51
|
Budisteanu M, Jurca C, Papuc SM, Focsa I, Riga D, Riga S, Jurca A, Arghir A. Treatment of Epilepsy Associated with Common Chromosomal Developmental Diseases. Open Life Sci 2020; 15:21-29. [PMID: 33987468 PMCID: PMC8114617 DOI: 10.1515/biol-2020-0003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/22/2019] [Indexed: 11/15/2022] Open
Abstract
Chromosomal diseases are heterogeneous conditions with complex phenotypes, which include also epileptic seizures. Each chromosomal syndrome has a range of specific characteristics regarding the type of seizures, EEG findings and specific response to antiepileptic drugs, significant in the context of the respective genetic etiology. Therefore, it is very important to know these particularities, in order to avoid an exacerbation of seizures or some side effects. In this paper we will present a review of the epileptic seizures and antiepileptic treatment in some of the most common chromosomal syndromes.
Collapse
Affiliation(s)
- Magdalena Budisteanu
- Prof. Dr. Alexandru Obregia” Clinical Hospital of Psychiatry, BucharestRomania
- ”Victor Babes“ National Institute of Pathology, BucharestRomania
- ”Titu Maiorescu” University – Faculty of Medicine, BucharestRomania
| | - Claudia Jurca
- University of Oradea, Faculty of Medicine and Pharmacy, Preclinical Department, OradeaRomania
| | | | - Ina Focsa
- ”Carol Davila” University of Pharmacy and Medicine, BucharestRomania
| | - Dan Riga
- Prof. Dr. Alexandru Obregia” Clinical Hospital of Psychiatry, BucharestRomania
| | - Sorin Riga
- Prof. Dr. Alexandru Obregia” Clinical Hospital of Psychiatry, BucharestRomania
| | - Alexandru Jurca
- University of Oradea, Faculty of Medicine and Pharmacy, Preclinical Department, OradeaRomania
| | - Aurora Arghir
- ”Victor Babes“ National Institute of Pathology, BucharestRomania
- ”Carol Davila” University of Pharmacy and Medicine, BucharestRomania
| |
Collapse
|
52
|
Berg EL, Pride MC, Petkova SP, Lee RD, Copping NA, Shen Y, Adhikari A, Fenton TA, Pedersen LR, Noakes LS, Nieman BJ, Lerch JP, Harris S, Born HA, Peters MM, Deng P, Cameron DL, Fink KD, Beitnere U, O'Geen H, Anderson AE, Dindot SV, Nash KR, Weeber EJ, Wöhr M, Ellegood J, Segal DJ, Silverman JL. Translational outcomes in a full gene deletion of ubiquitin protein ligase E3A rat model of Angelman syndrome. Transl Psychiatry 2020; 10:39. [PMID: 32066685 PMCID: PMC7026078 DOI: 10.1038/s41398-020-0720-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 12/17/2019] [Accepted: 01/02/2020] [Indexed: 12/17/2022] Open
Abstract
Angelman syndrome (AS) is a rare neurodevelopmental disorder characterized by developmental delay, impaired communication, motor deficits and ataxia, intellectual disabilities, microcephaly, and seizures. The genetic cause of AS is the loss of expression of UBE3A (ubiquitin protein ligase E6-AP) in the brain, typically due to a deletion of the maternal 15q11-q13 region. Previous studies have been performed using a mouse model with a deletion of a single exon of Ube3a. Since three splice variants of Ube3a exist, this has led to a lack of consistent reports and the theory that perhaps not all mouse studies were assessing the effects of an absence of all functional UBE3A. Herein, we report the generation and functional characterization of a novel model of Angelman syndrome by deleting the entire Ube3a gene in the rat. We validated that this resulted in the first comprehensive gene deletion rodent model. Ultrasonic vocalizations from newborn Ube3am-/p+ were reduced in the maternal inherited deletion group with no observable change in the Ube3am+/p- paternal transmission cohort. We also discovered Ube3am-/p+ exhibited delayed reflex development, motor deficits in rearing and fine motor skills, aberrant social communication, and impaired touchscreen learning and memory in young adults. These behavioral deficits were large in effect size and easily apparent in the larger rodent species. Low social communication was detected using a playback task that is unique to rats. Structural imaging illustrated decreased brain volume in Ube3am-/p+ and a variety of intriguing neuroanatomical phenotypes while Ube3am+/p- did not exhibit altered neuroanatomy. Our report identifies, for the first time, unique AS relevant functional phenotypes and anatomical markers as preclinical outcomes to test various strategies for gene and molecular therapies in AS.
Collapse
Affiliation(s)
- E L Berg
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, USA
| | - M C Pride
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, USA
| | - S P Petkova
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, USA
| | - R D Lee
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, USA
| | - N A Copping
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Y Shen
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, USA
| | - A Adhikari
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, USA
| | - T A Fenton
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, USA
| | - L R Pedersen
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, USA
| | - L S Noakes
- Mouse Imaging Centre, Toronto Centre for Phenogenomics, The Hospital for Sick Children, Toronto, ON, Canada
| | - B J Nieman
- Mouse Imaging Centre, Toronto Centre for Phenogenomics, The Hospital for Sick Children, Toronto, ON, Canada
| | - J P Lerch
- Wellcome Centre for Integrative Neuroimaging, The University of Oxford, Oxford, UK
| | - S Harris
- Department of Pediatrics and Neurology, Baylor College of Medicine, Houston, TX, USA
| | - H A Born
- Department of Pediatrics and Neurology, Baylor College of Medicine, Houston, TX, USA
| | - M M Peters
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, USA
| | - P Deng
- Stem Cell Program, Institute for Regenerative Cures, and Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, USA
| | - D L Cameron
- Stem Cell Program, Institute for Regenerative Cures, and Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, USA
| | - K D Fink
- Stem Cell Program, Institute for Regenerative Cures, and Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, USA
| | - U Beitnere
- MIND Institute, Genome Center, and Department of Biochemistry and Molecular Medicine, University of California Davis, Davis, CA, USA
| | - H O'Geen
- MIND Institute, Genome Center, and Department of Biochemistry and Molecular Medicine, University of California Davis, Davis, CA, USA
| | - A E Anderson
- Department of Pediatrics and Neurology, Baylor College of Medicine, Houston, TX, USA
| | - S V Dindot
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - K R Nash
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, USA
| | - E J Weeber
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, USA
| | - M Wöhr
- Behavioral Neuroscience, Experimental and Biological Psychology, Philipps-University of Marburg, Marburg, Germany
| | - J Ellegood
- Mouse Imaging Centre, Toronto Centre for Phenogenomics, The Hospital for Sick Children, Toronto, ON, Canada
| | - D J Segal
- MIND Institute, Genome Center, and Department of Biochemistry and Molecular Medicine, University of California Davis, Davis, CA, USA
| | - J L Silverman
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, USA.
| |
Collapse
|
53
|
Herber DL, Weeber EJ, D'Agostino DP, Duis J. Evaluation of the safety and tolerability of a nutritional Formulation in patients with ANgelman Syndrome (FANS): study protocol for a randomized controlled trial. Trials 2020; 21:60. [PMID: 31918761 PMCID: PMC6953273 DOI: 10.1186/s13063-019-3996-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Accepted: 12/13/2019] [Indexed: 01/12/2023] Open
Abstract
Background Ketogenic and low-glycemic-index diets are effective in treating drug-resistant seizures in children with Angelman syndrome. Cognition, mobility, sleep, and gastrointestinal health are intrinsically linked to seizure activity and overall quality of life. Ketogenic and low-glycemic diets restrict carbohydrate consumption and stabilize blood glucose levels. The ketogenic diet induces ketosis, a metabolic state where ketone bodies are preferentially used for fuel. The use of exogenous ketones in promoting ketosis in Angelman syndrome has not been previously studied. The study formulation evaluated herein contains the exogenous ketone beta-hydroxybutyrate to rapidly shift the body towards ketosis, resulting in enhanced metabolic efficiency. Methods/design This is a 16-week, randomized, double-blind, placebo-controlled, crossover study to assess the safety and tolerability of a nutritional formula containing exogenous ketones. It also examines the potential for exogenous ketones to improve the patient’s nutritional status which can impact the physiologic, symptomatic, and health outcome liabilities of living with Angelman syndrome. Discussion This manuscript outlines the rationale for a study designed to be the first to provide data on nutritional approaches for patients with Angelman syndrome using exogenous ketones. Trial registration ClinicalTrials.gov, ID: NCT03644693. Registered on 23 August 2018. Last updated on 23 August 2018.
