1
|
Stratton MS, López-Domínguez JA, Canella A, Ramsey JJ, Cortopassi GA. Differential effects of short-term and long-term ketogenic diet on gene expression in the aging mouse brain. J Nutr Health Aging 2025; 29:100427. [PMID: 39662156 DOI: 10.1016/j.jnha.2024.100427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 12/13/2024]
Abstract
BACKGROUND Aging is associated with multiple neurodegenerative conditions that severely limit quality of life and can shorten lifespan. Studies in rodents indicate that in addition to extending lifespan, the ketogenic diet (KD) improves cognitive function in aged animals, yet long term adherence to KD in Humans is poor. OBJECTIVES To broadly investigate what mechanisms might be activated in the brain in response to ketogenic diet. METHODS We conducted transcriptome wide analysis on whole brain samples from 13-month-old mice, 13-month-old mice fed a ketogenic diet for 1 month, 26-month-old mice, and 26-month-old mice fed a ketogenic diet for 14 months. RESULTS As expected, analysis of differently expressed genes between the old (26 month) vs younger mice (13 month) showed clear activation of inflammation and complement system pathways with aging. Analysis between the 26-month-old animals fed ketogenic diet for 14 months with 26-month-old animals fed control diet indicate that long-term KD resulted in activation of LRP, TCF7L2 (WNT pathway), and IGF1 signaling. There was also a significant increase in the expression of SOX2-dependent oligodendrocyte/myelination markers, though TCF7L2 and SOX2 dependent gene sets were largely overlapping. Remarkably, the effect of 1 month of ketogenic diet was minimal and there was no congruence between gene expression effects of short-term KD vs long-term KD. CONCLUSIONS This work informs target identification efforts for aging and neurodegenerative disorder therapeutics discovery while also establishing differential effects of short-term vs long-term KD on gene expression in the brain.
Collapse
Affiliation(s)
- Matthew S Stratton
- Department of Physiology & Cell Biology, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH 43210, United States.
| | - José Alberto López-Domínguez
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, United States
| | - Alessandro Canella
- Department of Physiology & Cell Biology, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH 43210, United States
| | - Jon J Ramsey
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, United States
| | - Gino A Cortopassi
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, United States
| |
Collapse
|
2
|
Elitt CM, Volpe JJ. Degenerative Disorders of the Newborn. VOLPE'S NEUROLOGY OF THE NEWBORN 2025:967-1007.e17. [DOI: 10.1016/b978-0-443-10513-5.00033-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
3
|
Fernandes MGF, Pernin F, Antel JP, Kennedy TE. From BBB to PPP: Bioenergetic requirements and challenges for oligodendrocytes in health and disease. J Neurochem 2025; 169:e16219. [PMID: 39253904 PMCID: PMC11657931 DOI: 10.1111/jnc.16219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 06/24/2024] [Accepted: 08/08/2024] [Indexed: 09/11/2024]
Abstract
Mature myelinating oligodendrocytes, the cells that produce the myelin sheath that insulates axons in the central nervous system, have distinct energetic and metabolic requirements compared to neurons. Neurons require substantial energy to execute action potentials, while the energy needs of oligodendrocytes are directed toward building the lipid-rich components of myelin and supporting neuronal metabolism by transferring glycolytic products to axons as additional fuel. The utilization of energy metabolites in the brain parenchyma is tightly regulated to meet the needs of different cell types. Disruption of the supply of metabolites can lead to stress and oligodendrocyte injury, contributing to various neurological disorders, including some demyelinating diseases. Understanding the physiological properties, structures, and mechanisms involved in oligodendrocyte energy metabolism, as well as the relationship between oligodendrocytes and neighboring cells, is crucial to investigate the underlying pathophysiology caused by metabolic impairment in these disorders. In this review, we describe the particular physiological properties of oligodendrocyte energy metabolism and the response of oligodendrocytes to metabolic stress. We delineate the relationship between oligodendrocytes and other cells in the context of the neurovascular unit, and the regulation of metabolite supply according to energetic needs. We focus on the specific bioenergetic requirements of oligodendrocytes and address the disruption of metabolic energy in demyelinating diseases. We encourage further studies to increase understanding of the significance of metabolic stress on oligodendrocyte injury, to support the development of novel therapeutic approaches for the treatment of demyelinating diseases.
Collapse
Affiliation(s)
- Milton Guilherme Forestieri Fernandes
- Neuroimmunological Diseases and Glia Biology Research Group, Department of Neurology and Neurosurgery, Montreal Neurological InstituteMcGill UniversityMontrealQuebecCanada
| | - Florian Pernin
- Neuroimmunological Diseases and Glia Biology Research Group, Department of Neurology and Neurosurgery, Montreal Neurological InstituteMcGill UniversityMontrealQuebecCanada
| | - Jack P. Antel
- Neuroimmunological Diseases and Glia Biology Research Group, Department of Neurology and Neurosurgery, Montreal Neurological InstituteMcGill UniversityMontrealQuebecCanada
| | - Timothy E. Kennedy
- Neuroimmunological Diseases and Glia Biology Research Group, Department of Neurology and Neurosurgery, Montreal Neurological InstituteMcGill UniversityMontrealQuebecCanada
| |
Collapse
|
4
|
Omori NE, Malys MK, Woo G, Mansor L. Exogenous ketone bodies and the ketogenic diet as a treatment option for neurodevelopmental disorders. Front Nutr 2024; 11:1485280. [PMID: 39749357 PMCID: PMC11693454 DOI: 10.3389/fnut.2024.1485280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 11/13/2024] [Indexed: 01/04/2025] Open
Abstract
Background Despite being the most prevalent neurodevelopmental disorders, there are comparatively few treatment options available to patients presenting with autism spectrum disorder (ASD) and attention deficit hyperactivity disorder (ADHD). The ketogenic diet has historically shown therapeutic utility in treating refractory epilepsy, an adjacent neuropsychiatric condition, in children, adolescents and adults. The following review explores preclinical and clinical literature focusing on the therapeutic potential of the ketogenic diet and exogenous ketone body supplementation in treating common neurodevelopmental disorders. Method A narrative review of extant literature was conducted across the domains of perinatal nutrition, ASD, and ADHD. Preclinical and clinical studies focusing on the effect of either the ketogenic diet or exogenous ketone supplementation as a treatment option were included for review. Results 14 preclinical and 10 clinical studies were included for discussion. Data supporting the use of a ketogenic intervention for neurodevelopmental disorders is mixed. High heterogeneity in study design was noted for preclinical models, ketogenic intervention, and outcomes measured. Conclusion Studies evaluating ketogenic interventions for neurodevelopmental disorders remain in their infancy in terms of both the depth and scope of available literature. The safety and tolerability of ketogenic diets and supplements means there would be value in exploring their effectiveness further in clinical studies.
Collapse
Affiliation(s)
- Naomi Elyse Omori
- Health Via Modern Nutrition Inc. (H.V.M.N.), San Francisco, CA, United States
| | - Mantas Kazimieras Malys
- Department of Psychological Medicine, King’s College London, Institute of Psychiatry, Psychology & Neuroscience, London, United Kingdom
| | - Geoffrey Woo
- Health Via Modern Nutrition Inc. (H.V.M.N.), San Francisco, CA, United States
| | - Latt Mansor
- Health Via Modern Nutrition Inc. (H.V.M.N.), San Francisco, CA, United States
| |
Collapse
|
5
|
Asadollahi E, Trevisiol A, Saab AS, Looser ZJ, Dibaj P, Ebrahimi R, Kusch K, Ruhwedel T, Möbius W, Jahn O, Lee JY, Don AS, Khalil MA, Hiller K, Baes M, Weber B, Abel ED, Ballabio A, Popko B, Kassmann CM, Ehrenreich H, Hirrlinger J, Nave KA. Oligodendroglial fatty acid metabolism as a central nervous system energy reserve. Nat Neurosci 2024; 27:1934-1944. [PMID: 39251890 PMCID: PMC11452346 DOI: 10.1038/s41593-024-01749-6] [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: 06/22/2023] [Accepted: 08/05/2024] [Indexed: 09/11/2024]
Abstract
Brain function requires a constant supply of glucose. However, the brain has no known energy stores, except for glycogen granules in astrocytes. In the present study, we report that continuous oligodendroglial lipid metabolism provides an energy reserve in white matter tracts. In the isolated optic nerve from young adult mice of both sexes, oligodendrocytes survive glucose deprivation better than astrocytes. Under low glucose, both axonal ATP levels and action potentials become dependent on fatty acid β-oxidation. Importantly, ongoing oligodendroglial lipid degradation feeds rapidly into white matter energy metabolism. Although not supporting high-frequency spiking, fatty acid β-oxidation in mitochondria and oligodendroglial peroxisomes protects axons from conduction blocks when glucose is limiting. Disruption of the glucose transporter GLUT1 expression in oligodendrocytes of adult mice perturbs myelin homeostasis in vivo and causes gradual demyelination without behavioral signs. This further suggests that the imbalance of myelin synthesis and degradation can underlie myelin thinning in aging and disease.
Collapse
Affiliation(s)
- Ebrahim Asadollahi
- Max Planck Institute for Multidisciplinary Sciences, Department of Neurogenetics, Göttingen, Germany.
| | - Andrea Trevisiol
- Max Planck Institute for Multidisciplinary Sciences, Department of Neurogenetics, Göttingen, Germany
- University of Toronto, Sunnybrook Health Sciences Centre, Department of Physical Sciences, North York, Ontario, Canada
| | - Aiman S Saab
- Max Planck Institute for Multidisciplinary Sciences, Department of Neurogenetics, Göttingen, Germany
- University of Zurich, Institute of Pharmacology and Toxicology, Zurich, Switzerland
| | - Zoe J Looser
- University of Zurich, Institute of Pharmacology and Toxicology, Zurich, Switzerland
| | - Payam Dibaj
- Max Planck Institute for Multidisciplinary Sciences, Department of Neurogenetics, Göttingen, Germany
- Center for Rare Diseases Göttingen, Department of Pediatrics and Pediatric Neurology, Georg August University Göttingen, Göttingen, Germany
| | - Reyhane Ebrahimi
- Max Planck Institute for Multidisciplinary Sciences, Department of Neurogenetics, Göttingen, Germany
| | - Kathrin Kusch
- Max Planck Institute for Multidisciplinary Sciences, Department of Neurogenetics, Göttingen, Germany
- University of Göttingen Medical School, Institute for Auditory Neuroscience and Inner Ear Lab, Göttingen, Germany
| | - Torben Ruhwedel
- Max Planck Institute for Multidisciplinary Sciences, Department of Neurogenetics, Göttingen, Germany
| | - Wiebke Möbius
- Max Planck Institute for Multidisciplinary Sciences, Department of Neurogenetics, Göttingen, Germany
| | - Olaf Jahn
- Max Planck Institute for Multidisciplinary Sciences, Department of Molecular Neurobiology, Neuroproteomics Group, Göttingen, Germany
- University Medical Center Göttingen, Department of Psychiatry and Psychotherapy, Translational Neuroproteomics Group, Göttingen, Germany
| | - Jun Yup Lee
- School of Medical Sciences and Charles Perkins Centre, The University of Sydney, Camperdown, New South Wales, Australia
| | - Anthony S Don
- School of Medical Sciences and Charles Perkins Centre, The University of Sydney, Camperdown, New South Wales, Australia
| | - Michelle-Amirah Khalil
- Department for Bioinformatics and Biochemistry, Braunschweig Integrated Center of System Biology, Technische Universität Braunschweig, Braunschweig, Germany
| | - Karsten Hiller
- Department for Bioinformatics and Biochemistry, Braunschweig Integrated Center of System Biology, Technische Universität Braunschweig, Braunschweig, Germany
| | - Myriam Baes
- Lab of Cell Metabolism, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Bruno Weber
- University of Zurich, Institute of Pharmacology and Toxicology, Zurich, Switzerland
| | - E Dale Abel
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine, Naples, Italy
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Brian Popko
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Celia M Kassmann
- Max Planck Institute for Multidisciplinary Sciences, Department of Neurogenetics, Göttingen, Germany
| | - Hannelore Ehrenreich
- Max Planck Institute for Multidisciplinary Sciences, Clinical Neuroscience, Göttingen, Germany
- Central Institute of Mental Health, Mannheim, Germany
| | - Johannes Hirrlinger
- Max Planck Institute for Multidisciplinary Sciences, Department of Neurogenetics, Göttingen, Germany
- Carl-Ludwig-Institute for Physiology, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Klaus-Armin Nave
- Max Planck Institute for Multidisciplinary Sciences, Department of Neurogenetics, Göttingen, Germany.
| |
Collapse
|
6
|
Briglia M, Allia F, Avola R, Signorini C, Cardile V, Romano GL, Giurdanella G, Malaguarnera R, Bellomo M, Graziano ACE. Diet and Nutrients in Rare Neurological Disorders: Biological, Biochemical, and Pathophysiological Evidence. Nutrients 2024; 16:3114. [PMID: 39339713 PMCID: PMC11435074 DOI: 10.3390/nu16183114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 09/12/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
Background/Objectives: Rare diseases are a wide and heterogeneous group of multisystem life-threatening or chronically debilitating clinical conditions with reduced life expectancy and a relevant mortality rate in childhood. Some of these disorders have typical neurological symptoms, presenting from birth to adulthood. Dietary patterns and nutritional compounds play key roles in the onset and progression of neurological disorders, and the impact of alimentary needs must be enlightened especially in rare neurological diseases. This work aims to collect the in vitro, in vivo, and clinical evidence on the effects of diet and of nutrient intake on some rare neurological disorders, including some genetic diseases, and rare brain tumors. Herein, those aspects are critically linked to the genetic, biological, biochemical, and pathophysiological hallmarks typical of each disorder. Methods: By searching the major web-based databases (PubMed, Web of Science Core Collection, DynaMed, and Clinicaltrials.gov), we try to sum up and improve our understanding of the emerging role of nutrition as both first-line therapy and risk factors in rare neurological diseases. Results: In line with the increasing number of consensus opinions suggesting that nutrients should receive the same attention as pharmacological treatments, the results of this work pointed out that a standard dietary recommendation in a specific rare disease is often limited by the heterogeneity of occurrent genetic mutations and by the variability of pathophysiological manifestation. Conclusions: In conclusion, we hope that the knowledge gaps identified here may inspire further research for a better evaluation of molecular mechanisms and long-term effects.
