1
|
Boltri M, Scalia A, Brusa F, Manzo F, Apicella E, Mendolicchio L. Keto therapy-unveiling the potential of ketogenic diet in psychiatric care: A scoping review. Nutrition 2025; 134:112710. [PMID: 40043464 DOI: 10.1016/j.nut.2025.112710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 01/28/2025] [Accepted: 02/05/2025] [Indexed: 04/12/2025]
Abstract
BACKGROUND Within the framework of gut-brain interactions, the ketogenic diet (KD), a high-fat, low-carbohydrate, and moderate-protein dietary intervention, has gained attention for its potential in psychiatric care. Renowned for its efficacy in managing obesity, KD has demonstrated effectiveness in facilitating weight loss and inducing favorable metabolic changes in the short term. With its established benefits in neurological disorders, KD is now being investigated as a potential therapeutic avenue for individuals with psychiatric conditions. OBJECTIVE This scoping review aims to summarize the latest studies on this topic, consider clinical implications, and suggest future research directions. METHODS Records were systematically (PRISMA-ScR guidelines) identified through PubMed, Scopus, and EBSCOhost searches. RESULTS A total of 58 studies were initially identified, with 13 meeting the eligibility criteria. While clinical trials remain limited, emerging evidence from case reports, case series, and pilot studies highlights the potential of a KD in reducing symptoms across psychiatric conditions, including mood disorders, psychotic disorders, alcohol use, and eating disorders. A KD shows potential in reducing cravings in eating and alcohol use disorders by stabilizing brain metabolism and modulating addictive behaviors, while its neuroprotective and anti-inflammatory effects may contribute to symptom improvement in mood and psychotic disorders. CONCLUSIONS Implementing a KD in patients with mental disorders seems to be a feasible and well-tolerated approach, resulting in psychiatric symptom reduction and improvements in metabolic health. Most interestingly, research suggests that KD can also be safely implemented in the care of patients with addictive-like eating disorders.
Collapse
Affiliation(s)
- Margherita Boltri
- I.R.C.C.S. Istituto Auxologico Italiano, Experimental Laboratory for Metabolic Neurosciences Research, Piancavallo, VCO, Italy; Psychology Department, Università Cattolica del Sacro Cuore, Milan, Italy.
| | - Alberto Scalia
- I.R.C.C.S. Istituto Auxologico Italiano, Experimental Laboratory for Metabolic Neurosciences Research, Piancavallo, VCO, Italy
| | - Federico Brusa
- I.R.C.C.S. Istituto Auxologico Italiano, Experimental Laboratory for Metabolic Neurosciences Research, Piancavallo, VCO, Italy
| | - Francesca Manzo
- I.R.C.C.S. Istituto Auxologico Italiano, Experimental Laboratory for Metabolic Neurosciences Research, Piancavallo, VCO, Italy
| | - Emanuela Apicella
- I.R.C.C.S. Istituto Auxologico Italiano, Experimental Laboratory for Metabolic Neurosciences Research, Piancavallo, VCO, Italy
| | - Leonardo Mendolicchio
- I.R.C.C.S. Istituto Auxologico Italiano, Experimental Laboratory for Metabolic Neurosciences Research, Piancavallo, VCO, Italy
| |
Collapse
|
2
|
Thieren L, Zanker HS, Droux J, Dalvi U, Wyss MT, Waag R, Germain PL, von Ziegler LM, Looser ZJ, Hösli L, Ravotto L, Abel ED, Bohacek J, Wegener S, Barros LF, El Amki M, Weber B, Saab AS. Astrocytic GLUT1 deletion in adult mice enhances glucose metabolism and resilience to stroke. Nat Commun 2025; 16:4190. [PMID: 40328784 PMCID: PMC12056070 DOI: 10.1038/s41467-025-59400-2] [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: 07/12/2024] [Accepted: 04/22/2025] [Indexed: 05/08/2025] Open
Abstract
Brain activity relies on a steady supply of blood glucose. Astrocytes express glucose transporter 1 (GLUT1), considered their primary route for glucose uptake to sustain metabolic and antioxidant support for neurons. While GLUT1 deficiency causes severe developmental impairments, its role in adult astrocytes remains unclear. Here, we show that astrocytes and neurons tolerate the inducible, astrocyte-specific deletion of GLUT1 in adulthood. Sensorimotor and memory functions remain intact in male GLUT1 cKO mice, indicating that GLUT1 loss does not impair behavior. Despite GLUT1 loss, two-photon glucose sensor imaging reveals that astrocytes maintain normal resting glucose levels but exhibit a more than two-fold increase in glucose consumption, indicating enhanced metabolic activity. Notably, male GLUT1 cKO mice display reduced infarct volumes following stroke, suggesting a neuroprotective effect of increased astrocytic glucose metabolism. Our findings reveal metabolic adaptability in astrocytes, ensuring glucose uptake and neuronal support despite the absence of their primary transporter.
Collapse
Affiliation(s)
- Laetitia Thieren
- University of Zurich, Institute of Pharmacology and Toxicology, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Henri S Zanker
- University of Zurich, Institute of Pharmacology and Toxicology, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Jeanne Droux
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Urvashi Dalvi
- University of Zurich, Institute of Pharmacology and Toxicology, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Matthias T Wyss
- University of Zurich, Institute of Pharmacology and Toxicology, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Rebecca Waag
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
- Lab of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Pierre-Luc Germain
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
- Lab of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Lab of Statistical Bioinformatics, University of Zurich, Zurich, Switzerland
| | - Lukas M von Ziegler
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
- Lab of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Zoe J Looser
- University of Zurich, Institute of Pharmacology and Toxicology, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Ladina Hösli
- University of Zurich, Institute of Pharmacology and Toxicology, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Luca Ravotto
- University of Zurich, Institute of Pharmacology and Toxicology, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - E Dale Abel
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Johannes Bohacek
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
- Lab of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Susanne Wegener
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | - L Felipe Barros
- Centro de Estudios Científicos (CECs), Valdivia, Chile
- Facultad de Medicina, Universidad San Sebastián, Valdivia, Chile
| | - Mohamad El Amki
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Bruno Weber
- University of Zurich, Institute of Pharmacology and Toxicology, Zurich, Switzerland.
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland.
| | - Aiman S Saab
- University of Zurich, Institute of Pharmacology and Toxicology, Zurich, Switzerland.
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
3
|
Shao C, Cheng L, Hong Y, Li Z, Li C, Ban X, Gu Z. Effect of different ratios of carbohydrate to fat intake on glucolipid metabolism in developing mice. Food Funct 2025; 16:3482-3492. [PMID: 40214409 DOI: 10.1039/d4fo04976d] [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: 05/07/2025]
Abstract
The manner of consumption of fats and carbohydrates affects the ability of glucolipid metabolism. Improper diets have an impact on health. Few studies have examined the impact of the ratio of two nutrient intakes on developing mice's health as opposed to the type. In this study, three-week-old C57BL/6J mice were fed four different types of chow: conventional (63.95% CHO, 15.75% FAT), medium-carbon (39.70% CHO, 40.00% FAT), low-carbon (1.57% CHO, 64.00% FAT), and ketogenic (1.00% CHO, 78.71% FAT) diets. The results showed that during the developing stage, all four groups of mice had no discernible changes in health. However, the mice in the medium-carbon diet group exhibited impaired liver slices and mRNA expression of inflammatory factors and had the least amount of serum inosine. Furthermore, the expression levels of genes related to gluconeogenesis and fat oxidation increased with increasing fat intake. The expression levels of fat synthesis-related enzymes were proportional to the carbohydrate intake. When the intake of either carbohydrates or fat was higher than that of another, the serum levels of inosine in mice increased, activating PPARγ. This activated PPARγ regulated the expression of mRNAs related to gluconeogenesis and lipid metabolism and suppressed inflammatory factors. Conversely, when the carbohydrate intake ratio was consistent with that of fat, the inosine content was low, and PPARγ expression was reduced. As a result, no inhibitory effect was observed on the growth of hepatic inflammatory factors. This study may provide some theoretical basis for selecting dietary patterns during the period of development, thus eliminating the medium-carbon diet option.
Collapse
Affiliation(s)
- Chongxuan Shao
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China.
| | - Li Cheng
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China.
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Yan Hong
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China.
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Zhaofeng Li
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China.
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Caiming Li
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China.
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Xiaofeng Ban
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China.
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Zhengbiao Gu
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China.
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
4
|
Mosharov EV, Rosenberg AM, Monzel AS, Osto CA, Stiles L, Rosoklija GB, Dwork AJ, Bindra S, Junker A, Zhang Y, Fujita M, Mariani MB, Bakalian M, Sulzer D, De Jager PL, Menon V, Shirihai OS, Mann JJ, Underwood MD, Boldrini M, Thiebaut de Schotten M, Picard M. A human brain map of mitochondrial respiratory capacity and diversity. Nature 2025; 641:749-758. [PMID: 40140564 DOI: 10.1038/s41586-025-08740-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 02/04/2025] [Indexed: 03/28/2025]
Abstract
Mitochondrial oxidative phosphorylation (OXPHOS) powers brain activity1,2, and mitochondrial defects are linked to neurodegenerative and neuropsychiatric disorders3,4. To understand the basis of brain activity and behaviour, there is a need to define the molecular energetic landscape of the brain5-10. Here, to bridge the scale gap between cognitive neuroscience and cell biology, we developed a physical voxelization approach to partition a frozen human coronal hemisphere section into 703 voxels comparable to neuroimaging resolution (3 × 3 × 3 mm). In each cortical and subcortical brain voxel, we profiled mitochondrial phenotypes, including OXPHOS enzyme activities, mitochondrial DNA and volume density, and mitochondria-specific respiratory capacity. We show that the human brain contains diverse mitochondrial phenotypes driven by both topology and cell types. Compared with white matter, grey matter contains >50% more mitochondria. Moreover, the mitochondria in grey matter are biochemically optimized for energy transformation, particularly among recently evolved cortical brain regions. Scaling these data to the whole brain, we created a backwards linear regression model that integrates several neuroimaging modalities11 to generate a brain-wide map of mitochondrial distribution and specialization. This model predicted mitochondrial characteristics in an independent brain region of the same donor brain. This approach and the resulting MitoBrainMap of mitochondrial phenotypes provide a foundation for exploring the molecular energetic landscape that enables normal brain function. This resource also relates to neuroimaging data and defines the subcellular basis for regionalized brain processes relevant to neuropsychiatric and neurodegenerative disorders. All data are available at http://humanmitobrainmap.bcblab.com .
