroenterology and Hepatology, Antwerp University, Antwerp, Belgium
- Department of Gastroenterology and Hepatology, Maria Middelares Hospital, Ghent, Belgium
| | - Roosmarijn E. Vandenbroucke
- Barriers in Inflammation, VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| |
Collapse
|
2
|
Chen J, Chen J, Yu C, Xia K, Yang B, Wang R, Li Y, Shi K, Zhang Y, Xu H, Zhang X, Wang J, Chen Q, Liang C. Metabolic reprogramming: a new option for the treatment of spinal cord injury. Neural Regen Res 2025; 20:1042-1057. [PMID: 38989936 PMCID: PMC11438339 DOI: 10.4103/nrr.nrr-d-23-01604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 02/27/2024] [Indexed: 07/12/2024] Open
Abstract
Spinal cord injuries impose a notably economic burden on society, mainly because of the severe after-effects they cause. Despite the ongoing development of various therapies for spinal cord injuries, their effectiveness remains unsatisfactory. However, a deeper understanding of metabolism has opened up a new therapeutic opportunity in the form of metabolic reprogramming. In this review, we explore the metabolic changes that occur during spinal cord injuries, their consequences, and the therapeutic tools available for metabolic reprogramming. Normal spinal cord metabolism is characterized by independent cellular metabolism and intercellular metabolic coupling. However, spinal cord injury results in metabolic disorders that include disturbances in glucose metabolism, lipid metabolism, and mitochondrial dysfunction. These metabolic disturbances lead to corresponding pathological changes, including the failure of axonal regeneration, the accumulation of scarring, and the activation of microglia. To rescue spinal cord injury at the metabolic level, potential metabolic reprogramming approaches have emerged, including replenishing metabolic substrates, reconstituting metabolic couplings, and targeting mitochondrial therapies to alter cell fate. The available evidence suggests that metabolic reprogramming holds great promise as a next-generation approach for the treatment of spinal cord injury. To further advance the metabolic treatment of the spinal cord injury, future efforts should focus on a deeper understanding of neurometabolism, the development of more advanced metabolomics technologies, and the design of highly effective metabolic interventions.
Collapse
Affiliation(s)
- Jiangjie Chen
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Orthopedics Research Institute of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang Province, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Jinyang Chen
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Orthopedics Research Institute of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang Province, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Chao Yu
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Orthopedics Research Institute of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang Province, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Kaishun Xia
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Orthopedics Research Institute of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang Province, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Biao Yang
- Qiandongnan Prefecture People's Hospital, Kaili, Guizhou Province, China
| | - Ronghao Wang
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Orthopedics Research Institute of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang Province, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Yi Li
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Orthopedics Research Institute of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang Province, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Kesi Shi
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Orthopedics Research Institute of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang Province, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Yuang Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Orthopedics Research Institute of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang Province, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Haibin Xu
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Orthopedics Research Institute of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang Province, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Xuesong Zhang
- Department of Orthopedics, Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jingkai Wang
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Orthopedics Research Institute of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang Province, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Qixin Chen
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Orthopedics Research Institute of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang Province, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Chengzhen Liang
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Orthopedics Research Institute of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang Province, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang Province, China
| |
Collapse
|
3
|
Furukawa K, Ikoma Y, Niino Y, Hiraoka Y, Tanaka K, Miyawaki A, Hirrlinger J, Matsui K. Dynamics of Neuronal and Astrocytic Energy Molecules in Epilepsy. J Neurochem 2025; 169:e70044. [PMID: 40108970 PMCID: PMC11923518 DOI: 10.1111/jnc.70044] [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: 11/07/2024] [Revised: 02/26/2025] [Accepted: 02/28/2025] [Indexed: 03/22/2025]
Abstract
The dynamics of energy molecules in the mouse brain during metabolic challenges induced by epileptic seizures were examined. A transgenic mouse line expressing a fluorescence resonance energy transfer (FRET)-based adenosine triphosphate (ATP) sensor, selectively expressed in the cytosol of neurons, was used. An optical fiber was inserted into the hippocampus, and changes in cytosolic ATP concentration were estimated using the fiber photometry method. To induce epileptic neuronal hyperactivity, a train of electrical stimuli was delivered to a bipolar electrode placed alongside the optical fiber. Although maintaining a steady cytosolic ATP concentration is crucial for cell survival, a single episode of epileptic neuronal hyperactivity drastically reduced neuronal ATP levels. Interestingly, the magnitude of ATP reduction did not increase with the exacerbation of epilepsy, but rather decreased. This suggests that the primary consumption of ATP during epileptic neuronal hyperactivity may not be solely directed toward restoring the Na+ and K+ ionic imbalance caused by action potential bursts. Cytosolic ATP concentration reflects the balance between supply and consumption. To investigate the metabolic flux leading to neuronal ATP production, a new FRET-based pyruvate sensor was developed and selectively expressed in the cytosol of astrocytes in transgenic mice. Upon epileptic neuronal hyperactivity, an increase in astrocytic pyruvate concentration was observed. Changes in the supply of energy molecules, such as glucose and oxygen, due to blood vessel constriction or dilation, as well as metabolic alterations in astrocyte function, may contribute to cytosolic ATP dynamics in neurons.
Collapse
Affiliation(s)
- Kota Furukawa
- Super-network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Yoko Ikoma
- Super-network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Yusuke Niino
- Laboratory for Cell Function Dynamics, RIKEN Center for Brain Science, Wako-City, Japan
| | - Yuichi Hiraoka
- Laboratory of Molecular Neuroscience, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo Institute of Technology, Tokyo, Japan
- Laboratory of Genome Editing for Biomedical Research, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo Institute of Technology, Tokyo, Japan
| | - Kohichi Tanaka
- Laboratory of Genome Editing for Biomedical Research, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo Institute of Technology, Tokyo, Japan
| | - Atsushi Miyawaki
- Laboratory for Cell Function Dynamics, RIKEN Center for Brain Science, Wako-City, Japan
- Biotechnological Optics Research Team, RIKEN Center for Advanced Photonics, Wako-City, Japan
| | - Johannes Hirrlinger
- Carl-Ludwig-Institute for Physiology, Faculty of Medicine, Leipzig University, Leipzig, Germany
- Department of Neurogenetics, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Ko Matsui
- Super-network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
- Super-network Brain Physiology, Graduate School of Medicine, Tohoku University, Sendai, Japan
| |
Collapse
|
4
|
Tutas J, Tolve M, Özer-Yildiz E, Ickert L, Klein I, Silverman Q, Liebsch F, Dethloff F, Giavalisco P, Endepols H, Georgomanolis T, Neumaier B, Drzezga A, Schwarz G, Thorens B, Gatto G, Frezza C, Kononenko NL. Autophagy regulator ATG5 preserves cerebellar function by safeguarding its glycolytic activity. Nat Metab 2025; 7:297-320. [PMID: 39815080 PMCID: PMC11860254 DOI: 10.1038/s42255-024-01196-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 11/29/2024] [Indexed: 01/18/2025]
Abstract
Dysfunctions in autophagy, a cellular mechanism for breaking down components within lysosomes, often lead to neurodegeneration. The specific mechanisms underlying neuronal vulnerability due to autophagy dysfunction remain elusive. Here we show that autophagy contributes to cerebellar Purkinje cell (PC) survival by safeguarding their glycolytic activity. Outside the conventional housekeeping role, autophagy is also involved in the ATG5-mediated regulation of glucose transporter 2 (GLUT2) levels during cerebellar maturation. Autophagy-deficient PCs exhibit GLUT2 accumulation on the plasma membrane, along with increased glucose uptake and alterations in glycolysis. We identify lysophosphatidic acid and serine as glycolytic intermediates that trigger PC death and demonstrate that the deletion of GLUT2 in ATG5-deficient mice mitigates PC neurodegeneration and rescues their ataxic gait. Taken together, this work reveals a mechanism for regulating GLUT2 levels in neurons and provides insights into the neuroprotective role of autophagy by controlling glucose homeostasis in the brain.
Collapse
Affiliation(s)
- Janine Tutas
- CECAD Excellence Center, University of Cologne, Cologne, Germany
- Center for Physiology and Pathophysiology, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Marianna Tolve
- CECAD Excellence Center, University of Cologne, Cologne, Germany
- Center for Physiology and Pathophysiology, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Ebru Özer-Yildiz
- CECAD Excellence Center, University of Cologne, Cologne, Germany
- Center for Physiology and Pathophysiology, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Lotte Ickert
- CECAD Excellence Center, University of Cologne, Cologne, Germany
- Center for Physiology and Pathophysiology, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Ines Klein
- Department of Neurology, University Hospital of Cologne, Cologne, Germany
| | - Quinn Silverman
- Department of Neurology, University Hospital of Cologne, Cologne, Germany
| | - Filip Liebsch
- Institute of Biochemistry, Department of Chemistry, University of Cologne, Cologne, Germany
| | | | | | - Heike Endepols
- Department of Nuclear Medicine, Faculty of Medicine, University Hospital Cologne, Cologne, Germany
- Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Forschungszentrum Jülich GmbH, Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Jülich, Germany
| | | | - Bernd Neumaier
- Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Forschungszentrum Jülich GmbH, Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Jülich, Germany
| | - Alexander Drzezga
- Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Forschungszentrum Jülich GmbH, Institute of Neuroscience and Medicine, Molecular Organization of the Brain (INM-2), Jülich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn-Cologne, Germany
| | - Guenter Schwarz
- Institute of Biochemistry, Department of Chemistry, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Bernard Thorens
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Graziana Gatto
- Department of Neurology, University Hospital of Cologne, Cologne, Germany
| | - Christian Frezza
- CECAD Excellence Center, University of Cologne, Cologne, Germany
- Institute for Genetics, Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany
| | - Natalia L Kononenko
- CECAD Excellence Center, University of Cologne, Cologne, Germany.
- Center for Physiology and Pathophysiology, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.
- Institute for Genetics, Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany.
| |
Collapse
|
5
|
Barros LF, Schirmeier S, Weber B. The Astrocyte: Metabolic Hub of the Brain. Cold Spring Harb Perspect Biol 2024; 16:a041355. [PMID: 38438188 PMCID: PMC11368191 DOI: 10.1101/cshperspect.a041355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Abstract
Astrocytic metabolism has taken center stage. Interposed between the neuron and the vasculature, astrocytes exert control over the fluxes of energy and building blocks required for neuronal activity and plasticity. They are also key to local detoxification and waste recycling. Whereas neurons are metabolically rigid, astrocytes can switch between different metabolic profiles according to local demand and the nutritional state of the organism. Their metabolic state even seems to be instructive for peripheral nutrient mobilization and has been implicated in information processing and behavior. Here, we summarize recent progress in our understanding of astrocytic metabolism and its effects on metabolic homeostasis and cognition.
Collapse
Affiliation(s)
- L Felipe Barros
- Centro de Estudios Científicos, Valdivia 5110465, Chile
- Universidad San Sebastián, Facultad de Medicina y Ciencia, Valdivia 5110693, Chile
| | - Stefanie Schirmeier
- Technische Universität Dresden, Department of Biology, 01217 Dresden, Germany
| | - Bruno Weber
- University of Zurich, Institute of Pharmacology and Toxicology, 8057 Zurich, Switzerland
| |
Collapse
|
6
|
Fernández-Moncada I, Lavanco G, Fundazuri UB, Bollmohr N, Mountadem S, Dalla Tor T, Hachaguer P, Julio-Kalajzic F, Gisquet D, Serrat R, Bellocchio L, Cannich A, Fortunato-Marsol B, Nasu Y, Campbell RE, Drago F, Cannizzaro C, Ferreira G, Bouzier-Sore AK, Pellerin L, Bolaños JP, Bonvento G, Barros LF, Oliet SHR, Panatier A, Marsicano G. A lactate-dependent shift of glycolysis mediates synaptic and cognitive processes in male mice. Nat Commun 2024; 15:6842. [PMID: 39122700 PMCID: PMC11316019 DOI: 10.1038/s41467-024-51008-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 07/16/2024] [Indexed: 08/12/2024] Open
Abstract
Astrocytes control brain activity via both metabolic processes and gliotransmission, but the physiological links between these functions are scantly known. Here we show that endogenous activation of astrocyte type-1 cannabinoid (CB1) receptors determines a shift of glycolysis towards the lactate-dependent production of D-serine, thereby gating synaptic and cognitive functions in male mice. Mutant mice lacking the CB1 receptor gene in astrocytes (GFAP-CB1-KO) are impaired in novel object recognition (NOR) memory. This phenotype is rescued by the gliotransmitter D-serine, by its precursor L-serine, and also by lactate and 3,5-DHBA, an agonist of the lactate receptor HCAR1. Such lactate-dependent effect is abolished when the astrocyte-specific phosphorylated-pathway (PP), which diverts glycolysis towards L-serine synthesis, is blocked. Consistently, lactate and 3,5-DHBA promoted the co-agonist binding site occupancy of CA1 post-synaptic NMDA receptors in hippocampal slices in a PP-dependent manner. Thus, a tight cross-talk between astrocytic energy metabolism and gliotransmission determines synaptic and cognitive processes.