Collapse
Affiliation(s)
- Marilena Briglia
- Department of Medicine and Surgery, “Kore” University of Enna, 94100 Enna, Italy; (M.B.); (F.A.); (R.A.); (G.L.R.); (R.M.); (M.B.)
| | - Fabio Allia
- Department of Medicine and Surgery, “Kore” University of Enna, 94100 Enna, Italy; (M.B.); (F.A.); (R.A.); (G.L.R.); (R.M.); (M.B.)
| | - Rosanna Avola
- Department of Medicine and Surgery, “Kore” University of Enna, 94100 Enna, Italy; (M.B.); (F.A.); (R.A.); (G.L.R.); (R.M.); (M.B.)
| | - Cinzia Signorini
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy;
| | - Venera Cardile
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy;
| | - Giovanni Luca Romano
- Department of Medicine and Surgery, “Kore” University of Enna, 94100 Enna, Italy; (M.B.); (F.A.); (R.A.); (G.L.R.); (R.M.); (M.B.)
| | - Giovanni Giurdanella
- Department of Medicine and Surgery, “Kore” University of Enna, 94100 Enna, Italy; (M.B.); (F.A.); (R.A.); (G.L.R.); (R.M.); (M.B.)
| | - Roberta Malaguarnera
- Department of Medicine and Surgery, “Kore” University of Enna, 94100 Enna, Italy; (M.B.); (F.A.); (R.A.); (G.L.R.); (R.M.); (M.B.)
| | - Maria Bellomo
- Department of Medicine and Surgery, “Kore” University of Enna, 94100 Enna, Italy; (M.B.); (F.A.); (R.A.); (G.L.R.); (R.M.); (M.B.)
| | - Adriana Carol Eleonora Graziano
- Department of Medicine and Surgery, “Kore” University of Enna, 94100 Enna, Italy; (M.B.); (F.A.); (R.A.); (G.L.R.); (R.M.); (M.B.)
| |
Collapse
|
7
|
Späte E, Zhou B, Sun T, Kusch K, Asadollahi E, Siems SB, Depp C, Werner HB, Saher G, Hirrlinger J, Möbius W, Nave KA, Goebbels S. Downregulated expression of lactate dehydrogenase in adult oligodendrocytes and its implication for the transfer of glycolysis products to axons. Glia 2024; 72:1374-1391. [PMID: 38587131 DOI: 10.1002/glia.24533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 03/25/2024] [Accepted: 03/26/2024] [Indexed: 04/09/2024]
Abstract
Oligodendrocytes and astrocytes are metabolically coupled to neuronal compartments. Pyruvate and lactate can shuttle between glial cells and axons via monocarboxylate transporters. However, lactate can only be synthesized or used in metabolic reactions with the help of lactate dehydrogenase (LDH), a tetramer of LDHA and LDHB subunits in varying compositions. Here we show that mice with a cell type-specific disruption of both Ldha and Ldhb genes in oligodendrocytes lack a pathological phenotype that would be indicative of oligodendroglial dysfunctions or lack of axonal metabolic support. Indeed, when combining immunohistochemical, electron microscopical, and in situ hybridization analyses in adult mice, we found that the vast majority of mature oligodendrocytes lack detectable expression of LDH. Even in neurodegenerative disease models and in mice under metabolic stress LDH was not increased. In contrast, at early development and in the remyelinating brain, LDHA was readily detectable in immature oligodendrocytes. Interestingly, by immunoelectron microscopy LDHA was particularly enriched at gap junctions formed between adjacent astrocytes and at junctions between astrocytes and oligodendrocytes. Our data suggest that oligodendrocytes metabolize lactate during development and remyelination. In contrast, for metabolic support of axons mature oligodendrocytes may export their own glycolysis products as pyruvate rather than lactate. Lacking LDH, these oligodendrocytes can also "funnel" lactate through their "myelinic" channels between gap junction-coupled astrocytes and axons without metabolizing it. We suggest a working model, in which the unequal cellular distribution of LDH in white matter tracts facilitates a rapid and efficient transport of glycolysis products among glial and axonal compartments.
Collapse
Affiliation(s)
- Erik Späte
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Baoyu Zhou
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Ting Sun
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Laboratory of Molecular Neurobiology, Department of Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Kathrin Kusch
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
| | - Ebrahim Asadollahi
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Sophie B Siems
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Constanze Depp
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Hauke B Werner
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Gesine Saher
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Johannes Hirrlinger
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Carl-Ludwig-Institute for Physiology, Faculty of Medicine, Leipzig University, Leipzig, Germany
| | - Wiebke Möbius
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Sandra Goebbels
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| |
Collapse
|
8
|
Vaia Y, Bruschi F, Tagi VM, Tosi M, Montanari C, Zuccotti G, Tonduti D, Verduci E. Microbiota gut-brain axis: implications for pediatric-onset leukodystrophies. Front Nutr 2024; 11:1417981. [PMID: 39070252 PMCID: PMC11272617 DOI: 10.3389/fnut.2024.1417981] [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: 04/15/2024] [Accepted: 07/02/2024] [Indexed: 07/30/2024] Open
Abstract
Neurodegenerative disorders are a group of diseases characterized by progressive degeneration of the nervous system, leading to a gradual loss of previously acquired motor, sensory and/or cognitive functions. Leukodystrophies are amongst the most frequent childhood-onset neurodegenerative diseases and primarily affect the white matter of the brain, often resulting in neuro-motor disability. Notably, gastrointestinal (GI) symptoms and complications, such as gastroesophageal reflux disease (GERD) and dysphagia, significantly impact patients' quality of life, highlighting the need for comprehensive management strategies. Gut dysbiosis, characterized by microbial imbalance, has been implicated in various GI disorders and neurodegenerative diseases. This narrative review explores the intricate relationship between GI symptoms, Gut Microbiota (GM), and neurodegeneration. Emerging evidence underscores the profound influence of GM on neurological functions via the microbiota gut-brain axis. Animal models have demonstrated alterations in GM composition associated with neuroinflammation and neurodegeneration. Our single-centre experience reveals a high prevalence of GI symptoms in leukodystrophy population, emphasizing the importance of gastroenterological assessment and nutritional intervention in affected children. The bidirectional relationship between GI disorders and neurodegeneration suggests a potential role of gut dysbiosis in disease progression. Prospective studies investigating the GM in leukodystrophies are essential to understand the role of gut-brain axis dysfunction in disease progression and identify novel therapeutic targets. In conclusion, elucidating the interplay between GI disorders, GM, and neurodegeneration holds promise for precision treatments aimed at improving patient outcomes and quality of life.
Collapse
Affiliation(s)
- Ylenia Vaia
- C.O.A.L.A. (Center for Diagnosis and Treatment of Leukodystrophies), Unit of Pediatric Neurology, V. Buzzi Children’s Hospital, Milan, Italy
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Fabio Bruschi
- C.O.A.L.A. (Center for Diagnosis and Treatment of Leukodystrophies), Unit of Pediatric Neurology, V. Buzzi Children’s Hospital, Milan, Italy
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Veronica Maria Tagi
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
- Department of Pediatrics, V. Buzzi Children's Hospital, Milan, Italy
| | - Martina Tosi
- Department of Pediatrics, V. Buzzi Children's Hospital, Milan, Italy
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Chiara Montanari
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
- Department of Pediatrics, V. Buzzi Children's Hospital, Milan, Italy
| | - Gianvincenzo Zuccotti
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
- Department of Pediatrics, V. Buzzi Children's Hospital, Milan, Italy
| | - Davide Tonduti
- C.O.A.L.A. (Center for Diagnosis and Treatment of Leukodystrophies), Unit of Pediatric Neurology, V. Buzzi Children’s Hospital, Milan, Italy
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Elvira Verduci
- Department of Health Sciences, University of Milan, Milan, Italy
- Metabolic Diseases Unit, Department of Pediatrics, V. Buzzi Children’s Hospital, University of Milan, Milan, Italy
| |
Collapse
|
9
|
Elitt MS, Tesar PJ. Pelizaeus-Merzbacher disease: on the cusp of myelin medicine. Trends Mol Med 2024; 30:459-470. [PMID: 38582621 PMCID: PMC11081862 DOI: 10.1016/j.molmed.2024.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 03/05/2024] [Accepted: 03/08/2024] [Indexed: 04/08/2024]
Abstract
Pelizaeus-Merzbacher disease (PMD) is caused by mutations in the proteolipid protein 1 (PLP1) gene encoding proteolipid protein (PLP). As a major component of myelin, mutated PLP causes progressive neurodegeneration and eventually death due to severe white matter deficits. Medical care has long been limited to symptomatic treatments, but first-in-class PMD therapies with novel mechanisms now stand poised to enter clinical trials. Here, we review PMD disease mechanisms and outline rationale for therapeutic interventions, including PLP1 suppression, cell transplantation, iron chelation, and intracellular stress modulation. We discuss available preclinical data and their implications on clinical development. With several novel treatments on the horizon, PMD is on the precipice of a new era in the diagnosis and treatment of patients suffering from this debilitating disease.
Collapse
Affiliation(s)
- Matthew S Elitt
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Paul J Tesar
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| |
Collapse
|
10
|
Trimarco A, Audano M, Marca RL, Cariello M, Falco M, Pedretti S, Imperato G, Cestaro A, Podini P, Dina G, Quattrini A, Massimino L, Caruso D, Mitro N, Taveggia C. Prostaglandin D2 synthase controls Schwann cells metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.29.582775. [PMID: 38496560 PMCID: PMC10942270 DOI: 10.1101/2024.02.29.582775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
We previously reported that in the absence of Prostaglandin D2 synthase (L-PGDS) peripheral nerves are hypomyelinated in development and that with aging they present aberrant myelin sheaths. We now demonstrate that L-PGDS expressed in Schwann cells is part of a coordinated program aiming at preserving myelin integrity. In vivo and in vitro lipidomic, metabolomic and transcriptomic analyses confirmed that myelin lipids composition, Schwann cells energetic metabolism and key enzymes controlling these processes are altered in the absence of L-PGDS. Moreover, Schwann cells undergo a metabolic rewiring and turn to acetate as the main energetic source. Further, they produce ketone bodies to ensure glial cell and neuronal survival. Importantly, we demonstrate that all these changes correlate with morphological myelin alterations and describe the first physiological pathway implicated in preserving PNS myelin. Collectively, we posit that myelin lipids serve as a reservoir to provide ketone bodies, which together with acetate represent the adaptive substrates Schwann cells can rely on to sustain the axo-glial unit and preserve the integrity of the PNS.
Collapse
|
11
|
Looser ZJ, Faik Z, Ravotto L, Zanker HS, Jung RB, Werner HB, Ruhwedel T, Möbius W, Bergles DE, Barros LF, Nave KA, Weber B, Saab AS. Oligodendrocyte-axon metabolic coupling is mediated by extracellular K + and maintains axonal health. Nat Neurosci 2024; 27:433-448. [PMID: 38267524 PMCID: PMC10917689 DOI: 10.1038/s41593-023-01558-3] [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: 11/08/2022] [Accepted: 12/13/2023] [Indexed: 01/26/2024]
Abstract
The integrity of myelinated axons relies on homeostatic support from oligodendrocytes (OLs). To determine how OLs detect axonal spiking and how rapid axon-OL metabolic coupling is regulated in the white matter, we studied activity-dependent calcium (Ca2+) and metabolite fluxes in the mouse optic nerve. We show that fast axonal spiking triggers Ca2+ signaling and glycolysis in OLs. OLs detect axonal activity through increases in extracellular potassium (K+) concentrations and activation of Kir4.1 channels, thereby regulating metabolite supply to axons. Both pharmacological inhibition and OL-specific inactivation of Kir4.1 reduce the activity-induced axonal lactate surge. Mice lacking oligodendroglial Kir4.1 exhibit lower resting lactate levels and altered glucose metabolism in axons. These early deficits in axonal energy metabolism are associated with late-onset axonopathy. Our findings reveal that OLs detect fast axonal spiking through K+ signaling, making acute metabolic coupling possible and adjusting the axon-OL metabolic unit to promote axonal health.
Collapse
Affiliation(s)
- Zoe J Looser
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Zainab Faik
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Luca Ravotto
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Henri S Zanker
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Ramona B Jung
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Hauke B Werner
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Torben Ruhwedel
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Wiebke Möbius
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Dwight E Bergles
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA
| | - L Felipe Barros
- Centro de Estudios Científicos (CECs), Valdivia, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Valdivia, Chile
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Bruno Weber
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Aiman S Saab
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
12
|
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
|
13
|
Kawade N, Yamanaka K. Novel insights into brain lipid metabolism in Alzheimer's disease: Oligodendrocytes and white matter abnormalities. FEBS Open Bio 2024; 14:194-216. [PMID: 37330425 PMCID: PMC10839347 DOI: 10.1002/2211-5463.13661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/07/2023] [Accepted: 06/14/2023] [Indexed: 06/19/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. A genome-wide association study has shown that several AD risk genes are involved in lipid metabolism. Additionally, epidemiological studies have indicated that the levels of several lipid species are altered in the AD brain. Therefore, lipid metabolism is likely changed in the AD brain, and these alterations might be associated with an exacerbation of AD pathology. Oligodendrocytes are glial cells that produce the myelin sheath, which is a lipid-rich insulator. Dysfunctions of the myelin sheath have been linked to white matter abnormalities observed in the AD brain. Here, we review the lipid composition and metabolism in the brain and myelin and the association between lipidic alterations and AD pathology. We also present the abnormalities in oligodendrocyte lineage cells and white matter observed in AD. Additionally, we discuss metabolic disorders, including obesity, as AD risk factors and the effects of obesity and dietary intake of lipids on the brain.