Collapse
Affiliation(s)
- Eugene V Mosharov
- Department of Psychiatry, Divisions of Molecular Therapeutics and Behavioral Medicine, Columbia University Irving Medical Center, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
| | - Ayelet M Rosenberg
- Department of Psychiatry, Divisions of Molecular Therapeutics and Behavioral Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Anna S Monzel
- Department of Psychiatry, Divisions of Molecular Therapeutics and Behavioral Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Corey A Osto
- Department of Medicine, Endocrinology, and Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Linsey Stiles
- Department of Medicine, Endocrinology, and Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Gorazd B Rosoklija
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Division of Molecular Imaging and Neuropathology, Columbia University Irving Medical Center, New York, NY, USA
| | - Andrew J Dwork
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Division of Molecular Imaging and Neuropathology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Snehal Bindra
- Department of Psychiatry, Divisions of Molecular Therapeutics and Behavioral Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Alex Junker
- Department of Psychiatry, Divisions of Molecular Therapeutics and Behavioral Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Ya Zhang
- Center for Translational and Computational Neuroimmunology, Neuroimmunology Division, Department of Neurology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Masashi Fujita
- Center for Translational and Computational Neuroimmunology, Neuroimmunology Division, Department of Neurology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Madeline B Mariani
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Division of Molecular Imaging and Neuropathology, Columbia University Irving Medical Center, New York, NY, USA
| | - Mihran Bakalian
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Division of Molecular Imaging and Neuropathology, Columbia University Irving Medical Center, New York, NY, USA
| | - David Sulzer
- Department of Psychiatry, Divisions of Molecular Therapeutics and Behavioral Medicine, Columbia University Irving Medical Center, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
- Departments of Neurology and Pharmacology, Columbia University Irving Medical Center, New York, NY, USA
| | - Philip L De Jager
- Center for Translational and Computational Neuroimmunology, Neuroimmunology Division, Department of Neurology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Vilas Menon
- Center for Translational and Computational Neuroimmunology, Neuroimmunology Division, Department of Neurology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Orian S Shirihai
- Department of Medicine, Endocrinology, and Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - J John Mann
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Division of Molecular Imaging and Neuropathology, Columbia University Irving Medical Center, New York, NY, USA
| | - Mark D Underwood
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Division of Molecular Imaging and Neuropathology, Columbia University Irving Medical Center, New York, NY, USA
| | - Maura Boldrini
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Division of Molecular Imaging and Neuropathology, Columbia University Irving Medical Center, New York, NY, USA
| | - Michel Thiebaut de Schotten
- Brain Connectivity and Behaviour Laboratory, Paris, France.
- Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives-UMR 5293, CNRS, CEA University of Bordeaux, Bordeaux, France.
| | - Martin Picard
- Department of Psychiatry, Divisions of Molecular Therapeutics and Behavioral Medicine, Columbia University Irving Medical Center, New York, NY, USA.
- New York State Psychiatric Institute, New York, NY, USA.
- Department of Neurology, H. Houston Merritt Center, Columbia Translational Neuroscience Initiative, Columbia University Irving Medical Center, New York, NY, USA.
- Robert N. Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, NY, USA.
| |
Collapse
|
5
|
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] [Download PDF] [Figures] [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 PhysiologyGraduate School of Life Sciences, Tohoku UniversitySendaiJapan
| | - Yoko Ikoma
- Super‐network Brain PhysiologyGraduate School of Life Sciences, Tohoku UniversitySendaiJapan
| | - Yusuke Niino
- Laboratory for Cell Function DynamicsRIKEN Center for Brain ScienceWako‐CityJapan
| | - Yuichi Hiraoka
- Laboratory of Molecular NeuroscienceMedical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo Institute of TechnologyTokyoJapan
- Laboratory of Genome Editing for Biomedical ResearchMedical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo Institute of TechnologyTokyoJapan
| | - Kohichi Tanaka
- Laboratory of Genome Editing for Biomedical ResearchMedical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo Institute of TechnologyTokyoJapan
| | - Atsushi Miyawaki
- Laboratory for Cell Function DynamicsRIKEN Center for Brain ScienceWako‐CityJapan
- Biotechnological Optics Research TeamRIKEN Center for Advanced PhotonicsWako‐CityJapan
| | - Johannes Hirrlinger
- Carl‐Ludwig‐Institute for Physiology, Faculty of MedicineLeipzig UniversityLeipzigGermany
- Department of NeurogeneticsMax‐Planck‐Institute for Multidisciplinary SciencesGöttingenGermany
| | - Ko Matsui
- Super‐network Brain PhysiologyGraduate School of Life Sciences, Tohoku UniversitySendaiJapan
- Super‐network Brain Physiology, Graduate School of MedicineTohoku UniversitySendaiJapan
| |
Collapse
|
6
|
Picard M, Monzel A, Devine J, Kapri D, Enriquez J, Trumpff C. A Quantitative Approach to Mapping Mitochondrial Specialization and Plasticity. RESEARCH SQUARE 2025:rs.3.rs-5961609. [PMID: 39989954 PMCID: PMC11844627 DOI: 10.21203/rs.3.rs-5961609/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Mitochondria are a diverse family of organelles that specialize to accomplish complimentary functions 1-3. All mitochondria share general features, but not all mitochondria are created equal 4.Here we develop a quantitative pipeline to define the degree of molecular specialization among different mitochondrial phenotypes - or mitotypes. By distilling hundreds of validated mitochondrial genes/proteins into 149 biologically interpretable MitoPathway scores (MitoCarta 3.0 5) the simple mitotyping pipeline allows investigators to quantify and interpret mitochondrial diversity and plasticity from transcriptomics or proteomics data across a variety of natural and experimental contexts. We show that mouse and human multi-organ mitotypes segregate along two main axes of mitochondrial specialization, contrasting anabolic (liver) and catabolic (brain) tissues. In cultured primary human fibroblasts exhibiting robust time-dependent and treatment-induced metabolic plasticity 6-8, we demonstrate how the mitotype of a given cell type recalibrates i) over time in parallel with hallmarks of aging, and ii) in response to genetic, pharmacological, and metabolic perturbations. Investigators can now use MitotypeExplorer.org and the associated code to visualize, quantify and interpret the multivariate space of mitochondrial biology.
Collapse
|
7
|
Monzel AS, Devine J, Kapri D, Enriquez JA, Trumpff C, Picard M. A Quantitative Approach to Mapping Mitochondrial Specialization and Plasticity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.03.635951. [PMID: 39975232 PMCID: PMC11838522 DOI: 10.1101/2025.02.03.635951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Mitochondria are a diverse family of organelles that specialize to accomplish complimentary functions1-3. All mitochondria share general features, but not all mitochondria are created equal4. Here we develop a quantitative pipeline to define the degree of molecular specialization among different mitochondrial phenotypes - or mitotypes. By distilling hundreds of validated mitochondrial genes/proteins into 149 biologically interpretable MitoPathway scores (MitoCarta 3.05) the simple mitotyping pipeline allows investigators to quantify and interpret mitochondrial diversity and plasticity from transcriptomics or proteomics data across a variety of natural and experimental contexts. We show that mouse and human multi-organ mitotypes segregate along two main axes of mitochondrial specialization, contrasting anabolic (liver) and catabolic (brain) tissues. In cultured primary human fibroblasts exhibiting robust time-dependent and treatment-induced metabolic plasticity6-8, we demonstrate how the mitotype of a given cell type recalibrates i) over time in parallel with hallmarks of aging, and ii) in response to genetic, pharmacological, and metabolic perturbations. Investigators can now use MitotypeExplorer.org and the associated code to visualize, quantify and interpret the multivariate space of mitochondrial biology.
Collapse
Affiliation(s)
- Anna S. Monzel
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Jack Devine
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Darshana Kapri
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Jose Antonio Enriquez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid 28029, Spain
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - Caroline Trumpff
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Martin Picard
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
- Department of Neurology, H. Houston Merritt Center, Neuromuscular Medicine Division, Columbia University Medical Center, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
- Robert N Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, NY, USA
| |
Collapse
|
8
|
Zhou C, Qu S. Application and Mechanism of Action of a Ketogenic Diet in Antiepileptic Therapy. ACS Chem Neurosci 2025; 16:284-291. [PMID: 39787038 DOI: 10.1021/acschemneuro.4c00695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025] Open
Abstract
Epilepsy is a chronic neurological disorder caused by abnormal discharges of neurons in the brain, which seriously affects the quality of life of patients. Although there are various drug treatments available, many epilepsy patients still experience seizures with the effect of drugs and develop refractory epilepsy. The ketogenic diet can treat drug-refractory epilepsy by regulating the body's metabolism and can enhance the quality of life by improving their cognition, behavior, and sleep quality. However, there is no unified conclusion on the mechanism through which the ketogenic diet plays a therapeutic role in epilepsy. This article provides a review of the possible mechanisms of how the ketogenic diet exerts a protective effect on epilepsy.
Collapse
Affiliation(s)
- Chang Zhou
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
- Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou, Guangdong 510515, P.R. China
| | - Shaogang Qu
- Department of Neurology, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, Jiangxi 341000, China
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
- Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
9
|
Rubio C, López-Landa A, Romo-Parra H, Rubio-Osornio M. Impact of the Ketogenic Diet on Neurological Diseases: A Review. Life (Basel) 2025; 15:71. [PMID: 39860011 PMCID: PMC11767209 DOI: 10.3390/life15010071] [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: 12/03/2024] [Revised: 12/28/2024] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND The ketogenic diet (KD), high in fat and low in carbohydrates, was introduced in the 1920s as a non-pharmacological treatment for refractory epilepsy. Although its mechanism of action is not fully understood, beneficial effects have been observed in neurological diseases such as epilepsy, Alzheimer's disease, and Parkinson's disease. OBJECTIVE This review examines the impact of the ketogenic diet and its molecular and neuroglial effects as a complementary therapy for neurological diseases. DISCUSSION KD is associated with neuroprotective and antioxidant effects that improve mitochondrial function, regulate neurotransmitter flow, and reduce neuroinflammation and oxidative stress. Glial cells play an essential role in the utilization of ketone bodies (KBs) within the central nervous system's metabolism, particularly during ketosis induced by the KD. Thus, the KD represents a broad and promising strategy that involves both neurons and glial cells, with a molecular impact on brain metabolism and neuroinflammatory homeostasis. CONCLUSION Multiple molecular mechanisms have been identified to explain the benefits of the KD in neurological diseases; however, further experimental and clinical studies are needed to address various molecular pathways in order to achieve conclusive results.