Collapse
Affiliation(s)
- Donna L Herber
- Disruptive Nutrition, LLC, 300 West Morgan Street, Suite 1510, Durham, NC, 27701, USA
| | - Edwin J Weeber
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, NJ, 07080-2449, USA
| | - Dominic P D'Agostino
- Morsani College of Medicine, Department of Molecular Pharmacology and Physiology, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, USA.,Institute for Human and Machine Cognition, Ocala, FL, USA
| | - Jessica Duis
- Division of Medical Genetics & Genomic Medicine, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, 37232-2578, USA.
| |
Collapse
|
54
|
Enkhtuy B, Kwon HE, Kim HD. Advances in Ketogenic Diet Therapies in Pediatric Epilepsy. ANNALS OF CHILD NEUROLOGY 2019. [DOI: 10.26815/acn.2019.00192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
55
|
Yang X. Towards an understanding of Angelman syndrome in mice studies. J Neurosci Res 2019; 98:1162-1173. [PMID: 31867793 DOI: 10.1002/jnr.24576] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 11/28/2019] [Accepted: 12/04/2019] [Indexed: 12/13/2022]
Abstract
Angelman syndrome (AS) is a rare neurodevelopmental disorder characterized by severe mental retardation, absence of speech, abnormal motor coordination, abnormal EEG, and spontaneous seizure. AS is caused by a deficiency in the ubiquitin ligase E3A (Ube3a) gene product, known to play a dual role as both ubiquitin ligase and transcription coactivator. In AS animal models, multiple Ube3a substrates are accumulated in neurons. So far, studies in mouse models have either aimed at re-expressing Ube3a or manipulating downstream signaling pathways. Reintroducing Ube3a in AS mice showed promising results but may have two caveats. First, it may cause an overdosage in the Ube3a expression, which in turn is known to contribute to autism spectrum disorders. Second, in mutation cases, the exogenous Ube3a may have to compete with the mutated endogenous form. Such two caveats left spaces for developing therapies or interventions directed to targets downstream Ube3a. Notably, Ube3a expression is dynamically regulated by neuronal activity and plays a crucial role in synaptic plasticity. The abnormal synaptic plasticity uncovered in AS mice has been frequently rescued, but circuits symptoms like seizure are resistant to treatment. Future investigations are needed to further clarify the function (s) of Ube3a during development. Here I reviewed the recently identified major Ube3a substrates and signaling pathways involved in AS pathology, the Ube3a expression, imprinting and evolution, the AS mouse models that have been generated and inspired therapeutic potentials, and finally proposed some future explorations to better understand the AS pathology.
Collapse
Affiliation(s)
- Xin Yang
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, China
| |
Collapse
|
56
|
Germeys C, Vandoorne T, Bercier V, Van Den Bosch L. Existing and Emerging Metabolomic Tools for ALS Research. Genes (Basel) 2019; 10:E1011. [PMID: 31817338 PMCID: PMC6947647 DOI: 10.3390/genes10121011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 11/23/2019] [Accepted: 12/03/2019] [Indexed: 12/12/2022] Open
Abstract
Growing evidence suggests that aberrant energy metabolism could play an important role in the pathogenesis of amyotrophic lateral sclerosis (ALS). Despite this, studies applying advanced technologies to investigate energy metabolism in ALS remain scarce. The rapidly growing field of metabolomics offers exciting new possibilities for ALS research. Here, we review existing and emerging metabolomic tools that could be used to further investigate the role of metabolism in ALS. A better understanding of the metabolic state of motor neurons and their surrounding cells could hopefully result in novel therapeutic strategies.
Collapse
Affiliation(s)
- Christine Germeys
- Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI), KU Leuven—University of Leuven, 3000 Leuven, Belgium; (C.G.); (T.V.); (V.B.)
- VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, 3000 Leuven, Belgium
| | - Tijs Vandoorne
- Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI), KU Leuven—University of Leuven, 3000 Leuven, Belgium; (C.G.); (T.V.); (V.B.)
- VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, 3000 Leuven, Belgium
| | - Valérie Bercier
- Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI), KU Leuven—University of Leuven, 3000 Leuven, Belgium; (C.G.); (T.V.); (V.B.)
- VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, 3000 Leuven, Belgium
| | - Ludo Van Den Bosch
- Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI), KU Leuven—University of Leuven, 3000 Leuven, Belgium; (C.G.); (T.V.); (V.B.)
- VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, 3000 Leuven, Belgium
| |
Collapse
|
57
|
Poff AM, Rho JM, D'Agostino DP. Ketone Administration for Seizure Disorders: History and Rationale for Ketone Esters and Metabolic Alternatives. Front Neurosci 2019; 13:1041. [PMID: 31680801 PMCID: PMC6803688 DOI: 10.3389/fnins.2019.01041] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 09/13/2019] [Indexed: 12/31/2022] Open
Abstract
The ketogenic diet (KD) is a high-fat, low-carbohydrate treatment for medically intractable epilepsy. One of the hallmark features of the KD is the production of ketone bodies which have long been believed, but not yet proven, to exert direct anti-seizure effects. The prevailing view has been that ketosis is an epiphenomenon during KD treatment, mostly due to clinical observations that blood ketone levels do not correlate well with seizure control. Nevertheless, there is increasing experimental evidence that ketone bodies alone can exert anti-seizure properties through a multiplicity of mechanisms, including but not limited to: (1) activation of inhibitory adenosine and ATP-sensitive potassium channels; (2) enhancement of mitochondrial function and reduction in oxidative stress; (3) attenuation of excitatory neurotransmission; and (4) enhancement of central γ-aminobutyric acid (GABA) synthesis. Other novel actions more recently reported include inhibition of inflammasome assembly and activation of peripheral immune cells, and epigenetic effects by decreasing the activity of histone deacetylases (HDACs). Collectively, the preclinical evidence to date suggests that ketone administration alone might afford anti-seizure benefits for patients with epilepsy. There are, however, pragmatic challenges in administering ketone bodies in humans, but prior concerns may largely be mitigated through the use of ketone esters or balanced ketone electrolyte formulations that can be given orally and induce elevated and sustained hyperketonemia to achieve therapeutic effects.
Collapse
Affiliation(s)
- Angela M Poff
- Laboratory of Metabolic Medicine, Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Jong M Rho
- Departments of Pediatrics, Clinical Neurosciences, Physiology and Pharmacology, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Division of Pediatric Neurology, Rady Children's Hospital-San Diego, University of California, San Diego, San Diego, CA, United States
| | - Dominic P D'Agostino
- Laboratory of Metabolic Medicine, Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States.,Institute for Human and Machine Cognition, Ocala, FL, United States
| |
Collapse
|
58
|
Sandusky-Beltran LA, Kovalenko A, Ma C, Calahatian JIT, Placides DS, Watler MD, Hunt JB, Darling AL, Baker JD, Blair LJ, Martin MD, Fontaine SN, Dickey CA, Lussier AL, Weeber EJ, Selenica MLB, Nash KR, Gordon MN, Morgan D, Lee DC. Spermidine/spermine-N 1-acetyltransferase ablation impacts tauopathy-induced polyamine stress response. Alzheimers Res Ther 2019; 11:58. [PMID: 31253191 PMCID: PMC6599347 DOI: 10.1186/s13195-019-0507-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 05/21/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND Tau stabilizes microtubules; however, in Alzheimer's disease (AD) and tauopathies, tau becomes hyperphosphorylated, aggregates, and results in neuronal death. Our group recently uncovered a unique interaction between polyamine metabolism and tau fate. Polyamines exert an array of physiological effects that support neuronal function and cognitive processing. Specific stimuli can elicit a polyamine stress response (PSR), resulting in altered central polyamine homeostasis. Evidence suggests that elevations in polyamines following a short-term stressor are beneficial; however, persistent stress and subsequent PSR activation may lead to maladaptive polyamine dysregulation, which is observed in AD, and may contribute to neuropathology and disease progression. METHODS Male and female mice harboring tau P301L mutation (rTg4510) were examined for a tau-induced central polyamine stress response (tau-PSR). The direct effect of tau-PSR byproducts on tau fibrillization and oligomerization were measured using a thioflavin T assay and a N2a split superfolder GFP-Tau (N2a-ssGT) cell line, respectively. To therapeutically target the tau-PSR, we bilaterally injected caspase 3-cleaved tau truncated at aspartate 421 (AAV9 Tau ΔD421) into the hippocampus and cortex of spermidine/spermine-N1-acetyltransferase (SSAT), a key regulator of the tau-PSR, knock out (SSAT-/-), and wild type littermates, and the effects on tau neuropathology, polyamine dysregulation, and behavior were measured. Lastly, cellular models were employed to further examine how SSAT repression impacted tau biology. RESULTS Tau induced a unique tau-PSR signature in rTg4510 mice, notably in the accumulation of acetylated spermidine. In vitro, higher-order polyamines prevented tau fibrillization but acetylated spermidine failed to mimic this effect and even promoted fibrillization and oligomerization. AAV9 Tau ΔD421 also elicited a unique tau-PSR in vivo, and targeted disruption of SSAT prevented the accumulation of acetylated polyamines and impacted several tau phospho-epitopes. Interestingly, SSAT knockout mice presented with altered behavior in the rotarod task, the elevated plus maze, and marble burying task, thus highlighting the impact of polyamine homeostasis within the brain. CONCLUSION These data represent a novel paradigm linking tau pathology and polyamine dysfunction and that targeting specific arms within the polyamine pathway may serve as new targets to mitigate certain components of the tau phenotype.