Collapse
Affiliation(s)
- Noe Kawade
- Department of Neuroscience and Pathobiology, Research Institute of Environmental MedicineNagoya UniversityJapan
- Department of Neuroscience and Pathobiology, Nagoya University Graduate School of MedicineNagoya UniversityJapan
| | - Koji Yamanaka
- Department of Neuroscience and Pathobiology, Research Institute of Environmental MedicineNagoya UniversityJapan
- Department of Neuroscience and Pathobiology, Nagoya University Graduate School of MedicineNagoya UniversityJapan
- Institute for Glyco‐core Research (iGCORE)Nagoya UniversityJapan
- Center for One Medicine Innovative Translational Research (COMIT)Nagoya UniversityJapan
| |
Collapse
|
14
|
Perrier S, Gauquelin L, Bernard G. Inherited white matter disorders: Hypomyelination (myelin disorders). HANDBOOK OF CLINICAL NEUROLOGY 2024; 204:197-223. [PMID: 39322379 DOI: 10.1016/b978-0-323-99209-1.00014-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
Hypomyelinating leukodystrophies are a subset of genetic white matter diseases characterized by insufficient myelin deposition during development. MRI patterns are used to identify hypomyelinating disorders, and genetic testing is used to determine the causal genes implicated in individual disease forms. Clinical course can range from severe, with patients manifesting neurologic symptoms in infancy or early childhood, to mild, with onset in adolescence or adulthood. This chapter discusses the most common hypomyelinating leukodystrophies, including X-linked Pelizaeus-Merzbacher disease and other PLP1-related disorders, autosomal recessive Pelizaeus-Merzbacher-like disease, and POLR3-related leukodystrophy. PLP1-related disorders are caused by hemizygous pathogenic variants in the proteolipid protein 1 (PLP1) gene, and encompass classic Pelizaeus-Merzbacher disease, the severe connatal form, PLP1-null syndrome, spastic paraplegia type 2, and hypomyelination of early myelinating structures. Pelizaeus-Merzbacher-like disease presents a similar clinical picture to Pelizaeus-Merzbacher disease, however, it is caused by biallelic pathogenic variants in the GJC2 gene, which encodes for the gap junction protein Connexin-47. POLR3-related leukodystrophy, or 4H leukodystrophy (hypomyelination, hypodontia, and hypogonadotropic hypogonadism), is caused by biallelic pathogenic variants in genes encoding specific subunits of the transcription enzyme RNA polymerase III. In this chapter, the clinical features, disease pathophysiology and genetics, imaging patterns, as well as supportive and future therapies are discussed for each disorder.
Collapse
Affiliation(s)
- Stefanie Perrier
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada; Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Laurence Gauquelin
- Division of Pediatric Neurology, Department of Pediatrics, CHUL et Centre Mère-Enfant Soleil du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Geneviève Bernard
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada; Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada; Departments of Pediatrics and Human Genetics, McGill University, Montréal, QC, Canada.
| |
Collapse
|
15
|
Corsello A, Trovato CM, Di Profio E, Cardile S, Campoy C, Zuccotti G, Verduci E, Diamanti A. Ketogenic Diet in Children and Adolescents: the Effects on Growth and Nutritional Status. Pharmacol Res 2023; 191:106780. [PMID: 37088260 DOI: 10.1016/j.phrs.2023.106780] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/12/2023] [Accepted: 04/21/2023] [Indexed: 04/25/2023]
Abstract
The ketogenic diet is known to be a possible adjuvant treatment in several medical conditions, such as in patients with severe or drug-resistant forms of epilepsy. Its use has recently been increasing among adolescents and young adults due to its supposed weight-loss effect, mediated by lipolysis and lowered insulin levels. However, there are still no precise indications on the possible use of ketogenic diets in pediatric age for weight loss. This approach has also recently been proposed for other types of disorder such as inherited metabolic disorders, Prader-Willi syndrome, and some specific types of cancers. Due to its unbalanced ratio of lipids, carbohydrates and proteins, a clinical evaluation of possible side effects with a strict evaluation of growth and nutritional status is essential in all patients following a long-term restrictive diet such as the ketogenic one. The prophylactic use of micronutrients supplementation should be considered before starting any ketogenic diet. Lastly, while there is sufficient literature on possible short-term side effects of ketogenic diets, their possible long-term impact on growth and nutritional status is not yet fully understood, especially when started in pediatric age.
Collapse
Affiliation(s)
- Antonio Corsello
- Department of Paediatrics, Vittore Buzzi Children's Hospital, University of Milan, Milan, Italy.
| | - Chiara Maria Trovato
- Hepatology Gastroenterology and Nutrition Unit, Bambino Gesù Children Hospital, Rome, Italy.
| | - Elisabetta Di Profio
- Department of Paediatrics, Vittore Buzzi Children's Hospital, University of Milan, Milan, Italy; Department of Health Sciences, University of Milan, Milan, Italy.
| | - Sabrina Cardile
- Hepatology Gastroenterology and Nutrition Unit, Bambino Gesù Children Hospital, Rome, Italy.
| | - Cristina Campoy
- Department of Paediatrics, School of Medicine, University of Granada, Granada, Spain; EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, Granada, Spain; Spanish Network of Biomedical Research in Epidemiology and Public Health (CIBERESP), Granada's node, Institute of Health Carlos III, Madrid, Spain.
| | - Gianvincenzo Zuccotti
- Department of Paediatrics, Vittore Buzzi Children's Hospital, University of Milan, Milan, Italy; Department of Biomedical and Clinical Sciences L. Sacco, University of Milan, Milan, Italy; Pediatric Clinical Research Center, Fondazione Romeo ed Enrica Invernizzi, University of Milan, Milan, Italy.
| | - Elvira Verduci
- Department of Paediatrics, Vittore Buzzi Children's Hospital, University of Milan, Milan, Italy; Department of Health Sciences, University of Milan, Milan, Italy.
| | - Antonella Diamanti
- Hepatology Gastroenterology and Nutrition Unit, Bambino Gesù Children Hospital, Rome, Italy.
| |
Collapse
|
16
|
Singh S, Mishra A, Murthy C, Inban P, Abdefatah Ali M, Yadav AS, Intsiful TA, O Omar ZT, Lakhra S, Khan DA. A Rare Case of Hypomyelinating Leukodystrophy and Its Management: A Case Report and Literature Review. Cureus 2023; 15:e36471. [PMID: 37090362 PMCID: PMC10117409 DOI: 10.7759/cureus.36471] [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] [Accepted: 03/21/2023] [Indexed: 04/25/2023] Open
Abstract
A subset of hereditary white matter disorders called hypomyelinating leukodystrophies (HLD) is characterized primarily by the absence of myelin deposition. Although the clinical presentation can be mild and the development of symptoms can occur in adolescence or adulthood, the majority of severe cases present during infancy and early childhood with significant neurological impairments. The clinical features vary from muscle stiffness to seizures and developmental delay. The detailed myelination process can be seen with magnetic resonance imaging (MRI), and many patients are diagnosed using MRI pattern recognition and next-generation sequencing (NGS) in most cases. Here, we report a case of an infant suffering from the hypomyelinating leukodystrophy-13 (HLD-13) variant, whose next-generation sequencing revealed a pathogenic homozygous variant.
Collapse
Affiliation(s)
- Sonali Singh
- Pediatrics, King George's Medical University, Lucknow, IND
| | - Anshika Mishra
- Pediatrics, King George's Medical University, Lucknow, IND
| | - Chinmayee Murthy
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Pugazhendi Inban
- Department of General Medicine, Government Medical College, Omandurar, Chennai, IND
| | | | - Anupam S Yadav
- Psychiatry and Behavioral Sciences, Ganesh Shankar Vidyarthi Memorial (GSVM) Medical College, Kanpur, IND
| | | | | | - Sakshi Lakhra
- Internal Medicine, All Saints School of Medicine, St. Roseau, DMA
| | - Dr Aadil Khan
- Department of Internal Medicine, Lala Lajpat Rai (LLR) Hospital, Kanpur, IND
| |
Collapse
|
17
|
Ketogenic Diet Modulates Neuroinflammation via Metabolites from Lactobacillus reuteri After Repetitive Mild Traumatic Brain Injury in Adolescent Mice. Cell Mol Neurobiol 2023; 43:907-923. [PMID: 35499776 DOI: 10.1007/s10571-022-01226-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 04/14/2022] [Indexed: 11/03/2022]
Abstract
Repetitive mild traumatic brain injury (rmTBI) is associated with a range of neural changes which is characterized by axonal injury and neuroinflammation. Ketogenic diet (KD) is regarded as a potential therapy for facilitating recovery after moderate-severe traumatic brain injury (TBI). However, its effect on rmTBI has not been fully studied. In this study, we evaluated the anti-neuroinflammation effects of KD after rmTBI in adolescent mice and explored the potential mechanisms. Experimentally, specific pathogen-free (SPF) adolescent male C57BL/6 mice received a sham surgery or repetitive mild controlled cortical impacts consecutively for 7 days. The uninjured mice received the standard diet, and the mice with rmTBI were fed either the standard diet or KD for 7 days. One week later, all mice were subjected to behavioral tests and experimental analysis. Results suggest that KD significantly increased blood beta-hydroxybutyrate (β-HB) levels and improved neurological function. KD also reduced white matter damage, microgliosis, and astrogliosis induced by rmTBI. Aryl hydrocarbon receptor (AHR) signaling pathway, which was mediated by indole-3-acetic acid (3-IAA) from Lactobacillus reuteri (L. reuteri) in gut and activated in microglia and astrocytes after rmTBI, was inhibited by KD. The expression level of the toll-like receptor 4 (TLR4)/myeloid differentiation primary response 88 (MyD88) in inflammatory cells, which mediates the NF-κB pathway, was also attenuated by KD. Taken together, our results indicated that KD can promote recovery following rmTBI in adolescent mice. KD may modulate neuroinflammation by altering L. reuteri in gut and its metabolites. The inhibition of indole/AHR pathway and the downregulation of TLR4/MyD88 may play a role in the beneficial effect of KD against neuroinflammation in rmTBI mice.
Collapse
|
18
|
Barnes-Vélez JA, Aksoy Yasar FB, Hu J. Myelin lipid metabolism and its role in myelination and myelin maintenance. Innovation (N Y) 2023; 4:100360. [PMID: 36588745 PMCID: PMC9800635 DOI: 10.1016/j.xinn.2022.100360] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 12/03/2022] [Indexed: 12/12/2022] Open
Abstract
Myelin is a specialized cell membrane indispensable for rapid nerve conduction. The high abundance of membrane lipids is one of myelin's salient features that contribute to its unique role as an insulator that electrically isolates nerve fibers across their myelinated surface. The most abundant lipids in myelin include cholesterol, glycosphingolipids, and plasmalogens, each playing critical roles in myelin development as well as function. This review serves to summarize the role of lipid metabolism in myelination and myelin maintenance, as well as the molecular determinants of myelin lipid homeostasis, with an emphasis on findings from genetic models. In addition, the implications of myelin lipid dysmetabolism in human diseases are highlighted in the context of hereditary leukodystrophies and neuropathies as well as acquired disorders such as Alzheimer's disease.
Collapse
Affiliation(s)
- Joseph A. Barnes-Vélez
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77054-1901, USA
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Science, Houston, TX 77225-0334, USA
- University of Puerto Rico Medical Sciences Campus, School of Medicine, San Juan, PR 00936-5067, USA
| | - Fatma Betul Aksoy Yasar
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77054-1901, USA
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Science, Houston, TX 77225-0334, USA
| | - Jian Hu
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77054-1901, USA
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Science, Houston, TX 77225-0334, USA
| |
Collapse
|
19
|
Sun W, Wen M, Liu M, Wang Q, Liu Q, Li L, Siebert HC, Loers G, Zhang R, Zhang N. Effect of β-hydroxybutyrate on behavioral alterations, molecular and morphological changes in CNS of multiple sclerosis mouse model. Front Aging Neurosci 2022; 14:1075161. [PMID: 36533180 PMCID: PMC9752847 DOI: 10.3389/fnagi.2022.1075161] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 11/14/2022] [Indexed: 09/29/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory and degenerative disease of central nervous system (CNS). Aging is the most significant risk factor for the progression of MS. Dietary modulation (such as ketogenic diet) and caloric restriction, can increase ketone bodies, especially β-hydroxybutyrate (BHB). Increased BHB has been reported to prevent or improve age-related disease. The present studies were performed to understand the therapeutic effect and potential mechanisms of exogenous BHB in cuprizone (CPZ)-induced demyelinating model. In this study, a continuous 35 days CPZ mouse model with or without BHB was established. The changes of behavior function, pathological hallmarks of CPZ, and intracellular signal pathways in mice were detected by Open feld test, Morris water maze, RT-PCR, immuno-histochemistry, and western blot. The results showed that BHB treatment improved behavioral performance, prevented myelin loss, decreased the activation of astrocyte as well as microglia, and up-regulated the neurotrophin brain-derived neurotrophic factor in both the corpus callosum and hippocampus. Meanwhile, BHB treatment increased the number of MCT1+ cells and APC+ oligodendrocytes. Furthermore, the treatment decreased the expression of HDAC3, PARP1, AIF and TRPA1 which is related to oligodendrocyte (OL) apoptosis in the corpus callosum, accompanied by increased expression of TrkB. This leads to an increased density of doublecortin (DCX)+ neuronal precursor cells and mature NeuN+ neuronal cells in the hippocampus. As a result, BHB treatment effectively promotes the generation of PDGF-Ra+ (oligodendrocyte precursor cells, OPCs), Sox2+ cells and GFAP+ (astrocytes), and decreased the production of GFAP+ TRAP1+ cells, and Oligo2+ TRAP1+ cells in the corpus callosum of mouse brain. Thus, our results demonstrate that BHB treatment efficiently supports OPC differentiation and decreases the OLs apoptosis in CPZ-intoxicated mice, partly by down-regulating the expression of TRPA1 and PARP, which is associated with the inhibition of the p38-MAPK/JNK/JUN pathway and the activation of ERK1/2, PI3K/AKT/mTOR signaling, supporting BHB treatment adjunctive nutritional therapy for the treatment of chronic demyelinating diseases, such as multiple sclerosis (MS).