Collapse
Affiliation(s)
- Carmen Rubio
- Neurophysiology Department, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Mexico City 14269, Mexico; (C.R.); (A.L.-L.); (H.R.-P.)
| | - Alejandro López-Landa
- Neurophysiology Department, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Mexico City 14269, Mexico; (C.R.); (A.L.-L.); (H.R.-P.)
- School of Medicine, Benemérita Universidad Autónoma de Puebla, Puebla City 72000, Mexico
| | - Hector Romo-Parra
- Neurophysiology Department, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Mexico City 14269, Mexico; (C.R.); (A.L.-L.); (H.R.-P.)
- Psychology Department, Universidad Iberoamericana, Mexico City 01376, Mexico
| | - Moisés Rubio-Osornio
- Neurochemistry Department, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Mexico City 14269, Mexico
| |
Collapse
|
10
|
Colombo G, Monsorno K, Paolicelli RC. Metabolic control of microglia in health and disease. HANDBOOK OF CLINICAL NEUROLOGY 2025; 209:143-159. [PMID: 40122622 DOI: 10.1016/b978-0-443-19104-6.00009-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Metabolic states within cells are tightly linked to functional outcomes and finely regulated by nutrient availability. A growing body of the literature supports the idea that various metabolites can influence cellular functions, such as cell differentiation, migration, and proliferation in different contexts, with ample evidence coming from the immune system. Additionally, certain functional programs can trigger significant metabolic changes within cells, which are crucial not only to meet high energy demands, but also to produce intermediate metabolites necessary to support specific tasks. Microglia, the resident innate immune cells of the central nervous system, are constantly active, surveying the brain parenchyma and providing support to neighboring cells in the brain. They exhibit high metabolic flexibility, capable of quickly undergoing metabolic reprogramming based on nutrient availability and functional requirements. In this chapter, we will discuss the major metabolic pathways within cells and provide examples of how relevant enzymes and metabolites can impact microglial function in physiologic and pathologic contexts.
Collapse
Affiliation(s)
- Gloria Colombo
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Katia Monsorno
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Rosa C Paolicelli
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
11
|
Chaudhari PS, Ermolaeva MA. Too old for healthy aging? Exploring age limits of longevity treatments. NPJ METABOLIC HEALTH AND DISEASE 2024; 2:37. [PMID: 39678297 PMCID: PMC11638076 DOI: 10.1038/s44324-024-00040-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 11/13/2024] [Indexed: 12/17/2024]
Abstract
It is well documented that aging elicits metabolic failures, while poor metabolism contributes to accelerated aging. Metabolism in general, and energy metabolism in particular are also effective entry points for interventions that extend lifespan and improve organ function during aging. In this review, we discuss common metabolic remedies for healthy aging from the angle of their potential age-specificity. We demonstrate that some well-known metabolic treatments are mostly effective in young and middle-aged organisms, while others maintain high efficacy independently of age. The mechanistic basis of presence or lack of the age limitations is laid out and discussed.
Collapse
Affiliation(s)
| | - Maria A. Ermolaeva
- Leibniz Institute on Aging – Fritz Lipmann Institute (FLI), Beutenbergstrasse 11, 07745 Jena, Germany
| |
Collapse
|
12
|
Neal ES, Xu W, Borges K. Metabolic aspects of genetic ion channel epilepsies. J Neurochem 2024; 168:3911-3935. [PMID: 37594756 PMCID: PMC11591411 DOI: 10.1111/jnc.15938] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 07/17/2023] [Accepted: 08/01/2023] [Indexed: 08/19/2023]
Abstract
Nowadays, particularly in countries with high incomes, individual mutations in people affected by genetic epilepsies are identified, and genetic therapies are being developed. In addition, drugs are being screened to directly target specific mutations, and personalised medicine is possible. However, people with epilepsy do not yet benefit from these advances, and many types of epilepsies are medication-resistant, including Dravet syndrome. Thus, in the meantime, alternative and effective treatment options are needed. There is increasing evidence that metabolic deficits contribute to epileptic seizures and that such metabolic impairments may be amenable to treatment, with metabolic treatment options like the ketogenic diet being employed with some success. However, the brain metabolic alterations that occur in ion channel epilepsies are not well-understood, nor how these may differ from epilepsies that are of acquired and unknown origins. Here, we provide an overview of studies investigating metabolic alterations in epilepsies caused by mutations in the SCN1A and KCNA1 genes, which are currently the most studied ion channel epilepsies in animal models. The metabolic changes found in these models are likely to contribute to seizures. A metabolic basis of these ion channel epilepsies is supported by human and/or animal studies that show beneficial effects of the ketogenic diet, which may be mediated by the provision of auxiliary brain fuel in the form of ketone bodies. Other potentially more preferred dietary therapies including medium-chain triglycerides and triheptanoin have also been tested in a limited number of studies, but their efficacies remain to be clearly established. The extent to which brain metabolism is affected in people with Dravet syndrome, KCNA1 epilepsy and the models thereof still requires clarification. This requires more experiments that yield functional insight into metabolism.
Collapse
Affiliation(s)
- Elliott S. Neal
- School of Biomedical SciencesThe University of QueenslandSt LuciaQueenslandAustralia
| | - Weizhi Xu
- School of Biomedical SciencesThe University of QueenslandSt LuciaQueenslandAustralia
| | - Karin Borges
- School of Biomedical SciencesThe University of QueenslandSt LuciaQueenslandAustralia
| |
Collapse
|
13
|
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
|
14
|
Wang W, Li Z, Yuan S, Du Z, Li J, Peng H, Ru S. A Potential Neurotoxic Mechanism: Bisphenol S-Induced Inhibition of Glucose Transporter 1 Leads to ATP Excitotoxicity in the Zebrafish Brain. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:15463-15474. [PMID: 39167196 DOI: 10.1021/acs.est.4c03870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
Many environmental pollutants have neurotoxic effects, but the initial molecular events involved in these effects are unclear. Here, zebrafish were exposed to the neurotoxicant bisphenol S (BPS, 1, 10, or 100 μg/L) from the embryonic stage to the larval stage to explore the ability of BPS to interfere with energy metabolism in the brain. BPS, which is similar to a glucose transporter 1 (GLUT1) inhibitor, inhibited GLUT1 function but increased mitochondrial activity in the brains of larval zebrafish. Interestingly, GLUT1 inhibitor treatment and BPS exposure did not reduce energy production in the brain; instead, they increased ATP production by inducing the preferential use of ketone bodies. Moreover, BPS promoted the protein expression of the purinergic 2X receptor but inhibited the purinergic 2Y-mediated phosphatidylinositol signaling pathway, indicating that excess ATP acts as a neurotransmitter to activate the purinergic 2X receptor under the BPS-induced restriction of GLUT1 function. BPS-induced inhibition of GLUT1 increased the number of neurons but promoted apoptosis by activating ATP-purinergic 2X receptors in the brain, causing ATP excitatory neurotoxicity. Our data reveal a potential neurotoxic mechanism induced by BPS that may represent a new adverse outcome pathway.
Collapse
Affiliation(s)
- Weiwei Wang
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Ze Li
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Shipeng Yuan
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Zehui Du
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Jiali Li
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Hongyuan Peng
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Shaoguo Ru
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| |
Collapse
|
15
|
Wculek SK, Forisch S, Miguel V, Sancho D. Metabolic homeostasis of tissue macrophages across the lifespan. Trends Endocrinol Metab 2024; 35:793-808. [PMID: 38763781 DOI: 10.1016/j.tem.2024.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 05/21/2024]
Abstract
Macrophages are present in almost all organs. Apart from being immune sentinels, tissue-resident macrophages (TRMs) have organ-specific functions that require a specialized cellular metabolism to maintain homeostasis. In addition, organ-dependent metabolic adaptations of TRMs appear to be fundamentally distinct in homeostasis and in response to a challenge, such as infection or injury. Moreover, TRM function becomes aberrant with advancing age, contributing to inflammaging and organ deterioration, and a metabolic imbalance may underlie TRM immunosenescence. Here, we outline current understanding of the particular metabolic states of TRMs across organs and the relevance for their function. Moreover, we discuss the concomitant aging-related decline in metabolic plasticity and functions of TRMs, highlighting potential novel therapeutic avenues to promote healthy aging.
Collapse
Affiliation(s)
- Stefanie K Wculek
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
| | - Stephan Forisch
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Verónica Miguel
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - David Sancho
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.
| |
Collapse
|
16
|
Ren Q, Fu J, Duan X, Sun L, Mu Z, Liang W, Li Y, Wang Z, Xiu S. The Effects of Ketogenic Diet on Brain Gene Expressions in Type 2 Diabetes Background. Neuroscience 2024; 549:101-109. [PMID: 38734303 DOI: 10.1016/j.neuroscience.2024.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/07/2024] [Accepted: 04/20/2024] [Indexed: 05/13/2024]
Abstract
Type 2 diabetes mellitus (T2DM) is a major risk factor of a number of neurodegenerative diseases (NDDs). Ketogenic diet (KD) has significant beneficial effects on glycemic control and may act effectively against NDDs, but the mechanism remains unclear. In this study, we aimed to investigate the potential effects of KD on gene expressions in the brains of T2DM model mice. Male db/db mice at the age of 9 weeks were fed with KD or normal diet to the age of 6 months, and the whole brains were subjected to mRNA-seq analysis for differentially expressed genes. KD significantly lowered fasting glucose and body weights in db/db mice (P < 0.05), and the expression of 189 genes in the brain were significantly changed (P < 0.05, |log2| > 1). Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses revealed that the differentially expressed genes upon KD are involved in inflammatory responses and the functions of biosynthesis. In inflammatory responses, NF-κB signaling pathway, viral protein interaction with cytokine and cytokine receptor, and cytokine-cytokine receptor interaction pathways were enriched, and in biosynthesis pathways, genes functioning in lipid and amino acid metabolism, protein synthesis, and energy metabolism were enriched. Moreover, consistent with the gene set enrichment analysis results, proteasomal activity measured biochemically were enhanced in KD-fed T2DM mice. These data may facilitate the understanding of how KD can be protective to the brain in T2DM background. KD could be a new strategy for the prevention of NDDs in T2DM patients.