Collapse
Affiliation(s)
| | - Gianluca Lavanco
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, ''G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Unai B Fundazuri
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Nasrin Bollmohr
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Sarah Mountadem
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Tommaso Dalla Tor
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy
| | - Pauline Hachaguer
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | | | - Doriane Gisquet
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Roman Serrat
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| | - Luigi Bellocchio
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Astrid Cannich
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | | | - Yusuke Nasu
- Department of Chemistry, School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- PRESTO, Japan Science and Technology Agency, Chiyoda-ku, Tokyo, Japan
| | - Robert E Campbell
- Department of Chemistry, School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- CERVO Brain Research Center and Department of Biochemistry, Microbiology, and Bioinformatics, Université Laval, Québec City, QC, Canada
| | - Filippo Drago
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy
| | - Carla Cannizzaro
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Guillaume Ferreira
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| | - Anne-Karine Bouzier-Sore
- Univ. Bordeaux, CNRS, Centre de Résonance Magnétique des Systèmes Biologiques, UMR 5536, F-33000, Bordeaux, France
| | - Luc Pellerin
- Université de Poitiers et CHU de Poitiers, INSERM, IRMETIST, U1313, Poitiers, France
| | - Juan P Bolaños
- Institute of Functional Biology and Genomics (IBFG), Universidad de Salamanca, CSIC, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Hospital Universitario de Salamanca, Salamanca, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - Gilles Bonvento
- Universite Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodegeneratives, Fontenay-aux-Roses, France
| | - L Felipe Barros
- Centro de Estudios Cientificos, Valdivia, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Valdivia, Chile
| | - Stephane H R Oliet
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Aude Panatier
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Giovanni Marsicano
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France.
| |
Collapse
|
7
|
Dienel GA, Rothman DL. In vivo calibration of genetically encoded metabolite biosensors must account for metabolite metabolism during calibration and cellular volume. J Neurochem 2024; 168:506-532. [PMID: 36726217 DOI: 10.1111/jnc.15775] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 01/21/2023] [Accepted: 01/28/2023] [Indexed: 02/03/2023]
Abstract
Isotopic assays of brain glucose utilization rates have been used for more than four decades to establish relationships between energetics, functional activity, and neurotransmitter cycling. Limitations of these methods include the relatively long time (1-60 min) for the determination of labeled metabolite levels and the lack of cellular resolution. Identification and quantification of fuels for neurons and astrocytes that support activation and higher brain functions are a major, unresolved issues. Glycolysis is preferentially up-regulated during activation even though oxygen level and supply are adequate, causing lactate concentrations to quickly rise during alerting, sensory processing, cognitive tasks, and memory consolidation. However, the fate of lactate (rapid release from brain or cell-cell shuttling coupled with local oxidation) is long disputed. Genetically encoded biosensors can determine intracellular metabolite concentrations and report real-time lactate level responses to sensory, behavioral, and biochemical challenges at the cellular level. Kinetics and time courses of cellular lactate concentration changes are informative, but accurate biosensor calibration is required for quantitative comparisons of lactate levels in astrocytes and neurons. An in vivo calibration procedure for the Laconic lactate biosensor involves intracellular lactate depletion by intravenous pyruvate-mediated trans-acceleration of lactate efflux followed by sensor saturation by intravenous infusion of high doses of lactate plus ammonium chloride. In the present paper, the validity of this procedure is questioned because rapid lactate-pyruvate interconversion in blood, preferential neuronal oxidation of both monocarboxylates, on-going glycolytic metabolism, and cellular volumes were not taken into account. Calibration pitfalls for the Laconic lactate biosensor also apply to other metabolite biosensors that are standardized in vivo by infusion of substrates that can be metabolized in peripheral tissues. We discuss how technical shortcomings negate the conclusion that Laconic sensor calibrations support the existence of an in vivo astrocyte-neuron lactate concentration gradient linked to lactate shuttling from astrocytes to neurons to fuel neuronal activity.
Collapse
Affiliation(s)
- Gerald A 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
| | - Douglas L Rothman
- Magnetic Resonance Research Center and Departments of Radiology and Biomedical Engineering, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
8
|
Schirmbeck GH, Seady M, Fróes FT, Taday J, Da Ré C, Souza JM, Gonçalves CA, Leite MC. Long-term LPS systemic administration leads to memory impairment and disturbance in astrocytic homeostasis. Neurotoxicology 2023; 99:322-331. [PMID: 38006911 DOI: 10.1016/j.neuro.2023.11.009] [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: 07/03/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 11/27/2023]
Abstract
Dementia is the most prevalent neurodegenerative disorder, characterized by progressive loss of memory and cognitive function. Inflammation is a major aspect in the progression of brain disorders, and inflammatory events have been associated with accelerated deterioration of cognitive function. In the present work, we investigated the impact of low-grade repeated inflammation stimuli induced by lipopolysaccharide (LPS) in hippocampal function and spatial memory. Adult male Wistar rats received a weekly injection of LPS (500 ug/kg) for sixteen weeks, eliciting systemic inflammation. Animals submitted to LPS presented impaired spatial memory and neuroinflammation. While neuronal synaptic markers such as synaptophysin and PSD-95 were unaltered, critical aspects of astrocyte homeostatic functions, such as glutamate uptake and glutathione content, were reduced. Also, glucose uptake and astrocyte lactate transporters were altered, suggesting a disturbance in the astrocyte-neuron coupling. Our present work demonstrates that long-term repeated systemic inflammation can lead to memory impairment and hippocampal metabolic disorders, especially regarding astrocyte function.
Collapse
Affiliation(s)
- Gabriel Henrique Schirmbeck
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Marina Seady
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Fernanda Telles Fróes
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Jéssica Taday
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Carollina Da Ré
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Jéssica Maria Souza
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Carlos Alberto Gonçalves
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Marina Concli Leite
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
| |
Collapse
|
9
|
Rabah Y, Francés R, Minatchy J, Guédon L, Desnous C, Plaçais PY, Preat T. Glycolysis-derived alanine from glia fuels neuronal mitochondria for memory in Drosophila. Nat Metab 2023; 5:2002-2019. [PMID: 37932430 PMCID: PMC10663161 DOI: 10.1038/s42255-023-00910-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 09/20/2023] [Indexed: 11/08/2023]
Abstract
Glucose is the primary source of energy for the brain; however, it remains controversial whether, upon neuronal activation, glucose is primarily used by neurons for ATP production or if it is partially oxidized in astrocytes, as proposed by the astrocyte-neuron lactate shuttle model for glutamatergic neurons. Thus, an in vivo picture of glucose metabolism during cognitive processes is missing. Here, we uncover in Drosophila melanogaster a glia-to-neuron alanine transfer involving alanine aminotransferase that sustains memory formation. Following associative conditioning, glycolysis in glial cells produces alanine, which is back-converted into pyruvate in cholinergic neurons of the olfactory memory center to uphold their increased mitochondrial needs. Alanine, as a mediator of glia-neuron coupling, could be an alternative to lactate in cholinergic systems. In parallel, a dedicated glial glucose transporter imports glucose specifically for long-term memory, by directly transferring it to neurons for use by the pentose phosphate pathway. Our results demonstrate in vivo the compartmentalization of glucose metabolism between neurons and glial cells during memory formation.
Collapse
Affiliation(s)
- Yasmine Rabah
- Energy & Memory, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
| | - Raquel Francés
- Energy & Memory, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
| | - Julia Minatchy
- Energy & Memory, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
| | - Laura Guédon
- Energy & Memory, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
| | - Coraline Desnous
- Energy & Memory, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
| | - Pierre-Yves Plaçais
- Energy & Memory, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France.
| | - Thomas Preat
- Energy & Memory, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France.
| |
Collapse
|
10
|
Barros LF. Glial metabolism checkpoints memory. Nat Metab 2023; 5:1852-1853. [PMID: 37932429 DOI: 10.1038/s42255-023-00886-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2023]
Affiliation(s)
- L Felipe Barros
- Centro de Estudios Científicos-CECs, Valdivia, Chile.
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Valdivia, Chile.
| |
Collapse
|
11
|
Guidetti OA, Speelman CP, Bouhlas P. Mapping between cognitive theories and psycho-physiological models of attention system performance. Cereb Cortex 2023; 33:10122-10138. [PMID: 37492014 PMCID: PMC10502801 DOI: 10.1093/cercor/bhad271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/27/2023] [Accepted: 06/28/2023] [Indexed: 07/27/2023] Open
Abstract
Declines in the capacity to sustain attention to repetitive, monotonous tasks is a phenomenon known as vigilance decrement (Endsley M, Kiris E. The out-of-the-loop performance problem and level of control in automation. 1995. Hum Factors. 37:32-64). This review compares cognitive theories with psycho-physiological models of vigilance decrement, and a gap is identified in mapping between the 2. That is, theories of vigilance decrement refer to "cognitive" resources; by contrast, psychophysiological models of the cerebral systems associated with attention explain performance functions according to neurochemical resources. A map does not currently exist in the literature that bridges the gap between cognitive theories of vigilance decrement and psychophysiological models of the human attention system. The link between "cognitive resource" theories of vigilance decrement and the psychophysiological models of attention performance is a gap in the literature that this review fills. This comprehensive review provides an expanded psychophysiological understanding of vigilance decrement that could help inform the management of declines in sustained attention capacity in operational settings. In addition, elucidating the link between cognitive theories of vigilance decrement and psychophysiological models of the human attention system might be used to treat and better understand pathologies such as attention-deficit hyperactivity disorder.
Collapse
Affiliation(s)
- Oliver A Guidetti
- Western Australian Department of the Premier and Cabinet, The Cyber Security Research Cooperative, Edith Cowan University, Building 30, 270 Joondalup Dr, Joondalup, Western Australia 6027, Australia
| | - Craig P Speelman
- Department of Psychology, Edith Cowan University, Building 30, 270 Joondalup Dr, Joondalup, Western Australia 6027, Australia
| | - Peter Bouhlas
- Western Australian Department of the Premier and Cabinet, Dumas House, 2 Havelock St, West Perth, Western Australia 6005, Australia
| |
Collapse
|
12
|
Barros LF, Ruminot I, Sandoval PY, San Martín A. Enlightening brain energy metabolism. Neurobiol Dis 2023:106211. [PMID: 37352985 DOI: 10.1016/j.nbd.2023.106211] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/06/2023] [Accepted: 06/20/2023] [Indexed: 06/25/2023] Open
Abstract
Brain tissue metabolism is distributed across several cell types and subcellular compartments, which activate at different times and with different temporal patterns. The introduction of genetically-encoded fluorescent indicators that are imaged using time-lapse microscopy has opened the possibility of studying brain metabolism at cellular and sub-cellular levels. There are indicators for sugars, monocarboxylates, Krebs cycle intermediates, amino acids, cofactors, and energy nucleotides, which inform about relative levels, concentrations and fluxes. This review offers a brief survey of the metabolic indicators that have been validated in brain cells, with some illustrative examples from the literature. Whereas only a small fraction of the metabolome is currently accessible to fluorescent probes, there are grounds to be optimistic about coming developments and the application of these tools to the study of brain disease.
Collapse
Affiliation(s)
- L F Barros
- Centro de Estudios Científicos (CECs), Valdivia, Chile; Facultad de Medicina y Ciencia, Universidad San Sebastián, Valdivia, Chile.
| | - I Ruminot
- Centro de Estudios Científicos (CECs), Valdivia, Chile; Facultad de Ciencias para el Cuidado de La Salud, Universidad San Sebastián, Valdivia, Chile
| | - P Y Sandoval
- Centro de Estudios Científicos (CECs), Valdivia, Chile; Facultad de Ciencias para el Cuidado de La Salud, Universidad San Sebastián, Valdivia, Chile
| | - A San Martín
- Centro de Estudios Científicos (CECs), Valdivia, Chile; Facultad de Ciencias para el Cuidado de La Salud, Universidad San Sebastián, Valdivia, Chile
| |
Collapse
|
13
|
Cauli B, Dusart I, Li D. Lactate as a determinant of neuronal excitability, neuroenergetics and beyond. Neurobiol Dis 2023:106207. [PMID: 37331530 DOI: 10.1016/j.nbd.2023.106207] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 06/13/2023] [Accepted: 06/15/2023] [Indexed: 06/20/2023] Open
Abstract
Over the last decades, lactate has emerged as important energy substrate for the brain fueling of neurons. A growing body of evidence now indicates that it is also a signaling molecule modulating neuronal excitability and activity as well as brain functions. In this review, we will briefly summarize how different cell types produce and release lactate. We will further describe different signaling mechanisms allowing lactate to fine-tune neuronal excitability and activity, and will finally discuss how these mechanisms could cooperate to modulate neuroenergetics and higher order brain functions both in physiological and pathological conditions.
Collapse
Affiliation(s)
- Bruno Cauli
- Sorbonne Université, CNRS, INSERM, Neurosciences Paris Seine - Institut de Biologie Paris Seine (NPS-IBPS), 9 quai Saint Bernard, 75005 Paris, France.
| | - Isabelle Dusart
- Sorbonne Université, CNRS, INSERM, Neurosciences Paris Seine - Institut de Biologie Paris Seine (NPS-IBPS), 9 quai Saint Bernard, 75005 Paris, France
| | - Dongdong Li
- Sorbonne Université, CNRS, INSERM, Neurosciences Paris Seine - Institut de Biologie Paris Seine (NPS-IBPS), 9 quai Saint Bernard, 75005 Paris, France
| |
Collapse
|
14
|
Astroglial CB1 receptors, energy metabolism, and gliotransmission: an integrated signaling system? Essays Biochem 2023; 67:49-61. [PMID: 36645029 DOI: 10.1042/ebc20220089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 12/06/2022] [Accepted: 12/07/2022] [Indexed: 01/17/2023]
Abstract
Astrocytes are key players in brain homeostasis and function. During the last years, several studies have cemented this notion by showing that these cells respond to neuronal signals and, via the release of molecules that modulate and support synaptic activity (gliotransmission) participates in the functions of the so-called tripartite synapse. Thus, besides their established control of brain metabolism, astrocytes can also actively control synaptic activity and behavior. Among the signaling pathways that shape the functions of astrocyte, the cannabinoid type-1 (CB1) receptor is emerging as a critical player in the control of both gliotransmission and the metabolic cooperation between astrocytes and neurons. In the present short review, we describe known and newly discovered properties of the astroglial CB1 receptors and their role in modulating brain function and behavior. Based on this evidence, we finally discuss how the functions and mode of actions of astrocyte CB1 receptors might represent a clear example of the inextricable relationship between energy metabolism and gliotransmission. These tight interactions will need to be taken into account for future research in astrocyte functions and call for a reinforcement of the theoretical and experimental bridges between studies on metabolic and synaptic functions of astrocytes.