Collapse
Affiliation(s)
- Wei Sun
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong, China
| | - Min Wen
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong, China
| | - Min Liu
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong, China
| | - Qingpeng Wang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong, China
| | - Quiqin Liu
- Shandong Donkey Industry, Technology Collaborative Innovation Center, Liaocheng University, Liaocheng, China
| | - Lanjie Li
- Shandong Donkey Industry, Technology Collaborative Innovation Center, Liaocheng University, Liaocheng, China
| | - Hans-Christian Siebert
- Schauenburgerstr, RI-B-NT - Research Institute of Bioinformatics and Nanotechnology, Kiel University, Kiel, Germany
| | - Gabriele Loers
- Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, University of Hamburg, Hamburg, Germany
| | - Ruiyan Zhang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong, China
| | - Ning Zhang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong, China
- Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
| |
Collapse
|
20
|
Field R, Field T, Pourkazemi F, Rooney K. Ketogenic diets and the nervous system: a scoping review of neurological outcomes from nutritional ketosis in animal studies. Nutr Res Rev 2022; 35:268-281. [PMID: 34180385 DOI: 10.1017/s0954422421000214] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Ketogenic diets have reported efficacy for neurological dysfunctions; however, there are limited published human clinical trials elucidating the mechanisms by which nutritional ketosis produces therapeutic effects. The purpose of this present study was to investigate animal models that report variations in nervous system function by changing from a standard animal diet to a ketogenic diet, synthesise these into broad themes, and compare these with mechanisms reported as targets in pain neuroscience to inform human chronic pain trials. METHODS An electronic search of seven databases was conducted in July 2020. Two independent reviewers screened studies for eligibility, and descriptive outcomes relating to nervous system function were extracted for a thematic analysis, then synthesised into broad themes. RESULTS In total, 170 studies from eighteen different disease models were identified and grouped into fourteen broad themes: alterations in cellular energetics and metabolism, biochemical, cortical excitability, epigenetic regulation, mitochondrial function, neuroinflammation, neuroplasticity, neuroprotection, neurotransmitter function, nociception, redox balance, signalling pathways, synaptic transmission and vascular supply. DISCUSSION The mechanisms presented centred around the reduction of inflammation and oxidative stress as well as a reduction in nervous system excitability. Given the multiple potential mechanisms presented, it is likely that many of these are involved synergistically and undergo adaptive processes within the human body, and controlled animal models that limit the investigation to a particular pathway in isolation may reach differing conclusions. Attention is required when translating this information to human chronic pain populations owing to the limitations outlined from the animal research.
Collapse
Affiliation(s)
- Rowena Field
- The University of Sydney, Faculty of Medicine and Health, Sydney, Australia
| | - Tara Field
- The New South Wales Ministry of Health (NSW Health), Sydney, Australia
| | | | - Kieron Rooney
- The University of Sydney, Faculty of Medicine and Health, Sydney, Australia
| |
Collapse
|
21
|
Molina-Gonzalez I, Miron VE, Antel JP. Chronic oligodendrocyte injury in central nervous system pathologies. Commun Biol 2022; 5:1274. [PMID: 36402839 PMCID: PMC9675815 DOI: 10.1038/s42003-022-04248-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 11/10/2022] [Indexed: 11/21/2022] Open
Abstract
Myelin, the membrane surrounding neuronal axons, is critical for central nervous system (CNS) function. Injury to myelin-forming oligodendrocytes (OL) in chronic neurological diseases (e.g. multiple sclerosis) ranges from sublethal to lethal, leading to OL dysfunction and myelin pathology, and consequent deleterious impacts on axonal health that drive clinical impairments. This is regulated by intrinsic factors such as heterogeneity and age, and extrinsic cellular and molecular interactions. Here, we discuss the responses of OLs to injury, and perspectives for therapeutic targeting. We put forward that targeting mature OL health in neurological disease is a promising therapeutic strategy to support CNS function.
Collapse
Affiliation(s)
- Irene Molina-Gonzalez
- grid.4305.20000 0004 1936 7988United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, Scotland UK ,grid.4305.20000 0004 1936 7988Centre for Discovery Brain Sciences, Chancellor’s Building, The University of Edinburgh, Edinburgh, Scotland UK ,grid.4305.20000 0004 1936 7988Medical Research Council Centre for Reproductive Health, The Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, Scotland UK
| | - Veronique E. Miron
- grid.4305.20000 0004 1936 7988United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, Scotland UK ,grid.4305.20000 0004 1936 7988Centre for Discovery Brain Sciences, Chancellor’s Building, The University of Edinburgh, Edinburgh, Scotland UK ,grid.4305.20000 0004 1936 7988Medical Research Council Centre for Reproductive Health, The Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, Scotland UK ,grid.415502.7Barlo Multiple Sclerosis Centre and Keenan Research Centre for Biomedical Science, Toronto, Canada ,grid.17063.330000 0001 2157 2938Department of Immunology, University of Toronto, Toronto, Canada
| | - Jack P. Antel
- grid.14709.3b0000 0004 1936 8649Neuroimmunology Unit, Montreal Neurological Institute, McGill University, Montreal, QC Canada
| |
Collapse
|
22
|
Düking T, Spieth L, Berghoff SA, Piepkorn L, Schmidke AM, Mitkovski M, Kannaiyan N, Hosang L, Scholz P, Shaib AH, Schneider LV, Hesse D, Ruhwedel T, Sun T, Linhoff L, Trevisiol A, Köhler S, Pastor AM, Misgeld T, Sereda M, Hassouna I, Rossner MJ, Odoardi F, Ischebeck T, de Hoz L, Hirrlinger J, Jahn O, Saher G. Ketogenic diet uncovers differential metabolic plasticity of brain cells. SCIENCE ADVANCES 2022; 8:eabo7639. [PMID: 36112685 PMCID: PMC9481126 DOI: 10.1126/sciadv.abo7639] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 08/01/2022] [Indexed: 06/15/2023]
Abstract
To maintain homeostasis, the body, including the brain, reprograms its metabolism in response to altered nutrition or disease. However, the consequences of these challenges for the energy metabolism of the different brain cell types remain unknown. Here, we generated a proteome atlas of the major central nervous system (CNS) cell types from young and adult mice, after feeding the therapeutically relevant low-carbohydrate, high-fat ketogenic diet (KD) and during neuroinflammation. Under steady-state conditions, CNS cell types prefer distinct modes of energy metabolism. Unexpectedly, the comparison with KD revealed distinct cell type-specific strategies to manage the altered availability of energy metabolites. Astrocytes and neurons but not oligodendrocytes demonstrated metabolic plasticity. Moreover, inflammatory demyelinating disease changed the neuronal metabolic signature in a similar direction as KD. Together, these findings highlight the importance of the metabolic cross-talk between CNS cells and between the periphery and the brain to manage altered nutrition and neurological disease.
Collapse
Affiliation(s)
- Tim Düking
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Lena Spieth
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Stefan A. Berghoff
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Lars Piepkorn
- Neuroproteomics Group, Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Translational Neuroproteomics Group, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | - Annika M. Schmidke
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Miso Mitkovski
- City Campus Light Microscopy Facility, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Nirmal Kannaiyan
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
| | - Leon Hosang
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Göttingen, Germany
| | - Patricia Scholz
- Department of Plant Biochemistry, Albrecht-von-Haller-Institute for Plant Sciences and Göttingen Center for Molecular Biosciences (GZMB), University of Göttingen, Göttingen, Germany
| | - Ali H. Shaib
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Institute for Neuro- and Sensory Physiology, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
| | - Lennart V. Schneider
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Dörte Hesse
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Neuroproteomics Group, Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Torben Ruhwedel
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Electron Microscopy Core Unit, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Ting Sun
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Lisa Linhoff
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
- Translational Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Andrea Trevisiol
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Susanne Köhler
- Carl-Ludwig-Institute for Physiology, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Adrian Marti Pastor
- Institute of Neuronal Cell Biology, Technische Universität München, Cluster for Systems Neurology (SyNergy), German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Thomas Misgeld
- Institute of Neuronal Cell Biology, Technische Universität München, Cluster for Systems Neurology (SyNergy), German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Michael Sereda
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
- Translational Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Imam Hassouna
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Moritz J. Rossner
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
| | - Francesca Odoardi
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Göttingen, Germany
| | - Till Ischebeck
- Department of Plant Biochemistry, Albrecht-von-Haller-Institute for Plant Sciences and Göttingen Center for Molecular Biosciences (GZMB), University of Göttingen, Göttingen, Germany
- Institute of Plant Biology and Biotechnology (IBBP), Green Biotechnology, University of Münster, Münster, Germany
| | - Livia de Hoz
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Neurowissenschafliches Forschungszentrum, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Johannes Hirrlinger
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Carl-Ludwig-Institute for Physiology, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Olaf Jahn
- Neuroproteomics Group, Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Translational Neuroproteomics Group, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | - Gesine Saher
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| |
Collapse
|
23
|
Lin WS, Lin SJ, Liao PY, Suresh D, Hsu TR, Wang PY. Role of Ketogenic Diets in Multiple Sclerosis and Related Animal Models: An Updated Review. Adv Nutr 2022; 13:2002-2014. [PMID: 35679067 PMCID: PMC9526852 DOI: 10.1093/advances/nmac065] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 04/25/2022] [Accepted: 06/02/2022] [Indexed: 01/28/2023] Open
Abstract
Prescribing a ketogenic diet (KD) is a century-old dietary intervention mainly used in the context of intractable epilepsy. The classic KD and its variants regained popularity in recent decades, and they are considered potentially beneficial in a variety of neurological conditions other than epilepsy. Many patients with multiple sclerosis (MS) have attempted diet modification for better control of their disease, although evidence thus far remains insufficient to recommend a specific diet for these patients. The results of 3 pilot clinical trials of KD therapy for MS, as well as several related studies, have been reported in recent years. The preliminary findings suggest that KD is safe, feasible, and potentially neuroprotective and disease-modifying for patients with MS. Research on corresponding rodent models has also lent support to the efficacy of KD in the prevention and treatment of experimental autoimmune encephalomyelitis and toxin-induced inflammatory demyelinating conditions in the brain. Furthermore, the animal studies have yielded mechanistic insights into the molecular mechanisms of KD action in relevant situations, paving the way for precision nutrition. Herein we review and synthesize recent advances and also identify unresolved issues, such as the roles of adipokines and gut microbiota, in this field. Hopefully this panoramic view of current understanding can inform future research directions and clinical practice with regard to KD in MS and related conditions.
Collapse
Affiliation(s)
| | - Shan-Ju Lin
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital Yunlin Branch, Yunlin, Taiwan
| | - Pei-Yin Liao
- Department of Dietetics, National Taiwan University Hospital Yunlin Branch, Yunlin, Taiwan
| | - Divya Suresh
- Department of Pediatrics, National Taiwan University Hospital Yunlin Branch, Yunlin, Taiwan
| | - Ting-Rong Hsu
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan,School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Pei-Yu Wang
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan,Neurobiology and Cognitive Science Center, National Taiwan University, Taipei, Taiwan,Program in Translational Medicine, National Taiwan University and Academia Sinica, Taipei, Taiwan,Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Taiwan University and Academia Sinica, Taipei, Taiwan,Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
24
|
Narine M, Colognato H. Current Insights Into Oligodendrocyte Metabolism and Its Power to Sculpt the Myelin Landscape. Front Cell Neurosci 2022; 16:892968. [PMID: 35573837 PMCID: PMC9097137 DOI: 10.3389/fncel.2022.892968] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 04/06/2022] [Indexed: 12/20/2022] Open
Abstract
Once believed to be part of the nervenkitt or "nerve glue" network in the central nervous system (CNS), oligodendroglial cells now have established roles in key neurological functions such as myelination, neuroprotection, and motor learning. More recently, oligodendroglia has become the subject of intense investigations aimed at understanding the contributions of its energetics to CNS physiology and pathology. In this review, we discuss the current understanding of oligodendroglial metabolism in regulating key stages of oligodendroglial development and health, its role in providing energy to neighboring cells such as neurons, as well as how alterations in oligodendroglial bioenergetics contribute to disease states. Importantly, we highlight how certain inputs can regulate oligodendroglial metabolism, including extrinsic and intrinsic mediators of cellular signaling, pharmacological compounds, and even dietary interventions. Lastly, we discuss emerging studies aimed at discovering the therapeutic potential of targeting components within oligodendroglial bioenergetic pathways.