Collapse
Affiliation(s)
- Qianxu Ren
- The National Clinical Research Center for Geriatric Disease, Department of Neurology, Advanced Innovation Center for Human Brain Protection, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Junling Fu
- Department of Endocrinology, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Xiaoye Duan
- Department of Endocrinology, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Lina Sun
- Department of Endocrinology, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Zhijing Mu
- Department of Endocrinology, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Wenping Liang
- The National Clinical Research Center for Geriatric Disease, Department of Neurology, Advanced Innovation Center for Human Brain Protection, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yu Li
- The National Clinical Research Center for Geriatric Disease, Department of Neurology, Advanced Innovation Center for Human Brain Protection, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zhe Wang
- The National Clinical Research Center for Geriatric Disease, Department of Neurology, Advanced Innovation Center for Human Brain Protection, Xuanwu Hospital, Capital Medical University, Beijing, China.
| | - Shuangling Xiu
- Department of Endocrinology, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.
| |
Collapse
|
17
|
Gao M, Kirk M, Lash E, Knight H, Michalopoulou M, Guess N, Browning M, Weich S, Burnet P, Jebb SA, Stevens R, Aveyard P. Evaluating the efficacy and mechanisms of a ketogenic diet as adjunctive treatment for people with treatment-resistant depression: A protocol for a randomised controlled trial. J Psychiatr Res 2024; 174:230-236. [PMID: 38653031 DOI: 10.1016/j.jpsychires.2024.04.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 04/09/2024] [Indexed: 04/25/2024]
Abstract
BACKGROUND One-third of people with depression do not respond to antidepressants, and, after two adequate courses of antidepressants, are classified as having treatment-resistant depression (TRD). Some case reports suggest that ketogenic diets (KDs) may improve some mental illnesses, and preclinical data indicate that KDs can influence brain reward signalling, anhedonia, cortisol, and gut microbiome which are associated with depression. To date, no trials have examined the clinical effect of a KD on TRD. METHODS This is a proof-of-concept randomised controlled trial to investigate the efficacy of a six-week programme of weekly dietitian counselling plus provision of KD meals, compared with an intervention involving similar dietetic contact time and promoting a healthy diet with increased vegetable consumption and reduction in saturated fat, plus food vouchers to purchase healthier items. At 12 weeks we will assess whether participants have continued to follow the assigned diet. The primary outcome is the difference between groups in the change in Patient Health Questionnaire-9 (PHQ-9) score from baseline to 6 weeks. PHQ-9 will be measured at weeks 2, 4, 6 and 12. The secondary outcomes are the differences between groups in the change in remission of depression, change in anxiety score, functioning ability, quality of life, cognitive performance, reward sensitivity, and anhedonia from baseline to 6 and 12 weeks. We will also assess whether changes in reward sensitivity, anhedonia, cortisol awakening response and gut microbiome may explain any changes in depression severity. DISCUSSION This study will test whether a ketogenic diet is an effective intervention to reduce the severity of depression, anxiety and improve quality of life and functioning ability for people with treatment-resistant depression.
Collapse
Affiliation(s)
- Min Gao
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK; NIHR Oxford Health Biomedical Research Centre, Warneford Hospital, Oxford, UK.
| | - Megan Kirk
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK; NIHR Oxford Health Biomedical Research Centre, Warneford Hospital, Oxford, UK
| | - Eva Lash
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK; NIHR Oxford Health Biomedical Research Centre, Warneford Hospital, Oxford, UK
| | - Heather Knight
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Moscho Michalopoulou
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Nicola Guess
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Michael Browning
- Department of Psychiatry, University of Oxford, Oxford, UK; Oxford Health NHS Foundation Trust, Oxford, UK
| | - Scott Weich
- School of Health and Related Research, University of Sheffield, Sheffield, UK
| | - Philip Burnet
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Susan A Jebb
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK; NIHR Oxford Health Biomedical Research Centre, Warneford Hospital, Oxford, UK; NIHR Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, UK
| | - Richard Stevens
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Paul Aveyard
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK; NIHR Oxford Health Biomedical Research Centre, Warneford Hospital, Oxford, UK; NIHR Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, UK
| |
Collapse
|
18
|
Sethi S, Wakeham D, Ketter T, Hooshmand F, Bjornstad J, Richards B, Westman E, Krauss RM, Saslow L. Ketogenic Diet Intervention on Metabolic and Psychiatric Health in Bipolar and Schizophrenia: A Pilot Trial. Psychiatry Res 2024; 335:115866. [PMID: 38547601 DOI: 10.1016/j.psychres.2024.115866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 03/15/2024] [Accepted: 03/17/2024] [Indexed: 04/14/2024]
Abstract
The ketogenic diet (KD, also known as metabolic therapy) has been successful in the treatment of obesity, type 2 diabetes, and epilepsy. More recently, this treatment has shown promise in the treatment of psychiatric illness. We conducted a 4-month pilot study to investigate the effects of a KD on individuals with schizophrenia or bipolar disorder with existing metabolic abnormalities. Twenty-three participants were enrolled in a single-arm trial. Results showcased improvements in metabolic health, with no participants meeting metabolic syndrome criteria by study conclusion. Adherent individuals experienced significant reduction in weight (12 %), BMI (12 %), waist circumference (13 %), and visceral adipose tissue (36 %). Observed biomarker enhancements in this population include a 27 % decrease in HOMA-IR, and a 25 % drop in triglyceride levels. In psychiatric measurements, participants with schizophrenia showed a 32 % reduction in Brief Psychiatric Rating Scale scores. Overall Clinical Global Impression (CGI) severity improved by an average of 31 %, and the proportion of participants that started with elevated symptomatology improved at least 1-point on CGI (79 %). Psychiatric outcomes across the cohort encompassed increased life satisfaction (17 %) and enhanced sleep quality (19 %). This pilot trial underscores the potential advantages of adjunctive ketogenic dietary treatment in individuals grappling with serious mental illness.
Collapse
Affiliation(s)
- Shebani Sethi
- Metabolic Psychiatry, Dept. of Psychiatry and Behavioral Sciences, Stanford Medicine, Stanford, CA, USA.
| | - Diane Wakeham
- Metabolic Psychiatry, Dept. of Psychiatry and Behavioral Sciences, Stanford Medicine, Stanford, CA, USA
| | - Terence Ketter
- Metabolic Psychiatry, Dept. of Psychiatry and Behavioral Sciences, Stanford Medicine, Stanford, CA, USA
| | - Farnaz Hooshmand
- Metabolic Psychiatry, Dept. of Psychiatry and Behavioral Sciences, Stanford Medicine, Stanford, CA, USA
| | - Julia Bjornstad
- Metabolic Psychiatry, Dept. of Psychiatry and Behavioral Sciences, Stanford Medicine, Stanford, CA, USA
| | - Blair Richards
- Department of Health Behavior and Biological Sciences, School of Nursing, University of Michigan, Ann Arbor, MI, USA
| | - Eric Westman
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Ronald M Krauss
- Department of Pediatrics and Medicine, University of California-San Francisco, San Francisco, CA, USA
| | - Laura Saslow
- Department of Health Behavior and Biological Sciences, School of Nursing, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
19
|
Kumar M, Bhatt B, Gusain C, Mahajan N, Bishnoi M. Sex-specific effects of ketogenic diet on anxiety-like behavior and neuroimmune response in C57Bl/6J mice. J Nutr Biochem 2024; 127:109591. [PMID: 38311044 DOI: 10.1016/j.jnutbio.2024.109591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/27/2024] [Accepted: 01/29/2024] [Indexed: 02/06/2024]
Abstract
The ketogenic diet (KD) has been shown to reduce anxiety and enhance cognitive functions in neurological diseases. However, the sex-specific effects of KD on anxiety-like behavior in healthy individuals and the underlying molecular mechanisms contributing to these effects, including neuroinflammation, are unelucidated. This study investigated the sex-specific effects of KD on anxiety-like behavior and the neuroimmune response in the prefrontal cortex (PFC) and hippocampus of healthy C57BL/6J male and female mice. Animals were fed either a control diet (CD- 17% fat, 65% carb, 18% protein) or a KD (80% fat, 5% carb, 15% protein) for 4 weeks. KD increased the levels of circulating β-hydroxybutyrate (BHB) both in males and females. However, PFC BHB levels were found to be elevated only in KD males. Moreover, KD did not affect the behavior of females but improved motor abilities and reduced anxiety levels in males. KD suppressed the mRNA expression of the pan microglial markers (Cd68, P2ry12) and induced morphological changes in the male PFC microglia. A sex-specific decrease in IL1β and an increase in IL-10 levels was found in the PFC of KD males. A similar trend was observed in the hippocampus of males where KD reduced the mRNA expression of P2ry12, Il1β, and cFos. Additionally, BHB increased the production of IL-10 whereas it decreased the production of IL1β from human microglia in in-vitro conditions. In summary, these results demonstrate that the anxiolytic and motor function enhancement abilities of KD are male-specific. Reduced pro-inflammatory and improved anti-inflammatory factors in the male PFC and hippocampus may underlie these effects.
