1
|
Ferraguto C, Piquemal-Lagoueillat M, Lemaire V, Moreau MM, Trazzi S, Uguagliati B, Ciani E, Bertrand SS, Louette E, Bontempi B, Pietropaolo S. Therapeutic efficacy of the BKCa channel opener chlorzoxazone in a mouse model of Fragile X syndrome. Neuropsychopharmacology 2024; 49:2032-2041. [PMID: 39223257 PMCID: PMC11480417 DOI: 10.1038/s41386-024-01956-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 06/30/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024]
Abstract
Fragile X syndrome (FXS) is an X-linked neurodevelopmental disorder characterized by several behavioral abnormalities, including hyperactivity, anxiety, sensory hyper-responsiveness, and autistic-like symptoms such as social deficits. Despite considerable efforts, effective pharmacological treatments are still lacking, prompting the need for exploring the therapeutic value of existing drugs beyond their original approved use. One such repurposed drug is chlorzoxazone which is classified as a large-conductance calcium-dependent potassium (BKCa) channel opener. Reduced BKCa channel functionality has been reported in FXS patients, suggesting that molecules activating these channels could serve as promising treatments for this syndrome. Here, we sought to characterize the therapeutic potential of chlorzoxazone using the Fmr1-KO mouse model of FXS which recapitulates the main phenotypes of FXS, including BKCa channel alterations. Chlorzoxazone, administered either acutely or chronically, rescued hyperactivity and acoustic hyper-responsiveness as well as impaired social interactions exhibited by Fmr1-KO mice. Chlorzoxazone was more efficacious in alleviating these phenotypes than gaboxadol and metformin, two repurposed treatments for FXS that do not target BKCa channels. Systemic administration of chlorzoxazone modulated the neuronal activity-dependent gene c-fos in selected brain areas of Fmr1-KO mice, corrected aberrant hippocampal dendritic spines, and was able to rescue impaired BKCa currents recorded from hippocampal and cortical neurons of these mutants. Collectively, these findings provide further preclinical support for BKCa channels as a valuable therapeutic target for treating FXS and encourage the repurposing of chlorzoxazone for clinical applications in FXS and other related neurodevelopmental diseases.
Collapse
Affiliation(s)
| | | | - Valerie Lemaire
- Univ. Bordeaux, CNRS, EPHE, INCIA, UMR 5287, Bordeaux, France
| | - Maïté M Moreau
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux, France
| | - Stefania Trazzi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Beatrice Uguagliati
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Elisabetta Ciani
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | | | | | - Bruno Bontempi
- Univ. Bordeaux, CNRS, EPHE, INCIA, UMR 5287, Bordeaux, France
| | | |
Collapse
|
2
|
Valencia ML, Sofela FA, Jongens TA, Sehgal A. Do metabolic deficits contribute to sleep disruption in monogenic intellectual disability syndromes? Trends Neurosci 2024; 47:583-592. [PMID: 39054162 DOI: 10.1016/j.tins.2024.06.006] [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: 03/13/2024] [Revised: 05/28/2024] [Accepted: 06/26/2024] [Indexed: 07/27/2024]
Abstract
Intellectual disability is defined as limitations in cognitive and adaptive behavior that often arise during development. Disordered sleep is common in intellectual disability and, given the importance of sleep for cognitive function, it may contribute to other behavioral phenotypes. Animal models of intellectual disability, in particular of monogenic intellectual disability syndromes (MIDS), recapitulate many disease phenotypes and have been invaluable for linking some of these phenotypes to specific molecular pathways. An emerging feature of MIDS, in both animal models and humans, is the prevalence of metabolic abnormalities, which could be relevant for behavior. Focusing on specific MIDS that have been molecularly characterized, we review sleep, circadian, and metabolic phenotypes in animal models and humans and propose that altered metabolic state contributes to the abnormal sleep/circadian phenotypes in MIDS.