Collapse
Affiliation(s)
- Leslie A. Sandusky-Beltran
- 0000 0001 2353 285Xgrid.170693.aByrd Alzheimer’s Institute, Department of Pharmaceutical Sciences, University of South Florida, 4001 E. Fletcher Ave, Tampa, FL 33613 USA
- 0000 0004 1936 8753grid.137628.9Neuroscience Institute, Department of Neuroscience and Physiology, New York University School of Medicine, 1 Park Avenue, New York, NY 10016 USA
| | - Andrii Kovalenko
- 0000 0001 2353 285Xgrid.170693.aByrd Alzheimer’s Institute, Department of Pharmaceutical Sciences, University of South Florida, 4001 E. Fletcher Ave, Tampa, FL 33613 USA
| | - Chao Ma
- 0000 0001 2353 285Xgrid.170693.aByrd Alzheimer’s Institute, Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL 33613 USA
| | - John Ivan T. Calahatian
- 0000 0001 2353 285Xgrid.170693.aByrd Alzheimer’s Institute, Department of Pharmaceutical Sciences, University of South Florida, 4001 E. Fletcher Ave, Tampa, FL 33613 USA
| | - Devon S. Placides
- 0000 0001 2353 285Xgrid.170693.aByrd Alzheimer’s Institute, Department of Pharmaceutical Sciences, University of South Florida, 4001 E. Fletcher Ave, Tampa, FL 33613 USA
| | - Mallory D. Watler
- 0000 0001 2353 285Xgrid.170693.aByrd Alzheimer’s Institute, Department of Pharmaceutical Sciences, University of South Florida, 4001 E. Fletcher Ave, Tampa, FL 33613 USA
| | - Jerry B. Hunt
- 0000 0001 2353 285Xgrid.170693.aByrd Alzheimer’s Institute, Department of Pharmaceutical Sciences, University of South Florida, 4001 E. Fletcher Ave, Tampa, FL 33613 USA
| | - April L. Darling
- 0000 0001 2353 285Xgrid.170693.aByrd Alzheimer’s Institute, Department of Molecular Medicine, University of South Florida, Tampa, FL 33613 USA
| | - Jeremy D. Baker
- 0000 0001 2353 285Xgrid.170693.aByrd Alzheimer’s Institute, Department of Molecular Medicine, University of South Florida, Tampa, FL 33613 USA
| | - Laura J. Blair
- 0000 0001 2353 285Xgrid.170693.aByrd Alzheimer’s Institute, Department of Molecular Medicine, University of South Florida, Tampa, FL 33613 USA
| | - Mackenzie D. Martin
- 0000 0001 2353 285Xgrid.170693.aByrd Alzheimer’s Institute, Department of Molecular Medicine, University of South Florida, Tampa, FL 33613 USA
| | - Sarah N. Fontaine
- 0000 0001 2353 285Xgrid.170693.aByrd Alzheimer’s Institute, Department of Molecular Medicine, University of South Florida, Tampa, FL 33613 USA
| | - Chad A. Dickey
- 0000 0001 2353 285Xgrid.170693.aByrd Alzheimer’s Institute, Department of Molecular Medicine, University of South Florida, Tampa, FL 33613 USA
| | - April L. Lussier
- 0000 0001 2353 285Xgrid.170693.aByrd Alzheimer’s Institute, Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL 33613 USA
| | - Edwin J. Weeber
- 0000 0001 2353 285Xgrid.170693.aByrd Alzheimer’s Institute, Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL 33613 USA
| | - Maj-Linda B. Selenica
- 0000 0001 2353 285Xgrid.170693.aByrd Alzheimer’s Institute, Department of Pharmaceutical Sciences, University of South Florida, 4001 E. Fletcher Ave, Tampa, FL 33613 USA
| | - Kevin R. Nash
- 0000 0001 2353 285Xgrid.170693.aByrd Alzheimer’s Institute, Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL 33613 USA
| | - Marcia N. Gordon
- 0000 0001 2353 285Xgrid.170693.aByrd Alzheimer’s Institute, Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL 33613 USA
- 0000 0001 2150 1785grid.17088.36Department of Translational Science & Molecular Medicine, Michigan State University, 400 Monroe Ave NW, Grand Rapids, MI 49503 USA
| | - Dave Morgan
- 0000 0001 2353 285Xgrid.170693.aByrd Alzheimer’s Institute, Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL 33613 USA
- 0000 0001 2150 1785grid.17088.36Department of Translational Science & Molecular Medicine, Michigan State University, 400 Monroe Ave NW, Grand Rapids, MI 49503 USA
| | - Daniel C. Lee
- 0000 0001 2353 285Xgrid.170693.aByrd Alzheimer’s Institute, Department of Pharmaceutical Sciences, University of South Florida, 4001 E. Fletcher Ave, Tampa, FL 33613 USA
| |
Collapse
|
59
|
Deemer SE, Davis RAH, Gower BA, Koutnik AP, Poff AM, Dickinson SL, Allison DB, D'Agostino DP, Plaisance EP. Concentration-Dependent Effects of a Dietary Ketone Ester on Components of Energy Balance in Mice. Front Nutr 2019; 6:56. [PMID: 31119133 PMCID: PMC6504762 DOI: 10.3389/fnut.2019.00056] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 04/11/2019] [Indexed: 12/30/2022] Open
Abstract
Objectives: Exogenous ketones may provide therapeutic benefit in treatment of obesity. Administration of the ketone ester (KE) R,S-1,3-butanediol acetoacetate diester (BD-AcAc2) decreases body weight in mice, but effects on energy balance have not been extensively characterized. The purpose of this investigation was to explore concentration-dependent effects of BD-AcAc2 on energy intake and expenditure in mice. Methods: Forty-two male C57BL/6J mice were randomly assigned to one of seven isocaloric diets (n = 6 per group): (1) Control (CON, 0% KE by kcals); (2) KE5 (5% KE); (3) KE10 (10% KE); (4) KE15 (15% KE); (5) KE20 (20% KE); (6) KE25 (25% KE); and (7) KE30 (30% KE) for 3 weeks. Energy intake and body weight were measured daily. Fat mass (FM), lean body mass (LBM), and energy expenditure (EE) were measured at completion of the study. Differences among groups were compared to CON using ANOVA and ANCOVA. Results: Mean energy intake was similar between CON and each concentration of KE, except KE30 which was 12% lower than CON (P < 0.01). KE25 and KE30 had lower body weight and FM compared to CON, while only KE30 had lower LBM (P < 0.03). Adjusted resting and total EE were lower in KE30 compared to CON (P < 0.03), but similar for all other groups. Conclusions: A diet comprised of 30% energy from BD-AcAc2 results in lower energy intake, coincident with lower body weight and whole animal adiposity; while KE20 and KE25 have significantly lower body weight and adiposity effects independent of changes in energy intake or expenditure.
Collapse
Affiliation(s)
- Sarah E. Deemer
- Nutrition Obesity Research Center, University of Alabama, Birmingham, AL, United States
| | - Rachel A. H. Davis
- Nutrition Obesity Research Center, University of Alabama, Birmingham, AL, United States
- Department of Nutrition Sciences, University of Alabama, Birmingham, AL, United States
| | - Barbara A. Gower
- Nutrition Obesity Research Center, University of Alabama, Birmingham, AL, United States
- Department of Nutrition Sciences, University of Alabama, Birmingham, AL, United States
| | - Andrew P. Koutnik
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, United States
| | - Angela M. Poff
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, United States
| | | | - David B. Allison
- School of Public Health, Indiana University Bloomington, Bloomington, IN, United States
| | - Dominic P. D'Agostino
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, United States
| | - Eric P. Plaisance
- Nutrition Obesity Research Center, University of Alabama, Birmingham, AL, United States
- Department of Nutrition Sciences, University of Alabama, Birmingham, AL, United States
- Department of Human Studies, University of Alabama, Birmingham, AL, United States
| |
Collapse
|
60
|
Fogle KJ, Smith AR, Satterfield SL, Gutierrez AC, Hertzler JI, McCardell CS, Shon JH, Barile ZJ, Novak MO, Palladino MJ. Ketogenic and anaplerotic dietary modifications ameliorate seizure activity in Drosophila models of mitochondrial encephalomyopathy and glycolytic enzymopathy. Mol Genet Metab 2019; 126:439-447. [PMID: 30683556 PMCID: PMC6536302 DOI: 10.1016/j.ymgme.2019.01.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 01/14/2019] [Accepted: 01/15/2019] [Indexed: 12/13/2022]
Abstract
Seizures are a feature not only of the many forms of epilepsy, but also of global metabolic diseases such as mitochondrial encephalomyopathy (ME) and glycolytic enzymopathy (GE). Modern anti-epileptic drugs (AEDs) are successful in many cases, but some patients are refractory to existing AEDs, which has led to a surge in interest in clinically managed dietary therapy such as the ketogenic diet (KD). This high-fat, low-carbohydrate diet causes a cellular switch from glycolysis to fatty acid oxidation and ketone body generation, with a wide array of downstream effects at the genetic, protein, and metabolite level that may mediate seizure protection. We have recently shown that a Drosophila model of human ME (ATP61) responds robustly to the KD; here, we have investigated the mechanistic importance of the major metabolic consequences of the KD in the context of this bioenergetics disease: ketogenesis, reduction of glycolysis, and anaplerosis. We have found that reduction of glycolysis does not confer seizure protection, but that dietary supplementation with ketone bodies or the anaplerotic lipid triheptanoin, which directly replenishes the citric acid cycle, can mimic the success of the ketogenic diet even in the presence of standard carbohydrate levels. We have also shown that the proper functioning of the citric acid cycle is crucial to the success of the KD in the context of ME. Furthermore, our data reveal that multiple seizure models, in addition to ATP61, are treatable with the ketogenic diet. Importantly, one of these mutants is TPIsugarkill, which models human glycolytic enzymopathy, an incurable metabolic disorder with severe neurological consequences. Overall, these studies reveal widespread success of the KD in Drosophila, further cementing its status as an excellent model for studies of KD treatment and mechanism, and reveal key insights into the therapeutic potential of dietary therapy against neuronal hyperexcitability in epilepsy and metabolic disease.