Collapse
Affiliation(s)
- Mohit Kumar
- Centre for Excellence in Functional Foods, Food & Nutrition Biotechnology Division, National Agri-Food Biotechnology Institute, S.A.S Nagar, Sector 81 (Knowledge City), Punjab, India; Adjunct faculty, Regional Centre for Biotechnology, Faridabad, Haryana, India.
| | - Babita Bhatt
- Centre for Excellence in Functional Foods, Food & Nutrition Biotechnology Division, National Agri-Food Biotechnology Institute, S.A.S Nagar, Sector 81 (Knowledge City), Punjab, India
| | - Chitralekha Gusain
- Centre for Excellence in Functional Foods, Food & Nutrition Biotechnology Division, National Agri-Food Biotechnology Institute, S.A.S Nagar, Sector 81 (Knowledge City), Punjab, India
| | - Nayan Mahajan
- Centre for Excellence in Functional Foods, Food & Nutrition Biotechnology Division, National Agri-Food Biotechnology Institute, S.A.S Nagar, Sector 81 (Knowledge City), Punjab, India
| | - Mahendra Bishnoi
- Centre for Excellence in Functional Foods, Food & Nutrition Biotechnology Division, National Agri-Food Biotechnology Institute, S.A.S Nagar, Sector 81 (Knowledge City), Punjab, India
| |
Collapse
|
20
|
Mosharov EV, Rosenberg AM, Monzel AS, Osto CA, Stiles L, Rosoklija GB, Dwork AJ, Bindra S, Zhang Y, Fujita M, Mariani MB, Bakalian M, Sulzer D, De Jager PL, Menon V, Shirihai OS, Mann JJ, Underwood M, Boldrini M, Thiebaut de Schotten M, Picard M. A Human Brain Map of Mitochondrial Respiratory Capacity and Diversity. RESEARCH SQUARE 2024:rs.3.rs-4047706. [PMID: 38562777 PMCID: PMC10984021 DOI: 10.21203/rs.3.rs-4047706/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Mitochondrial oxidative phosphorylation (OxPhos) powers brain activity1,2, and mitochondrial defects are linked to neurodegenerative and neuropsychiatric disorders3,4, underscoring the need to define the brain's molecular energetic landscape5-10. To bridge the cognitive neuroscience and cell biology scale gap, we developed a physical voxelization approach to partition a frozen human coronal hemisphere section into 703 voxels comparable to neuroimaging resolution (3×3×3 mm). In each cortical and subcortical brain voxel, we profiled mitochondrial phenotypes including OxPhos enzyme activities, mitochondrial DNA and volume density, and mitochondria-specific respiratory capacity. We show that the human brain contains a diversity of mitochondrial phenotypes driven by both topology and cell types. Compared to white matter, grey matter contains >50% more mitochondria. We show that the more abundant grey matter mitochondria also are biochemically optimized for energy transformation, particularly among recently evolved cortical brain regions. Scaling these data to the whole brain, we created a backward linear regression model integrating several neuroimaging modalities11, thereby generating a brain-wide map of mitochondrial distribution and specialization that predicts mitochondrial characteristics in an independent brain region of the same donor brain. This new approach and the resulting MitoBrainMap of mitochondrial phenotypes provide a foundation for exploring the molecular energetic landscape that enables normal brain functions, relating it to neuroimaging data, and defining the subcellular basis for regionalized brain processes relevant to neuropsychiatric and neurodegenerative disorders.
Collapse
Affiliation(s)
- Eugene V. Mosharov
- Department of Psychiatry, Divisions of Molecular Therapeutics and Behavioral Medicine, Columbia University Irving Medical Center, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
| | - Ayelet M Rosenberg
- Department of Psychiatry, Divisions of Molecular Therapeutics and Behavioral Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Anna S Monzel
- Department of Psychiatry, Divisions of Molecular Therapeutics and Behavioral Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Corey A. Osto
- Department of Medicine, Endocrinology, and Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Linsey Stiles
- Department of Medicine, Endocrinology, and Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Gorazd B. Rosoklija
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Division of Molecular Imaging and Neuropathology, Columbia University Irving Medical Center, New York, NY, USA
| | - Andrew J. Dwork
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Division of Molecular Imaging and Neuropathology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Snehal Bindra
- Department of Psychiatry, Divisions of Molecular Therapeutics and Behavioral Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Ya Zhang
- Center for Translational & Computational Neuroimmunology, Neuroimmunology Division, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Masashi Fujita
- Center for Translational & Computational Neuroimmunology, Neuroimmunology Division, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Madeline B Mariani
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Division of Molecular Imaging and Neuropathology, Columbia University Irving Medical Center, New York, NY, USA
| | - Mihran Bakalian
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Division of Molecular Imaging and Neuropathology, Columbia University Irving Medical Center, New York, NY, USA
| | - David Sulzer
- Department of Psychiatry, Divisions of Molecular Therapeutics and Behavioral Medicine, Columbia University Irving Medical Center, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
- Departments of Neurology and Pharmacology, Columbia University Irving Medical Center, New York, NY, USA; Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Philip L. De Jager
- Center for Translational & Computational Neuroimmunology, Neuroimmunology Division, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Vilas Menon
- Center for Translational & Computational Neuroimmunology, Neuroimmunology Division, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Orian S Shirihai
- Department of Medicine, Endocrinology, and Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA
| | - J. John Mann
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Division of Molecular Imaging and Neuropathology, Columbia University Irving Medical Center, New York, NY, USA
| | - Mark Underwood
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Division of Molecular Imaging and Neuropathology, Columbia University Irving Medical Center, New York, NY, USA
| | - Maura Boldrini
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Division of Molecular Imaging and Neuropathology, Columbia University Irving Medical Center, New York, NY, USA
| | - Michel Thiebaut de Schotten
- Brain Connectivity and Behavior Laboratory, Paris, France; Groupe d’Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives-UMR 5293, CNRS, CEA University of Bordeaux, France
| | - Martin Picard
- Department of Psychiatry, Divisions of Molecular Therapeutics and Behavioral Medicine, Columbia University Irving Medical Center, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
- Department of Neurology, H. Houston Merritt Center, Columbia Translational Neuroscience Initiative, Columbia University Irving Medical Center, New York, NY, USA
- Robert N Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, NY, USA
| |
Collapse
|
21
|
Mosharov EV, Rosenberg AM, Monzel AS, Osto CA, Stiles L, Rosoklija GB, Dwork AJ, Bindra S, Zhang Y, Fujita M, Mariani MB, Bakalian M, Sulzer D, De Jager PL, Menon V, Shirihai OS, Mann JJ, Underwood M, Boldrini M, de Schotten MT, Picard M. A Human Brain Map of Mitochondrial Respiratory Capacity and Diversity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.05.583623. [PMID: 38496679 PMCID: PMC10942385 DOI: 10.1101/2024.03.05.583623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Mitochondrial oxidative phosphorylation (OxPhos) powers brain activity1,2, and mitochondrial defects are linked to neurodegenerative and neuropsychiatric disorders3,4, underscoring the need to define the brain's molecular energetic landscape5-10. To bridge the cognitive neuroscience and cell biology scale gap, we developed a physical voxelization approach to partition a frozen human coronal hemisphere section into 703 voxels comparable to neuroimaging resolution (3×3×3 mm). In each cortical and subcortical brain voxel, we profiled mitochondrial phenotypes including OxPhos enzyme activities, mitochondrial DNA and volume density, and mitochondria-specific respiratory capacity. We show that the human brain contains a diversity of mitochondrial phenotypes driven by both topology and cell types. Compared to white matter, grey matter contains >50% more mitochondria. We show that the more abundant grey matter mitochondria also are biochemically optimized for energy transformation, particularly among recently evolved cortical brain regions. Scaling these data to the whole brain, we created a backward linear regression model integrating several neuroimaging modalities11, thereby generating a brain-wide map of mitochondrial distribution and specialization that predicts mitochondrial characteristics in an independent brain region of the same donor brain. This new approach and the resulting MitoBrainMap of mitochondrial phenotypes provide a foundation for exploring the molecular energetic landscape that enables normal brain functions, relating it to neuroimaging data, and defining the subcellular basis for regionalized brain processes relevant to neuropsychiatric and neurodegenerative disorders.
Collapse
Affiliation(s)
- Eugene V. Mosharov
- Department of Psychiatry, Divisions of Molecular Therapeutics and Behavioral Medicine, Columbia University Irving Medical Center, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
| | - Ayelet M Rosenberg
- Department of Psychiatry, Divisions of Molecular Therapeutics and Behavioral Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Anna S Monzel
- Department of Psychiatry, Divisions of Molecular Therapeutics and Behavioral Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Corey A. Osto
- Department of Medicine, Endocrinology, and Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Linsey Stiles
- Department of Medicine, Endocrinology, and Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Gorazd B. Rosoklija
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Division of Molecular Imaging and Neuropathology, Columbia University Irving Medical Center, New York, NY, USA
| | - Andrew J. Dwork
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Division of Molecular Imaging and Neuropathology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Snehal Bindra
- Department of Psychiatry, Divisions of Molecular Therapeutics and Behavioral Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Ya Zhang
- Center for Translational & Computational Neuroimmunology, Neuroimmunology Division, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Masashi Fujita
- Center for Translational & Computational Neuroimmunology, Neuroimmunology Division, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Madeline B Mariani
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Division of Molecular Imaging and Neuropathology, Columbia University Irving Medical Center, New York, NY, USA
| | - Mihran Bakalian
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Division of Molecular Imaging and Neuropathology, Columbia University Irving Medical Center, New York, NY, USA
| | - David Sulzer
- Department of Psychiatry, Divisions of Molecular Therapeutics and Behavioral Medicine, Columbia University Irving Medical Center, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
- Departments of Neurology and Pharmacology, Columbia University Irving Medical Center, New York, NY, USA; Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Philip L. De Jager
- Center for Translational & Computational Neuroimmunology, Neuroimmunology Division, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Vilas Menon
- Center for Translational & Computational Neuroimmunology, Neuroimmunology Division, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Orian S Shirihai
- Department of Medicine, Endocrinology, and Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA
| | - J. John Mann
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Division of Molecular Imaging and Neuropathology, Columbia University Irving Medical Center, New York, NY, USA
| | - Mark Underwood
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Division of Molecular Imaging and Neuropathology, Columbia University Irving Medical Center, New York, NY, USA
| | - Maura Boldrini
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Division of Molecular Imaging and Neuropathology, Columbia University Irving Medical Center, New York, NY, USA
| | - Michel Thiebaut de Schotten
- Brain Connectivity and Behavior Laboratory, Paris, France; Groupe d’Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives-UMR 5293, CNRS, CEA University of Bordeaux, France
| | - Martin Picard
- Department of Psychiatry, Divisions of Molecular Therapeutics and Behavioral Medicine, Columbia University Irving Medical Center, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
- Department of Neurology, H. Houston Merritt Center, Columbia Translational Neuroscience Initiative, Columbia University Irving Medical Center, New York, NY, USA
- Robert N Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, NY, USA
| |
Collapse
|
22
|
Liu Y, Fan L, Yang H, Wang D, Liu R, Shan T, Xia X. Ketogenic therapy towards precision medicine for brain diseases. Front Nutr 2024; 11:1266690. [PMID: 38450235 PMCID: PMC10915067 DOI: 10.3389/fnut.2024.1266690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 01/29/2024] [Indexed: 03/08/2024] Open
Abstract
Precision nutrition and nutrigenomics are emerging in the development of therapies for multiple diseases. The ketogenic diet (KD) is the most widely used clinical diet, providing high fat, low carbohydrate, and adequate protein. KD produces ketones and alters the metabolism of patients. Growing evidence suggests that KD has therapeutic effects in a wide range of neuronal diseases including epilepsy, neurodegeneration, cancer, and metabolic disorders. Although KD is considered to be a low-side-effect diet treatment, its therapeutic mechanism has not yet been fully elucidated. Also, its induced keto-response among different populations has not been elucidated. Understanding the ketone metabolism in health and disease is critical for the development of KD-associated therapeutics and synergistic therapy under any physiological background. Here, we review the current advances and known heterogeneity of the KD response and discuss the prospects for KD therapy from a precision nutrition perspective.