Collapse
Affiliation(s)
- Mariela Lopez Valencia
- Chronobiology and Sleep Institute, Perelman Medical School of University of Pennsylvania, Philadelphia, PA, USA
| | - Folasade A Sofela
- Chronobiology and Sleep Institute, Perelman Medical School of University of Pennsylvania, Philadelphia, PA, USA
| | - Thomas A Jongens
- Chronobiology and Sleep Institute, Perelman Medical School of University of Pennsylvania, Philadelphia, PA, USA; Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Autism Spectrum Program of Excellence, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Amita Sehgal
- Chronobiology and Sleep Institute, Perelman Medical School of University of Pennsylvania, Philadelphia, PA, USA; Howard Hughes Medical Institute, Philadelphia, PA, USA.
| |
Collapse
|
3
|
D'Antoni S, Spatuzza M, Bonaccorso CM, Catania MV. Role of fragile X messenger ribonucleoprotein 1 in the pathophysiology of brain disorders: a glia perspective. Neurosci Biobehav Rev 2024; 162:105731. [PMID: 38763180 DOI: 10.1016/j.neubiorev.2024.105731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 05/21/2024]
Abstract
Fragile X messenger ribonucleoprotein 1 (FMRP) is a widely expressed RNA binding protein involved in several steps of mRNA metabolism. Mutations in the FMR1 gene encoding FMRP are responsible for fragile X syndrome (FXS), a leading genetic cause of intellectual disability and autism spectrum disorder, and fragile X-associated tremor-ataxia syndrome (FXTAS), a neurodegenerative disorder in aging men. Although FMRP is mainly expressed in neurons, it is also present in glial cells and its deficiency or altered expression can affect functions of glial cells with implications for the pathophysiology of brain disorders. The present review focuses on recent advances on the role of glial subtypes, astrocytes, oligodendrocytes and microglia, in the pathophysiology of FXS and FXTAS, and describes how the absence or reduced expression of FMRP in these cells can impact on glial and neuronal functions. We will also briefly address the role of FMRP in radial glial cells and its effects on neural development, and gliomas and will speculate on the role of glial FMRP in other brain disorders.
Collapse
Affiliation(s)
- S D'Antoni
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), Via Paolo Gaifami 18, Catania 95126, Italy
| | - M Spatuzza
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), Via Paolo Gaifami 18, Catania 95126, Italy
| | - C M Bonaccorso
- Oasi Research Institute - IRCCS, via Conte Ruggero 73, Troina 94018, Italy
| | - M V Catania
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), Via Paolo Gaifami 18, Catania 95126, Italy.
| |
Collapse
|
4
|
Westmark PR, Lyon G, Gutierrez A, Boeck B, Van Hammond O, Ripp N, Pagan-Torres NA, Brower J, Held PK, Scarlett C, Westmark CJ. Effects of Soy Protein Isolate on Fragile X Phenotypes in Mice. Nutrients 2024; 16:284. [PMID: 38257177 PMCID: PMC10819477 DOI: 10.3390/nu16020284] [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: 11/28/2023] [Revised: 01/08/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
Obesity is a pediatric epidemic that is more prevalent in children with developmental disabilities. We hypothesize that soy protein-based diets increase weight gain and alter neurobehavioral outcomes. Our objective herein was to test matched casein- and soy protein-based purified ingredient diets in a mouse model of fragile X syndrome, Fmr1KO mice. The experimental methods included assessment of growth; 24-7 activity levels; motor coordination; learning and memory; blood-based amino acid, phytoestrogen and glucose levels; and organ weights. The primary outcome measure was body weight. We find increased body weight in male Fmr1KO from postnatal day 6 (P6) to P224, male wild type (WT) from P32-P39, female Fmr1KO from P6-P18 and P168-P224, and female Fmr1HET from P9-P18 as a function of soy. Activity at the beginning of the light and dark cycles increased in female Fmr1HET and Fmr1KO mice fed soy. We did not find significant differences in rotarod or passive avoidance behavior as a function of genotype or diet. Several blood-based amino acids and phytoestrogens were significantly altered in response to soy. Liver weight was increased in WT and adipose tissue in Fmr1KO mice fed soy. Activity levels at the beginning of the light cycle and testes weight were greater in Fmr1KO versus WT males irrespective of diet. DEXA analysis at 8-months-old indicated increased fat mass and total body area in Fmr1KO females and lean mass and bone mineral density in Fmr1KO males fed soy. Overall, dietary consumption of soy protein isolate by C57BL/6J mice caused increased growth, which could be attributed to increased lean mass in males and fat mass in females. There were sex-specific differences with more pronounced effects in Fmr1KO versus WT and in males versus females.