Collapse
Affiliation(s)
- Keri J Fogle
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Pittsburgh Institute for Neurodegenerative Diseases (PIND), University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA.
| | - Amber R Smith
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Pittsburgh Institute for Neurodegenerative Diseases (PIND), University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Sidney L Satterfield
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Pittsburgh Institute for Neurodegenerative Diseases (PIND), University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Alejandra C Gutierrez
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Pittsburgh Institute for Neurodegenerative Diseases (PIND), University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - J Ian Hertzler
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Pittsburgh Institute for Neurodegenerative Diseases (PIND), University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Caleb S McCardell
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Pittsburgh Institute for Neurodegenerative Diseases (PIND), University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Joy H Shon
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Pittsburgh Institute for Neurodegenerative Diseases (PIND), University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Zackery J Barile
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Pittsburgh Institute for Neurodegenerative Diseases (PIND), University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Molly O Novak
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Pittsburgh Institute for Neurodegenerative Diseases (PIND), University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Michael J Palladino
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Pittsburgh Institute for Neurodegenerative Diseases (PIND), University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| |
Collapse
|
61
|
Çubukçu D, Güzel O, Arslan N. Effect of Ketogenic Diet on Motor Functions and Daily Living Activities of Children With Multidrug-Resistant Epilepsy: A Prospective Study. J Child Neurol 2018; 33:718-723. [PMID: 30062917 DOI: 10.1177/0883073818786558] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
AIMS To investigate the effect of ketogenic diet on motor function and daily living activities in children with epilepsy. METHODS A total of 62 children (median age 5.0 years) were enrolled. Motor function was assessed using the Gross Motor Function Measure (GMFM), and daily living activities and cognitive functions were assessed using the Functional Independence Measure (WeeFIM) before treatment and 3, 6, and 12 months after ketogenic diet treatment. RESULTS Significant improvement in total GMFM and WeeFIM scores ( P < .001) were found during the 12 months of ketogenic diet treatment. There was a positive correlation between total GMFM scores and WeeFIM scores at baseline (r= 0.792, P = .0001), and at 3 (r= 0.780, P = .0001), 6 (r= 0.744, P = .0001), and 12 months (r= 0.692, P = .0001) of treatment. Both the responder (50 patients, 80.7%) and nonresponder (12 patients, 19.3%) patient groups showed significantly higher GMFM and WeeFIM scores at 12 months of treatment compared to baseline values. A ≥50% reduction in seizure frequency was observed in 77.4%, 72.6%, and 80.7% of the patients after 3, 6, and 12 months of treatment, respectively. CONCLUSION Ketogenic diet treatment improves motor functions and daily living activities in children with epilepsy during the 12 months of treatment.
Collapse
Affiliation(s)
- Duygu Çubukçu
- 1 Department of Physical Medicine and Rehabilitation, Behçet Uz Children Hospital, Izmir, Turkey
| | - Orkide Güzel
- 2 Division of Pediatric Neurology, Behçet Uz Children Hospital, Izmir, Turkey.,3 Mediterranean Ketogenic Diet Center, Izmir, Turkey
| | - Nur Arslan
- 4 Department of Pediatrics, Division of Pediatric Metabolism and Nutrition, Dokuz Eylul University, Izmir, Turkey.,5 Izmir Biomedicine and Genome Center (iBG-izmir), Dokuz Eylul University, Izmir, Turkey
| |
Collapse
|
62
|
Investigating owner use of dietary supplements in dogs with idiopathic epilepsy. Res Vet Sci 2018; 119:276-284. [PMID: 30064067 DOI: 10.1016/j.rvsc.2018.07.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 06/01/2018] [Accepted: 07/21/2018] [Indexed: 12/29/2022]
Abstract
Epilepsy is the most common chronic neurological disorder in dogs. Some diets have been shown to have a positive impact upon the seizure activity in dogs with idiopathic epilepsy (IE), while other diets and dietary supplements (DS), although marketed as providing health benefits, lack conclusive scientific evidence on their actual beneficial effects. A web-based owner questionnaire was designed to assess how and why owners of dogs with IE use different dietary regimes and DS. The study cohort, with 297 valid responses, consisted mainly of pure-breed (82.5%) male neutered (52.9%) dogs. Over two-thirds of owners (67.7%) changed their dog's diet after their dog received a diagnosis of IE. Nearly half of the owners (45.8%) reported giving DS, the most common being coconut oil or derived medium-chain triglyceride oil (71.3%). Some owner justifications of DS use included improvement of seizure frequency (88.2%), seizure severity (61.8%) and protection from potential drug side effects (62.5%). Many owners give DS to their dog with IE. The pharmacokinetic properties of anti-epileptic drugs, such as efficacy, absorption and clearance can be influenced by other medications, diets and possibly by DS. We propose that use of DS should be considered and monitored by veterinary surgeons in epilepsy management.
Collapse
|
63
|
Sada N, Inoue T. Electrical Control in Neurons by the Ketogenic Diet. Front Cell Neurosci 2018; 12:208. [PMID: 30061816 PMCID: PMC6054928 DOI: 10.3389/fncel.2018.00208] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 06/26/2018] [Indexed: 01/01/2023] Open
Abstract
The ketogenic diet is used as a diet treatment for drug-resistant epilepsy, but there are no antiepileptic drugs based on the ketogenic diet. The ketogenic diet changes energy metabolites (ketone bodies, glucose and lactate) in the brain, which consequently changes electrical activities in neurons and ultimately suppresses seizures in epileptic patients. In order to elucidate the antiseizure effects of the ketogenic diet, it is important to clarify the mechanism by which these metabolic changes are converted to electrical changes in neurons. In this review, we summarize electrophysiological studies focusing on electrical control in neurons by the ketogenic diet. Recent studies have identified electrical regulators driven by the ketogenic diet: ion channels (ATP-sensitive K+ channels and voltage-dependent Ca2+ channels), synaptic receptors (AMPA-type glutamate receptors and adenosine A1 receptors), neurotransmitter transporters (vesicular glutamate transporters), and others (BCL-2-associated agonist of cell death and lactate dehydrogenase). Thus, the ketogenic diet presumably elicits neuronal inhibition via the combined actions of these molecules. From the viewpoint of drug development, these molecules are valuable as targets for the development of new antiepileptic drugs. Drug therapy to mimic the ketogenic diet may be feasible in the future, through the combination of multiple antiepileptic drugs targeting these molecules.
Collapse
Affiliation(s)
- Nagisa Sada
- Department of Biophysical Chemistry, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
- Department of Hygiene, Kawasaki Medical School, Kurashiki, Japan
| | - Tsuyoshi Inoue
- Department of Biophysical Chemistry, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
64
|
Abstract
PURPOSE OF REVIEW High-fat, low-carbohydrate ketogenic diets have been used for almost a century for the treatment of epilepsy. Used traditionally for the treatment of refractory pediatric epilepsies, in recent years the use of ketogenic diets has experienced a revival to include the treatment of adulthood epilepsies as well as conditions ranging from autism to chronic pain and cancer. Despite the ability of ketogenic diet therapy to suppress seizures refractory to antiepileptic drugs and reports of lasting seizure freedom, the underlying mechanisms are poorly understood. This review explores new insights into mechanisms mobilized by ketogenic diet therapies. RECENT FINDINGS Ketogenic diets act through a combination of mechanisms, which are linked to the effects of ketones and glucose restriction, and to interactions with receptors, channels, and metabolic enzymes. Decanoic acid, a component of medium-chain triclycerides, contributes to seizure control through direct α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor inhibition, whereas drugs targeting lactate dehydrogenase reduce seizures through inhibition of a metabolic pathway. Ketogenic diet therapy also affects DNA methylation, a novel epigenetic mechanism of the diet. SUMMARY Ketogenic diet therapy combines several beneficial mechanisms that provide broad benefits for the treatment of epilepsy with the potential to not only suppress seizures but also to modify the course of the epilepsy.