Collapse
Affiliation(s)
- Yang Liu
- Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, China
- Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, China
| | - Linlin Fan
- Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, China
| | - Haoying Yang
- Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, China
| | - Danli Wang
- Zhoushan People’s Hospital, Zhoushan, China
| | - Runhan Liu
- Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, China
| | - Tikun Shan
- Neurosurgery Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Xue Xia
- Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, China
- School of Environmental and Life Sciences, College of Engineering, Science and Environment, University of Newcastle, Callaghan, NSW, Australia
| |
Collapse
|
23
|
Cantando I, Centofanti C, D’Alessandro G, Limatola C, Bezzi P. Metabolic dynamics in astrocytes and microglia during post-natal development and their implications for autism spectrum disorders. Front Cell Neurosci 2024; 18:1354259. [PMID: 38419654 PMCID: PMC10899402 DOI: 10.3389/fncel.2024.1354259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 02/02/2024] [Indexed: 03/02/2024] Open
Abstract
Autism Spectrum Disorder (ASD) is a complex neurodevelopmental condition characterized by elusive underlying mechanisms. Recent attention has focused on the involvement of astrocytes and microglia in ASD pathology. These glial cells play pivotal roles in maintaining neuronal homeostasis, including the regulation of metabolism. Emerging evidence suggests a potential association between ASD and inborn errors of metabolism. Therefore, gaining a comprehensive understanding of the functions of microglia and astrocytes in ASD is crucial for the development of effective therapeutic interventions. This review aims to provide a summary of the metabolism of astrocytes and microglia during post-natal development and the evidence of disrupted metabolic pathways in ASD, with particular emphasis on those potentially important for the regulation of neuronal post-natal maturation by astrocytes and microglia.
Collapse
Affiliation(s)
- Iva Cantando
- Department of Fundamental Neurosciences (DNF), University of Lausanne, Lausanne, Switzerland
| | - Cristiana Centofanti
- Department of Fundamental Neurosciences (DNF), University of Lausanne, Lausanne, Switzerland
| | - Giuseppina D’Alessandro
- Department of Physiology and Pharmacology, University of Rome Sapienza, Rome, Italy
- Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed Via Atinese 18, Pozzilli, Italy
| | - Cristina Limatola
- Department of Physiology and Pharmacology, University of Rome Sapienza, Rome, Italy
- Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed Via Atinese 18, Pozzilli, Italy
| | - Paola Bezzi
- Department of Fundamental Neurosciences (DNF), University of Lausanne, Lausanne, Switzerland
- Department of Physiology and Pharmacology, University of Rome Sapienza, Rome, Italy
| |
Collapse
|
24
|
Lin X, Wang S, Gao Y. The effects of intermittent fasting for patients with multiple sclerosis (MS): a systematic review. Front Nutr 2024; 10:1328426. [PMID: 38303903 PMCID: PMC10832063 DOI: 10.3389/fnut.2023.1328426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 12/27/2023] [Indexed: 02/03/2024] Open
Abstract
Some studies have investigated the impact of intermittent fasting (IF) for patients with multiple sclerosis (MS). We aimed to conduct a comprehensive systematic review to analyze and summarize all clinical studies concerning the effects of IF on patients with MS. We conducted an exhaustive review of information available in the Embase, Cochrane, and PubMed databases up until 1 September 2023. All clinical research relating to the impacts of IF for patients with MS were included. In total, this systematic review encompassed 5 studies, which included four RCTs and one pilot study. Each study involved was assessed of high quality. The results from these studies demonstrate that IF protocols could potentially serve as an effective dietary strategy for managing symptoms and improving the quality of life in individuals afflicted with MS. In conclusion, IF might be a potential beneficial dietary intervention for MS. However, the number of trials in this field is relatively limited. The large-scale clinical trials to investigate the effects of IF for MS are urgently needed, which may be helpful to manage this intricate neuroimmune disorder. Systematic review registration https://inplasy.com, identifier INPLASY2023100021.
Collapse
Affiliation(s)
- Xiaoxiao Lin
- Department of Geriatrics, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Senile Chronic Diseases, Hangzhou, Zhejiang, China
| | - Shuai Wang
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yue Gao
- Department of Geriatrics, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Senile Chronic Diseases, Hangzhou, Zhejiang, China
| |
Collapse
|
25
|
Makhlouf M, Souza DG, Kurian S, Bellaver B, Ellis H, Kuboki A, Al-Naama A, Hasnah R, Venturin GT, Costa da Costa J, Venugopal N, Manoel D, Mennella J, Reisert J, Tordoff MG, Zimmer ER, Saraiva LR. Short-term consumption of highly processed diets varying in macronutrient content impair the sense of smell and brain metabolism in mice. Mol Metab 2024; 79:101837. [PMID: 37977411 PMCID: PMC10724696 DOI: 10.1016/j.molmet.2023.101837] [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: 09/12/2023] [Revised: 10/29/2023] [Accepted: 11/10/2023] [Indexed: 11/19/2023] Open
Abstract
OBJECTIVE Food processing greatly contributed to increased food safety, diversity, and accessibility. However, the prevalence of highly palatable and highly processed food in our modern diet has exacerbated obesity rates and contributed to a global health crisis. While accumulating evidence suggests that chronic consumption of such foods is detrimental to sensory and neural physiology, it is unclear whether its short-term intake has adverse effects. Here, we assessed how short-term consumption (<2 months) of three diets varying in composition and macronutrient content influence olfaction and brain metabolism in mice. METHODS The diets tested included a grain-based standard chow diet (CHOW; 54% carbohydrate, 32% protein, 14% fat; #8604 Teklad Rodent diet , Envigo Inc.), a highly processed control diet (hpCTR; 70% carbohydrate, 20% protein, 10% fat; #D12450B, Research Diets Inc.), and a highly processed high-fat diet (hpHFD; 20% carbohydrate, 20% protein, 60% fat; #D12492, Research Diets Inc.). We performed behavioral and metabolic phenotyping, electro-olfactogram (EOG) recordings, brain glucose metabolism imaging, and mitochondrial respirometry in different brain regions. We also performed RNA-sequencing (RNA-seq) in the nose and across several brain regions, and conducted differential expression analysis, gene ontology, and network analysis. RESULTS We show that short-term consumption of the two highly processed diets, but not the grain-based diet, regardless of macronutrient content, adversely affects odor-guided behaviors, physiological responses to odorants, transcriptional profiles in the olfactory mucosa and brain regions, and brain glucose metabolism and mitochondrial respiration. CONCLUSIONS Even short periods of highly processed food consumption are sufficient to cause early olfactory and brain abnormalities, which has the potential to alter food choices and influence the risk of developing metabolic disease.
Collapse
Affiliation(s)
| | - Débora G Souza
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | | | - Bruna Bellaver
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Hillary Ellis
- Monell Chemical Senses Center, 3500 Market Street, Philadelphia, PA 19104, USA
| | - Akihito Kuboki
- Monell Chemical Senses Center, 3500 Market Street, Philadelphia, PA 19104, USA
| | | | - Reem Hasnah
- Sidra Medicine, PO Box 26999, Doha, Qatar; College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Gianina Teribele Venturin
- Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Jaderson Costa da Costa
- Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | | | | | - Julie Mennella
- Monell Chemical Senses Center, 3500 Market Street, Philadelphia, PA 19104, USA
| | - Johannes Reisert
- Monell Chemical Senses Center, 3500 Market Street, Philadelphia, PA 19104, USA
| | - Michael G Tordoff
- Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Eduardo R Zimmer
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil; Department of Pharmacology, UFRGS, Porto Alegre, Brazil; Graduate Program in Biological Sciences: Pharmacology and Therapeutics, UFRGS, Porto Alegre, Brazil; McGill Centre for Studies in Aging, Montreal, Canada.
| | - Luis R Saraiva
- Sidra Medicine, PO Box 26999, Doha, Qatar; Monell Chemical Senses Center, 3500 Market Street, Philadelphia, PA 19104, USA; College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar.