Collapse
|
15
|
Barros LF, Ruminot I, Sotelo-Hitschfeld T, Lerchundi R, Fernández-Moncada I. Metabolic Recruitment in Brain Tissue. Annu Rev Physiol 2023; 85:115-135. [PMID: 36270291 DOI: 10.1146/annurev-physiol-021422-091035] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Information processing imposes urgent metabolic demands on neurons, which have negligible energy stores and restricted access to fuel. Here, we discuss metabolic recruitment, the tissue-level phenomenon whereby active neurons harvest resources from their surroundings. The primary event is the neuronal release of K+ that mirrors workload. Astrocytes sense K+ in exquisite fashion thanks to their unique coexpression of NBCe1 and α2β2 Na+/K+ ATPase, and within seconds switch to Crabtree metabolism, involving GLUT1, aerobic glycolysis, transient suppression of mitochondrial respiration, and lactate export. The lactate surge serves as a secondary recruiter by inhibiting glucose consumption in distant cells. Additional recruiters are glutamate, nitric oxide, and ammonium, which signal over different spatiotemporal domains. The net outcome of these events is that more glucose, lactate, and oxygen are made available. Metabolic recruitment works alongside neurovascular coupling and various averaging strategies to support the inordinate dynamic range of individual neurons.
Collapse
Affiliation(s)
- L F Barros
- Centro de Estudios Científicos (CECs), Valdivia, Chile; .,Facultad de Medicina y Ciencia, Universidad San Sebastián, Valdivia, Chile;
| | - I Ruminot
- Centro de Estudios Científicos (CECs), Valdivia, Chile; .,Facultad de Medicina y Ciencia, Universidad San Sebastián, Valdivia, Chile;
| | - T Sotelo-Hitschfeld
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - R Lerchundi
- Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), MIRCen, Fontenay-aux-Roses, France
| | - I Fernández-Moncada
- NeuroCentre Magendie, INSERM U1215, University of Bordeaux, Bordeaux, France
| |
Collapse
|
16
|
Understanding ayahuasca effects in major depressive disorder treatment through in vitro metabolomics and bioinformatics. Anal Bioanal Chem 2023:10.1007/s00216-023-04556-3. [PMID: 36717401 DOI: 10.1007/s00216-023-04556-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/27/2022] [Accepted: 01/19/2023] [Indexed: 02/01/2023]
Abstract
Emerging insights from metabolomic-based studies of major depression disorder (MDD) are mainly related to biochemical processes such as energy or oxidative stress, in addition to neurotransmission linked to specific metabolite intermediates. Hub metabolites represent nodes in the biochemical network playing a critical role in integrating the information flow in cells between metabolism and signaling pathways. Limited technical-scientific studies have been conducted to understand the effects of ayahuasca (Aya) administration in the metabolism considering MDD molecular context. Therefore, this work aims to investigate an in vitro primary astrocyte model by untargeted metabolomics of two cellular subfractions: secretome and intracellular content after pre-defined Aya treatments, based on DMT concentration. Mass spectrometry (MS)-based metabolomics data revealed significant hub metabolites, which were used to predict biochemical pathway alterations. Branched-chain amino acid (BCAA) metabolism, and vitamin B6 and B3 metabolism were associated to Aya treatment, as "housekeeping" pathways. Dopamine synthesis was overrepresented in the network results when considering the lowest tested DMT concentration (1 µmol L-1). Building reaction networks containing significant and differential metabolites, such as nicotinamide, L-DOPA, and L-leucine, is a useful approach to guide on dose decision and pathway selection in further analytical and molecular studies.
Collapse
|
17
|
Aburto C, Galaz A, Bernier A, Sandoval PY, Holtheuer-Gallardo S, Ruminot I, Soto-Ojeda I, Hertenstein H, Schweizer JA, Schirmeier S, Pástor TP, Mardones GA, Barros LF, San Martín A. Single-Fluorophore Indicator to Explore Cellular and Sub-cellular Lactate Dynamics. ACS Sens 2022; 7:3278-3286. [PMID: 36306435 DOI: 10.1021/acssensors.2c00731] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Lactate is an energy substrate and an intercellular signal, which can be monitored in intact cells with the genetically encoded FRET indicator Laconic. However, the structural complexity, need for sophisticated equipment, and relatively small fluorescent change limit the use of FRET indicators for subcellular targeting and development of high-throughput screening methodologies. Using the bacterial periplasmic binding protein TTHA0766 from Thermus thermophilus, we have now developed a single-fluorophore indicator for lactate, CanlonicSF. This indicator exhibits a maximal fluorescence change of 200% and a KD of ∼300 μM. The fluorescence is not affected by other monocarboxylates. The lactate indicator was not significantly affected by Ca2+ at the physiological concentrations prevailing in the cytosol, endoplasmic reticulum, and extracellular space, but was affected by Ca2+ in the low micromolar range. Targeting the indicator to the endoplasmic reticulum revealed for the first time sub-cellular lactate dynamics. Its improved lactate-induced fluorescence response permitted the development of a multiwell plate assay to screen for inhibitors of the monocarboxylate transporters MCTs, a pharmaceutical target for cancer and inflammation. The functionality of the indicator in living tissue was demonstrated in the brain of Drosophila melanogaster larvae. CanlonicSF is well suited to explore lactate dynamics with sub-cellular resolution in intact systems.
Collapse
Affiliation(s)
- Camila Aburto
- Centro de Estudios Científicos (CECs), Avenida Arturo Prat 514, Postal Code 5110466 Valdivia, Chile.,Universidad Austral de Chile, Isla Teja s/n, Postal Code 5110566 Valdivia, Chile
| | - Alex Galaz
- Centro de Estudios Científicos (CECs), Avenida Arturo Prat 514, Postal Code 5110466 Valdivia, Chile
| | - Angelo Bernier
- Centro de Estudios Científicos (CECs), Avenida Arturo Prat 514, Postal Code 5110466 Valdivia, Chile.,Universidad Austral de Chile, Isla Teja s/n, Postal Code 5110566 Valdivia, Chile
| | - Pamela Yohana Sandoval
- Centro de Estudios Científicos (CECs), Avenida Arturo Prat 514, Postal Code 5110466 Valdivia, Chile.,Facultad de Medicina y Ciencia, Universidad San Sebastián, Postal Code 5110773 Valdivia, Chile
| | - Sebastián Holtheuer-Gallardo
- Centro de Estudios Científicos (CECs), Avenida Arturo Prat 514, Postal Code 5110466 Valdivia, Chile.,Universidad Austral de Chile, Isla Teja s/n, Postal Code 5110566 Valdivia, Chile
| | - Iván Ruminot
- Centro de Estudios Científicos (CECs), Avenida Arturo Prat 514, Postal Code 5110466 Valdivia, Chile.,Facultad de Medicina y Ciencia, Universidad San Sebastián, Postal Code 5110773 Valdivia, Chile
| | - Ignacio Soto-Ojeda
- Centro de Estudios Científicos (CECs), Avenida Arturo Prat 514, Postal Code 5110466 Valdivia, Chile.,Universidad Austral de Chile, Isla Teja s/n, Postal Code 5110566 Valdivia, Chile
| | - Helen Hertenstein
- Department of Biology, Technische Universität Dresden, Postal Code 01062 Dresden, Germany
| | | | - Stefanie Schirmeier
- Department of Biology, Technische Universität Dresden, Postal Code 01062 Dresden, Germany
| | - Tammy Paulina Pástor
- Department of Physiology, School of Medicine, Universidad Austral de Chile, Isla Teja s/n, Postal Code 5110566 Valdivia, Chile
| | - Gonzalo Antonio Mardones
- Department of Physiology, School of Medicine, Universidad Austral de Chile, Isla Teja s/n, Postal Code 5110566 Valdivia, Chile
| | - Luis Felipe Barros
- Centro de Estudios Científicos (CECs), Avenida Arturo Prat 514, Postal Code 5110466 Valdivia, Chile.,Facultad de Medicina y Ciencia, Universidad San Sebastián, Postal Code 5110773 Valdivia, Chile
| | - Alejandro San Martín
- Centro de Estudios Científicos (CECs), Avenida Arturo Prat 514, Postal Code 5110466 Valdivia, Chile.,Facultad de Medicina y Ciencia, Universidad San Sebastián, Postal Code 5110773 Valdivia, Chile
| |
Collapse
|
18
|
Andersen JV, Schousboe A, Verkhratsky A. Astrocyte energy and neurotransmitter metabolism in Alzheimer's disease: integration of the glutamate/GABA-glutamine cycle. Prog Neurobiol 2022; 217:102331. [PMID: 35872221 DOI: 10.1016/j.pneurobio.2022.102331] [Citation(s) in RCA: 121] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 07/14/2022] [Accepted: 07/19/2022] [Indexed: 02/06/2023]
Abstract
Astrocytes contribute to the complex cellular pathology of Alzheimer's disease (AD). Neurons and astrocytes function in close collaboration through neurotransmitter recycling, collectively known as the glutamate/GABA-glutamine cycle, which is essential to sustain neurotransmission. Neurotransmitter recycling is intimately linked to astrocyte energy metabolism. In the course of AD, astrocytes undergo extensive metabolic remodeling, which may profoundly affect the glutamate/GABA-glutamine cycle. The consequences of altered astrocyte function and metabolism in relation to neurotransmitter recycling are yet to be comprehended. Metabolic alterations of astrocytes in AD deprive neurons of metabolic support, thereby contributing to synaptic dysfunction and neurodegeneration. In addition, several astrocyte-specific components of the glutamate/GABA-glutamine cycle, including glutamine synthesis and synaptic neurotransmitter uptake, are perturbed in AD. Integration of the complex astrocyte biology within the context of AD is essential for understanding the fundamental mechanisms of the disease, while restoring astrocyte metabolism may serve as an approach to arrest or even revert clinical progression of AD.
Collapse
Affiliation(s)
- Jens V Andersen
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark.
| | - Arne Schousboe
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK; Achucarro Center for Neuroscience, IKERBASQUE, 48011 Bilbao, Spain; Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102 Vilnius, Lithuania.
| |
Collapse
|
19
|
Becker-Krail DD, Ketchesin KD, Burns JN, Zong W, Hildebrand MA, DePoy LM, Vadnie CA, Tseng GC, Logan RW, Huang YH, McClung CA. Astrocyte Molecular Clock Function in the Nucleus Accumbens Is Important for Reward-Related Behavior. Biol Psychiatry 2022; 92:68-80. [PMID: 35461698 PMCID: PMC9232937 DOI: 10.1016/j.biopsych.2022.02.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 01/25/2022] [Accepted: 02/11/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Substance use disorders are associated with disruptions in circadian rhythms. Both human and animal work have shown the integral role for circadian clocks in the modulation of reward behaviors. Astrocytes have emerged as key regulators of circadian rhythmicity. However, no studies to date have identified the role of circadian astrocyte function in the nucleus accumbens (NAc), a hub for reward regulation, or determined the importance of these rhythms for reward-related behavior. METHODS Using astrocyte-specific RNA sequencing across time of day, we first characterized diurnal variation of the NAc astrocyte transcriptome. We then investigated the functional significance of this circadian regulation through viral-mediated disruption of molecular clock function in NAc astrocytes, followed by assessment of reward-related behaviors, metabolic-related molecular assays, and whole-cell electrophysiology in the NAc. RESULTS Strikingly, approximately 43% of the astrocyte transcriptome has a diurnal rhythm, and key metabolic pathways were enriched among the top rhythmic genes. Moreover, mice with a viral-mediated loss of molecular clock function in NAc astrocytes show a significant increase in locomotor response to novelty, exploratory drive, operant food self-administration, and motivation. At the molecular level, these animals also show disrupted metabolic gene expression, along with significant downregulation of both lactate and glutathione levels in the NAc. Loss of NAc astrocyte clock function also significantly altered glutamatergic signaling onto neighboring medium spiny neurons, alongside upregulated glutamate-related gene expression. CONCLUSIONS Taken together, these findings demonstrate a novel role for astrocyte circadian molecular clock function in the regulation of the NAc and reward-related behaviors.