Collapse
|
65
|
Simeone TA, Simeone KA, Stafstrom CE, Rho JM. Do ketone bodies mediate the anti-seizure effects of the ketogenic diet? Neuropharmacology 2018; 133:233-241. [PMID: 29325899 PMCID: PMC5858992 DOI: 10.1016/j.neuropharm.2018.01.011] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Revised: 11/27/2017] [Accepted: 01/07/2018] [Indexed: 01/01/2023]
Abstract
Although the mechanisms underlying the anti-seizure effects of the high-fat ketogenic diet (KD) remain unclear, a long-standing question has been whether ketone bodies (i.e., β-hydroxybutyrate, acetoacetate and acetone), either alone or in combination, contribute mechanistically. The traditional belief has been that while ketone bodies reflect enhanced fatty acid oxidation and a general shift toward intermediary metabolism, they are not likely to be the key mediators of the KD's clinical effects, as blood levels of β-hydroxybutyrate do not correlate consistently with improved seizure control. Against this unresolved backdrop, new data support ketone bodies as having anti-seizure actions. Specifically, β-hydroxybutyrate has been shown to interact with multiple novel molecular targets such as histone deacetylases, hydroxycarboxylic acid receptors on immune cells, and the NLRP3 inflammasome. Clearly, as a diet-based therapy is expected to render a broad array of biochemical, molecular, and cellular changes, no single mechanism can explain how the KD works. Specific metabolic substrates or enzymes are only a few of many important factors influenced by the KD that can collectively influence brain hyperexcitability and hypersynchrony. This review summarizes recent novel experimental findings supporting the anti-seizure and neuroprotective properties of ketone bodies.
Collapse
Affiliation(s)
- Timothy A Simeone
- Department of Pharmacology, Creighton University School of Medicine, Omaha, NE, USA
| | - Kristina A Simeone
- Department of Pharmacology, Creighton University School of Medicine, Omaha, NE, USA
| | - Carl E Stafstrom
- Department of Neurology, and Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jong M Rho
- Department of Pediatrics, Department of Clinical Neurosciences, and Department of Physiology and Pharmacology, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
66
|
Harvey CJDC, Schofield GM, Williden M. The use of nutritional supplements to induce ketosis and reduce symptoms associated with keto-induction: a narrative review. PeerJ 2018; 6:e4488. [PMID: 29576959 PMCID: PMC5858534 DOI: 10.7717/peerj.4488] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 02/20/2018] [Indexed: 12/21/2022] Open
Abstract
Background Adaptation to a ketogenic diet (keto-induction) can cause unpleasant symptoms, and this can reduce tolerability of the diet. Several methods have been suggested as useful for encouraging entry into nutritional ketosis (NK) and reducing symptoms of keto-induction. This paper reviews the scientific literature on the effects of these methods on time-to-NK and on symptoms during the keto-induction phase. Methods PubMed, Science Direct, CINAHL, MEDLINE, Alt Health Watch, Food Science Source and EBSCO Psychology and Behavioural Sciences Collection electronic databases were searched online. Various purported ketogenic supplements were searched along with the terms “ketogenic diet”, “ketogenic”, “ketosis” and ketonaemia (/ ketonemia). Additionally, author names and reference lists were used for further search of the selected papers for related references. Results Evidence, from one mouse study, suggests that leucine doesn’t significantly increase beta-hydroxybutyrate (BOHB) but the addition of leucine to a ketogenic diet in humans, while increasing the protein-to-fat ratio of the diet, doesn’t reduce ketosis. Animal studies indicate that the short chain fatty acids acetic acid and butyric acid, increase ketone body concentrations. However, only one study has been performed in humans. This demonstrated that butyric acid is more ketogenic than either leucine or an 8-chain monoglyceride. Medium-chain triglycerides (MCTs) increase BOHB in a linear, dose-dependent manner, and promote both ketonaemia and ketogenesis. Exogenous ketones promote ketonaemia but may inhibit ketogenesis. Conclusions There is a clear ketogenic effect of supplemental MCTs; however, it is unclear whether they independently improve time to NK and reduce symptoms of keto-induction. There is limited research on the potential for other supplements to improve time to NK and reduce symptoms of keto-induction. Few studies have specifically evaluated symptoms and adverse effects of a ketogenic diet during the induction phase. Those that have typically were not designed to evaluate these variables as primary outcomes, and thus, more research is required to elucidate the role that supplementation might play in encouraging ketogenesis, improve time to NK, and reduce symptoms associated with keto-induction.
Collapse
Affiliation(s)
- Cliff J D C Harvey
- Human Potential Centre, Auckland University of Technology, Auckland, New Zealand
| | - Grant M Schofield
- Human Potential Centre, Auckland University of Technology, Auckland, New Zealand
| | - Micalla Williden
- Human Potential Centre, Auckland University of Technology, Auckland, New Zealand
| |
Collapse
|
67
|
Kovács Z, D'Agostino DP, Ari C. Anxiolytic Effect of Exogenous Ketone Supplementation Is Abolished by Adenosine A1 Receptor Inhibition in Wistar Albino Glaxo/Rijswijk Rats. Front Behav Neurosci 2018. [PMID: 29520223 PMCID: PMC5827672 DOI: 10.3389/fnbeh.2018.00029] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Anxiety disorders are one of the most common mental health problems worldwide, but the exact pathophysiology remains largely unknown. It has been demonstrated previously that administration of exogenous ketone supplement KSMCT (ketone salt/KS + medium chain triglyceride/MCT oil) by intragastric gavage for 7 days decreased the anxiety level in genetically absence epileptic Wistar Albino Glaxo/Rijswijk (WAG/Rij) rats. To investigate the potential role of the adenosinergic system in the pathomechanism of anxiety we tested whether the inhibition of adenosine A1 receptors (A1Rs) influence the anxiolytic effect of the exogenous ketone supplement. As A1Rs may mediate such an effect, in the present study we used a specific A1R antagonist, DPCPX (1,3-dipropyl-8-cyclopentylxanthine) to test whether it modulates the anxiolytic effect of sub-chronically (7 days) applied KSMCT in the previously tested animal model by using elevated plus maze (EPM) test. We administered KSMCT (2.5 g/kg/day) alone by intragastric gavage and in combination with intraperitoneally (i.p.) injected of DPCPX in two doses (lower: 0.15 mg/kg, higher: 0.25 mg/kg). Control groups represented i.p saline and water gavage with or without i.p. DPCPX administration (2.5 g/kg/day). After treatments, the level of blood glucose and beta-hydroxybutyrate (βHB), as well as body weight were recorded. KSMCT alone significantly increased the time spent in the open arms and decreased the time spent in the closed arms, supporting our previous results. Injection of lower dose of DPCPX decreased, while higher dose of DPCPX abolished the effect of KSMCT administration on EPM. Blood βHB levels were significantly increased after administration of KSMCT, while DPCPX did not change the KSMCT induced increase in blood βHB levels. These results demonstrate that A1R inhibition modified (decreased) the anti-anxiety effect of KSMCT administration implying that the adenosinergic system, likely via A1Rs, may modulate the exogenous ketone supplement induced anxiolytic influence.