Collapse
Affiliation(s)
- Pamela R. Westmark
- Department of Neurology, University of Wisconsin, Madison, WI 53706, USA;
| | - Greg Lyon
- Undergraduate Research Scholars Program, University of Wisconsin, Madison, WI 53706, USA; (G.L.); (O.V.H.)
| | - Alejandra Gutierrez
- Molecular Environmental Toxicology Master’s Program, University of Wisconsin, Madison, WI 53706, USA;
| | - Brynne Boeck
- Neurology Undergraduate Research, University of Wisconsin, Madison, WI 53706, USA; (B.B.); (N.R.)
| | - Olivia Van Hammond
- Undergraduate Research Scholars Program, University of Wisconsin, Madison, WI 53706, USA; (G.L.); (O.V.H.)
| | - Nathan Ripp
- Neurology Undergraduate Research, University of Wisconsin, Madison, WI 53706, USA; (B.B.); (N.R.)
| | - Nicole Arianne Pagan-Torres
- Molecular Environmental Toxicology Summer Research Opportunities Program, University of Wisconsin, Madison, WI 53706, USA;
| | - James Brower
- Wisconsin State Laboratory of Hygiene, University of Wisconsin, Madison, WI 53706, USA; (J.B.); (P.K.H.)
| | - Patrice K. Held
- Wisconsin State Laboratory of Hygiene, University of Wisconsin, Madison, WI 53706, USA; (J.B.); (P.K.H.)
| | - Cameron Scarlett
- School of Pharmacy, University of Wisconsin, Madison, WI 53706, USA;
| | - Cara J. Westmark
- Department of Neurology and Molecular Environmental Toxicology Center, University of Wisconsin, Madison, WI 53706, USA
| |
Collapse
|
5
|
Cinquina V, Keimpema E, Pollak DD, Harkany T. Adverse effects of gestational ω-3 and ω-6 polyunsaturated fatty acid imbalance on the programming of fetal brain development. J Neuroendocrinol 2023; 35:e13320. [PMID: 37497857 PMCID: PMC10909496 DOI: 10.1111/jne.13320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 05/18/2023] [Accepted: 06/10/2023] [Indexed: 07/28/2023]
Abstract
Obesity is a key medical challenge of our time. The increasing number of children born to overweight or obese women is alarming. During pregnancy, the circulation of the mother and her fetus interact to maintain the uninterrupted availability of essential nutrients for fetal organ development. In doing so, the mother's dietary preference determines the amount and composition of nutrients reaching the fetus. In particular, the availability of polyunsaturated fatty acids (PUFAs), chiefly their ω-3 and ω-6 subclasses, can change when pregnant women choose a specific diet. Here, we provide a succinct overview of PUFA biochemistry, including exchange routes between ω-3 and ω-6 PUFAs, the phenotypes, and probable neurodevelopmental disease associations of offspring born to mothers consuming specific PUFAs, and their mechanistic study in experimental models to typify signaling pathways, transcriptional, and epigenetic mechanisms by which PUFAs can imprint long-lasting modifications to brain structure and function. We emphasize that the ratio, rather than the amount of individual ω-3 or ω-6 PUFAs, might underpin physiologically correct cellular differentiation programs, be these for neurons or glia, during pregnancy. Thereupon, the PUFA-driven programming of the brain is contextualized for childhood obesity, metabolic, and endocrine illnesses.
Collapse
Affiliation(s)
- Valentina Cinquina
- Department of Molecular NeurosciencesCenter for Brain Research, Medical University of ViennaViennaAustria
| | - Erik Keimpema
- Department of Molecular NeurosciencesCenter for Brain Research, Medical University of ViennaViennaAustria
| | - Daniela D. Pollak
- Department of Neurophysiology and NeuropharmacologyCenter for Physiology and Pharmacology, Medical University of ViennaViennaAustria
| | - Tibor Harkany
- Department of Molecular NeurosciencesCenter for Brain Research, Medical University of ViennaViennaAustria
- Deaprtment of NeuroscienceBiomedicum 7D, Karolinska InstitutetStockholmSweden
| |
Collapse
|
6
|
Caires CRS, Bossolani-Martins AL. Which form of environmental enrichment is most effective in rodent models of autism? Behav Processes 2023; 211:104915. [PMID: 37451559 DOI: 10.1016/j.beproc.2023.104915] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 07/10/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Environmental enrichment (EE) is known to produce experience-dependent changes in the brains and behaviors of rodents, and it has therefore been widely used to study neurodevelopmental disorders, including autism. Current studies show significant protocol variation, such as the presence of running wheels, number of cagemates, duration of enrichment, and the age of the animals at the beginning and end of the enrichment interventions. EE has been shown to have prominent positive effects in animal models of idiopathic and syndromic autism, but little is known about the ideal type of EE and the most efficient protocols for reversing autism spectrum disorder (ASD) behaviors modeled in rodents. This review presents evidence that social enrichment is the most effective way to rescue typical behaviors, and that variables such as onset, duration, and type of induction in the ASD model are important for EE success. Understanding which EE protocols are most beneficial for reversing ASD behaviors modeled in rodents opens up possibilities for the potential treatment of neuropsychiatric disorders characterized by behavioral deficits, such as autism.