Collapse
Affiliation(s)
- Darius D Becker-Krail
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Kyle D Ketchesin
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jennifer N Burns
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Wei Zong
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Mariah A Hildebrand
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Lauren M DePoy
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Chelsea A Vadnie
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - George C Tseng
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ryan W Logan
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts
| | - Yanhua H Huang
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Colleen A McClung
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
20
|
Gupta S, Moreno AJ, Wang D, Leon J, Chen C, Hahn O, Poon Y, Greenberg K, David N, Wyss-Coray T, Raftery D, Promislow DEL, Dubal DB. KL1 Domain of Longevity Factor Klotho Mimics the Metabolome of Cognitive Stimulation and Enhances Cognition in Young and Aging Mice. J Neurosci 2022; 42:4016-4025. [PMID: 35428698 PMCID: PMC9097772 DOI: 10.1523/jneurosci.2458-21.2022] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/02/2022] [Accepted: 03/31/2022] [Indexed: 11/22/2022] Open
Abstract
Cognitive deficits are a major biomedical challenge-and engagement of the brain in stimulating tasks improves cognition in aged individuals (Wilson et al., 2002; Gates et al., 2011) and rodents (Aidil-Carvalho et al., 2017), through unknown mechanisms. Whether cognitive stimulation alters specific metabolic pathways in the brain is unknown. Understanding which metabolic processes are involved in cognitive stimulation is important because it could lead to pharmacologic intervention that promotes biological effects of a beneficial behavior, toward the goal of effective medical treatments for cognitive deficits. Here we show using male mice that cognitive stimulation induced metabolic remodeling of the mouse hippocampus, and that pharmacologic treatment with the longevity hormone α-klotho (KL), mediated by its KL1 domain, partially mimicked this alteration. The shared, metabolic signature shared between cognitive stimulation and treatment with KL or KL1 closely correlated with individual mouse cognitive performance, indicating a link between metabolite levels and learning and memory. Importantly, the treatment of mice with KL1, an endogenous circulating factor that more closely mimicked cognitive stimulation than KL, acutely increased synaptic plasticity, a substrate of cognition. KL1 also improved cognition, itself, in young mice and countered deficits in old mice. Our data show that treatments or interventions mimicking the hippocampal metabolome of cognitive stimulation can enhance brain functions. Further, we identify the specific domain by which klotho promotes brain functions, through KL1, a metabolic mimic of cognitive stimulation.SIGNIFICANCE STATEMENT Cognitive deficits are a major biomedical challenge without truly effective pharmacologic treatments. Engaging the brain through cognitive tasks benefits cognition. Mimicking the effects of such beneficial behaviors through pharmacological treatment represents a highly valuable medical approach to treating cognitive deficits. We demonstrate that brain engagement through cognitive stimulation induces metabolic remodeling of the hippocampus that was acutely recapitulated by the longevity factor klotho, mediated by its KL1 domain. Treatment with KL1, a close mimic of cognitive stimulation, enhanced cognition and countered cognitive aging. Our findings shed light on how cognition metabolically alters the brain and provide a plausible therapeutic intervention for mimicking these alterations that, in turn, improves cognition in the young and aging brain.
Collapse
Affiliation(s)
- Shweta Gupta
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California 94143-1207
| | - Arturo J Moreno
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California 94143-1207
| | - Dan Wang
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California 94143-1207
| | - Julio Leon
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California 94143-1207
| | - Chen Chen
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California 94143-1207
| | - Oliver Hahn
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California 94305-5101
| | - Yan Poon
- Unity Biotechnology, Inc, South San Francisco 94080
| | | | | | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California 94305-5101
- Veterans Administration Palo Alto Healthcare System, Palo Alto, California 94304-1207
- Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, California 94305-5235
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California 94305-5235
| | - Daniel Raftery
- Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center, University of Washington, Seattle, Washington 98109-4714
- Fred Hutchinson Cancer Research Center, Seattle, Washington 98109-1024
| | - Daniel E L Promislow
- Department of Lab Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington 98195-7470
- Department of Biology, University of Washington, Seattle, Washington 98195-1800
| | - Dena B Dubal
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California 94143-1207
| |
Collapse
|
21
|
Wang R, Zhou R, Chen Z, Gao S, Zhou F. The Glial Cells Respond to Spinal Cord Injury. Front Neurol 2022; 13:844497. [PMID: 35599739 PMCID: PMC9120539 DOI: 10.3389/fneur.2022.844497] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 03/08/2022] [Indexed: 12/24/2022] Open
Abstract
It is been over 100 years since glial cells were discovered by Virchow. Since then, a great deal of research was carried out to specify these further roles and properties of glial cells in central nervous system (CNS). As it is well-known that glial cells, such as astrocytes, microglia, oligodendrocytes (OLs), and oligodendrocyte progenitor cells (OPCs) play an important role in supporting and enabling the effective nervous system function in CNS. After spinal cord injury (SCI), these glial cells play different roles in SCI and repair. In this review, we will discuss in detail about the role of glial cells in the healthy CNS and how they respond to SCI.
Collapse
|
22
|
Abstract
The energy cost of information processing is thought to be chiefly neuronal, with a minor fraction attributed to glial cells. However, there is compelling evidence that astrocytes capture synaptic K+ using their Na+/K+ ATPase, and not solely through Kir4.1 channels as was once thought. When this active buffering is taken into account, the cost of astrocytes rises by >200%. Gram-per-gram, astrocytes turn out to be as expensive as neurons. This conclusion is supported by 3D reconstruction of the neuropil showing similar mitochondrial densities in neurons and astrocytes, by cell-specific transcriptomics and proteomics, and by the rates of the tricarboxylic acid cycle. Possible consequences for reactive astrogliosis and brain disease are discussed.
Collapse
Affiliation(s)
- L F Barros
- Centro de Estudios Científicos - CECs, Valdivia, Chile
| |
Collapse
|
23
|
Yeo HC, Park SY, Tan T, Ng SK, Lakshmanan M, Lee DY. Combined multivariate statistical and flux balance analyses uncover media bottlenecks to the growth and productivity of CHO cell cultures. Biotechnol Bioeng 2022; 119:1740-1754. [PMID: 35435243 DOI: 10.1002/bit.28104] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 03/16/2022] [Accepted: 04/03/2022] [Indexed: 11/06/2022]
Abstract
Chinese hamster ovary (CHO) cells are widely used for producing recombinant proteins. To enhance their productivity and product quality, media reformulation has been a key strategy, albeit with several technical challenges, due to the myriad of complex molecular mechanisms underlying media effects on culture performance. Thus, it is imperative to characterize metabolic bottlenecks under various media conditions systematically. To do so, we combined partial least square regression (PLS-R) with the flux balance analysis of a genome-scale metabolic model to elucidate the physiological states and metabolic behaviors of human alpha-1 antitrypsin producing CHO-DG44 cells grown in one commercial and another two in-house media under development. At the onset, PLS-R was used to identify metabolite exchanges that were correlated to specific growth and productivity. Then, by comparing metabolic states described by resultant flux distributions under two of the media conditions, we found sub-optimal level of four nutrients and two metabolic wastes, which plausibly hindered cellular growth and productivity; mechanistically, lactate and ammonia recycling were modulated by glutamine and asparagine metabolisms in the media conditions, and also by hitherto unsuspected folate and choline supplements. Our work demonstrated how multivariate statistical analysis can be synergistically combined with metabolic modelling to uncover the mechanistic elements underlying differing media performance. It thus paved the way for the systematic identification of nutrient targets for medium reformulation to enhance recombinant protein production in CHO cells. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Hock Chuan Yeo
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01, Singapore, 138668.,Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, #07-01, Singapore, 138671
| | - Seo-Young Park
- School of Chemical Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Tessa Tan
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01, Singapore, 138668
| | - Say Kong Ng
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01, Singapore, 138668
| | - Meiyappan Lakshmanan
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01, Singapore, 138668
| | - Dong-Yup Lee
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01, Singapore, 138668.,School of Chemical Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| |
Collapse
|
24
|
Recent behavioral findings of pathophysiological involvement of lactate in the central nervous system. Biochim Biophys Acta Gen Subj 2022; 1866:130137. [DOI: 10.1016/j.bbagen.2022.130137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/24/2022] [Accepted: 03/25/2022] [Indexed: 11/19/2022]
|
25
|
San Martín A, Arce-Molina R, Aburto C, Baeza-Lehnert F, Barros LF, Contreras-Baeza Y, Pinilla A, Ruminot I, Rauseo D, Sandoval PY. Visualizing physiological parameters in cells and tissues using genetically encoded indicators for metabolites. Free Radic Biol Med 2022; 182:34-58. [PMID: 35183660 DOI: 10.1016/j.freeradbiomed.2022.02.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/08/2022] [Accepted: 02/10/2022] [Indexed: 02/07/2023]
Abstract
The study of metabolism is undergoing a renaissance. Since the year 2002, over 50 genetically-encoded fluorescent indicators (GEFIs) have been introduced, capable of monitoring metabolites with high spatial/temporal resolution using fluorescence microscopy. Indicators are fusion proteins that change their fluorescence upon binding a specific metabolite. There are indicators for sugars, monocarboxylates, Krebs cycle intermediates, amino acids, cofactors, and energy nucleotides. They permit monitoring relative levels, concentrations, and fluxes in living systems. At a minimum they report relative levels and, in some cases, absolute concentrations may be obtained by performing ad hoc calibration protocols. Proper data collection, processing, and interpretation are critical to take full advantage of these new tools. This review offers a survey of the metabolic indicators that have been validated in mammalian systems. Minimally invasive, these indicators have been instrumental for the purposes of confirmation, rebuttal and discovery. We envision that this powerful technology will foster metabolic physiology.
Collapse
Affiliation(s)
- A San Martín
- Centro de Estudios Científicos (CECs), Valdivia, Chile.
| | - R Arce-Molina
- Centro de Estudios Científicos (CECs), Valdivia, Chile
| | - C Aburto
- Centro de Estudios Científicos (CECs), Valdivia, Chile; Universidad Austral de Chile, Valdivia, Chile
| | | | - L F Barros
- Centro de Estudios Científicos (CECs), Valdivia, Chile
| | - Y Contreras-Baeza
- Centro de Estudios Científicos (CECs), Valdivia, Chile; Universidad Austral de Chile, Valdivia, Chile
| | - A Pinilla
- Centro de Estudios Científicos (CECs), Valdivia, Chile; Universidad Austral de Chile, Valdivia, Chile
| | - I Ruminot
- Centro de Estudios Científicos (CECs), Valdivia, Chile
| | - D Rauseo
- Centro de Estudios Científicos (CECs), Valdivia, Chile; Universidad Austral de Chile, Valdivia, Chile
| | - P Y Sandoval
- Centro de Estudios Científicos (CECs), Valdivia, Chile
| |
Collapse
|
26
|
Beard E, Lengacher S, Dias S, Magistretti PJ, Finsterwald C. Astrocytes as Key Regulators of Brain Energy Metabolism: New Therapeutic Perspectives. Front Physiol 2022; 12:825816. [PMID: 35087428 PMCID: PMC8787066 DOI: 10.3389/fphys.2021.825816] [Citation(s) in RCA: 106] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 12/20/2021] [Indexed: 12/11/2022] Open
Abstract
Astrocytes play key roles in the regulation of brain energy metabolism, which has a major impact on brain functions, including memory, neuroprotection, resistance to oxidative stress and homeostatic tone. Energy demands of the brain are very large, as they continuously account for 20–25% of the whole body’s energy consumption. Energy supply of the brain is tightly linked to neuronal activity, providing the origin of the signals detected by the widely used functional brain imaging techniques such as functional magnetic resonance imaging and positron emission tomography. In particular, neuroenergetic coupling is regulated by astrocytes through glutamate uptake that triggers astrocytic aerobic glycolysis and leads to glucose uptake and lactate release, a mechanism known as the Astrocyte Neuron Lactate Shuttle. Other neurotransmitters such as noradrenaline and Vasoactive Intestinal Peptide mobilize glycogen, the reserve for glucose exclusively localized in astrocytes, also resulting in lactate release. Lactate is then transferred to neurons where it is used, after conversion to pyruvate, as a rapid energy substrate, and also as a signal that modulates neuronal excitability, homeostasis, and the expression of survival and plasticity genes. Importantly, glycolysis in astrocytes and more generally cerebral glucose metabolism progressively deteriorate in aging and age-associated neurodegenerative diseases such as Alzheimer’s disease. This decreased glycolysis actually represents a common feature of several neurological pathologies. Here, we review the critical role of astrocytes in the regulation of brain energy metabolism, and how dysregulation of astrocyte-mediated metabolic pathways is involved in brain hypometabolism. Further, we summarize recent efforts at preclinical and clinical stages to target brain hypometabolism for the development of new therapeutic interventions in age-related neurodegenerative diseases.
Collapse
|
27
|
Bekdash R, Quejada JR, Ueno S, Kawano F, Morikawa K, Klein AD, Matsumoto K, Lee TC, Nakanishi K, Chalan A, Lee TM, Liu R, Homma S, Lin CS, Yelshanskaya MV, Sobolevsky AI, Goda K, Yazawa M. GEM-IL: A highly responsive fluorescent lactate indicator. CELL REPORTS METHODS 2021; 1:100092. [PMID: 35475001 PMCID: PMC9017230 DOI: 10.1016/j.crmeth.2021.100092] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 07/26/2021] [Accepted: 09/15/2021] [Indexed: 12/22/2022]
Abstract
Lactate metabolism has been shown to have increasingly important implications in cellular functions as well as in the development and pathophysiology of disease. The various roles as a signaling molecule and metabolite have led to interest in establishing a new method to detect lactate changes in live cells. Here we report our development of a genetically encoded metabolic indicator specifically for probing lactate (GEM-IL) based on superfolder fluorescent proteins and mutagenesis. With improvements in its design, specificity, and sensitivity, GEM-IL allows new applications compared with the previous lactate indicators, Laconic and Green Lindoblum. We demonstrate the functionality of GEM-IL to detect differences in lactate changes in human oncogenic neural progenitor cells and mouse primary ventricular myocytes. The development and application of GEM-IL show promise for enhancing our understanding of lactate dynamics and roles.