Collapse
Affiliation(s)
- Zsolt Kovács
- Savaria Department of Biology, Eötvös Loránd University (ELTE), Budapest, Hungary
| | - Dominic P D'Agostino
- Department of Molecular Pharmacology and Physiology, Metabolic Medicine Research Laboratory, Morsani College of Medicine, University of South Florida, Tampa, FL, United States.,Institute for Human and Machine Cognition, Ocala, FL, United States
| | - Csilla Ari
- Department of Molecular Pharmacology and Physiology, Metabolic Medicine Research Laboratory, Morsani College of Medicine, University of South Florida, Tampa, FL, United States.,Department of Psychology, Hyperbaric Neuroscience Research Laboratory, University of South Florida, Tampa, FL, United States
| |
Collapse
|
68
|
Veyrat-Durebex C, Reynier P, Procaccio V, Hergesheimer R, Corcia P, Andres CR, Blasco H. How Can a Ketogenic Diet Improve Motor Function? Front Mol Neurosci 2018; 11:15. [PMID: 29434537 PMCID: PMC5790787 DOI: 10.3389/fnmol.2018.00015] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 01/10/2018] [Indexed: 12/12/2022] Open
Abstract
A ketogenic diet (KD) is a normocaloric diet composed by high fat (80-90%), low carbohydrate, and low protein consumption that induces fasting-like effects. KD increases ketone body (KBs) production and its concentration in the blood, providing the brain an alternative energy supply that enhances oxidative mitochondrial metabolism. In addition to its profound impact on neuro-metabolism and bioenergetics, the neuroprotective effect of specific polyunsaturated fatty acids and KBs involves pleiotropic mechanisms, such as the modulation of neuronal membrane excitability, inflammation, or reactive oxygen species production. KD is a therapy that has been used for almost a century to treat medically intractable epilepsy and has been increasingly explored in a number of neurological diseases. Motor function has also been shown to be improved by KD and/or medium-chain triglyceride diets in rodent models of Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and spinal cord injury. These studies have proposed that KD may induce a modification in synaptic morphology and function, involving ionic channels, glutamatergic transmission, or synaptic vesicular cycling machinery. However, little is understood about the molecular mechanisms underlying the impact of KD on motor function and the perspectives of its use to acquire the neuromuscular effects. The aim of this review is to explore the conditions through which KD might improve motor function. First, we will describe the main consequences of KD exposure in tissues involved in motor function. Second, we will report and discuss the relevance of KD in pre-clinical and clinical trials in the major diseases presenting motor dysfunction.
Collapse
Affiliation(s)
- Charlotte Veyrat-Durebex
- Département de Biochimie et Génétique, Centre Hospitalier Universitaire, Angers, France
- INSERM 1083, CNRS, Equipe Mitolab, Institut MITOVASC, UMR 6015, Université d’Angers, Angers, France
| | - Pascal Reynier
- Département de Biochimie et Génétique, Centre Hospitalier Universitaire, Angers, France
- INSERM 1083, CNRS, Equipe Mitolab, Institut MITOVASC, UMR 6015, Université d’Angers, Angers, France
| | - Vincent Procaccio
- Département de Biochimie et Génétique, Centre Hospitalier Universitaire, Angers, France
- INSERM 1083, CNRS, Equipe Mitolab, Institut MITOVASC, UMR 6015, Université d’Angers, Angers, France
| | | | - Philippe Corcia
- INSERM U930, Université François Rabelais de Tours, Tours, France
- Service de Neurologie, Centre Hospitalier Universitaire de Tours, Tours, France
| | - Christian R. Andres
- INSERM U930, Université François Rabelais de Tours, Tours, France
- Laboratoire de Biochimie et Biologie Moléculaire, Centre Hospitalier Universitaire de Tours, Tours, France
| | - Hélène Blasco
- INSERM 1083, CNRS, Equipe Mitolab, Institut MITOVASC, UMR 6015, Université d’Angers, Angers, France
- INSERM U930, Université François Rabelais de Tours, Tours, France
- Laboratoire de Biochimie et Biologie Moléculaire, Centre Hospitalier Universitaire de Tours, Tours, France
| |
Collapse
|
69
|
Copping NA, Christian SGB, Ritter DJ, Islam MS, Buscher N, Zolkowska D, Pride MC, Berg EL, LaSalle JM, Ellegood J, Lerch JP, Reiter LT, Silverman JL, Dindot SV. Neuronal overexpression of Ube3a isoform 2 causes behavioral impairments and neuroanatomical pathology relevant to 15q11.2-q13.3 duplication syndrome. Hum Mol Genet 2017; 26:3995-4010. [PMID: 29016856 PMCID: PMC5886211 DOI: 10.1093/hmg/ddx289] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 06/21/2017] [Accepted: 07/10/2017] [Indexed: 01/07/2023] Open
Abstract
Maternally derived copy number gains of human chromosome 15q11.2-q13.3 (Dup15q syndrome or Dup15q) cause intellectual disability, epilepsy, developmental delay, hypotonia, speech impairments, and minor dysmorphic features. Dup15q syndrome is one of the most common and penetrant chromosomal abnormalities observed in individuals with autism spectrum disorder (ASD). Although ∼40 genes are located in the 15q11.2-q13.3 region, overexpression of the ubiquitin-protein E3A ligase (UBE3A) gene is thought to be the predominant molecular cause of the phenotypes observed in Dup15q syndrome. The UBE3A gene demonstrates maternal-specific expression in neurons and loss of maternal UBE3A causes Angelman syndrome, a neurodevelopmental disorder with some overlapping neurological features to Dup15q. To directly test the hypothesis that overexpression of UBE3A is an important underlying molecular cause of neurodevelopmental dysfunction, we developed and characterized a mouse overexpressing Ube3a isoform 2 in excitatory neurons. Ube3a isoform 2 is conserved between mouse and human and known to play key roles in neuronal function. Transgenic mice overexpressing Ube3a isoform 2 in excitatory forebrain neurons exhibited increased anxiety-like behaviors, learning impairments, and reduced seizure thresholds. However, these transgenic mice displayed normal social approach, social interactions, and repetitive motor stereotypies that are relevant to ASD. Reduced forebrain, hippocampus, striatum, amygdala, and cortical volume were also observed. Altogether, these findings show neuronal overexpression of Ube3a isoform 2 causes phenotypes translatable to neurodevelopmental disorders.
Collapse
Affiliation(s)
- Nycole A Copping
- MIND Institute, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | | | - Dylan J Ritter
- MIND Institute, School of Medicine, University of California, Davis, Sacramento, CA, USA
- Texas A&M, College Station, TX, USA
| | - M Saharul Islam
- MIND Institute, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Nathalie Buscher
- MIND Institute, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Dorota Zolkowska
- MIND Institute, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Michael C Pride
- MIND Institute, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Elizabeth L Berg
- MIND Institute, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Janine M LaSalle
- MIND Institute, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Jacob Ellegood
- The Hospital for Sick Children, Mouse Imaging Centre, Toronto, ON, Canada
| | - Jason P Lerch
- The Hospital for Sick Children, Mouse Imaging Centre, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Lawrence T Reiter
- Departments of Neurology, Pediatrics and Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Jill L Silverman
- MIND Institute, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | | |
Collapse
|
70
|
Paleologou E, Ismayilova N, Kinali M. Use of the Ketogenic Diet to Treat Intractable Epilepsy in Mitochondrial Disorders. J Clin Med 2017; 6:E56. [PMID: 28587136 PMCID: PMC5483866 DOI: 10.3390/jcm6060056] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/16/2017] [Accepted: 05/22/2017] [Indexed: 12/31/2022] Open
Abstract
Mitochondrial disorders are a clinically heterogeneous group of disorders that are caused by defects in the respiratory chain, the metabolic pathway of the adenosine tri-phosphate (ATP) production system. Epilepsy is a common and important feature of these disorders and its management can be challenging. Epileptic seizures in the context of mitochondrial disease are usually treated with conventional anti-epileptic medication, apart from valproic acid. However, in accordance with the treatment of intractable epilepsy where there are limited treatment options, the ketogenic diet (KD) has been considered as an alternative therapy. The use of the KD and its more palatable formulations has shown promising results. It is especially indicated and effective in the treatment of mitochondrial disorders due to complex I deficiency. Further research into the mechanism of action and the neuroprotective properties of the KD will allow more targeted therapeutic strategies and thus optimize the treatment of both epilepsy in the context of mitochondrial disorders but also in other neurodegenerative disorders.
Collapse
Affiliation(s)
- Eleni Paleologou
- Chelsea and Westmister Hospital, 369 Fulham road, Chelsea, London SW10 9NH, UK.
| | - Naila Ismayilova
- Chelsea and Westmister Hospital, 369 Fulham road, Chelsea, London SW10 9NH, UK.
| | - Maria Kinali
- Chelsea and Westmister Hospital, 369 Fulham road, Chelsea, London SW10 9NH, UK.