Collapse
Affiliation(s)
- Cássia Regina Suzuki Caires
- Laboratory of Experimental Physiology, Faculty of Medicine of São Jose do Rio Preto - FAMERP, Av. Brg. Faria Lima, 5416 - Vila São Pedro, São José do Rio Preto, SP, Brazil.
| | - Ana Luiza Bossolani-Martins
- Federal University of Mato Grosso do Sul - UFMS, Av. Pedro Pedrossian, 725 - Universitário, Paranaíba, MS, Brazil.
| |
Collapse
|
7
|
Talvio K, Wagner VA, Minkeviciene R, Kirkwood JS, Kulinich AO, Umemori J, Bhatia A, Hur M, Käkelä R, Ethell IM, Castrén ML. An iPSC-derived astrocyte model of fragile X syndrome exhibits dysregulated cholesterol homeostasis. Commun Biol 2023; 6:789. [PMID: 37516746 PMCID: PMC10387075 DOI: 10.1038/s42003-023-05147-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 07/14/2023] [Indexed: 07/31/2023] Open
Abstract
Cholesterol is an essential membrane structural component and steroid hormone precursor, and is involved in numerous signaling processes. Astrocytes regulate brain cholesterol homeostasis and they supply cholesterol to the needs of neurons. ATP-binding cassette transporter A1 (ABCA1) is the main cholesterol efflux transporter in astrocytes. Here we show dysregulated cholesterol homeostasis in astrocytes generated from human induced pluripotent stem cells (iPSCs) derived from males with fragile X syndrome (FXS), which is the most common cause of inherited intellectual disability. ABCA1 levels are reduced in FXS human and mouse astrocytes when compared with controls. Accumulation of cholesterol associates with increased desmosterol and polyunsaturated phospholipids in the lipidome of FXS mouse astrocytes. Abnormal astrocytic responses to cytokine exposure together with altered anti-inflammatory and cytokine profiles of human FXS astrocyte secretome suggest contribution of inflammatory factors to altered cholesterol homeostasis. Our results demonstrate changes of astrocytic lipid metabolism, which can critically regulate membrane properties and affect cholesterol transport in FXS astrocytes, providing target for therapy in FXS.