Collapse
Affiliation(s)
- Mohanlall Narine
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, United States
- Department of Neurobiology, & Behavior, Stony Brook University, Stony Brook, NY, United States
| | - Holly Colognato
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, United States
| |
Collapse
|
25
|
Duan R, Ji H, Yan H, Wang J, Zhang Y, Zhang Q, Li D, Cao B, Gu Q, Wu Y, Jiang Y, Li M, Wang J. Genotype-phenotype correlation and natural history analyses in a Chinese cohort with pelizaeus-merzbacher disease. Orphanet J Rare Dis 2022; 17:137. [PMID: 35346287 PMCID: PMC8962489 DOI: 10.1186/s13023-022-02267-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 02/20/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The natural history and genotype-phenotype correlation of Pelizaeus-Merzbacher disease (PMD) of Chinese patients has been rarely reported. METHOD Patients who met the criteria for PMD were enrolled in our study. Genomic analysis was conducted by multiplex ligation probe amplification (MLPA) and Sanger or whole-exome sequencing (WES). Natural history differences and genotype-phenotype correlations were analyzed. RESULT A total of 111 patients were enrolled in our follow-up study. The median follow-up interval was 53 m (1185). Among PMD patients, developmental delay was the most common sign, and nystagmus and hypotonia were the most common initial symptoms observed. A total of 78.4% of the patients were able to control their head, and 72.1% could speak words. However, few of the patients could stand (9.0%) or walk (4.5%) by themselves. Nystagmus improved in more than half of the patients, and hypotonia sometimes deteriorated to movement disorders. More PLP1 point mutations patients were categorized into severe group, while more patients with PLP1 duplications were categorized into mild group (p < 0.001). Compared to patients in mild groups, those in the severe group had earlier disease onset and had acquired fewer skills at a later age. CONCLUSION PMD patients have early disease onset with nystagmus and hypotonia followed by decreased nystagmus and movement disorders, such as spasticit. Patients with PLP1 duplication were more likely to be categorized into the mild group, whereas patients with point mutations were more likely to be categorized into the severe group.
Collapse
Affiliation(s)
- Ruoyu Duan
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Haoran Ji
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Huifang Yan
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Junyu Wang
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Yu Zhang
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Qian Zhang
- Department of Children's Development and Rehabilitation, Peking University First Hospital, Beijing, 100034, China
| | - Dongxiao Li
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Binbin Cao
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Qiang Gu
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Ye Wu
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Yuwu Jiang
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Ming Li
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China.
- Department of Children's Development and Rehabilitation, Peking University First Hospital, Beijing, 100034, China.
| | - Jingmin Wang
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China.
| |
Collapse
|
26
|
Meschkat M, Steyer AM, Weil MT, Kusch K, Jahn O, Piepkorn L, Agüi-Gonzalez P, Phan NTN, Ruhwedel T, Sadowski B, Rizzoli SO, Werner HB, Ehrenreich H, Nave KA, Möbius W. White matter integrity in mice requires continuous myelin synthesis at the inner tongue. Nat Commun 2022; 13:1163. [PMID: 35246535 PMCID: PMC8897471 DOI: 10.1038/s41467-022-28720-y] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 01/24/2022] [Indexed: 12/18/2022] Open
Abstract
Myelin, the electrically insulating sheath on axons, undergoes dynamic changes over time. However, it is composed of proteins with long lifetimes. This raises the question how such a stable structure is renewed. Here, we study the integrity of myelinated tracts after experimentally preventing the formation of new myelin in the CNS of adult mice, using an inducible Mbp null allele. Oligodendrocytes survive recombination, continue to express myelin genes, but they fail to maintain compacted myelin sheaths. Using 3D electron microscopy and mass spectrometry imaging we visualize myelin-like membranes failing to incorporate adaxonally, most prominently at juxta-paranodes. Myelinoid body formation indicates degradation of existing myelin at the abaxonal side and the inner tongue of the sheath. Thinning of compact myelin and shortening of internodes result in the loss of about 50% of myelin and axonal pathology within 20 weeks post recombination. In summary, our data suggest that functional axon-myelin units require the continuous incorporation of new myelin membranes. Myelin is formed of proteins of long half-lives. The mechanisms of renewal of such a stable structure are unclear. Here, the authors show that myelin integrity requires continuous myelin synthesis at the inner tongue, contributing to the maintenance of a functional axon-myelin unit.
Collapse
Affiliation(s)
- Martin Meschkat
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Electron Microscopy Core Unit, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Göttingen Graduate Center for Neurosciences, Biophysics, and Molecular Biosciences (GGNB), Göttingen, Germany.,DFG Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany.,Abberior Instruments GmbH, Göttingen, Germany
| | - Anna M Steyer
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Electron Microscopy Core Unit, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,DFG Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany.,Imaging Centre, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Marie-Theres Weil
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Electron Microscopy Core Unit, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,DFG Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Kathrin Kusch
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
| | - Olaf Jahn
- DFG Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany.,Proteomics Group, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Translational Neuroproteomics Group, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | - Lars Piepkorn
- Proteomics Group, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Translational Neuroproteomics Group, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | - Paola Agüi-Gonzalez
- Department of Neuro- and Sensory Physiology, University Medical Center Göttingen, Center for Biostructural Imaging of Neurodegeneration, Göttingen, Germany
| | - Nhu Thi Ngoc Phan
- Department of Neuro- and Sensory Physiology, University Medical Center Göttingen, Center for Biostructural Imaging of Neurodegeneration, Göttingen, Germany.,Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | - Torben Ruhwedel
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Electron Microscopy Core Unit, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Boguslawa Sadowski
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Electron Microscopy Core Unit, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,DFG Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Silvio O Rizzoli
- DFG Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany.,Department of Neuro- and Sensory Physiology, University Medical Center Göttingen, Center for Biostructural Imaging of Neurodegeneration, Göttingen, Germany.,Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Hauke B Werner
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Hannelore Ehrenreich
- DFG Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany.,Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,DFG Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Wiebke Möbius
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany. .,Electron Microscopy Core Unit, Max Planck Institute of Experimental Medicine, Göttingen, Germany. .,DFG Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany. .,Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany.
| |
Collapse
|
27
|
Berghoff SA, Spieth L, Saher G. Local cholesterol metabolism orchestrates remyelination. Trends Neurosci 2022; 45:272-283. [DOI: 10.1016/j.tins.2022.01.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 01/07/2022] [Accepted: 01/21/2022] [Indexed: 12/19/2022]
|
28
|
Poeta E, Petralla S, Babini G, Renzi B, Celauro L, Magnifico MC, Barile SN, Masotti M, De Chirico F, Massenzio F, Viggiano L, Palmieri L, Virgili M, Lasorsa FM, Monti B. Histone Acetylation Defects in Brain Precursor Cells: A Potential Pathogenic Mechanism Causing Proliferation and Differentiation Dysfunctions in Mitochondrial Aspartate-Glutamate Carrier Isoform 1 Deficiency. Front Cell Neurosci 2022; 15:773709. [PMID: 35095421 PMCID: PMC8790092 DOI: 10.3389/fncel.2021.773709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 11/29/2021] [Indexed: 11/13/2022] Open
Abstract
Mitochondrial aspartate-glutamate carrier isoform 1 (AGC1) deficiency is an ultra-rare genetic disease characterized by global hypomyelination and brain atrophy, caused by mutations in the SLC25A12 gene leading to a reduction in AGC1 activity. In both neuronal precursor cells and oligodendrocytes precursor cells (NPCs and OPCs), the AGC1 determines reduced proliferation with an accelerated differentiation of OPCs, both associated with gene expression dysregulation. Epigenetic regulation of gene expression through histone acetylation plays a crucial role in the proliferation/differentiation of both NPCs and OPCs and is modulated by mitochondrial metabolism. In AGC1 deficiency models, both OPCs and NPCs show an altered expression of transcription factors involved in the proliferation/differentiation of brain precursor cells (BPCs) as well as a reduction in histone acetylation with a parallel alteration in the expression and activity of histone acetyltransferases (HATs) and histone deacetylases (HDACs). In this study, histone acetylation dysfunctions have been dissected in in vitro models of AGC1 deficiency OPCs (Oli-Neu cells) and NPCs (neurospheres), in physiological conditions and following pharmacological treatments. The inhibition of HATs by curcumin arrests the proliferation of OPCs leading to their differentiation, while the inhibition of HDACs by suberanilohydroxamic acid (SAHA) has only a limited effect on proliferation, but it significantly stimulates the differentiation of OPCs. In NPCs, both treatments determine an alteration in the commitment toward glial cells. These data contribute to clarifying the molecular and epigenetic mechanisms regulating the proliferation/differentiation of OPCs and NPCs. This will help to identify potential targets for new therapeutic approaches that are able to increase the OPCs pool and to sustain their differentiation toward oligodendrocytes and to myelination/remyelination processes in AGC1 deficiency, as well as in other white matter neuropathologies.
Collapse
Affiliation(s)
- Eleonora Poeta
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Sabrina Petralla
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Giorgia Babini
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Brunaldo Renzi
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Luigi Celauro
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Maria Chiara Magnifico
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy
| | - Simona Nicole Barile
- CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, Bari, Italy
| | - Martina Masotti
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | | | - Francesca Massenzio
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Luigi Viggiano
- Department of Biology, University of Bari Aldo Moro, Bari, Italy
| | - Luigi Palmieri
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy,CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, Bari, Italy
| | - Marco Virgili
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Francesco Massimo Lasorsa
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy,CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, Bari, Italy,*Correspondence: Francesco Massimo Lasorsa,
| | - Barbara Monti
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy,Barbara Monti,
| |
Collapse
|
29
|
Sawaguchi S, Tago K, Oizumi H, Ohbuchi K, Yamamoto M, Mizoguchi K, Miyamoto Y, Yamauchi J. Hypomyelinating Leukodystrophy 7 (HLD7)-Associated Mutation of POLR3A Is Related to Defective Oligodendroglial Cell Differentiation, Which Is Ameliorated by Ibuprofen. Neurol Int 2021; 14:11-33. [PMID: 35076634 PMCID: PMC8788570 DOI: 10.3390/neurolint14010002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/14/2021] [Accepted: 12/21/2021] [Indexed: 01/13/2023] Open
Abstract
Hypomyelinating leukodystrophy 7 (HLD7) is an autosomal recessive oligodendroglial cell-related myelin disease, which is associated with some nucleotide mutations of the RNA polymerase 3 subunit a (polr3a) gene. POLR3A is composed of the catalytic core of RNA polymerase III synthesizing non-coding RNAs, such as rRNA and tRNA. Here, we show that an HLD7-associated nonsense mutation of Arg140-to-Ter (R140X) primarily localizes POLR3A proteins as protein aggregates into lysosomes in mouse oligodendroglial FBD-102b cells, whereas the wild type proteins are not localized in lysosomes. Expression of the R140X mutant proteins, but not the wild type proteins, in cells decreased signaling through the mechanistic target of rapamycin (mTOR), controlling signal transduction around lysosomes. While cells harboring the wild type constructs exhibited phenotypes with widespread membranes with myelin marker protein expression following the induction of differentiation, cells harboring the R140X mutant constructs did not exhibit them. Ibuprofen, a non-steroidal anti-inflammatory drug (NSAID), which is also known as an mTOR signaling activator, ameliorated defects in differentiation with myelin marker protein expression and the related signaling in cells harboring the R140X mutant constructs. Collectively, HLD7-associated POLR3A mutant proteins are localized in lysosomes where they decrease mTOR signaling, inhibiting cell morphological differentiation. Importantly, ibuprofen reverses undifferentiated phenotypes. These findings may reveal some of the pathological mechanisms underlying HLD7 and their amelioration at the molecular and cellular levels.
Collapse
Affiliation(s)
- Sui Sawaguchi
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan; (S.S.); (Y.M.)
| | - Kenji Tago
- Department of Biochemistry, Jichi Medical University, Shimotsuke 321-0498, Japan;
| | - Hiroaki Oizumi
- Tsumura Research Laboratories, Tsumura & Co., Inashiki 200-1192, Japan; (H.O.); (K.O.); (M.Y.); (K.M.)
| | - Katsuya Ohbuchi
- Tsumura Research Laboratories, Tsumura & Co., Inashiki 200-1192, Japan; (H.O.); (K.O.); (M.Y.); (K.M.)
| | - Masahiro Yamamoto
- Tsumura Research Laboratories, Tsumura & Co., Inashiki 200-1192, Japan; (H.O.); (K.O.); (M.Y.); (K.M.)
| | - Kazushige Mizoguchi
- Tsumura Research Laboratories, Tsumura & Co., Inashiki 200-1192, Japan; (H.O.); (K.O.); (M.Y.); (K.M.)
| | - Yuki Miyamoto
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan; (S.S.); (Y.M.)
- Department of Pharmacology, National Research Institute for Child Health and Development, Setagaya, Tokyo 157-8535, Japan
| | - Junji Yamauchi
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan; (S.S.); (Y.M.)
- Department of Pharmacology, National Research Institute for Child Health and Development, Setagaya, Tokyo 157-8535, Japan
- Correspondence: ; Tel.: +81-42-676-7164; Fax: +81-42-676-8841
| |
Collapse
|
30
|
Suchkov S, Seifi Salmi T, Bai CH, Alizargar J, Wu JP. Ketogenic Diet Is Good for Aging-Related Sarcopenic Obesity. ROLE OF OBESITY IN HUMAN HEALTH AND DISEASE 2021. [DOI: 10.5772/intechopen.96028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2025]
Abstract
Sarcopenic obesity is a skeletal muscle weight loss disease. It has happened at an elderly age. A ketogenic diet is a low-carbohydrate (5%), moderate protein (15%), and a higher-fat diet (80%) can help sarcopenic obese patients burn their fat more effectively. It has many benefits for muscle and fat weight loss. A ketogenic diet can be especially useful for losing excess body fat without hunger and for improving type 2 diabetes. That is because of only a few carbohydrates in the diet, the liver converts fat into fatty acids and ketones. Ketone bodies can replace higher ATP energy. This diet forces the human body to burn fat. This is a good way to lose fat weight without restriction.