Collapse
Affiliation(s)
- Ramsey Bekdash
- Columbia Stem Cell Initiative, Columbia University, New York, NY 10032, USA
- Department of Rehabilitation and Regenerative Medicine, Vagelos College of Physicians and Surgeons, Columbia University, 650 West 168th Street, BB1108/BB1109D, New York, NY 10032, USA
- Department of Molecular Pharmacology and Therapeutics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Jose R. Quejada
- Columbia Stem Cell Initiative, Columbia University, New York, NY 10032, USA
- Department of Rehabilitation and Regenerative Medicine, Vagelos College of Physicians and Surgeons, Columbia University, 650 West 168th Street, BB1108/BB1109D, New York, NY 10032, USA
- Department of Molecular Pharmacology and Therapeutics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Shunnosuke Ueno
- Columbia Stem Cell Initiative, Columbia University, New York, NY 10032, USA
- Department of Rehabilitation and Regenerative Medicine, Vagelos College of Physicians and Surgeons, Columbia University, 650 West 168th Street, BB1108/BB1109D, New York, NY 10032, USA
- Department of Chemistry, University of Tokyo, Tokyo 113-0033, Japan
| | - Fuun Kawano
- Columbia Stem Cell Initiative, Columbia University, New York, NY 10032, USA
- Department of Rehabilitation and Regenerative Medicine, Vagelos College of Physicians and Surgeons, Columbia University, 650 West 168th Street, BB1108/BB1109D, New York, NY 10032, USA
| | - Kumi Morikawa
- Columbia Stem Cell Initiative, Columbia University, New York, NY 10032, USA
- Department of Rehabilitation and Regenerative Medicine, Vagelos College of Physicians and Surgeons, Columbia University, 650 West 168th Street, BB1108/BB1109D, New York, NY 10032, USA
| | - Alison D. Klein
- Columbia Stem Cell Initiative, Columbia University, New York, NY 10032, USA
- Department of Rehabilitation and Regenerative Medicine, Vagelos College of Physicians and Surgeons, Columbia University, 650 West 168th Street, BB1108/BB1109D, New York, NY 10032, USA
| | - Kenji Matsumoto
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Tetz C. Lee
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Koki Nakanishi
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Amy Chalan
- Columbia Stem Cell Initiative, Columbia University, New York, NY 10032, USA
- Department of Rehabilitation and Regenerative Medicine, Vagelos College of Physicians and Surgeons, Columbia University, 650 West 168th Street, BB1108/BB1109D, New York, NY 10032, USA
| | - Teresa M. Lee
- Columbia Stem Cell Initiative, Columbia University, New York, NY 10032, USA
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Rui Liu
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Shunichi Homma
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Chyuan-Sheng Lin
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
- Transgenic Mouse Shared Resource, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10032, USA
| | - Maria V. Yelshanskaya
- Department of Biochemistry and Molecular Biophysics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Alexander I. Sobolevsky
- Department of Biochemistry and Molecular Biophysics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Keisuke Goda
- Department of Chemistry, University of Tokyo, Tokyo 113-0033, Japan
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- Institute of Technological Sciences, Wuhan University, Hubei 430072, China
| | - Masayuki Yazawa
- Columbia Stem Cell Initiative, Columbia University, New York, NY 10032, USA
- Department of Rehabilitation and Regenerative Medicine, Vagelos College of Physicians and Surgeons, Columbia University, 650 West 168th Street, BB1108/BB1109D, New York, NY 10032, USA
- Department of Molecular Pharmacology and Therapeutics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| |
Collapse
|
28
|
Karagiannis A, Gallopin T, Lacroix A, Plaisier F, Piquet J, Geoffroy H, Hepp R, Naudé J, Le Gac B, Egger R, Lambolez B, Li D, Rossier J, Staiger JF, Imamura H, Seino S, Roeper J, Cauli B. Lactate is an energy substrate for rodent cortical neurons and enhances their firing activity. eLife 2021; 10:e71424. [PMID: 34766906 PMCID: PMC8651295 DOI: 10.7554/elife.71424] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 11/09/2021] [Indexed: 12/12/2022] Open
Abstract
Glucose is the mandatory fuel for the brain, yet the relative contribution of glucose and lactate for neuronal energy metabolism is unclear. We found that increased lactate, but not glucose concentration, enhances the spiking activity of neurons of the cerebral cortex. Enhanced spiking was dependent on ATP-sensitive potassium (KATP) channels formed with KCNJ11 and ABCC8 subunits, which we show are functionally expressed in most neocortical neuronal types. We also demonstrate the ability of cortical neurons to take-up and metabolize lactate. We further reveal that ATP is produced by cortical neurons largely via oxidative phosphorylation and only modestly by glycolysis. Our data demonstrate that in active neurons, lactate is preferred to glucose as an energy substrate, and that lactate metabolism shapes neuronal activity in the neocortex through KATP channels. Our results highlight the importance of metabolic crosstalk between neurons and astrocytes for brain function.
Collapse
Affiliation(s)
- Anastassios Karagiannis
- Sorbonne Université, CNRS, INSERM, Neurosciences Paris Seine - Institut de Biologie Paris Seine (NPS-IBPS)ParisFrance
| | - Thierry Gallopin
- Brain Plasticity Unit, CNRS UMR 8249, CNRS, ESPCI ParisParisFrance
| | - Alexandre Lacroix
- Sorbonne Université, CNRS, INSERM, Neurosciences Paris Seine - Institut de Biologie Paris Seine (NPS-IBPS)ParisFrance
| | - Fabrice Plaisier
- Sorbonne Université, CNRS, INSERM, Neurosciences Paris Seine - Institut de Biologie Paris Seine (NPS-IBPS)ParisFrance
| | - Juliette Piquet
- Sorbonne Université, CNRS, INSERM, Neurosciences Paris Seine - Institut de Biologie Paris Seine (NPS-IBPS)ParisFrance
| | - Hélène Geoffroy
- Brain Plasticity Unit, CNRS UMR 8249, CNRS, ESPCI ParisParisFrance
| | - Régine Hepp
- Sorbonne Université, CNRS, INSERM, Neurosciences Paris Seine - Institut de Biologie Paris Seine (NPS-IBPS)ParisFrance
| | - Jérémie Naudé
- Sorbonne Université, CNRS, INSERM, Neurosciences Paris Seine - Institut de Biologie Paris Seine (NPS-IBPS)ParisFrance
| | - Benjamin Le Gac
- Sorbonne Université, CNRS, INSERM, Neurosciences Paris Seine - Institut de Biologie Paris Seine (NPS-IBPS)ParisFrance
| | - Richard Egger
- Institute for Neurophysiology, Goethe University FrankfurtFrankfurtGermany
| | - Bertrand Lambolez
- Sorbonne Université, CNRS, INSERM, Neurosciences Paris Seine - Institut de Biologie Paris Seine (NPS-IBPS)ParisFrance
| | - Dongdong Li
- Sorbonne Université, CNRS, INSERM, Neurosciences Paris Seine - Institut de Biologie Paris Seine (NPS-IBPS)ParisFrance
| | - Jean Rossier
- Sorbonne Université, CNRS, INSERM, Neurosciences Paris Seine - Institut de Biologie Paris Seine (NPS-IBPS)ParisFrance
- Brain Plasticity Unit, CNRS UMR 8249, CNRS, ESPCI ParisParisFrance
| | - Jochen F Staiger
- Institute for Neuroanatomy, University Medical Center Göttingen, Georg-August- University GöttingenGoettingenGermany
| | - Hiromi Imamura
- Graduate School of Biostudies, Kyoto UniversityKyotoJapan
| | - Susumu Seino
- Division of Molecular and Metabolic Medicine, Kobe University Graduate School of MedicineHyogoJapan
| | - Jochen Roeper
- Institute for Neurophysiology, Goethe University FrankfurtFrankfurtGermany
| | - Bruno Cauli
- Sorbonne Université, CNRS, INSERM, Neurosciences Paris Seine - Institut de Biologie Paris Seine (NPS-IBPS)ParisFrance
| |
Collapse
|
29
|
Horvat A, Zorec R, Vardjan N. Lactate as an Astroglial Signal Augmenting Aerobic Glycolysis and Lipid Metabolism. Front Physiol 2021; 12:735532. [PMID: 34658920 PMCID: PMC8514727 DOI: 10.3389/fphys.2021.735532] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 09/06/2021] [Indexed: 01/16/2023] Open
Abstract
Astrocytes, heterogeneous neuroglial cells, contribute to metabolic homeostasis in the brain by providing energy substrates to neurons. In contrast to predominantly oxidative neurons, astrocytes are considered primarily as glycolytic cells. They take up glucose from the circulation and in the process of aerobic glycolysis (despite the normal oxygen levels) produce L-lactate, which is then released into the extracellular space via lactate transporters and possibly channels. Astroglial L-lactate can enter neurons, where it is used as a metabolic substrate, or exit the brain via the circulation. Recently, L-lactate has also been considered to be a signaling molecule in the brain, but the mechanisms of L-lactate signaling and how it contributes to the brain function remain to be fully elucidated. Here, we provide an overview of L-lactate signaling mechanisms in the brain and present novel insights into the mechanisms of L-lactate signaling via G-protein coupled receptors (GPCRs) with the focus on astrocytes. We discuss how increased extracellular L-lactate upregulates cAMP production in astrocytes, most likely viaL-lactate-sensitive Gs-protein coupled GPCRs. This activates aerobic glycolysis, enhancing L-lactate production and accumulation of lipid droplets, suggesting that L-lactate augments its own production in astrocytes (i.e., metabolic excitability) to provide more L-lactate for neurons and that astrocytes in conditions of increased extracellular L-lactate switch to lipid metabolism.
Collapse
Affiliation(s)
- Anemari Horvat
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.,Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia
| | - Robert Zorec
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.,Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia
| | - Nina Vardjan
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.,Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia
| |
Collapse
|
30
|
Salcedo C, Andersen JV, Vinten KT, Pinborg LH, Waagepetersen HS, Freude KK, Aldana BI. Functional Metabolic Mapping Reveals Highly Active Branched-Chain Amino Acid Metabolism in Human Astrocytes, Which Is Impaired in iPSC-Derived Astrocytes in Alzheimer's Disease. Front Aging Neurosci 2021; 13:736580. [PMID: 34603012 PMCID: PMC8484639 DOI: 10.3389/fnagi.2021.736580] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 08/04/2021] [Indexed: 01/04/2023] Open
Abstract
The branched-chain amino acids (BCAAs) leucine, isoleucine, and valine are important nitrogen donors for synthesis of glutamate, the main excitatory neurotransmitter in the brain. The glutamate carbon skeleton originates from the tricarboxylic acid (TCA) cycle intermediate α-ketoglutarate, while the amino group is derived from nitrogen donors such as the BCAAs. Disturbances in neurotransmitter homeostasis, mainly of glutamate, are strongly implicated in the pathophysiology of Alzheimer's disease (AD). The divergent BCAA metabolism in different cell types of the human brain is poorly understood, and so is the involvement of astrocytic and neuronal BCAA metabolism in AD. The goal of this study is to provide the first functional characterization of BCAA metabolism in human brain tissue and to investigate BCAA metabolism in AD pathophysiology using astrocytes and neurons derived from human-induced pluripotent stem cells (hiPSCs). Mapping of BCAA metabolism was performed using mass spectrometry and enriched [15N] and [13C] isotopes of leucine, isoleucine, and valine in acutely isolated slices of surgically resected cerebral cortical tissue from human brain and in hiPSC-derived brain cells carrying mutations in either amyloid precursor protein (APP) or presenilin-1 (PSEN-1). We revealed that both human astrocytes of acutely isolated cerebral cortical slices and hiPSC-derived astrocytes were capable of oxidatively metabolizing the carbon skeleton of BCAAs, particularly to support glutamine synthesis. Interestingly, hiPSC-derived astrocytes with APP and PSEN-1 mutations exhibited decreased amino acid synthesis of glutamate, glutamine, and aspartate derived from leucine metabolism. These results clearly demonstrate that there is an active BCAA metabolism in human astrocytes, and that leucine metabolism is selectively impaired in astrocytes derived from the hiPSC models of AD. This impairment in astrocytic BCAA metabolism may contribute to neurotransmitter and energetic imbalances in the AD brain.
Collapse
Affiliation(s)
- Claudia Salcedo
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens V Andersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kasper Tore Vinten
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lars H Pinborg
- Epilepsy Clinic and Neurobiology Research Unit, Copenhagen University Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Helle S Waagepetersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kristine K Freude
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Blanca I Aldana
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
31
|
Bonvento G, Bolaños JP. Astrocyte-neuron metabolic cooperation shapes brain activity. Cell Metab 2021; 33:1546-1564. [PMID: 34348099 DOI: 10.1016/j.cmet.2021.07.006] [Citation(s) in RCA: 234] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 05/11/2021] [Accepted: 07/03/2021] [Indexed: 12/12/2022]
Abstract
The brain has almost no energy reserve, but its activity coordinates organismal function, a burden that requires precise coupling between neurotransmission and energy metabolism. Deciphering how the brain accomplishes this complex task is crucial to understand central facets of human physiology and disease mechanisms. Each type of neural cell displays a peculiar metabolic signature, forcing the intercellular exchange of metabolites that serve as both energy precursors and paracrine signals. The paradigm of this biological feature is the astrocyte-neuron couple, in which the glycolytic metabolism of astrocytes contrasts with the mitochondrial oxidative activity of neurons. Astrocytes generate abundant mitochondrial reactive oxygen species and shuttle to neurons glycolytically derived metabolites, such as L-lactate and L-serine, which sustain energy needs, conserve redox status, and modulate neurotransmitter-receptor activity. Conversely, early disruption of this metabolic cooperation may contribute to the initiation or progression of several neurological diseases, thus requiring innovative therapies to preserve brain energetics.