Collapse
Affiliation(s)
- Karo Talvio
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Victoria A Wagner
- Division of Biomedical Sciences, and Neuroscience Graduate Program, School of Medicine, University of California Riverside, Riverside, CA, USA
| | - Rimante Minkeviciene
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jay S Kirkwood
- Metabolomics Core Facility, Institute for Integrative Genome Biology, University of California Riverside, Riverside, CA, USA
| | - Anna O Kulinich
- Division of Biomedical Sciences, and Neuroscience Graduate Program, School of Medicine, University of California Riverside, Riverside, CA, USA
| | - Juzoh Umemori
- Gene and Cell Technology, A.I.Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Anil Bhatia
- Metabolomics Core Facility, Institute for Integrative Genome Biology, University of California Riverside, Riverside, CA, USA
| | - Manhoi Hur
- Metabolomics Core Facility, Institute for Integrative Genome Biology, University of California Riverside, Riverside, CA, USA
| | - Reijo Käkelä
- Helsinki University Lipidomics Unit, HiLIPID, Helsinki Institute of Life Science, HiLIFE, Biocenter Finland (Metabolomics), and Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Iryna M Ethell
- Division of Biomedical Sciences, and Neuroscience Graduate Program, School of Medicine, University of California Riverside, Riverside, CA, USA
| | - Maija L Castrén
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
8
|
Premoli M, Fyke W, Bellocchio L, Lemaire V, Wolley-Roberts M, Bontempi B, Pietropaolo S. Early Administration of the Phytocannabinoid Cannabidivarin Prevents the Neurobehavioral Abnormalities Associated with the Fmr1-KO Mouse Model of Fragile X Syndrome. Cells 2023; 12:1927. [PMID: 37566006 PMCID: PMC10416983 DOI: 10.3390/cells12151927] [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: 06/17/2023] [Revised: 07/14/2023] [Accepted: 07/18/2023] [Indexed: 08/12/2023] Open
Abstract
Phytocannabinoids, including the non-addictive cannabis component cannabidivarin (CBDV), have been reported to hold therapeutic potential in several neurodevelopmental disorders (NDDs). Nonetheless, the therapeutic value of phytocannabinoids for treating Fragile X syndrome (FXS), a major NDD, remains unexplored. Here, we characterized the neurobehavioral effects of CBDV at doses of 20 or 100 mg/kg in the Fmr1-knockout (Fmr1-KO) mouse model of FXS using two temporally different intraperitoneal regimens: subchronic 10-day delivery during adulthood (Study 1: rescue treatment) or chronic 5-week delivery at adolescence (Study 2: preventive treatment). Behavioral tests assessing FXS-like abnormalities included anxiety, locomotor, cognitive, social and sensory alterations. Expression of inflammatory and plasticity markers was investigated in the hippocampus and prefrontal cortex. When administered during adulthood (Study 1), the effects of CBDV were marginal, rescuing at the lower dose only the acoustic hyper-responsiveness of Fmr1-KO mice and at both doses their altered hippocampal expression of neurotrophins. When administered during adolescence (Study 2), CBDV at both doses prevented the cognitive, social and acoustic alterations of adult Fmr1-KO mice and modified the expression of several inflammatory brain markers in both wild-type littermates and mutants. These findings warrant the therapeutic potential of CBDV for preventing neurobehavioral alterations associated with FXS, highlighting the relevance of its early administration.
Collapse
Affiliation(s)
- Marika Premoli
- CNRS, EPHE, INCIA, UMR 5287, Univ. Bordeaux, 33000 Bordeaux, France
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - William Fyke
- CNRS, EPHE, INCIA, UMR 5287, Univ. Bordeaux, 33000 Bordeaux, France
- Graduate Program in Neural and Behavioral Science, SUNY Downstate Medical Center, 450 Clarkson Avenue, Brooklyn, NY 11203, USA
| | - Luigi Bellocchio
- INSERM, U1215 NeuroCentre Magendie, Group Endocannabinoids and Neuroadaptation, University of Bordeaux, 33077 Bordeaux, France
| | - Valerie Lemaire
- CNRS, EPHE, INCIA, UMR 5287, Univ. Bordeaux, 33000 Bordeaux, France
| | | | - Bruno Bontempi
- CNRS, EPHE, INCIA, UMR 5287, Univ. Bordeaux, 33000 Bordeaux, France
| | | |
Collapse
|
9
|
Subashi E, Lemaire V, Petroni V, Pietropaolo S. The Impact of Mild Chronic Stress and Maternal Experience in the Fmr1 Mouse Model of Fragile X Syndrome. Int J Mol Sci 2023; 24:11398. [PMID: 37511156 PMCID: PMC10380347 DOI: 10.3390/ijms241411398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/02/2023] [Accepted: 07/09/2023] [Indexed: 07/30/2023] Open
Abstract
Fragile X syndrome (FXS) is a pervasive developmental disorder and the most common monogenic cause of autism spectrum disorder (ASD). Female heterozygous (HET) carriers play a major role in the transmission of the pathology and present several FXS- and ASD-like behavioral alterations. Despite their clear genetic origins, FXS symptoms are known to be modulated by environmental factors, e.g., exposure to chronic stress, especially during critical life periods, such as pregnancy. Pregnancy, together with pups' care, constitutes maternal experience, i.e., another powerful environmental factor affecting several neurobehavioral functions in females. Here we investigated the impact of maternal experience on the long-term effects of stress in Fmr1-HET female mice. Our findings demonstrated that the behavioral abnormalities of HET females, i.e., hyperactivity and memory deficits, were unaffected by stress or maternal experience. In contrast, stress, independently of maternal experience, induced the appearance of cognitive deficits in WT mice. Maternal experience increased anxiety levels in all mice and enhanced their corticosterone levels, concomitantly promoting the effects of stress on social communication and adrenal glands. In translational terms, these results advance our understanding of the environmental modulation of the behavioral alterations observed in FXS female carriers and highlight the long-term impact of maternal experience and its interactions with chronic stress.