Collapse
|
31
|
Kapoor D, Garg D, Sharma S. Emerging Role of the Ketogenic Dietary Therapies beyond Epilepsy in Child Neurology. Ann Indian Acad Neurol 2021; 24:470-480. [PMID: 34728937 PMCID: PMC8513984 DOI: 10.4103/aian.aian_20_21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 01/19/2021] [Accepted: 01/23/2021] [Indexed: 01/18/2023] Open
Abstract
Ketogenic dietary therapies (KDTs) have been in use for refractory paediatric epilepsy for a century now. Over time, KDTs themselves have undergone various modifications to improve tolerability and clinical feasibility, including the Modified Atkins diet (MAD), medium chain triglyceride (MCT) diet and the low glycaemic index treatment (LGIT). Animal and observational studies indicate numerous benefits of KDTs in paediatric neurological conditions apart from their evident benefits in childhood intractable epilepsy, including neurodevelopmental disorders such as autism spectrum disorder, rarer neurogenetic conditions such as Rett syndrome, Fragile X syndrome and Kabuki syndrome, neurodegenerative conditions such as Pelizaeus-Merzbacher disease, and other conditions such as stroke and migraine. A large proportion of the evidence is derived from individual case reports, case series and some small clinical trials, emphasising the vast scope for research in this avenue. The term 'neuroketotherapeutics' has been coined recently to encompass the rapid strides in this field. In the 100th year of its use for paediatric epilepsy, this review covers the role of the KDTs in non-epilepsy neurological conditions among children.
Collapse
Affiliation(s)
- Dipti Kapoor
- Department of Pediatrics (Neurology Division), Lady Hardinge Medical College and Kalawati Saran Children's Hospital, New Delhi, India
| | - Divyani Garg
- Department of Neurology, Lady Hardinge Medical College and Smt. Sucheta Kriplani Hospital, New Delhi, India
| | - Suvasini Sharma
- Department of Pediatrics (Neurology Division), Lady Hardinge Medical College and Kalawati Saran Children's Hospital, New Delhi, India
| |
Collapse
|
32
|
Leclercq S, Le Roy T, Furgiuele S, Coste V, Bindels LB, Leyrolle Q, Neyrinck AM, Quoilin C, Amadieu C, Petit G, Dricot L, Tagliatti V, Cani PD, Verbeke K, Colet JM, Stärkel P, de Timary P, Delzenne NM. Gut Microbiota-Induced Changes in β-Hydroxybutyrate Metabolism Are Linked to Altered Sociability and Depression in Alcohol Use Disorder. Cell Rep 2021; 33:108238. [PMID: 33053357 DOI: 10.1016/j.celrep.2020.108238] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 08/04/2020] [Accepted: 09/16/2020] [Indexed: 12/18/2022] Open
Abstract
Patients with alcohol use disorder (AUD) present with important emotional, cognitive, and social impairments. The gut microbiota has been recently shown to regulate brain functions and behavior but convincing evidence of its role in AUD is lacking. Here, we show that gut dysbiosis is associated with metabolic alterations that affect behavioral (depression, sociability) and neurobiological (myelination, neurotransmission, inflammation) processes involved in alcohol addiction. By transplanting the gut microbiota from AUD patients to mice, we point out that the production of ethanol by specific bacterial genera and the reduction of lipolysis are associated with a lower hepatic synthesis of β-hydroxybutyrate (BHB), which thereby prevents the neuroprotective effect of BHB. We confirm these results in detoxified AUD patients, in which we observe a persisting ethanol production in the feces as well as correlations among low plasma BHB levels and social impairments, depression, or brain white matter alterations.
Collapse
Affiliation(s)
- Sophie Leclercq
- Institute of Neuroscience, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Tiphaine Le Roy
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; WELBIO (Walloon Excellence in Life Sciences and Biotechnology), Louvain Drug Research Institute, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Sonia Furgiuele
- Laboratory of Human Biology & Toxicology, UMONS, 7000 Mons, Belgium
| | - Valentin Coste
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Laure B Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Quentin Leyrolle
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Audrey M Neyrinck
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Caroline Quoilin
- Institute of Neuroscience, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Camille Amadieu
- Institute of Neuroscience, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Géraldine Petit
- Institute of Neuroscience, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Laurence Dricot
- Institute of Neuroscience, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | | | - Patrice D Cani
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; WELBIO (Walloon Excellence in Life Sciences and Biotechnology), Louvain Drug Research Institute, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Kristin Verbeke
- Translational Research Center in Gastrointestinal Disorders, KU Leuven, 3000 Leuven, Belgium
| | - Jean-Marie Colet
- Laboratory of Human Biology & Toxicology, UMONS, 7000 Mons, Belgium
| | - Peter Stärkel
- Laboratory of Hepato-Gastroenterology, Institute of Experimental and Clinical Research, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; Department of Hepatogastroenterology, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Philippe de Timary
- Institute of Neuroscience, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; Department of Adult Psychiatry, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium.
| | - Nathalie M Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium.
| |
Collapse
|
33
|
Sams E. Oligodendrocytes in the aging brain. Neuronal Signal 2021; 5:NS20210008. [PMID: 34290887 PMCID: PMC8264650 DOI: 10.1042/ns20210008] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 12/22/2022] Open
Abstract
More than half of the human brain volume is made up of white matter: regions where axons are coated in myelin, which primarily functions to increase the conduction speed of axon potentials. White matter volume significantly decreases with age, correlating with cognitive decline. Much research in the field of non-pathological brain aging mechanisms has taken a neuron-centric approach, with relatively little attention paid to other neural cells. This review discusses white matter changes, with focus on oligodendrocyte lineage cells and their ability to produce and maintain myelin to support normal brain homoeostasis. Improved understanding of intrinsic cellular changes, general senescence mechanisms, intercellular interactions and alterations in extracellular environment which occur with aging and impact oligodendrocyte cells is paramount. This may lead to strategies to support oligodendrocytes in aging, for example by supporting myelin synthesis, protecting against oxidative stress and promoting the rejuvenation of the intrinsic regenerative potential of progenitor cells. Ultimately, this will enable the protection of white matter integrity thus protecting cognitive function into the later years of life.
Collapse
Affiliation(s)
- Eleanor Catherine Sams
- Blizard Institute, Barts and The London School of Medicine and Dentistry Centre for Neuroscience, Surgery and Trauma, Blizard Institute, 4 Newark Street, Whitechapel E1 2AT, London
| |
Collapse
|
34
|
Zhang N, Liu C, Zhang R, Jin L, Yin X, Zheng X, Siebert HC, Li Y, Wang Z, Loers G, Petridis AK. Amelioration of clinical course and demyelination in the cuprizone mouse model in relation to ketogenic diet. Food Funct 2021; 11:5647-5663. [PMID: 32539054 DOI: 10.1039/c9fo02944c] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Ketogenic diet (KD) is defined as a high-fat, low-carbohydrate diet with appropriate amounts of protein, which has broad neuroprotective effects. However, the mechanisms of ameliorating the demyelination and of the neuroprotective effects of KD have not yet been completely elucidated. Therefore, the present study investigated the protection mechanism of KD treatment in the cuprizone (bis-cyclohexanone oxalydihydrazone, CPZ)-induced demyelination mice model, with special emphasis on neuroinflammation. After the KD treatment, an increased ketone body level in the blood of mice was detected, and a significant increase in the distance traveled within the central area was observed in the open field test, which reflected the increased exploration and decreased anxiety of mice that received CPZ. The results of Luxol fast blue and myelin basic protein (MBP) immunohistochemistry staining for the evaluation of the myelin content within the corpus callosum revealed a noticeable increase in the number of myelinated fibers and myelin score after KD administration in these animals. Concomitant, the protein expressions of glial fibrillary acidic protein (GFAP, an astrocyte marker), ionized calcium-binding adaptor molecule 1 (Iba-1, a microglial marker), CD68 (an activated microglia marker) and CD16/32 (a M1 microglial marker) were down-regulated, while the expression of oligodendrocyte lineage transcription factor 2 (OLIG2, an oligodendrocyte precursor cells marker) was up-regulated by the KD treatment. In addition, the KD treatment not only reduced the level of the C-X-C motif chemokine 10 (CXCL10), which is correlated to the recruitment of activated microglia, but also inhibited the production of proinflammatory cytokines, including interleukin 1β (IL-1β) and tumor necrosis factor-α (TNF-α), which are closely correlated to the M1 phenotype microglia. It is noteworthy, that the expression levels of histone deacetylase 3 (HADC3) and nod-like receptor pyrin domain containing 3 (NLRP3) significantly decreased after KD administration. In conclusion, these data demonstrate that KD decreased the reactive astrocytes and activated the microglia in the corpus callosum, and that KD inhibited the HADC3 and NLRP3 inflammasome signaling pathway in CPZ-treated mice. This suggests that the inhibition of the HADC3 and NLRP3 signaling pathway may be a novel mechanism by which KD exerts its protective actions for the treatment of demyelinating diseases.
Collapse
Affiliation(s)
- Ning Zhang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong 252000, China.
| | - Chunhong Liu
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong 252000, China.
| | - Ruiyan Zhang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong 252000, China.
| | - Li Jin
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong 252000, China.
| | - Xiaohan Yin
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong 252000, China.
| | - Xuexing Zheng
- Department of Virology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.
| | - Hans-Christian Siebert
- RI-B-NT - Research Institute of Bioinformatics and Nanotechnology, Schauenburgerstr. 116, 24118 Kiel, Germany
| | - Yubao Li
- College of agriculture, Liaocheng University, Liaocheng, Shandong 252000, China
| | - Zhengping Wang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong 252000, China.
| | - Gabriele Loers
- Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, University of Hamburg, Falkenried 94, 20251 Hamburg, Germany
| | - Athanasios K Petridis
- Neurosurgical Department, Heinrich Heine University of Düsseldorf, Moorenstraße 5, 40255 Düsseldorf, Germany
| |
Collapse
|
35
|
Hypomyelinating Leukodystrophy 15 (HLD15)-Associated Mutation of EPRS1 Leads to Its Polymeric Aggregation in Rab7-Positive Vesicle Structures, Inhibiting Oligodendroglial Cell Morphological Differentiation. Polymers (Basel) 2021; 13:polym13071074. [PMID: 33805425 PMCID: PMC8037150 DOI: 10.3390/polym13071074] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/24/2021] [Accepted: 03/26/2021] [Indexed: 01/28/2023] Open
Abstract
Pelizaeus–Merzbacher disease (PMD), also known as hypomyelinating leukodystrophy 1 (HLD1), is an X-linked recessive disease affecting in the central nervous system (CNS). The gene responsible for HLD1 encodes proteolipid protein 1 (plp1), which is the major myelin structural protein produced by oligodendroglial cells (oligodendrocytes). HLD15 is an autosomal recessive disease affecting the glutamyl-prolyl-aminoacyl-tRNA synthetase 1 (eprs1) gene, whose product, the EPRS1 protein, is a bifunctional aminoacyl-tRNA synthetase that is localized throughout cell bodies and that catalyzes the aminoacylation of glutamic acid and proline tRNA species. Here, we show that the HLD15-associated nonsense mutation of Arg339-to-Ter (R339X) localizes EPRS1 proteins as polymeric aggregates into Rab7-positive vesicle structures in mouse oligodendroglial FBD-102b cells. Wild-type proteins, in contrast, are distributed throughout the cell bodies. Expression of the R339X mutant proteins, but not the wild-type proteins, in cells induces strong signals regulating Rab7. Whereas cells expressing the wild-type proteins exhibited phenotypes with myelin web-like structures bearing processes following the induction of differentiation, cells expressing the R339X mutant proteins did not. These results indicate that HLD15-associated EPRS1 mutant proteins are localized in Rab7-positive vesicle structures where they modulate Rab7 regulatory signaling, inhibiting cell morphological differentiation. These findings may reveal some of the molecular and cellular pathological mechanisms underlying HLD15.
Collapse
|
36
|
Tepavčević V. Oligodendroglial Energy Metabolism and (re)Myelination. Life (Basel) 2021; 11:238. [PMID: 33805670 PMCID: PMC7998845 DOI: 10.3390/life11030238] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/04/2021] [Accepted: 03/09/2021] [Indexed: 12/12/2022] Open
Abstract
Central nervous system (CNS) myelin has a crucial role in accelerating the propagation of action potentials and providing trophic support to the axons. Defective myelination and lack of myelin regeneration following demyelination can both lead to axonal pathology and neurodegeneration. Energy deficit has been evoked as an important contributor to various CNS disorders, including multiple sclerosis (MS). Thus, dysregulation of energy homeostasis in oligodendroglia may be an important contributor to myelin dysfunction and lack of repair observed in the disease. This article will focus on energy metabolism pathways in oligodendroglial cells and highlight differences dependent on the maturation stage of the cell. In addition, it will emphasize that the use of alternative energy sources by oligodendroglia may be required to save glucose for functions that cannot be fulfilled by other metabolites, thus ensuring sufficient energy input for both myelin synthesis and trophic support to the axons. Finally, it will point out that neuropathological findings in a subtype of MS lesions likely reflect defective oligodendroglial energy homeostasis in the disease.
Collapse
Affiliation(s)
- Vanja Tepavčević
- Achucarro Basque Center for Neuroscience, University of the Basque Country, Parque Cientifico de la UPV/EHU, Barrio Sarriena s/n, Edificio Sede, Planta 3, 48940 Leioa, Spain
| |
Collapse
|
37
|
Mu J, Wang T, Li M, Guan T, Guo Y, Zhang X, Zhang G, Kong J. Ketogenic diet protects myelin and axons in diffuse axonal injury. Nutr Neurosci 2021; 25:1534-1547. [PMID: 33487123 DOI: 10.1080/1028415x.2021.1875300] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
BACKGROUND Ketogenic diet (KD) has been identified as a potential therapy to enhance recovery after traumatic brain injury (TBI). Diffuse axonal injury (DAI) is a common type of traumatic brain injury that is characterized by delayed axonal disconnection. Previous studies showed that demyelination resulting from oligodendrocyte damage contributes to axonal degeneration in DAI. AIM The present study tests a hypothesis that ketone bodies from the ketogenic diet confers protection for myelin and attenuates degeneration of demyelinated axon in DAI. METHODS A modified Marmarou's model of DAI was induced in adult rats. The DAI rats were fed with KD and analyzed with western blot, transmission electron microscope, ELISA test and immunohistochemistry. Meanwhile, a co-culture of primary oligodendrocytes and neurons was treated with ketone body β-hydroxybutryate (βHB) to test for its effects on the myelin-axon unit. RESULTS Here we report that rats fed with KD showed an increased fatty acid metabolism and ketonemia. This dietary intervention significantly reduced demyelination and attenuated axonal damage in rats following DAI, likely through inhibition of DAI-induced excessive mitochondrial fission and promoting mitochondrial fusion. In an in vitro model of myelination, the ketone body βHB increased myelination significantly and reduced axonal degeneration induced by glucose deprivation (GD). βHB robustly increased cell viability, inhibited GD-induced collapse of mitochondrial membrane potential and attenuated death of oligodendrocytes. CONCLUSION Ketone bodies protect myelin-forming oligodendrocytes and reduce axonal damage. Ketogenic diet maybe a promising therapy for DAI.