Collapse
Affiliation(s)
- Gilles Bonvento
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France.
| | - Juan P Bolaños
- Institute of Functional Biology and Genomics (IBFG), Universidad de Salamanca, CSIC, Salamanca, Spain; Centro de Investigación Biomédica en Red sobre Fragilidad y Envejecimiento Saludable (CIBERFES), Institute of Biomedical Research of Salamanca, Salamanca, Spain
| |
Collapse
|
32
|
A O, U M, Lf B, A GC. Energy metabolism in childhood neurodevelopmental disorders. EBioMedicine 2021; 69:103474. [PMID: 34256347 PMCID: PMC8324816 DOI: 10.1016/j.ebiom.2021.103474] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/30/2021] [Accepted: 06/18/2021] [Indexed: 12/24/2022] Open
Abstract
Whereas energy function in the aging brain and their related neurodegenerative diseases has been explored in some detail, there is limited knowledge about molecular mechanisms and brain networks of energy metabolism during infancy and childhood. In this review we describe current insights on physiological brain energetics at prenatal and neonatal stages, and in childhood. We then describe the main groups of inborn errors of energy metabolism affecting the brain. Of note, scarce basic neuroscience research in this field limits the opportunity for these disorders to provide paradigms of energy utilization during neurodevelopment. Finally, we report energy metabolism disturbances in well-known non-metabolic neurodevelopmental disorders. As energy metabolism is a fundamental biological function, brain energy utilization is likely altered in most neuropediatric diseases. Precise knowledge on mechanisms of brain energy disturbance will open the possibility of metabolic modulation therapies regardless of disease etiology.
Collapse
Affiliation(s)
- Oyarzábal A
- Neurometabolic Unit and Laboratory of Synaptic Metabolism. IPR, CIBERER (ISCIII) and MetabERN, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Musokhranova U
- Neurometabolic Unit and Laboratory of Synaptic Metabolism. IPR, CIBERER (ISCIII) and MetabERN, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Barros Lf
- Center for Scientific Studies - CECs, Valdivia 5110466, Chile
| | - García-Cazorla A
- Neurometabolic Unit and Laboratory of Synaptic Metabolism. IPR, CIBERER (ISCIII) and MetabERN, Hospital Sant Joan de Déu, Barcelona, Spain.
| |
Collapse
|
33
|
Pires Monteiro S, Voogd E, Muzzi L, De Vecchis G, Mossink B, Levers M, Hassink G, Van Putten M, Le Feber J, Hofmeijer J, Frega M. Neuroprotective effect of hypoxic preconditioning and neuronal activation in a in vitro human model of the ischemic penumbra. J Neural Eng 2021; 18:036016. [PMID: 33724235 DOI: 10.1088/1741-2552/abe68a] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE In ischemic stroke, treatments to protect neurons from irreversible damage are urgently needed. Studies in animal models have shown that neuroprotective treatments targeting neuronal silencing improve brain recovery, but in clinical trials none of these were effective in patients. This failure of translation poses doubts on the real efficacy of treatments tested and on the validity of animal models for human stroke. Here, we established a human neuronal model of the ischemic penumbra by using human induced pluripotent stem cells and we provided an in-depth characterization of neuronal responses to hypoxia and treatment strategies at the network level. APPROACH We generated neurons from induced pluripotent stem cells derived from healthy donor and we cultured them on micro-electrode arrays. We measured the electrophysiological activity of human neuronal networks under controlled hypoxic conditions. We tested the effect of different treatment strategies on neuronal network functionality. MAIN RESULTS Human neuronal networks are vulnerable to hypoxia reflected by a decrease in activity and synchronicity under low oxygen conditions. We observe that full, partial or absent recovery depend on the timing of re-oxygenation and we provide a critical time threshold that, if crossed, is associated with irreversible impairments. We found that hypoxic preconditioning improves resistance to a second hypoxic insult. Finally, in contrast to previously tested, ineffective treatments, we show that stimulatory treatments counteracting neuronal silencing during hypoxia, such as optogenetic stimulation, are neuroprotective. SIGNIFICANCE We presented a human neuronal model of the ischemic penumbra and we provided insights that may offer the basis for novel therapeutic approaches for patients after stroke. The use of human neurons might improve drug discovery and translation of findings to patients and might open new perspectives for personalized investigations.
Collapse
Affiliation(s)
- Sara Pires Monteiro
- Department of Clinical Neurophysiology, University of Twente, 7522 NB Enschede, The Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Chowdhury HH. Differences in cytosolic glucose dynamics in astrocytes and adipocytes measured by FRET-based nanosensors. Biophys Chem 2020; 261:106377. [PMID: 32302866 DOI: 10.1016/j.bpc.2020.106377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 04/08/2020] [Accepted: 04/08/2020] [Indexed: 11/17/2022]
Abstract
The cellular response to fluctuations in blood glucose levels consists of integrative regulation of cell glucose uptake and glucose utilization in the cytosol, resulting in altered levels of glucose in the cytosol. Cytosolic glucose is difficult to be measured in the intact tissue, however recently methods have become available that allow measurements of glucose in single living cells with fluorescence resonance energy transfer (FRET) based protein sensors. By studying the dynamics of cytosolic glucose levels in different experimental settings, we can gain insights into the properties of plasma membrane permeability to glucose and glucose utilization in the cytosol, and how these processes are modulated by different environmental conditions, agents and enzymes. In this review, we compare the cytosolic regulation of glucose in adipocytes and astrocytes - two important regulators of energy balance and glucose homeostasis in whole body and brain, respectively, with particular emphasis on the data obtained with FRET based protein sensors as well as other biochemical and molecular approaches.
Collapse
Affiliation(s)
- Helena H Chowdhury
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, University of Ljubljana, Faculty of Medicine, 1000 Ljubljana, Slovenia; Laboratory of Cell Engineering, Celica Biomedical, 1000 Ljubljana, Slovenia.
| |
Collapse
|
35
|
Fluid Brain Glycolysis: Limits, Speed, Location, Moonlighting, and the Fates of Glycogen and Lactate. Neurochem Res 2020; 45:1328-1334. [DOI: 10.1007/s11064-020-03005-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 02/26/2020] [Accepted: 02/28/2020] [Indexed: 01/08/2023]
|
36
|
Zuend M, Saab AS, Wyss MT, Ferrari KD, Hösli L, Looser ZJ, Stobart JL, Duran J, Guinovart JJ, Barros LF, Weber B. Arousal-induced cortical activity triggers lactate release from astrocytes. Nat Metab 2020; 2:179-191. [PMID: 32694692 DOI: 10.1038/s42255-020-0170-4] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Accepted: 01/15/2020] [Indexed: 01/01/2023]
Abstract
It has been suggested that, in states of arousal, release of noradrenaline and β-adrenergic signalling affect long-term memory formation by stimulating astrocytic lactate production from glycogen. However, the temporal relationship between cortical activity and cellular lactate fluctuations upon changes in arousal remains to be fully established. Also, the role of β-adrenergic signalling and brain glycogen metabolism on neural lactate dynamics in vivo is still unknown. Here, we show that an arousal-induced increase in cortical activity triggers lactate release into the extracellular space, and this correlates with a fast and prominent lactate dip in astrocytes. The immediate drop in astrocytic lactate concentration and the parallel increase in extracellular lactate levels underline an activity-dependent lactate release from astrocytes. Moreover, when β-adrenergic signalling is blocked or the brain is depleted of glycogen, the arousal-evoked cellular lactate surges are significantly reduced. We provide in vivo evidence that cortical activation upon arousal triggers lactate release from astrocytes, a rise in intracellular lactate levels mediated by β-adrenergic signalling and the mobilization of lactate from glycogen stores.
Collapse
Affiliation(s)
- Marc Zuend
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Aiman S Saab
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Matthias T Wyss
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Kim David Ferrari
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Ladina Hösli
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Zoe J Looser
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Jillian L Stobart
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Jordi Duran
- Institute for Research in Biomedicine, The Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Madrid, Spain
| | - Joan J Guinovart
- Institute for Research in Biomedicine, The Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Madrid, Spain
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona, Barcelona, Spain
| | | | - Bruno Weber
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
37
|
Barros LF, Ruminot I, San Martín A, Lerchundi R, Fernández-Moncada I, Baeza-Lehnert F. Aerobic Glycolysis in the Brain: Warburg and Crabtree Contra Pasteur. Neurochem Res 2020; 46:15-22. [PMID: 31981059 DOI: 10.1007/s11064-020-02964-w] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 01/10/2020] [Accepted: 01/16/2020] [Indexed: 12/20/2022]
Abstract
Information processing is onerous. Curiously, active brain tissue does not fully oxidize glucose and instead generates a local surplus of lactate, a phenomenon termed aerobic glycolysis. Why engage in inefficient ATP production by glycolysis when energy demand is highest and oxygen is plentiful? Aerobic glycolysis is associated to classic biochemical effects known by the names of Pasteur, Warburg and Crabtree. Here we discuss these three interdependent phenomena in brain cells, in light of high-resolution data of neuronal and astrocytic metabolism in culture, tissue slices and in vivo, acquired with genetically-encoded fluorescent sensors. These sensors are synthetic proteins that can be targeted to specific cell types and subcellular compartments, which change their fluorescence in response to variations in metabolite concentration. A major site of acute aerobic glycolysis is the astrocyte. In this cell, a Crabtree effect triggered by K+ coincides with a Warburg effect mediated by NO, superimposed on a slower longer-lasting Warburg effect caused by glutamate and possibly by NH4+. The compounded outcome is that more fuel (lactate) and more oxygen are made available to neurons, on demand. Meanwhile neurons consume both glucose and lactate, maintaining a strict balance between glycolysis and respiration, commanded by the Na+ pump. We conclude that activity-dependent Warburg and Crabtree effects in brain tissue, and the resulting aerobic glycolysis, do not reflect inefficient energy generation but the marshalling of astrocytes for the purpose of neuronal ATP generation. It remains to be seen whether neurons contribute to aerobic glycolysis under physiological conditions.
Collapse
Affiliation(s)
- L Felipe Barros
- Centro de Estudios Científicos-CECs, 5110466, Valdivia, Chile.
| | - Iván Ruminot
- Centro de Estudios Científicos-CECs, 5110466, Valdivia, Chile
| | | | | | | | | |
Collapse
|
38
|
Bernardini A, Wolf A, Brockmeier U, Riffkin H, Metzen E, Acker-Palmer A, Fandrey J, Acker H. Carotid body type I cells engage flavoprotein and Pin1 for oxygen sensing. Am J Physiol Cell Physiol 2020; 318:C719-C731. [PMID: 31967857 DOI: 10.1152/ajpcell.00320.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Carotid body (CB) type I cells sense the blood Po2 and generate a nervous signal for stimulating ventilation and circulation when blood oxygen levels decline. Three oxygen-sensing enzyme complexes may be used for this purpose: 1) mitochondrial electron transport chain metabolism, 2) heme oxygenase 2 (HO-2)-generating CO, and/or 3) an NAD(P)H oxidase (NOX). We hypothesize that intracellular redox changes are the link between the sensor and nervous signals. To test this hypothesis type I cell autofluorescence of flavoproteins (Fp) and NAD(P)H within the mouse CB ex vivo was recorded as Fp/(Fp+NAD(P)H) redox ratio. CB type I cell redox ratio transiently declined with the onset of hypoxia. Upon reoxygenation, CB type I cells showed a significantly increased redox ratio. As a control organ, the non-oxygen-sensing sympathetic superior cervical ganglion (SCG) showed a continuously reduced redox ratio upon hypoxia. CN-, diphenyleneiodonium, or reactive oxygen species influenced chemoreceptor discharge (CND) with subsequent loss of O2 sensitivity and inhibited hypoxic Fp reduction only in the CB but not in SCG Fp, indicating a specific role of Fp in the oxygen-sensing process. Hypoxia-induced changes in CB type I cell redox ratio affected peptidyl prolyl isomerase Pin1, which is believed to colocalize with the NADPH oxidase subunit p47phox in the cell membrane to trigger the opening of potassium channels. We postulate that hypoxia-induced changes in the Fp-mediated redox ratio of the CB regulate the Pin1/p47phox tandem to alter type I cell potassium channels and therewith CND.
Collapse
Affiliation(s)
- André Bernardini
- Institute of Physiology, University of Duisburg-Essen, Essen, Germany
| | - Alexandra Wolf
- Institute of Physiology, University of Duisburg-Essen, Essen, Germany
| | - Ulf Brockmeier
- Institute of Physiology, University of Duisburg-Essen, Essen, Germany
| | - Helena Riffkin
- Institute of Physiology, University of Duisburg-Essen, Essen, Germany
| | - Eric Metzen
- Institute of Physiology, University of Duisburg-Essen, Essen, Germany
| | - Amparo Acker-Palmer
- Institute for Cell Biology and Neuroscience, Goethe University, Frankfurt, Germany
| | - Joachim Fandrey
- Institute of Physiology, University of Duisburg-Essen, Essen, Germany
| | - Helmut Acker
- Institute of Physiology, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
39
|
González-Gutiérrez A, Ibacache A, Esparza A, Barros LF, Sierralta J. Neuronal lactate levels depend on glia-derived lactate during high brain activity in Drosophila. Glia 2019; 68:1213-1227. [PMID: 31876077 DOI: 10.1002/glia.23772] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 12/10/2019] [Accepted: 12/10/2019] [Indexed: 01/13/2023]
Abstract
Lactate/pyruvate transport between glial cells and neurons is thought to play an important role in how brain cells sustain the high-energy demand that neuronal activity requires. However, the in vivo mechanisms and characteristics that underlie the transport of monocarboxylates are poorly described. Here, we use Drosophila expressing genetically encoded FRET sensors to provide an ex vivo characterization of the transport of monocarboxylates in motor neurons and glial cells from the larval ventral nerve cord. We show that lactate/pyruvate transport in glial cells is coupled to protons and is more efficient than in neurons. Glial cells maintain higher levels of intracellular lactate generating a positive gradient toward neurons. Interestingly, during high neuronal activity, raised lactate in motor neurons is dependent on transfer from glial cells mediated in part by the previously described monocarboxylate transporter Chaski, providing support for in vivo glia-to-neuron lactate shuttling during neuronal activity.