| |
Collapse
|
26
|
Nomura M, Murad NF, Madhavan SS, Eap B, Garcia TY, Aguirre CG, Verdin E, Ellerby L, Furman D, Newman JC. A ketogenic diet reduces age-induced chronic neuroinflammation in mice Running title: ketogenic diet and brain inflammaging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.01.569598. [PMID: 38106160 PMCID: PMC10723274 DOI: 10.1101/2023.12.01.569598] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Beta-hydroxybutyrate (BHB) is a ketone body synthesized during fasting or strenuous exercise. Our previous study demonstrated that a cyclic ketogenic diet (KD), which induces BHB levels similar to fasting every other week, reduces midlife mortality and improves memory in aging mice. BHB actively regulates gene expression and inflammatory activation through non-energetic signaling pathways. Neither of these activities has been well-characterized in the brain and they may represent mechanisms by which BHB affects brain function during aging. First, we analyzed hepatic gene expression in an aging KD-treated mouse cohort using bulk RNA-seq. In addition to the downregulation of TOR pathway activity, cyclic KD reduces inflammatory gene expression in the liver. We observed via flow cytometry that KD also modulates age-related systemic T cell functions. Next, we investigated whether BHB affects brain cells transcriptionally in vitro. Gene expression analysis in primary human brain cells (microglia, astrocytes, neurons) using RNA-seq shows that BHB causes a mild level of inflammation in all three cell types. However, BHB inhibits the more pronounced LPS-induced inflammatory gene activation in microglia. Furthermore, we confirmed that BHB similarly reduces LPS-induced inflammation in primary mouse microglia and bone marrow-derived macrophages (BMDMs). BHB is recognized as an inhibitor of histone deacetylase (HDAC), an inhibitor of NLRP3 inflammasome, and an agonist of the GPCR Hcar2. Nevertheless, in microglia, BHB's anti-inflammatory effects are independent of these known mechanisms. Finally, we examined the brain gene expression of 12-month-old male mice fed with one-week and one-year cyclic KD. While a one-week KD increases inflammatory signaling, a one-year cyclic KD reduces neuroinflammation induced by aging. In summary, our findings demonstrate that BHB mitigates the microglial response to inflammatory stimuli, like LPS, possibly leading to decreased chronic inflammation in the brain after long-term KD treatment in aging mice.
Collapse
Affiliation(s)
| | | | - Sidharth S Madhavan
- Buck Institute for Research on Aging, Novato, CA, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Brenda Eap
- Buck Institute for Research on Aging, Novato, CA, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | | | - Carlos Galicia Aguirre
- Buck Institute for Research on Aging, Novato, CA, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Eric Verdin
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Lisa Ellerby
- Buck Institute for Research on Aging, Novato, CA, USA
| | - David Furman
- Buck Institute for Research on Aging, Novato, CA, USA
| | - John C Newman
- Buck Institute for Research on Aging, Novato, CA, USA
- Division of Geriatrics, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
27
|
Chatterjee M, Özdemir S, Kunadt M, Koel-Simmelink M, Boiten W, Piepkorn L, Pham TV, Chiasserini D, Piersma SR, Knol JC, Möbius W, Mollenhauer B, van der Flier WM, Jimenez CR, Teunissen CE, Jahn O, Schneider A. C1q is increased in cerebrospinal fluid-derived extracellular vesicles in Alzheimer's disease: A multi-cohort proteomics and immuno-assay validation study. Alzheimers Dement 2023; 19:4828-4840. [PMID: 37023079 DOI: 10.1002/alz.13066] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 02/03/2023] [Accepted: 02/07/2023] [Indexed: 04/07/2023]
Abstract
INTRODUCTION Extracellular vesicles (EVs) may propagate and modulate Alzheimer's disease (AD) pathology. We aimed to comprehensively characterize the proteome of cerebrospinal fluid (CSF) EVs to identify proteins and pathways altered in AD. METHODS CSF EVs were isolated by ultracentrifugation (Cohort 1) or Vn96 peptide (Cohort 2) from non-neurodegenerative controls (n = 15, 16) and AD patients (n = 22, 20, respectively). EVs were subjected to untargeted quantitative mass spectrometry-based proteomics. Results were validated by enzyme-linked immunosorbent assay (ELISA) in Cohorts 3 and 4, consisting of controls (n = 16, n = 43, (Cohort3, Cohort4)), and patients with AD (n = 24, n = 100). RESULTS We found > 30 differentially expressed proteins in AD CSF EVs involved in immune-regulation. Increase of C1q levels in AD compared to non-demented controls was validated by ELISA (∼ 1.5 fold, p (Cohort 3) = 0.03, p (Cohort 4) = 0.005). DISCUSSION EVs may be utilized as a potential biomarker and may play a so far unprecedented role in immune-regulation in AD.
Collapse
Affiliation(s)
| | - Selcuk Özdemir
- German Center for Neurodegenerative Diseases, DZNE, Bonn, Germany
| | - Marcel Kunadt
- Department of Psychiatry and Psychotherapy, University Medical Center, Goettingen, Germany
| | - Marleen Koel-Simmelink
- Clinical Chemistry Department, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Walter Boiten
- Clinical Chemistry Department, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Lars Piepkorn
- Department of Psychiatry and Psychotherapy, University Medical Center, Goettingen, Germany
- Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Thang V Pham
- OncoProteomics Laboratory, Department Medical Oncology, 1098 XH Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, The Netherlands
| | - Davide Chiasserini
- OncoProteomics Laboratory, Department Medical Oncology, 1098 XH Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, The Netherlands
- Department of Experimental Medicine, Section of Physiology and Biochemistry, University of Perugia, Perugia, Italy
| | - Sander R Piersma
- OncoProteomics Laboratory, Department Medical Oncology, 1098 XH Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, The Netherlands
| | - Jaco C Knol
- OncoProteomics Laboratory, Department Medical Oncology, 1098 XH Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, The Netherlands
| | - Wiebke Möbius
- Department of Neurogenetics, Electron Microscopy, City Campus, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Brit Mollenhauer
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
- Paracelsus-Elena-Klinik, Kassel, Germany
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Neurology, Epidemiology and Data Science, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Connie R Jimenez
- OncoProteomics Laboratory, Department Medical Oncology, 1098 XH Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, The Netherlands
| | - Charlotte E Teunissen
- Clinical Chemistry Department, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Olaf Jahn
- Department of Psychiatry and Psychotherapy, University Medical Center, Goettingen, Germany
- Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Anja Schneider
- German Center for Neurodegenerative Diseases, DZNE, Bonn, Germany
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
28
|
Brockhoff JD, Bereswill S, Heimesaat MM. The impact of ketogenic diet on the onset and progression of multiple sclerosis. Eur J Microbiol Immunol (Bp) 2023; 13:29-36. [PMID: 37665667 PMCID: PMC10578139 DOI: 10.1556/1886.2023.00020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 08/21/2023] [Indexed: 09/06/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disease of the central nervous system (CNS) characterized by inflammation and neurodegeneration. Current research suggests that diet may influence disease course, severity of symptoms, and quality of life in MS patients. The ketogenic diet (KD) has been used for more than a century as a therapeutic approach for various medical conditions. It was originally developed in the 1920s as a treatment option for epilepsy, and especially in the last 30 years, has gained popularity for its potential benefits in a variety of neurological conditions other than epilepsy. This prompted us to perform a literature survey regarding the effect of KD on the onset and progression of MS. The here reviewed 15 original research articles including in vitro, preclinical, and clinical studies provide evidence for the safety and feasibility of the KD in MS, showing potential neuroprotective effects and positive impacts on cellular metabolism and disease outcome. Since the literature is limited and most studies were conducted with low numbers of MS patients and rather exploratory in nature, further studies with larger cohorts are needed to gain a better understanding of the mechanisms by which the improvements of the MS disease course are achieved.
Collapse
Affiliation(s)
- Jurij D. Brockhoff
- Gastrointestinal Microbiology Research Group, Institute of Microbiology, Infectious Diseases and Immunology, Charité - University Medicine Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Stefan Bereswill
- Gastrointestinal Microbiology Research Group, Institute of Microbiology, Infectious Diseases and Immunology, Charité - University Medicine Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Markus M. Heimesaat
- Gastrointestinal Microbiology Research Group, Institute of Microbiology, Infectious Diseases and Immunology, Charité - University Medicine Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
29
|
Fehsel K. Why Is Iron Deficiency/Anemia Linked to Alzheimer's Disease and Its Comorbidities, and How Is It Prevented? Biomedicines 2023; 11:2421. [PMID: 37760862 PMCID: PMC10526115 DOI: 10.3390/biomedicines11092421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/23/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Impaired iron metabolism has been increasingly observed in many diseases, but a deeper, mechanistic understanding of the cellular impact of altered iron metabolism is still lacking. In addition, deficits in neuronal energy metabolism due to reduced glucose import were described for Alzheimer's disease (AD) and its comorbidities like obesity, depression, cardiovascular disease, and type 2 diabetes mellitus. The aim of this review is to present the molecular link between both observations. Insufficient cellular glucose uptake triggers increased ferritin expression, leading to depletion of the cellular free iron pool and stabilization of the hypoxia-induced factor (HIF) 1α. This transcription factor induces the expression of the glucose transporters (Glut) 1 and 3 and shifts the cellular metabolism towards glycolysis. If this first line of defense is not adequate for sufficient glucose supply, further reduction of the intracellular iron pool affects the enzymes of the mitochondrial electron transport chain and activates the AMP-activated kinase (AMPK). This enzyme triggers the translocation of Glut4 to the plasma membrane as well as the autophagic recycling of cell components in order to mobilize energy resources. Moreover, AMPK activates the autophagic process of ferritinophagy, which provides free iron urgently needed as a cofactor for the synthesis of heme- and iron-sulfur proteins. Excessive activation of this pathway ends in ferroptosis, a special iron-dependent form of cell death, while hampered AMPK activation steadily reduces the iron pools, leading to hypoferremia with iron sequestration in the spleen and liver. Long-lasting iron depletion affects erythropoiesis and results in anemia of chronic disease, a common condition in patients with AD and its comorbidities. Instead of iron supplementation, drugs, diet, or phytochemicals that improve energy supply and cellular glucose uptake should be administered to counteract hypoferremia and anemia of chronic disease.