Collapse
Affiliation(s)
- Jiao Mu
- Department of Forensic Medicine, Hebei North University, Zhangjiakou, People's Republic of China.,Department of Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, People's Republic of China.,Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| | - Tingting Wang
- Department of Forensic Medicine, Hebei North University, Zhangjiakou, People's Republic of China
| | - Meiyu Li
- Department of Forensic Medicine, Hebei North University, Zhangjiakou, People's Republic of China
| | - Teng Guan
- Department of Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, People's Republic of China
| | - Ying Guo
- Department of Forensic Medicine, Hebei North University, Zhangjiakou, People's Republic of China.,Department of Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, People's Republic of China
| | - Xiaoli Zhang
- Department of Life Science Research Center, Hebei North University, Zhangjiakou, People's Republic of China
| | - Guohui Zhang
- Department of Forensic Medicine, Hebei North University, Zhangjiakou, People's Republic of China
| | - Jiming Kong
- Department of Forensic Medicine, Hebei North University, Zhangjiakou, People's Republic of China.,Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
38
|
Abstract
Hypomyelinating leukodystrophies constitute a subset of genetic white matter disorders characterized by a primary lack of myelin deposition. Most patients with severe hypomyelination present in infancy or early childhood and develop severe neurological deficits, but the clinical presentation can also be mild with onset of symptoms in adolescence or adulthood. MRI can be used to visualize the process of myelination in detail, and MRI pattern recognition can provide a clinical diagnosis in many patients. Next-generation sequencing provides a definitive diagnosis in 80-90% of patients. Genes associated with hypomyelination include those that encode structural myelin proteins but also many that encode proteins involved in RNA translation and some lysosomal proteins. The precise pathomechanisms remain to be elucidated. Improved understanding of the process of myelination, the metabolic axonal support functions of myelin and the proposed contribution of myelin to CNS plasticity provide possible explanations as to why almost all patients with hypomyelination experience slow clinical decline after a long phase of stability. In this Review, we provide an overview of the hypomyelinating leukodystrophies, the advances in our understanding of myelin biology and of the genes involved in these disorders, and the insights these advances have provided into their clinical presentations and evolution.
Collapse
|
39
|
Gruenenfelder FI, McLaughlin M, Griffiths IR, Garbern J, Thomson G, Kuzman P, Barrie JA, McCulloch ML, Penderis J, Stassart R, Nave KA, Edgar JM. Neural stem cells restore myelin in a demyelinating model of Pelizaeus-Merzbacher disease. Brain 2020; 143:1383-1399. [PMID: 32419025 PMCID: PMC7462093 DOI: 10.1093/brain/awaa080] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 01/20/2020] [Accepted: 02/05/2020] [Indexed: 12/13/2022] Open
Abstract
Pelizaeus-Merzbacher disease is a fatal X-linked leukodystrophy caused by mutations in the PLP1 gene, which is expressed in the CNS by oligodendrocytes. Disease onset, symptoms and mortality span a broad spectrum depending on the nature of the mutation and thus the degree of CNS hypomyelination. In the absence of an effective treatment, direct cell transplantation into the CNS to restore myelin has been tested in animal models of severe forms of the disease with failure of developmental myelination, and more recently, in severely affected patients with early disease onset due to point mutations in the PLP1 gene, and absence of myelin by MRI. In patients with a PLP1 duplication mutation, the most common cause of Pelizaeus-Merzbacher disease, the pathology is poorly defined because of a paucity of autopsy material. To address this, we examined two elderly patients with duplication of PLP1 in whom the overall syndrome, including end-stage pathology, indicated a complex disease involving dysmyelination, demyelination and axonal degeneration. Using the corresponding Plp1 transgenic mouse model, we then tested the capacity of transplanted neural stem cells to restore myelin in the context of PLP overexpression. Although developmental myelination and axonal coverage by endogenous oligodendrocytes was extensive, as assessed using electron microscopy (n = 3 at each of four end points) and immunostaining (n = 3 at each of four end points), wild-type neural precursors, transplanted into the brains of the newborn mutants, were able to effectively compete and replace the defective myelin (n = 2 at each of four end points). These data demonstrate the potential of neural stem cell therapies to restore normal myelination and protect axons in patients with PLP1 gene duplication mutation and further, provide proof of principle for the benefits of stem cell transplantation for other fatal leukodystrophies with 'normal' developmental myelination.
Collapse
Affiliation(s)
- Fredrik I Gruenenfelder
- School of Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - Mark McLaughlin
- School of Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - Ian R Griffiths
- School of Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - James Garbern
- Department of Neurology and Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
| | - Gemma Thomson
- School of Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - Peter Kuzman
- Department of Neuropathology, University Clinic Leipzig, D-04103 Leipzig, Germany
| | - Jennifer A Barrie
- School of Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK.,Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, G12 8TA, UK
| | - Maj-Lis McCulloch
- School of Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - Jacques Penderis
- School of Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - Ruth Stassart
- Department of Neuropathology, University Clinic Leipzig, D-04103 Leipzig, Germany
| | - Klaus-Armin Nave
- Max Planck Institute for Experimental Medicine, D-37075 Goettingen, Germany
| | - Julia M Edgar
- School of Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK.,Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, G12 8TA, UK.,Max Planck Institute for Experimental Medicine, D-37075 Goettingen, Germany
| |
Collapse
|
40
|
Structural myelin defects are associated with low axonal ATP levels but rapid recovery from energy deprivation in a mouse model of spastic paraplegia. PLoS Biol 2020; 18:e3000943. [PMID: 33196637 PMCID: PMC7704050 DOI: 10.1371/journal.pbio.3000943] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 11/30/2020] [Accepted: 10/22/2020] [Indexed: 11/19/2022] Open
Abstract
In several neurodegenerative disorders, axonal pathology may originate from impaired oligodendrocyte-to-axon support of energy substrates. We previously established transgenic mice that allow measuring axonal ATP levels in electrically active optic nerves. Here, we utilize this technique to explore axonal ATP dynamics in the Plpnull/y mouse model of spastic paraplegia. Optic nerves from Plpnull/y mice exhibited lower and more variable basal axonal ATP levels and reduced compound action potential (CAP) amplitudes, providing a missing link between axonal pathology and a role of oligodendrocytes in brain energy metabolism. Surprisingly, when Plpnull/y optic nerves are challenged with transient glucose deprivation, both ATP levels and CAP decline slower, but recover faster upon reperfusion of glucose. Structurally, myelin sheaths display an increased frequency of cytosolic channels comprising glucose and monocarboxylate transporters, possibly facilitating accessibility of energy substrates to the axon. These data imply that complex metabolic alterations of the axon–myelin unit contribute to the phenotype of Plpnull/y mice. Imaging of ATP dynamics in the optic nerve axons of mice lacking the major myelin protein PLP (a model of spastic paraplegia) reveals complex alterations in the metabolic interaction between oligodendrocytes and axons, associated with structural deficits of myelin.
Collapse
|
41
|
Luetic GG, Menichini ML, Deri N, Steinberg J, Carrá A, Cristiano E, Patrucco L, Curbelo MC, Rojas JI. High birth weight and risk of multiple sclerosis: A multicentre study in Argentina. Mult Scler Relat Disord 2020; 47:102628. [PMID: 33220566 DOI: 10.1016/j.msard.2020.102628] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 10/11/2020] [Accepted: 11/10/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND Multiple sclerosis (MS) is now recognized as a multifactorial disease in which genetic and environmental factors intervene. Considerable efforts have been made to identify external risk factors present in childhood, adolescence and youth, though only a few perinatal risk factors have been positively associated with MS. Previously, we found an association between high birth weight and MS in male patients in a small study in Argentina. The present research was designed to further assess the association between high birth weight and MS in a larger sample of patients, using an extensive and validated general population database as control. METHODS We present an analytical observational, multicentre, population-based, and case-control study. A total of 637 patients (cases) with confirmed MS diagnosis attending five MS specialized centres in Argentina were included. Birth weight (BW) data was recalled by the patient's mother, which is a validated approach. A two-way comparison was performed. First, we used the standard categories of high, adequate and low BW in grams. Then, we applied the weight percentile distribution to provide reproducible results for further research. For a proper assessment and comparison of variables, we adopted the guidelines of the American Academy of Pediatrics for neonate classification according to gestational weeks and to BW in grams. The neonate's BW distribution of the general population was used as control. For the purposes of the study, we adapted Urquía's et al. curves, which are based on an extensive database of all the live births registered in the country from 2003 to 2007. To measure the magnitude of the proportional differences between low, adequate and high BW, the odds ratio (OR) and their 95% confidence interval (CI) were estimated. The mean BW and percentile values for each sex were compared using a z-Normal test. The respective MS patients and general population BW distribution curves by sex were compared between each other. RESULTS Cases and controls were comparable in their demographic, geographic and environmental characteristics. Males showed higher BW than females both in the MS patients and the general population groups. When we applied the sex stratified analysis separately, we found that males in the MS group showed an almost seven times higher risk of high birth weight than males from the general population (OR 6.58 [95% CI 4.81-8.99]). Female patients showed an almost five times higher risk of high BW than their respective controls (OR 4.5 [95% CI 3.06-6.58]). The comparison based on the BW percentile distribution confirmed that MS patients showed higher BW than the general population. This result reached statistical significance from the 75th percentile onwards for both sexes. CONCLUSION In summary, our findings suggested that high BW could be one of the earliest risk factors for MS in life. If this results were reproduced in other centres, high birth weight would emerge as a novel and very early risk factor, potentially modifiable in utero or immediately postpartum, representing a unique opportunity to prevent the disease in future generations.
Collapse
Affiliation(s)
- Geraldine G Luetic
- Instituto de Neurociencias de Rosario, San Lorenzo 3598, Rosario, Santa Fe, 2000, Argentina.
| | - Maria L Menichini
- Instituto de Neurociencias de Rosario, San Lorenzo 3598, Rosario, Santa Fe, 2000, Argentina
| | - Norma Deri
- Centro de Investigaciones Diabaid, CABA, Argentina
| | - Judith Steinberg
- Sección de enfermedades desmielinizantes, Hospital Británico; CABA, Argentina
| | - Adriana Carrá
- Sección de enfermedades desmielinizantes, Hospital Británico; CABA, Argentina
| | | | | | - Maria C Curbelo
- Sección de enfermedades desmielinizantes, Hospital Británico; CABA, Argentina; Hospital Municipal Sofía Santamarina, Buenos Aires, Argentina
| | - Juan I Rojas
- Centro de Esclerosis Múltiple de Buenos Aires, CABA, Argentina
| |
Collapse
|
42
|
Montani L. Lipids in regulating oligodendrocyte structure and function. Semin Cell Dev Biol 2020; 112:114-122. [PMID: 32912639 DOI: 10.1016/j.semcdb.2020.07.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/17/2020] [Accepted: 07/29/2020] [Indexed: 12/15/2022]
Abstract
Oligodendrocytes enwrap central nervous system axons with myelin, a lipid enriched highly organized multi-layer membrane structure that allows for fast long-distance saltatory conduction of neuronal impulses. Myelin has an extremely high lipid content (∼80 % of its dry weight) and a peculiar lipid composition, with a 2:2:1 cholesterol:phospholipid:glycolipid ratio. Inherited neurodegenerative diseases of the lipids (caused by mutations in lipogenic enzymes) often present oligodendrocyte and/or myelin defects which contribute to the overall disease pathophysiology. These phenomena triggered an increasing number of studies over the functions lipid exert to shape and maintain myelin, and brought to the finding that lipids are more than only structural building blocks. They act as signaling molecules to drive proliferation and differentiation of oligodendrocyte progenitor cells, as well as proliferation of premyelinating oligodendrocytes, and their maturation into myelinating ones. Here, we summarize key findings in these areas, while presenting the main related human diseases. Despite many advances in the field, various questions remain open which we briefly discuss. This article is part of a special issue entitled "Role of Lipids in CNS Cell Physiology and Pathology".
Collapse
Affiliation(s)
- Laura Montani
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology, ETH Zürich, Zürich, CH-8093, Switzerland.
| |
Collapse
|
43
|
Bouçanova F, Pollmeier G, Sandor K, Morado Urbina C, Nijssen J, Médard JJ, Bartesaghi L, Pellerin L, Svensson CI, Hedlund E, Chrast R. Disrupted function of lactate transporter MCT1, but not MCT4, in Schwann cells affects the maintenance of motor end-plate innervation. Glia 2020; 69:124-136. [PMID: 32686211 DOI: 10.1002/glia.23889] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 07/02/2020] [Accepted: 07/03/2020] [Indexed: 12/20/2022]
Abstract
Recent studies in neuron-glial metabolic coupling have shown that, in the CNS, astrocytes and oligodendrocytes support neurons with energy-rich lactate/pyruvate via monocarboxylate transporters (MCTs). The presence of such transporters in the PNS, in both Schwann cells and neurons, has prompted us to question if a similar interaction may be present. Here we describe the generation and characterization of conditional knockout mouse models where MCT1 or MCT4 is specifically deleted in Schwann cells (named MCT1 and MCT4 cKO). We show that MCT1 cKO and MCT4 cKO mice develop normally and that myelin in the PNS is preserved. However, MCT1 expressed by Schwann cells is necessary for long-term maintenance of motor end-plate integrity as revealed by disrupted neuromuscular innervation in mutant mice, while MCT4 appears largely dispensable for the support of motor neurons. Concomitant to detected structural alterations, lumbar motor neurons from MCT1 cKO mice show transcriptional changes affecting cytoskeletal components, transcriptional regulators, and mitochondria related transcripts, among others. Together, our data indicate that MCT1 plays a role in Schwann cell-mediated maintenance of motor end-plate innervation thus providing further insight into the emerging picture of the biology of the axon-glia metabolic crosstalk.