Collapse
Affiliation(s)
- Andrés González-Gutiérrez
- Department of Neuroscience and Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Andrés Ibacache
- Department of Neuroscience and Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Andrés Esparza
- Department of Neuroscience and Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | | | - Jimena Sierralta
- Department of Neuroscience and Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| |
Collapse
|
40
|
Contreras-Baeza Y, Ceballo S, Arce-Molina R, Sandoval PY, Alegría K, Barros LF, San Martín A. MitoToxy assay: A novel cell-based method for the assessment of metabolic toxicity in a multiwell plate format using a lactate FRET nanosensor, Laconic. PLoS One 2019; 14:e0224527. [PMID: 31671132 PMCID: PMC6822764 DOI: 10.1371/journal.pone.0224527] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 10/15/2019] [Indexed: 12/13/2022] Open
Abstract
Mitochondrial toxicity is a primary source of pre-clinical drug attrition, black box warning and post-market drug withdrawal. Methods that detect mitochondrial toxicity as early as possible during the drug development process are required. Here we introduce a new method for detecting mitochondrial toxicity based on MDA-MB-231 cells stably expressing the genetically encoded FRET lactate indicator, Laconic. The method takes advantage of the high cytosolic lactate accumulation observed during mitochondrial stress, regardless of the specific toxicity mechanism, explained by compensatory glycolytic activation. Using a standard multi-well plate reader, dose-response curve experiments allowed the sensitivity of the methodology to detect metabolic toxicity induced by classical mitochondrial toxicants. Suitability for high-throughput screening applications was evaluated resulting in a Z’-factor > 0.5 and CV% < 20 inter-assay variability. A pilot screening allowed sensitive detection of commercial drugs that were previously withdrawn from the market due to liver/cardiac toxicity issues, such as camptothecin, ciglitazone, troglitazone, rosiglitazone, and terfenadine, in ten minutes. We envisage that the availability of this technology, based on a fluorescent genetically encoded indicator, will allow direct assessment of mitochondrial metabolism, and will make the early detection of mitochondrial toxicity in the drug development process possible, saving time and resources.
Collapse
Affiliation(s)
| | | | - Robinson Arce-Molina
- Centro de Estudios Científicos (CECs), Valdivia, Chile
- Universidad Austral de Chile (UACh), Valdivia, Chile
| | | | - Karin Alegría
- Centro de Estudios Científicos (CECs), Valdivia, Chile
| | | | | |
Collapse
|
41
|
Martano G, Borroni EM, Lopci E, Cattaneo MG, Mattioli M, Bachi A, Decimo I, Bifari F. Metabolism of Stem and Progenitor Cells: Proper Methods to Answer Specific Questions. Front Mol Neurosci 2019; 12:151. [PMID: 31249511 PMCID: PMC6584756 DOI: 10.3389/fnmol.2019.00151] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 05/28/2019] [Indexed: 01/01/2023] Open
Abstract
Stem cells can stay quiescent for a long period of time or proliferate and differentiate into multiple lineages. The activity of stage-specific metabolic programs allows stem cells to best adapt their functions in different microenvironments. Specific cellular phenotypes can be, therefore, defined by precise metabolic signatures. Notably, not only cellular metabolism describes a defined cellular phenotype, but experimental evidence now clearly indicate that also rewiring cells towards a particular cellular metabolism can drive their cellular phenotype and function accordingly. Cellular metabolism can be studied by both targeted and untargeted approaches. Targeted analyses focus on a subset of identified metabolites and on their metabolic fluxes. In addition, the overall assessment of the oxygen consumption rate (OCR) gives a measure of the overall cellular oxidative metabolism and mitochondrial function. Untargeted approach provides a large-scale identification and quantification of the whole metabolome with the aim to describe a metabolic fingerprinting. In this review article, we overview the methodologies currently available for the study of invitro stem cell metabolism, including metabolic fluxes, fingerprint analyses, and single-cell metabolomics. Moreover, we summarize available approaches for the study of in vivo stem cell metabolism. For all of the described methods, we highlight their specificities and limitations. In addition, we discuss practical concerns about the most threatening steps, including metabolic quenching, sample preparation and extraction. A better knowledge of the precise metabolic signature defining specific cell population is instrumental to the design of novel therapeutic strategies able to drive undifferentiated stem cells towards a selective and valuable cellular phenotype.
Collapse
Affiliation(s)
| | - Elena Monica Borroni
- Humanitas Clinical and Research Center, Rozzano, Italy.,Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Egesta Lopci
- Nuclear Medicine Unit, Humanitas Clinical and Research Hospital-IRCCS, Rozzano, Italy
| | - Maria Grazia Cattaneo
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Milena Mattioli
- Laboratory of Cell Metabolism and Regenerative Medicine, Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Angela Bachi
- IFOM-FIRC Institute of Molecular Oncology, Milan, Italy
| | - Ilaria Decimo
- Laboratory of Pharmacology, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Francesco Bifari
- Laboratory of Cell Metabolism and Regenerative Medicine, Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| |
Collapse
|
42
|
Andersen JV, Skotte NH, Aldana BI, Nørremølle A, Waagepetersen HS. Enhanced cerebral branched-chain amino acid metabolism in R6/2 mouse model of Huntington's disease. Cell Mol Life Sci 2019; 76:2449-2461. [PMID: 30830240 PMCID: PMC11105563 DOI: 10.1007/s00018-019-03051-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 01/23/2019] [Accepted: 02/19/2019] [Indexed: 01/01/2023]
Abstract
Huntington's disease (HD) is a hereditary and fatal disease causing profound neurodegeneration. Deficits in cerebral energy and neurotransmitter metabolism have been suggested to play a central role in the neuronal dysfunction and death associated with HD. The branched-chain amino acids (BCAAs), leucine, isoleucine and valine, are important for cerebral nitrogen homeostasis, neurotransmitter recycling and can be utilized as energy substrates in the tricarboxylic acid (TCA) cycle. Reduced levels of BCAAs in HD have been validated by several reports. However, it is still unknown how cerebral BCAA metabolism is regulated in HD. Here we investigate the metabolism of leucine and isoleucine in the R6/2 mouse model of HD. Acutely isolated cerebral cortical and striatal slices of control and R6/2 mice were incubated in media containing 15N- or 13C-labeled leucine or isoleucine and slice extracts were analyzed by gas chromatography-mass spectrometry (GC-MS) to determine isotopic enrichment of derived metabolites. Elevated BCAA transamination was found from incubations with [15N]leucine and [15N]isoleucine, in both cerebral cortical and striatal slices of R6/2 mice compared to controls. Metabolism of [U-13C]leucine and [U-13C]isoleucine, entering oxidative metabolism as acetyl CoA, was maintained in R6/2 mice. However, metabolism of [U-13C]isoleucine, entering the TCA cycle as succinyl CoA, was elevated in both cerebral cortical and striatal slices of R6/2 mice, suggesting enhanced metabolic flux via this anaplerotic pathway. To support the metabolic studies, expression of enzymes in the BCAA metabolic pathway was assessed from a proteomic resource. Several enzymes related to BCAA metabolism were found to exhibit augmented expression in the R6/2 brain, particularly related to isoleucine metabolism, suggesting an increase in the BCAA metabolic machinery. Our results show that the capacity for cerebral BCAA metabolism, predominantly of isoleucine, is amplified in the R6/2 brain and indicates that perturbations in cerebral BCAA homeostasis could have functional consequences for HD pathology.
Collapse
Affiliation(s)
- Jens V Andersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
| | - Niels H Skotte
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Blanca I Aldana
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
| | - Anne Nørremølle
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Helle S Waagepetersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark.
| |
Collapse
|
43
|
Rubio-Villena C, Viana R, Bonet J, Garcia-Gimeno MA, Casado M, Heredia M, Sanz P. Astrocytes: new players in progressive myoclonus epilepsy of Lafora type. Hum Mol Genet 2019; 27:1290-1300. [PMID: 29408991 DOI: 10.1093/hmg/ddy044] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 01/30/2018] [Indexed: 12/16/2022] Open
Abstract
Lafora disease (LD) is a fatal form of progressive myoclonus epilepsy characterized by the accumulation of insoluble poorly branched glycogen-like inclusions named Lafora bodies (LBs) in the brain and peripheral tissues. In the brain, since its first discovery in 1911, it was assumed that these glycogen inclusions were only present in affected neurons. Mouse models of LD have been obtained recently, and we and others have been able to report the accumulation of glycogen inclusions in the brain of LD animals, what recapitulates the hallmark of the disease. In this work we present evidence indicating that, although in mouse models of LD glycogen inclusions co-localize with neurons, as originally established, most of them co-localize with astrocytic markers such as glial fibrillary acidic protein (GFAP) and glutamine synthase. In addition, we have observed that primary cultures of astrocytes from LD mouse models accumulate higher levels of glycogen than controls. These results suggest that astrocytes may play a crucial role in the pathophysiology of Lafora disease, as the accumulation of glycogen inclusions in these cells may affect their regular functionality leading them to a possible neuronal dysfunction.
Collapse
Affiliation(s)
- Carla Rubio-Villena
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas (IBV-CSIC), Valencia, Spain
| | - Rosa Viana
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas (IBV-CSIC), Valencia, Spain
| | - Jose Bonet
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas (IBV-CSIC), Valencia, Spain
| | | | - Marta Casado
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas (IBV-CSIC), Valencia, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), (Group CB06/04/1069) Madrid, Spain
| | - Miguel Heredia
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), (Group U742) Valencia, Spain
| | - Pascual Sanz
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas (IBV-CSIC), Valencia, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), (Group U742) Valencia, Spain
| |
Collapse
|
44
|
Carter SF, Herholz K, Rosa-Neto P, Pellerin L, Nordberg A, Zimmer ER. Astrocyte Biomarkers in Alzheimer's Disease. Trends Mol Med 2019; 25:77-95. [PMID: 30611668 DOI: 10.1016/j.molmed.2018.11.006] [Citation(s) in RCA: 208] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 11/30/2018] [Accepted: 11/30/2018] [Indexed: 01/01/2023]
Abstract
Astrocytic contributions to Alzheimer's disease (AD) progression were, until recently, largely overlooked. Astrocytes are integral to normal brain function and astrocyte reactivity is an early feature of AD, potentially providing a promising target for preclinical diagnosis and treatment. Several in vivo AD biomarkers already exist, but presently there is a paucity of specific and sensitive in vivo astrocyte biomarkers that can accurately measure preclinical AD. Measuring monoamine oxidase-B with neuroimaging and glial fibrillary acidic protein from bodily fluids are biomarkers that are currently available. Developing novel, more specific, and sensitive astrocyte biomarkers will make it possible to pharmaceutically target chemical pathways that preserve beneficial astrocytic functions in response to AD pathology. This review discusses astrocyte biomarkers in the context of AD.
Collapse
Affiliation(s)
- Stephen F Carter
- Wolfson Molecular Imaging Centre, Division of Neuroscience and Experimental Psychology, University of Manchester, Manchester, United Kingdom
| | - Karl Herholz
- Wolfson Molecular Imaging Centre, Division of Neuroscience and Experimental Psychology, University of Manchester, Manchester, United Kingdom
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, Canada; Douglas Hospital Research Centre, Montreal, Canada; Montreal Neurological Institute, Montreal, Canada
| | - Luc Pellerin
- Département de Physiologie, Université de Lausanne, Lausanne, Switzerland; Centre de Résonance Magnétique des Systèmes Biologiques, UMR5536 CNRS, LabEx TRAIL-IBIO, Université de Bordeaux, Bordeaux Cedex 33760, France
| | - Agneta Nordberg
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Stockholm, Sweden; Theme Aging, Karolinska University Hospital, Huddinge, Sweden
| | - Eduardo R Zimmer
- Department of Pharmacology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Graduate Program in Biological Sciences: Pharmacology and Therapeutics, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Brain Institute (BraIns) of Rio Grande do Sul, Porto Alegre, Brazil; Website: www.zimmer-lab.org.
| |
Collapse
|
45
|
Zhang Z, Chen W, Zhao Y, Yang Y. Spatiotemporal Imaging of Cellular Energy Metabolism with Genetically-Encoded Fluorescent Sensors in Brain. Neurosci Bull 2018; 34:875-886. [PMID: 29679217 PMCID: PMC6129245 DOI: 10.1007/s12264-018-0229-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 03/24/2018] [Indexed: 12/21/2022] Open
Abstract
The brain has very high energy requirements and consumes 20% of the oxygen and 25% of the glucose in the human body. Therefore, the molecular mechanism underlying how the brain metabolizes substances to support neural activity is a fundamental issue for neuroscience studies. A well-known model in the brain, the astrocyte-neuron lactate shuttle, postulates that glucose uptake and glycolytic activity are enhanced in astrocytes upon neuronal activation and that astrocytes transport lactate into neurons to fulfill their energy requirements. Current evidence for this hypothesis has yet to reach a clear consensus, and new concepts beyond the shuttle hypothesis are emerging. The discrepancy is largely attributed to the lack of a critical method for real-time monitoring of metabolic dynamics at cellular resolution. Recent advances in fluorescent protein-based sensors allow the generation of a sensitive, specific, real-time readout of subcellular metabolites and fill the current technological gap. Here, we summarize the development of genetically encoded metabolite sensors and their applications in assessing cell metabolism in living cells and in vivo, and we believe that these tools will help to address the issue of elucidating neural energy metabolism.