| |
Collapse
|
71
|
Sidorov MS, Deck GM, Dolatshahi M, Thibert RL, Bird LM, Chu CJ, Philpot BD. Delta rhythmicity is a reliable EEG biomarker in Angelman syndrome: a parallel mouse and human analysis. J Neurodev Disord 2017; 9:17. [PMID: 28503211 PMCID: PMC5422949 DOI: 10.1186/s11689-017-9195-8] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 04/21/2017] [Indexed: 01/11/2023] Open
Abstract
Background Clinicians have qualitatively described rhythmic delta activity as a prominent EEG abnormality in individuals with Angelman syndrome, but this phenotype has yet to be rigorously quantified in the clinical population or validated in a preclinical model. Here, we sought to quantitatively measure delta rhythmicity and evaluate its fidelity as a biomarker. Methods We quantified delta oscillations in mouse and human using parallel spectral analysis methods and measured regional, state-specific, and developmental changes in delta rhythms in a patient population. Results Delta power was broadly increased and more dynamic in both the Angelman syndrome mouse model, relative to wild-type littermates, and in children with Angelman syndrome, relative to age-matched neurotypical controls. Enhanced delta oscillations in children with Angelman syndrome were present during wakefulness and sleep, were generalized across the neocortex, and were more pronounced at earlier ages. Conclusions Delta rhythmicity phenotypes can serve as reliable biomarkers for Angelman syndrome in both preclinical and clinical settings. Electronic supplementary material The online version of this article (doi:10.1186/s11689-017-9195-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Michael S Sidorov
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27599 USA.,Carolina Institute for Developmental Disabilities, University of North Carolina, Chapel Hill, NC 27599 USA.,Neuroscience Center, University of North Carolina, Chapel Hill, NC 27599 USA
| | - Gina M Deck
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114 USA.,Harvard Medical School, Boston, MA 02215 USA.,Present Address: The Neurology Foundation, Rhode Island Hospital and Warren Alpert School of Medicine at Brown University, Providence, RI 02903 USA
| | - Marjan Dolatshahi
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114 USA.,Harvard Medical School, Boston, MA 02215 USA
| | - Ronald L Thibert
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114 USA
| | - Lynne M Bird
- Department of Pediatrics, University of California, San Diego, CA USA.,Division of Dysmorphology/Genetics, Rady Children's Hospital, San Diego, CA USA
| | - Catherine J Chu
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114 USA.,Harvard Medical School, Boston, MA 02215 USA
| | - Benjamin D Philpot
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27599 USA.,Carolina Institute for Developmental Disabilities, University of North Carolina, Chapel Hill, NC 27599 USA.,Neuroscience Center, University of North Carolina, Chapel Hill, NC 27599 USA
| |
Collapse
|
72
|
Schoeler NE, Bell G, Yuen A, Kapelner AD, Heales SJR, Cross JH, Sisodiya S. An examination of biochemical parameters and their association with response to ketogenic dietary therapies. Epilepsia 2017; 58:893-900. [PMID: 28369834 DOI: 10.1111/epi.13729] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2017] [Indexed: 12/25/2022]
Abstract
OBJECTIVE In the absence of specific metabolic disorders, accurate predictors of response to ketogenic dietary therapies (KDTs) for treating epilepsy are largely unknown. We hypothesized that specific biochemical parameters would be associated with the effectiveness of KDT in humans with epilepsy. The parameters tested were β-hydroxybutyrate, acetoacetate, nonesterified fatty acids, free and acylcarnitine profile, glucose, and glucose-ketone index (GKI). METHODS Biochemical results from routine blood tests conducted at baseline prior to initiation of KDT and at 3-month follow-up were obtained from 13 adults and 215 children with KDT response data from participating centers. One hundred thirty-two (57%) of 228 participants had some data at both baseline and 3 months; 52 (23%) of 228 had data only at baseline; 22 (10%) of 228 had data only at 3 months; and 22 (10%) of 228 had no data. KDT response was defined as ≥50% seizure reduction at 3-month follow-up. RESULTS Acetyl carnitine at baseline was significantly higher in responders (p < 0.007). It was not associated with response at 3-month follow-up. There was a trend for higher levels of free carnitine and other acylcarnitine esters at baseline and at 3-month follow-up in KDT responders. There was also a trend for greater differences in levels of propionyl carnitine and in β-hydroxybutyrate measured at baseline and 3-month follow-up in KDT responders. No other biochemical parameters were associated with response at any time point. SIGNIFICANCE Our finding that certain carnitine fractions, in particular baseline acetyl carnitine, are positively associated with greater efficacy of KDT is consistent with the theory that alterations in energy metabolism may play a role in the mechanisms of action of KDT.
Collapse
Affiliation(s)
- Natasha E Schoeler
- UCL Great Ormond Street Institute of Child Health, London, United Kingdom.,Department of Clinical and Experimental Epilepsy, NIHR University College London Hospitals Biomedical Research Centre, UCL Institute of Neurology, London, United Kingdom
| | - Gail Bell
- Epilepsy Society, Chalfont St Peter, United Kingdom
| | - Alan Yuen
- Epilepsy Society, Chalfont St Peter, United Kingdom
| | - Adam D Kapelner
- Department of Mathematics, Queens College, The City University of New York (CUNY), New York, New York, U.S.A
| | - Simon J R Heales
- Genetics and Genomic Medicine, UCL Institute of Child Health, London, United Kingdom.,Chemical Pathology, Great Ormond Street Hospital for Children, London, United Kingdom.,Neurometabolic Unit, National Hospital for Neurology and Neurosurgery, London, United Kingdom
| | - J Helen Cross
- UCL Great Ormond Street Institute of Child Health, London, United Kingdom.,Great Ormond Street Hospital for Children, London, United Kingdom.,Young Epilepsy, Lingfield, United Kingdom
| | - Sanjay Sisodiya
- Department of Clinical and Experimental Epilepsy, NIHR University College London Hospitals Biomedical Research Centre, UCL Institute of Neurology, London, United Kingdom.,Epilepsy Society, Chalfont St Peter, United Kingdom
| |
Collapse
|
73
|
Cheng N, Rho JM, Masino SA. Metabolic Dysfunction Underlying Autism Spectrum Disorder and Potential Treatment Approaches. Front Mol Neurosci 2017; 10:34. [PMID: 28270747 PMCID: PMC5318388 DOI: 10.3389/fnmol.2017.00034] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 01/30/2017] [Indexed: 12/14/2022] Open
Abstract
Autism spectrum disorder (ASD) is characterized by deficits in sociability and communication, and increased repetitive and/or restrictive behaviors. While the etio-pathogenesis of ASD is unknown, clinical manifestations are diverse and many possible genetic and environmental factors have been implicated. As such, it has been a great challenge to identify key neurobiological mechanisms and to develop effective treatments. Current therapies focus on co-morbid conditions (such as epileptic seizures and sleep disturbances) and there is no cure for the core symptoms. Recent studies have increasingly implicated mitochondrial dysfunction in ASD. The fact that mitochondria are an integral part of diverse cellular functions and are susceptible to many insults could explain how a wide range of factors can contribute to a consistent behavioral phenotype in ASD. Meanwhile, the high-fat, low-carbohydrate ketogenic diet (KD), used for nearly a century to treat medically intractable epilepsy, has been shown to enhance mitochondrial function through a multiplicity of mechanisms and affect additional molecular targets that may address symptoms and comorbidities of ASD. Here, we review the evidence for the use of metabolism-based therapies such as the KD in the treatment of ASD as well as emerging co-morbid models of epilepsy and autism. Future research directions aimed at validating such therapeutic approaches and identifying additional and novel mechanistic targets are also discussed.
Collapse
Affiliation(s)
- Ning Cheng
- Departments of Pediatrics, University of CalgaryCalgary, AB, Canada
| | - Jong M. Rho
- Departments of Pediatrics, University of CalgaryCalgary, AB, Canada
- Clinical Neurosciences, University of CalgaryCalgary, AB, Canada
- Physiology and Pharmacology, Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of CalgaryCalgary, AB, Canada
| | - Susan A. Masino
- Neuroscience Program, Department of Psychology, Trinity CollegeHartford, CT, USA
| |
Collapse
|
74
|
Abstract
Ketone body metabolism is a central node in physiological homeostasis. In this review, we discuss how ketones serve discrete fine-tuning metabolic roles that optimize organ and organism performance in varying nutrient states and protect from inflammation and injury in multiple organ systems. Traditionally viewed as metabolic substrates enlisted only in carbohydrate restriction, observations underscore the importance of ketone bodies as vital metabolic and signaling mediators when carbohydrates are abundant. Complementing a repertoire of known therapeutic options for diseases of the nervous system, prospective roles for ketone bodies in cancer have arisen, as have intriguing protective roles in heart and liver, opening therapeutic options in obesity-related and cardiovascular disease. Controversies in ketone metabolism and signaling are discussed to reconcile classical dogma with contemporary observations.
Collapse
Affiliation(s)
- Patrycja Puchalska
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL 32827, USA
| | - Peter A Crawford
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL 32827, USA.
| |
Collapse
|
75
|
Ciarlone SL, Wang X, Rogawski MA, Weeber EJ. Effects of the synthetic neurosteroid ganaxolone on seizure activity and behavioral deficits in an Angelman syndrome mouse model. Neuropharmacology 2016; 116:142-150. [PMID: 27986596 DOI: 10.1016/j.neuropharm.2016.12.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 12/02/2016] [Accepted: 12/12/2016] [Indexed: 11/25/2022]
Abstract
Angelman syndrome (AS) is a rare neurogenetic disorder characterized by severe developmental delay, motor impairments, and epilepsy. GABAergic dysfunction is believed to contribute to many of the phenotypic deficits seen in AS. We hypothesized that restoration of inhibitory tone mediated by extrasynaptic GABAA receptors could provide therapeutic benefit. Here, we report that ganaxolone, a synthetic neurosteroid that acts as a positive allosteric modulator of synaptic and extrasynaptic GABAA receptors, was anxiolytic, anticonvulsant, and improved motor deficits in the Ube3a-deficient mouse model of AS when administered by implanted mini-pump for 3 days or 4 weeks. Treatment for 4 weeks also led to recovery of spatial working memory and hippocampal synaptic plasticity deficits. This study demonstrates that ganaxolone ameliorates many of the behavioral abnormalities in the adult AS mouse, and tolerance did not occur to the therapeutic effects of the drug. The results support clinical studies to investigate ganaxolone as a symptomatic treatment for AS.