Collapse
Affiliation(s)
- Karin Fehsel
- Neurobiochemical Research Unit, Department of Psychiatry, Medical Faculty, Heinrich-Heine-University, 240629 Düsseldorf, Germany
| |
Collapse
|
30
|
Scandella V, Petrelli F, Moore DL, Braun SMG, Knobloch M. Neural stem cell metabolism revisited: a critical role for mitochondria. Trends Endocrinol Metab 2023; 34:446-461. [PMID: 37380501 DOI: 10.1016/j.tem.2023.05.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/29/2023] [Accepted: 05/30/2023] [Indexed: 06/30/2023]
Abstract
Metabolism has emerged as a key regulator of stem cell behavior. Mitochondria are crucial metabolic organelles that are important for differentiated cells, yet considered less so for stem cells. However, recent studies have shown that mitochondria influence stem cell maintenance and fate decisions, inviting a revised look at this topic. In this review, we cover the current literature addressing the role of mitochondrial metabolism in mouse and human neural stem cells (NSCs) in the embryonic and adult brain. We summarize how mitochondria are implicated in fate regulation and how substrate oxidation affects NSC quiescence. We further explore single-cell RNA sequencing (scRNA-seq) data for metabolic signatures of adult NSCs, highlight emerging technologies reporting on metabolic signatures, and discuss mitochondrial metabolism in other stem cells.
Collapse
Affiliation(s)
- Valentina Scandella
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Francesco Petrelli
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Darcie L Moore
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, USA
| | - Simon M G Braun
- Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland
| | - Marlen Knobloch
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
31
|
García-Velázquez L, Massieu L. The proteomic effects of ketone bodies: implications for proteostasis and brain proteinopathies. Front Mol Neurosci 2023; 16:1214092. [PMID: 37575967 PMCID: PMC10413579 DOI: 10.3389/fnmol.2023.1214092] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 07/07/2023] [Indexed: 08/15/2023] Open
Abstract
A growing body of evidence supports the beneficial effects of the ketone bodies (KBs), acetoacetate and β-hydroxybutyrate (BHB), on diverse physiological processes and diseases. Hence, KBs have been suggested as therapeutic tools for neurodegenerative diseases. KBs are an alternative fuel during fasting and starvation as they can be converted to Ac-CoA to produce ATP. A ketogenic diet (KD), enriched in fats and low in carbohydrates, induces KB production in the liver and favors their use in the brain. BHB is the most abundant KB in the circulation; in addition to its role as energy fuel, it exerts many actions that impact the set of proteins in the cell and tissue. BHB can covalently bind to proteins in lysine residues as a new post-translational modification (PTM) named β-hydroxybutyrylation (Kbhb). Kbhb has been identified in many proteins where Kbhb sites can be critical for binding to other proteins or cofactors. Kbhb is mostly found in proteins involved in chromatin structure, DNA repair, regulation of spliceosome, transcription, and oxidative phosphorylation. Histones are the most studied family of proteins with this PTM, and H3K9bhb is the best studied histone mark. Their target genes are mainly related to cell metabolism, chromatin remodeling and the control of circadian rhythms. The role of Kbhb on physiological processes is poorly known, but it might link KB metabolism to cell signaling and genome regulation. BHB also impacts the proteome by influencing proteostasis. This KB can modulate the Unfolded Protein Response (UPR) and autophagy, two processes involved in the maintenance of protein homeostasis through the clearance of accumulated unfolded and damaged proteins. BHB can support proteostasis and regulate the UPR to promote metabolism adaptation in the liver and prevent cell damage in the brain. Also, BHB stimulates autophagy aiding to the degradation of accumulated proteins. Protein aggregation is common to proteinopathies like Alzheimer's (AD) and Parkinson's (PD) diseases, where the KD and BHB treatment have shown favorable effects. In the present review, the current literature supporting the effects of KBs on proteome conformation and proteostasis is discussed, as well as its possible impact on AD and PD.
Collapse
Affiliation(s)
| | - Lourdes Massieu
- Department of Molecular Neuropathology, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México (UNAM), México City, Mexico
| |
Collapse
|
32
|
Li Z, Jiang Y, Long C, Peng Q, Yue R. The gut microbiota-astrocyte axis: Implications for type 2 diabetic cognitive dysfunction. CNS Neurosci Ther 2023; 29 Suppl 1:59-73. [PMID: 36601656 PMCID: PMC10314112 DOI: 10.1111/cns.14077] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/20/2022] [Accepted: 12/18/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Diabetic cognitive dysfunction (DCD) is one of the most insidious complications of type 2 diabetes mellitus, which can seriously affect the ability to self-monitoring of blood glucose and the quality of life in the elderly. Previous pathological studies of cognitive dysfunction have focused on neuronal dysfunction, characterized by extracellular beta-amyloid deposition and intracellular tau hyperphosphorylation. In recent years, astrocytes have been recognized as a potential therapeutic target for cognitive dysfunction and important participants in the central control of metabolism. The disorder of gut microbiota and their metabolites have been linked to a series of metabolic diseases such as diabetes mellitus. The imbalance of intestinal flora has the effect of promoting the occurrence and deterioration of several diabetes-related complications. Gut microbes and their metabolites can drive astrocyte activation. AIMS We reviewed the pathological progress of DCD related to the "gut microbiota-astrocyte" axis in terms of peripheral and central inflammation, intestinal and blood-brain barrier (BBB) dysfunction, systemic and brain energy metabolism disorders to deepen the pathological research progress of DCD and explore the potential therapeutic targets. CONCLUSION "Gut microbiota-astrocyte" axis, unique bidirectional crosstalk in the brain-gut axis, mediates the intermediate pathological process of neurocognitive dysfunction secondary to metabolic disorders in diabetes mellitus.
Collapse
Affiliation(s)
- Zi‐Han Li
- Hospital of Chengdu University of Traditional Chinese MedicineChengduChina
| | - Ya‐Yi Jiang
- Hospital of Chengdu University of Traditional Chinese MedicineChengduChina
| | - Cai‐Yi Long
- Hospital of Chengdu University of Traditional Chinese MedicineChengduChina
| | - Qian Peng
- Hospital of Chengdu University of Traditional Chinese MedicineChengduChina
| | - Ren‐Song Yue
- Hospital of Chengdu University of Traditional Chinese MedicineChengduChina
| |
Collapse
|
33
|
Li S, Sheng ZH. Oligodendrocyte-derived transcellular signaling regulates axonal energy metabolism. Curr Opin Neurobiol 2023; 80:102722. [PMID: 37028201 PMCID: PMC10225329 DOI: 10.1016/j.conb.2023.102722] [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: 11/22/2022] [Revised: 02/28/2023] [Accepted: 03/07/2023] [Indexed: 04/08/2023]
Abstract
The unique morphology and functionality of central nervous system (CNS) neurons necessitate specialized mechanisms to maintain energy metabolism throughout long axons and extensive terminals. Oligodendrocytes (OLs) enwrap CNS axons with myelin sheaths in a multilamellar fashion. Apart from their well-established function in action potential propagation, OLs also provide intercellular metabolic support to axons by transferring energy metabolites and delivering exosomes consisting of proteins, lipids, and RNAs. OL-derived metabolic support is crucial for the maintenance of axonal integrity; its dysfunction has emerged as an important player in neurological disorders that are associated with axonal energy deficits and degeneration. In this review, we discuss recent advances in how these transcellular signaling pathways maintain axonal energy metabolism in health and neurological disorders.
Collapse
Affiliation(s)
- Sunan Li
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health, Room 2B-215, 35 Convent Drive, Bethesda, MD 20892-3706, USA. https://twitter.com/@sunan_li
| | - Zu-Hang Sheng
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health, Room 2B-215, 35 Convent Drive, Bethesda, MD 20892-3706, USA.
| |
Collapse
|
34
|
Zyla-Jackson K, Walton DA, Plafker KS, Kovats S, Georgescu C, Brush RS, Tytanic M, Agbaga MP, Plafker SM. Dietary protection against the visual and motor deficits induced by experimental autoimmune encephalomyelitis. Front Neurol 2023; 14:1113954. [PMID: 36937529 PMCID: PMC10017782 DOI: 10.3389/fneur.2023.1113954] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 02/13/2023] [Indexed: 03/06/2023] Open
Abstract
Introduction Five to eight percent of the world population currently suffers from at least one autoimmune disorder. Despite multiple immune modulatory therapies for autoimmune demyelinating diseases of the central nervous system, these treatments can be limiting for subsets of patients due to adverse effects and expense. To circumvent these barriers, we investigated a nutritional intervention in mice undergoing experimental autoimmune encephalomyelitis (EAE), a model of autoimmune-mediated demyelination that induces visual and motor pathologies similar to those experienced by people with multiple sclerosis (MS). Methods EAE was induced in female and male mice and the impact of limiting dietary carbohydrates by feeding a ketogenic diet (KD) enriched in medium chain triglycerides (MCTs), alpha-linolenic acid (an omega-3 fatty acid), and fiber was evaluated in both a preventive regimen (prior to immunization with MOG antigen) and an interventional regimen (following the onset of symptoms). Motor scores were assigned daily and visual acuity was measured using optokinetic tracking. Immunohistochemical analyses of optic nerves were done to assess inflammatory infiltrates and myelination status. Fatty acid and cytokine profiling from blood were performed to evaluate systemic inflammatory status. Results The KD was efficacious when fed as a preventive regimen as well as when initiated as an interventional regimen following symptom onset. The KD minimally impacted body weight during the experimental time course, increased circulating ketones, prevented motor and ocular deficits, preserved myelination of the optic nerve, and reduced infiltration of immune cells to optic nerves. The KD also increased anti-inflammatory-associated omega-3 fatty acids in the plasma and reduced select cytokines in the circulation associated with EAE-mediated pathological inflammation. Discussion In light of ongoing clinical trials using dietary strategies to treat people with MS, these findings support that a KD enriched in MCTs, omega-3 fatty acids, and fiber promotes a systemic anti-inflammatory milieu and ameliorates autoimmune-induced demyelinating visual and motor deficits.
Collapse
Affiliation(s)
- Katarzyna Zyla-Jackson
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Dorothy A. Walton
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Kendra S. Plafker
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Susan Kovats
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Constantin Georgescu
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Richard S. Brush
- Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Madison Tytanic
- Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Martin-Paul Agbaga
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Scott M. Plafker
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- *Correspondence: Scott M. Plafker
| |
Collapse
|