Collapse
Affiliation(s)
- Zhuo Zhang
- Synthetic Biology and Biotechnology Laboratory, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, Shanghai, 200237, China
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Weicai Chen
- Synthetic Biology and Biotechnology Laboratory, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, Shanghai, 200237, China
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Yuzheng Zhao
- Synthetic Biology and Biotechnology Laboratory, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, Shanghai, 200237, China.
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China.
| | - Yi Yang
- Synthetic Biology and Biotechnology Laboratory, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, Shanghai, 200237, China.
- Optogenetics and Synthetic Biology Interdisciplinary Research Center, CAS Center for Excellence in Brain Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
46
|
Piquet J, Toussay X, Hepp R, Lerchundi R, Le Douce J, Faivre É, Guiot E, Bonvento G, Cauli B. Supragranular Pyramidal Cells Exhibit Early Metabolic Alterations in the 3xTg-AD Mouse Model of Alzheimer's Disease. Front Cell Neurosci 2018; 12:216. [PMID: 30072874 PMCID: PMC6060432 DOI: 10.3389/fncel.2018.00216] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 06/29/2018] [Indexed: 01/23/2023] Open
Abstract
The impairment of cerebral glucose utilization is an early and predictive biomarker of Alzheimer’s disease (AD) that is likely to contribute to memory and cognition disorders during the progression of the pathology. Yet, the cellular and molecular mechanisms underlying these metabolic alterations remain poorly understood. Here we studied the glucose metabolism of supragranular pyramidal cells at an early presymptomatic developmental stage in non-transgenic (non-Tg) and 3xTg-AD mice, a mouse model of AD replicating numerous hallmarks of the disease. We performed both intracellular glucose imaging with a genetically encoded fluorescence resonance energy transfer (FRET)-based glucose biosensor and transcriptomic profiling of key molecular elements of glucose metabolism with single-cell multiplex RT-PCR (scRT-mPCR). We found that juvenile pyramidal cells exhibit active glycolysis and pentose phosphate pathway at rest that are respectively enhanced and impaired in 3xTg-AD mice without alteration of neuronal glucose uptake or transcriptional modification. Given the importance of glucose metabolism for neuronal survival, these early alterations could initiate or at least contribute to the later neuronal dysfunction of pyramidal cells in AD.
Collapse
Affiliation(s)
- Juliette Piquet
- UPMC Univ Paris 06, INSERM, CNRS, Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), Sorbonne Université, Paris, France
| | - Xavier Toussay
- UPMC Univ Paris 06, INSERM, CNRS, Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), Sorbonne Université, Paris, France
| | - Régine Hepp
- UPMC Univ Paris 06, INSERM, CNRS, Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), Sorbonne Université, Paris, France
| | - Rodrigo Lerchundi
- CNRS UMR 9199, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), Département de la Recherche Fondamentale (DRF), Institut de Biologie François Jacob, Molecular Imaging Center (MIRCen), Université Paris-Sud, Université Paris-Saclay, Paris, France
| | - Juliette Le Douce
- CNRS UMR 9199, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), Département de la Recherche Fondamentale (DRF), Institut de Biologie François Jacob, Molecular Imaging Center (MIRCen), Université Paris-Sud, Université Paris-Saclay, Paris, France
| | - Émilie Faivre
- CNRS UMR 9199, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), Département de la Recherche Fondamentale (DRF), Institut de Biologie François Jacob, Molecular Imaging Center (MIRCen), Université Paris-Sud, Université Paris-Saclay, Paris, France
| | - Elvire Guiot
- UPMC Univ Paris 06, INSERM, CNRS, Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), Sorbonne Université, Paris, France
| | - Gilles Bonvento
- CNRS UMR 9199, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), Département de la Recherche Fondamentale (DRF), Institut de Biologie François Jacob, Molecular Imaging Center (MIRCen), Université Paris-Sud, Université Paris-Saclay, Paris, France
| | - Bruno Cauli
- UPMC Univ Paris 06, INSERM, CNRS, Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), Sorbonne Université, Paris, France
| |
Collapse
|
47
|
Lenting K, Khurshed M, Peeters TH, van den Heuvel CNAM, van Lith SAM, de Bitter T, Hendriks W, Span PN, Molenaar RJ, Botman D, Verrijp K, Heerschap A, Ter Laan M, Kusters B, van Ewijk A, Huynen MA, van Noorden CJF, Leenders WPJ. Isocitrate dehydrogenase 1-mutated human gliomas depend on lactate and glutamate to alleviate metabolic stress. FASEB J 2018; 33:557-571. [PMID: 30001166 DOI: 10.1096/fj.201800907rr] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Diffuse gliomas often carry point mutations in isocitrate dehydrogenase ( IDH1mut), resulting in metabolic stress. Although IDHmut gliomas are difficult to culture in vitro, they thrive in the brain via diffuse infiltration, suggesting brain-specific tumor-stroma interactions that can compensate for IDH-1 deficits. To elucidate the metabolic adjustments in clinical IDHmut gliomas that contribute to their malignancy, we applied a recently developed method of targeted quantitative RNA next-generation sequencing to 66 clinical gliomas and relevant orthotopic glioma xenografts, with and without the endogenous IDH-1R132H mutation. Datasets were analyzed in R using Manhattan plots to calculate distance between expression profiles, Ward's method to perform unsupervised agglomerative clustering, and the Mann Whitney U test and Fisher's exact tests for supervised group analyses. The significance of transcriptome data was investigated by protein analysis, in situ enzymatic activity mapping, and in vivo magnetic resonance spectroscopy of orthotopic IDH1mut- and IDHwt-glioma xenografts. Gene set enrichment analyses of clinical IDH1mut gliomas strongly suggest a role for catabolism of lactate and the neurotransmitter glutamate, whereas, in IDHwt gliomas, processing of glucose and glutamine are the predominant metabolic pathways. Further evidence of the differential metabolic activity in these cancers comes from in situ enzymatic mapping studies and preclinical in vivo magnetic resonance spectroscopy imaging. Our data support an evolutionary model in which IDHmut glioma cells exist in symbiosis with supportive neuronal cells and astrocytes as suppliers of glutamate and lactate, possibly explaining the diffuse nature of these cancers. The dependency on glutamate and lactate opens the way for novel approaches in the treatment of IDHmut gliomas.-Lenting, K., Khurshed, M., Peeters, T. H., van den Heuvel, C. N. A. M., van Lith, S. A. M., de Bitter, T., Hendriks, W., Span, P. N., Molenaar, R. J., Botman, D., Verrijp, K., Heerschap, A., ter Laan, M., Kusters, B., van Ewijk, A., Huynen, M. A., van Noorden, C. J. F., Leenders, W. P. J. Isocitrate dehydrogenase 1-mutated human gliomas depend on lactate and glutamate to alleviate metabolic stress.
Collapse
Affiliation(s)
- Krissie Lenting
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands.,Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mohammed Khurshed
- Department of Medical Biology, Cancer Center Amsterdam, Academic Medical Centre, Amsterdam, The Netherlands
| | - Tom H Peeters
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Corina N A M van den Heuvel
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands.,Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Sanne A M van Lith
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Tessa de Bitter
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Wiljan Hendriks
- Department of Cell Biology, Radboud Institute of Molecular Life Sciences, Nijmegen, The Netherlands
| | - Paul N Span
- Radiotherapy and Oncoimmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Remco J Molenaar
- Department of Medical Biology, Cancer Center Amsterdam, Academic Medical Centre, Amsterdam, The Netherlands
| | - Dennis Botman
- Department of Medical Biology, Cancer Center Amsterdam, Academic Medical Centre, Amsterdam, The Netherlands
| | - Kiek Verrijp
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Arend Heerschap
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mark Ter Laan
- Department of Neurosurgery, Radboud University Medical Center, Nijmegen, The Netherlands; and
| | - Benno Kusters
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Anne van Ewijk
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Martijn A Huynen
- Center for Molecular and Biomolecular Informatics, Radboud Institute of Molecular Life Sciences, Nijmegen, The Netherlands
| | - Cornelis J F van Noorden
- Department of Medical Biology, Cancer Center Amsterdam, Academic Medical Centre, Amsterdam, The Netherlands
| | - William P J Leenders
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands.,Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
48
|
Barros LF, Bolaños JP, Bonvento G, Bouzier-Sore AK, Brown A, Hirrlinger J, Kasparov S, Kirchhoff F, Murphy AN, Pellerin L, Robinson MB, Weber B. Current technical approaches to brain energy metabolism. Glia 2018; 66:1138-1159. [PMID: 29110344 PMCID: PMC5903992 DOI: 10.1002/glia.23248] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 09/14/2017] [Accepted: 10/04/2017] [Indexed: 12/19/2022]
Abstract
Neuroscience is a technology-driven discipline and brain energy metabolism is no exception. Once satisfied with mapping metabolic pathways at organ level, we are now looking to learn what it is exactly that metabolic enzymes and transporters do and when, where do they reside, how are they regulated, and how do they relate to the specific functions of neurons, glial cells, and their subcellular domains and organelles, in different areas of the brain. Moreover, we aim to quantify the fluxes of metabolites within and between cells. Energy metabolism is not just a necessity for proper cell function and viability but plays specific roles in higher brain functions such as memory processing and behavior, whose mechanisms need to be understood at all hierarchical levels, from isolated proteins to whole subjects, in both health and disease. To this aim, the field takes advantage of diverse disciplines including anatomy, histology, physiology, biochemistry, bioenergetics, cellular biology, molecular biology, developmental biology, neurology, and mathematical modeling. This article presents a well-referenced synopsis of the technical side of brain energy metabolism research. Detail and jargon are avoided whenever possible and emphasis is given to comparative strengths, limitations, and weaknesses, information that is often not available in regular articles.
Collapse
Affiliation(s)
- L Felipe Barros
- Centro de Estudios Científicos (CECs), Valdivia, 5110466, Chile
| | - Juan P Bolaños
- Instituto de Biologia Funcional y Genomica-CSIC, Universidad de Salamanca, CIBERFES, Salamanca, 37007, Spain
| | - Gilles Bonvento
- Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Département de la Recherche Fondamentale (DRF), Institut de Biologie François Jacob, Molecular Imaging Research Center (MIRCen), CNRS UMR 9199, Université Paris-Sud, Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Anne-Karine Bouzier-Sore
- Centre de Résonance Magnétique des Systèmes Biologiques UMR 5536, CNRS-Université Bordeaux 146 rue Léo-Saignat, Bordeaux, France
| | - Angus Brown
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Johannes Hirrlinger
- Carl Ludwig Institute of Physiology, University of Leipzig, Liebigstr. 27, D-04103, Leipzig, Germany
- Department of Neurogenetics, Max-Planck-Institute for Experimental Medicine, Hermann-Rein-Str. 3, Göttingen, D-37075, Germany
| | - Sergey Kasparov
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, University Walk, BS8 1TD, United Kingdom
- Baltic Federal University, Kalinigrad, Russian Federation
| | - Frank Kirchhoff
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, Building 48, Homburg, 66421, Germany
| | - Anne N Murphy
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, 92093
| | - Luc Pellerin
- Département de Physiologie, 7 rue du Bugnon, Lausanne, CH1005, Switzerland
| | - Michael B Robinson
- Department of Pediatrics, and Department of Systems Pharmacology and Translational Therapeutics, Children's Hospital of Philadelphia Research Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Bruno Weber
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, Zurich, Switzerland
| |
Collapse
|
49
|
Abstract
Lactate in the brain has long been associated with ischaemia; however, more recent evidence shows that it can be found there under physiological conditions. In the brain, lactate is formed predominantly in astrocytes from glucose or glycogen in response to neuronal activity signals. Thus, neurons and astrocytes show tight metabolic coupling. Lactate is transferred from astrocytes to neurons to match the neuronal energetic needs, and to provide signals that modulate neuronal functions, including excitability, plasticity and memory consolidation. In addition, lactate affects several homeostatic functions. Overall, lactate ensures adequate energy supply, modulates neuronal excitability levels and regulates adaptive functions in order to set the 'homeostatic tone' of the nervous system.
Collapse
|
50
|
Neuronal control of astrocytic respiration through a variant of the Crabtree effect. Proc Natl Acad Sci U S A 2018; 115:1623-1628. [PMID: 29378955 DOI: 10.1073/pnas.1716469115] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Aerobic glycolysis is a phenomenon that in the long term contributes to synaptic formation and growth, is reduced by normal aging, and correlates with amyloid beta deposition. Aerobic glycolysis starts within seconds of neural activity and it is not obvious why energetic efficiency should be compromised precisely when energy demand is highest. Using genetically encoded FRET nanosensors and real-time oxygen measurements in culture and in hippocampal slices, we show here that astrocytes respond to physiological extracellular K+ with an acute rise in cytosolic ATP and a parallel inhibition of oxygen consumption, explained by glycolytic stimulation via the Na+-bicarbonate cotransporter NBCe1. This control of mitochondrial respiration via glycolysis modulation is reminiscent of a phenomenon previously described in proliferating cells, known as the Crabtree effect. Fast brain aerobic glycolysis may be interpreted as a strategy whereby neurons manipulate neighboring astrocytes to obtain oxygen, thus maximizing information processing.
Collapse
|