Collapse
Affiliation(s)
- Stephanie L Ciarlone
- USF Health Byrd Alzheimer's Institute, Tampa, FL, USA; Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, USA
| | - Xinming Wang
- USF Health Byrd Alzheimer's Institute, Tampa, FL, USA
| | - Michael A Rogawski
- Departments of Neurology and Pharmacology, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Edwin J Weeber
- USF Health Byrd Alzheimer's Institute, Tampa, FL, USA; Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, USA.
| |
Collapse
|
76
|
Ari C, Kovács Z, Juhasz G, Murdun C, Goldhagen CR, Koutnik AP, Poff AM, Kesl SL, D'Agostino DP. Exogenous Ketone Supplements Reduce Anxiety-Related Behavior in Sprague-Dawley and Wistar Albino Glaxo/Rijswijk Rats. Front Mol Neurosci 2016; 9:137. [PMID: 27999529 PMCID: PMC5138218 DOI: 10.3389/fnmol.2016.00137] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 11/22/2016] [Indexed: 12/16/2022] Open
Abstract
Nutritional ketosis has been proven effective for seizure disorders and other neurological disorders. The focus of this study was to determine the effects of ketone supplementation on anxiety-related behavior in Sprague-Dawley (SPD) and Wistar Albino Glaxo/Rijswijk (WAG/Rij) rats. We tested exogenous ketone supplements added to food and fed chronically for 83 days in SPD rats and administered sub-chronically for 7 days in both rat models by daily intragastric gavage bolus followed by assessment of anxiety measures on elevated plus maze (EPM). The groups included standard diet (SD) or SD + ketone supplementation. Low-dose ketone ester (LKE; 1,3-butanediol-acetoacetate diester, ~10 g/kg/day, LKE), high dose ketone ester (HKE; ~25 g/kg/day, HKE), beta-hydroxybutyrate-mineral salt (βHB-S; ~25 g/kg/day, KS) and βHB-S + medium chain triglyceride (MCT; ~25 g/kg/day, KSMCT) were used as ketone supplementation for chronic administration. To extend our results, exogenous ketone supplements were also tested sub-chronically on SPD rats (KE, KS and KSMCT; 5 g/kg/day) and on WAG/Rij rats (KE, KS and KSMCT; 2.5 g/kg/day). At the end of treatments behavioral data collection was conducted manually by a blinded observer and with a video-tracking system, after which blood βHB and glucose levels were measured. Ketone supplementation reduced anxiety on EPM as measured by less entries to closed arms (sub-chronic KE and KS: SPD rats and KSMCT: WAG/Rij rats), more time spent in open arms (sub-chronic KE: SPD and KSMCT: WAG/Rij rats; chronic KSMCT: SPD rats), more distance traveled in open arms (chronic KS and KSMCT: SPD rats) and by delayed latency to entrance to closed arms (chronic KSMCT: SPD rats), when compared to control. Our data indicates that chronic and sub-chronic ketone supplementation not only elevated blood βHB levels in both animal models, but reduced anxiety-related behavior. We conclude that ketone supplementation may represent a promising anxiolytic strategy through a novel means of inducing nutritional ketosis.
Collapse
Affiliation(s)
- Csilla Ari
- Department of Molecular Pharmacology and Physiology, Hyperbaric Biomedical Research Laboratory, Morsani College of Medicine, University of South Florida Tampa, FL, USA
| | - Zsolt Kovács
- Department of Zoology, University of West Hungary Szombathely, Hungary
| | - Gabor Juhasz
- Proteomics Laboratory, Eotvos Lorand University Budapest, Hungary
| | - Cem Murdun
- Department of Molecular Pharmacology and Physiology, Hyperbaric Biomedical Research Laboratory, Morsani College of Medicine, University of South Florida Tampa, FL, USA
| | - Craig R Goldhagen
- Department of Molecular Pharmacology and Physiology, Hyperbaric Biomedical Research Laboratory, Morsani College of Medicine, University of South Florida Tampa, FL, USA
| | - Andrew P Koutnik
- Department of Molecular Pharmacology and Physiology, Hyperbaric Biomedical Research Laboratory, Morsani College of Medicine, University of South Florida Tampa, FL, USA
| | - Angela M Poff
- Department of Molecular Pharmacology and Physiology, Hyperbaric Biomedical Research Laboratory, Morsani College of Medicine, University of South Florida Tampa, FL, USA
| | - Shannon L Kesl
- Department of Molecular Pharmacology and Physiology, Hyperbaric Biomedical Research Laboratory, Morsani College of Medicine, University of South Florida Tampa, FL, USA
| | - Dominic P D'Agostino
- Department of Molecular Pharmacology and Physiology, Hyperbaric Biomedical Research Laboratory, Morsani College of Medicine, University of South Florida Tampa, FL, USA
| |
Collapse
|
77
|
Tefera TW, Tan KN, McDonald TS, Borges K. Alternative Fuels in Epilepsy and Amyotrophic Lateral Sclerosis. Neurochem Res 2016; 42:1610-1620. [PMID: 27868154 DOI: 10.1007/s11064-016-2106-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 11/08/2016] [Accepted: 11/09/2016] [Indexed: 12/12/2022]
Abstract
This review summarises the recent findings on metabolic treatments for epilepsy and Amyotrophic Lateral Sclerosis (ALS) in honour of Professor Ursula Sonnewald. The metabolic impairments in rodent models of these disorders as well as affected patients are being discussed. In both epilepsy and ALS, there are defects in glucose uptake and reduced tricarboxylic acid (TCA) cycling, at least in part due to reduced amounts of C4 TCA cycle intermediates. In addition there are impairments in glycolysis in ALS. A reduction in glucose uptake can be addressed by providing the brain with alternative fuels, such as ketones or medium-chain triglycerides. As anaplerotic fuels, such as the triglyceride of heptanoate, triheptanoin, refill the TCA cycle C4/C5 intermediate pool that is deficient, they are ideal to boost TCA cycling and thus the oxidative metabolism of all fuels.
Collapse
Affiliation(s)
- Tesfaye W Tefera
- Department of Pharmacology, School of Biomedical Sciences, The University of Queensland, Skerman Building 65, St Lucia, QLD, 4072, Australia
| | - Kah Ni Tan
- Department of Pharmacology, School of Biomedical Sciences, The University of Queensland, Skerman Building 65, St Lucia, QLD, 4072, Australia
| | - Tanya S McDonald
- Department of Pharmacology, School of Biomedical Sciences, The University of Queensland, Skerman Building 65, St Lucia, QLD, 4072, Australia
| | - Karin Borges
- Department of Pharmacology, School of Biomedical Sciences, The University of Queensland, Skerman Building 65, St Lucia, QLD, 4072, Australia.
| |
Collapse
|
78
|
Tan WH, Bird LM. Angelman syndrome: Current and emerging therapies in 2016. AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2016; 172:384-401. [PMID: 27860204 DOI: 10.1002/ajmg.c.31536] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Angelman syndrome (AS) is a severe neurodevelopmental disorder caused by a loss of the maternally-inherited UBE3A; the paternal UBE3A is silenced in neurons by a mechanism involving an antisense transcript (UBE3A-AS) at the unmethylated paternal locus. We reviewed all published information on the clinical trials that have been completed as well as the publicly available information on ongoing trials of therapies in AS. To date, all clinical trials that strove to improve neurodevelopment in AS have been unsuccessful. Attempts at hypermethylating the maternal locus through dietary compounds were ineffective. The results of an 8-week open-label trial using minocycline as a matrix metalloproteinase-9 inhibitor were inconclusive, while a subsequent randomized placebo-controlled trial suggested that treatment with minocycline for 8 weeks did not result in any neurodevelopmental gains. A 1-year randomized placebo-controlled trial using levodopa to alter the phosphorylation of calcium/calmodulin-dependent kinase II did not lead to any improvement in neurodevelopment. Topoisomerase inhibitors and antisense oligonucleotides are being developed to directly inhibit UBE3A-AS. Artificial transcription factors are being developed to "super activate" UBE3A or inhibit UBE3A-AS. Other strategies targeting specific pathways are briefly discussed. We also reviewed the medications that are currently used to treat seizures and sleep disturbances, which are two of the more common complications of AS. © 2016 Wiley Periodicals, Inc.
Collapse
|