Collapse
Affiliation(s)
- Filipa Bouçanova
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Gill Pollmeier
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Katalin Sandor
- Department of Physiology and Pharmacology and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Carlos Morado Urbina
- Department of Physiology and Pharmacology and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jik Nijssen
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Jean-Jacques Médard
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Luca Bartesaghi
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Luc Pellerin
- Department of Physiology, University of Lausanne, Lausanne, Switzerland.,Centre de Résonance Magnétique des Systèmes Biologiques, UMR5536 CNRS, LabEx TRAIL-IBIO, Université de Bordeaux, Bordeaux Cedex, France.,Inserm U1082, Université de Poitiers, Poitiers Cedex, France
| | - Camilla I Svensson
- Department of Physiology and Pharmacology and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Eva Hedlund
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Roman Chrast
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
44
|
Dietary influence on central nervous system myelin production, injury, and regeneration. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165779. [DOI: 10.1016/j.bbadis.2020.165779] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/19/2020] [Accepted: 03/22/2020] [Indexed: 02/07/2023]
|
45
|
Bouçanova F, Chrast R. Metabolic Interaction Between Schwann Cells and Axons Under Physiological and Disease Conditions. Front Cell Neurosci 2020; 14:148. [PMID: 32547370 PMCID: PMC7274022 DOI: 10.3389/fncel.2020.00148] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 05/04/2020] [Indexed: 12/24/2022] Open
Abstract
Recent research into axon-glial interactions in the nervous system has made a compelling case that glial cells have a relevant role in the metabolic support of axons, and that, in the case of myelinating cells, this role is independent of myelination itself. In this mini-review article, we summarize some of those observations and focus on Schwann cells (SC), drawing parallels between glia of the central and peripheral nervous systems (PNS), pointing out limitations in current knowledge, and discussing its potential clinical relevance. First, we introduce SC, their development and main roles, and follow with an evolutionary perspective of glial metabolic function. Then we provide evidence of the myelin-independent aspects of axonal support and their coupling to neuronal metabolism. Finally, we address the opportunity to use SC-axon metabolic interactions as therapeutic targets to treat peripheral neuropathies.
Collapse
Affiliation(s)
- Filipa Bouçanova
- Department of Neuroscience, KarolinskaInstitutet, Stockholm, Sweden.,Department of Clinical Neuroscience, KarolinskaInstitutet, Stockholm, Sweden
| | - Roman Chrast
- Department of Neuroscience, KarolinskaInstitutet, Stockholm, Sweden.,Department of Clinical Neuroscience, KarolinskaInstitutet, Stockholm, Sweden
| |
Collapse
|
46
|
β-hydroxybutyrate and its metabolic effects on age-associated pathology. Exp Mol Med 2020; 52:548-555. [PMID: 32269287 PMCID: PMC7210293 DOI: 10.1038/s12276-020-0415-z] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 02/04/2020] [Accepted: 02/27/2020] [Indexed: 12/17/2022] Open
Abstract
Aging is a universal process that renders individuals vulnerable to many diseases. Although this process is irreversible, dietary modulation and caloric restriction are often considered to have antiaging effects. Dietary modulation can increase and maintain circulating ketone bodies, especially β-hydroxybutyrate (β-HB), which is one of the most abundant ketone bodies in human circulation. Increased β-HB has been reported to prevent or improve the symptoms of various age-associated diseases. Indeed, numerous studies have reported that a ketogenic diet or ketone ester administration alleviates symptoms of neurodegenerative diseases, cardiovascular diseases, and cancers. Considering the potential of β-HB and the intriguing data emerging from in vivo and in vitro experiments as well as clinical trials, this therapeutic area is worthy of attention. In this review, we highlight studies that focus on the identified targets of β-HB and the cellular signals regulated by β-HB with respect to alleviation of age-associated ailments. Boosting levels of a byproduct of fatty acid breakdown may help alleviate the symptoms of age-associated health conditions. When the body is low on glucose, it breaks down fatty acids for energy, generating byproduct metabolites called ketones. The ketone β-hydroxybutyrate (β-HB) regulates cellular signaling and gene and protein expression. There are indications that ketogenic diets or ketone administration, which increase β-BH may prevent ageing-associated progression of illnesses like cardiovascular and neurodegenerative diseases and cancer. Young-min Han and co-workers at Georgia State University in Atlanta, USA, reviewed current understanding of β-BH and its molecular targets. β-BH is a potent metabolite small enough to filter through cell membranes and circulate throughout the body, including the brain, influencing signaling pathways. Further investigations into associated molecular mechanisms will verify the metabolite’s potential as a therapeutic agent.
Collapse
|
47
|
Poitelon Y, Kopec AM, Belin S. Myelin Fat Facts: An Overview of Lipids and Fatty Acid Metabolism. Cells 2020; 9:cells9040812. [PMID: 32230947 PMCID: PMC7226731 DOI: 10.3390/cells9040812] [Citation(s) in RCA: 192] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 12/11/2022] Open
Abstract
Myelin is critical for the proper function of the nervous system and one of the most complex cell–cell interactions of the body. Myelination allows for the rapid conduction of action potentials along axonal fibers and provides physical and trophic support to neurons. Myelin contains a high content of lipids, and the formation of the myelin sheath requires high levels of fatty acid and lipid synthesis, together with uptake of extracellular fatty acids. Recent studies have further advanced our understanding of the metabolism and functions of myelin fatty acids and lipids. In this review, we present an overview of the basic biology of myelin lipids and recent insights on the regulation of fatty acid metabolism and functions in myelinating cells. In addition, this review may serve to provide a foundation for future research characterizing the role of fatty acids and lipids in myelin biology and metabolic disorders affecting the central and peripheral nervous system.
Collapse
|
48
|
Luan CJ, Guo W, Chen L, Wei XW, He Y, Chen Y, Dang SY, Prior R, Li X, Kuang Y, Wang ZG, Van Den Bosch L, Gu MM. CMT2Q-causing mutation in the Dhtkd1 gene lead to sensory defects, mitochondrial accumulation and altered metabolism in a knock-in mouse model. Acta Neuropathol Commun 2020; 8:32. [PMID: 32169121 PMCID: PMC7071680 DOI: 10.1186/s40478-020-00901-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 02/21/2020] [Indexed: 12/30/2022] Open
Abstract
Charcot-Marie-Tooth disease (CMT) is a group of inherited neurological disorders of the peripheral nervous system. CMT is subdivided into two main types: a demyelinating form, known as CMT1, and an axonal form, known as CMT2. Nearly 30 genes have been identified as a cause of CMT2. One of these is the 'dehydrogenase E1 and transketolase domain containing 1' (DHTKD1) gene. We previously demonstrated that a nonsense mutation [c.1455 T > G (p.Y485*)] in exon 8 of DHTKD1 is one of the disease-causing mutations in CMT2Q (MIM 615025). The aim of the current study was to investigate whether human disease-causing mutations in the Dhtkd1 gene cause CMT2Q phenotypes in a mouse model in order to investigate the physiological function and pathogenic mechanisms associated with mutations in the Dhtkd1 gene in vivo. Therefore, we generated a knock-in mouse model with the Dhtkd1Y486* point mutation. We observed that the Dhtkd1 expression level in sciatic nerve of knock-in mice was significantly lower than in wild-type mice. Moreover, a histopathological phenotype was observed, reminiscent of a peripheral neuropathy, including reduced large axon diameter and abnormal myelination in peripheral nerves. The knock-in mice also displayed clear sensory defects, while no abnormalities in the motor performance were observed. In addition, accumulation of mitochondria and an elevated energy metabolic state was observed in the knock-in mice. Taken together, our study indicates that the Dhtkd1Y486* knock-in mice partially recapitulate the clinical phenotypes of CMT2Q patients and we hypothesize that there might be a compensatory effect from the elevated metabolic state in the knock-in mice that enables them to maintain their normal locomotor function.
Collapse
|
49
|
Siems SB, Jahn O, Eichel MA, Kannaiyan N, Wu LMN, Sherman DL, Kusch K, Hesse D, Jung RB, Fledrich R, Sereda MW, Rossner MJ, Brophy PJ, Werner HB. Proteome profile of peripheral myelin in healthy mice and in a neuropathy model. eLife 2020; 9:e51406. [PMID: 32130108 PMCID: PMC7056269 DOI: 10.7554/elife.51406] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 02/19/2020] [Indexed: 02/07/2023] Open
Abstract
Proteome and transcriptome analyses aim at comprehending the molecular profiles of the brain, its cell-types and subcellular compartments including myelin. Despite the relevance of the peripheral nervous system for normal sensory and motor capabilities, analogous approaches to peripheral nerves and peripheral myelin have fallen behind evolving technical standards. Here we assess the peripheral myelin proteome by gel-free, label-free mass-spectrometry for deep quantitative coverage. Integration with RNA-Sequencing-based developmental mRNA-abundance profiles and neuropathy disease genes illustrates the utility of this resource. Notably, the periaxin-deficient mouse model of the neuropathy Charcot-Marie-Tooth 4F displays a highly pathological myelin proteome profile, exemplified by the discovery of reduced levels of the monocarboxylate transporter MCT1/SLC16A1 as a novel facet of the neuropathology. This work provides the most comprehensive proteome resource thus far to approach development, function and pathology of peripheral myelin, and a straightforward, accurate and sensitive workflow to address myelin diversity in health and disease.
Collapse
Affiliation(s)
- Sophie B Siems
- Department of Neurogenetics, Max Planck Institute of Experimental MedicineGöttingenGermany
| | - Olaf Jahn
- Proteomics Group, Max Planck Institute of Experimental MedicineGöttingenGermany
| | - Maria A Eichel
- Department of Neurogenetics, Max Planck Institute of Experimental MedicineGöttingenGermany
| | - Nirmal Kannaiyan
- Department of Psychiatry and Psychotherapy, University Hospital, LMU MunichMunichGermany
| | - Lai Man N Wu
- Centre for Discovery Brain Sciences, University of EdinburghEdinburghUnited Kingdom
| | - Diane L Sherman
- Centre for Discovery Brain Sciences, University of EdinburghEdinburghUnited Kingdom
| | - Kathrin Kusch
- Department of Neurogenetics, Max Planck Institute of Experimental MedicineGöttingenGermany
| | - Dörte Hesse
- Proteomics Group, Max Planck Institute of Experimental MedicineGöttingenGermany
| | - Ramona B Jung
- Department of Neurogenetics, Max Planck Institute of Experimental MedicineGöttingenGermany
| | - Robert Fledrich
- Department of Neurogenetics, Max Planck Institute of Experimental MedicineGöttingenGermany
- Institute of Anatomy, University of LeipzigLeipzigGermany
| | - Michael W Sereda
- Department of Neurogenetics, Max Planck Institute of Experimental MedicineGöttingenGermany
- Department of Clinical Neurophysiology, University Medical CenterGöttingenGermany
| | - Moritz J Rossner
- Department of Psychiatry and Psychotherapy, University Hospital, LMU MunichMunichGermany
| | - Peter J Brophy
- Centre for Discovery Brain Sciences, University of EdinburghEdinburghUnited Kingdom
| | - Hauke B Werner
- Department of Neurogenetics, Max Planck Institute of Experimental MedicineGöttingenGermany
| |
Collapse
|
50
|
Sauma S, Casaccia P. Does the gut microbiota contribute to the oligodendrocyte progenitor niche? Neurosci Lett 2019; 715:134574. [PMID: 31669313 DOI: 10.1016/j.neulet.2019.134574] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/15/2019] [Accepted: 10/18/2019] [Indexed: 12/29/2022]
Abstract
The past decade has seen a growing number of studies on the relationship between the gut microbiota and the brain. This mini-review will focus on the unexpected findings linking the microbiome to myelination. We first address the temporal correlation between the acquisition of a gut microbiota in the developing organism and developmental myelination. We then review the factors impacting the composition of the child's gut microbiota, ranging from maternal stress to modality of delivery and from breastfeeding and diet composition to antibiotic treatment early in life. We discuss the topic of gut-brain communication with an emphasis on myelination, and propose the concept that gut microbes produce metabolites which may constitute a "metabolic" niche. Distinct bacterial communities may create very different "niches", some permissive and others inhibitory for myelin generation or maintenance. We speculate that this concept of "gut dysbiosis" may also in part explain the reduced myelin content detected in several neurological and psychiatric disorders. We conclude by envisioning intervention with probiotics and prebiotics to favor the formation of a microbial metabolic "niche" favoring myelin production to promote brain health.
Collapse
Affiliation(s)
- Sami Sauma
- Neuroscience Initiative, Advanced Science Research Center, The Graduate Center at The City University of New York, New York, NY, USA; Graduate Program in Biology The Graduate Center at The City University of New York, New York, NY, USA
| | - Patrizia Casaccia
- Neuroscience Initiative, Advanced Science Research Center, The Graduate Center at The City University of New York, New York, NY, USA; Graduate Program in Biology The Graduate Center at The City University of New York, New York, NY, USA.
| |
Collapse
|