Collapse
Affiliation(s)
- Enejda Subashi
- University Bordeaux, CNRS, EPHE, INCIA, UMR 5287, F-33000 Bordeaux, France
| | - Valerie Lemaire
- University Bordeaux, CNRS, EPHE, INCIA, UMR 5287, F-33000 Bordeaux, France
| | - Valeria Petroni
- University Bordeaux, CNRS, EPHE, INCIA, UMR 5287, F-33000 Bordeaux, France
| | | |
Collapse
|
10
|
Turpin V, Schaffhauser M, Thabault M, Aubert A, Joffre C, Balado E, Longueville JE, Francheteau M, Burucoa C, Pichon M, Layé S, Jaber M. Mice prenatally exposed to valproic acid do not show autism-related disorders when fed with polyunsaturated fatty acid-enriched diets. Sci Rep 2023; 13:11235. [PMID: 37433863 DOI: 10.1038/s41598-023-38423-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 07/07/2023] [Indexed: 07/13/2023] Open
Abstract
Dietary supplementations with n-3 polyunsaturated fatty acid (PUFA) have been explored in autism spectrum disorder (ASD) but their efficiency and potential in ameliorating cardinal symptoms of the disease remain elusive. Here, we compared a n-3 long-chain (LC) PUFA dietary supplementation (n-3 supp) obtained from fatty fish with a n-3 PUFA precursor diet (n-3 bal) obtained from plant oils in the valproic acid (VPA, 450 mg/kg at E12.5) ASD mouse model starting from embryonic life, throughout lactation and until adulthood. Maternal and offspring behaviors were investigated as well as several VPA-induced ASD biological features: cerebellar Purkinje cell (PC) number, inflammatory markers, gut microbiota, and peripheral and brain PUFA composition. Developmental milestones were delayed in the n-3 supp group compared to the n-3 bal group in both sexes. Whatever the diet, VPA-exposed offspring did not show ASD characteristic alterations in social behavior, stereotypies, PC number, or gut microbiota dysbiosis while global activity, gait, peripheral and brain PUFA levels as well as cerebellar TNF-alpha levels were differentially altered by diet and treatment according to sex. The current study provides evidence of beneficial effects of n-3 PUFA based diets, including one without LCPUFAs, on preventing several behavioral and cellular symptoms related to ASD.
Collapse
Affiliation(s)
- Valentine Turpin
- Université de Poitiers, Inserm, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, France
| | - Maud Schaffhauser
- Université de Poitiers, Inserm, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, France
| | - Mathieu Thabault
- Université de Poitiers, Inserm, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, France
| | - Agnès Aubert
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeurO, UMR 1286, Bordeaux, France
| | - Corinne Joffre
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeurO, UMR 1286, Bordeaux, France
| | - Eric Balado
- Université de Poitiers, Inserm, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, France
| | - Jean-Emmanuel Longueville
- Université de Poitiers, Inserm, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, France
| | - Maureen Francheteau
- Université de Poitiers, Inserm, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, France
| | - Christophe Burucoa
- Université de Poitiers, Inserm, PHAR2, Poitiers, France
- CHU de Poitiers, Poitiers, France
| | - Maxime Pichon
- Université de Poitiers, Inserm, PHAR2, Poitiers, France
- CHU de Poitiers, Poitiers, France
| | - Sophie Layé
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeurO, UMR 1286, Bordeaux, France
| | - Mohamed Jaber
- Université de Poitiers, Inserm, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, France.
- CHU de Poitiers, Poitiers, France.
| |
Collapse
|
11
|
Alam S, Westmark CJ, McCullagh EA. Diet in treatment of autism spectrum disorders. Front Neurosci 2023; 16:1031016. [PMID: 37492195 PMCID: PMC10364988 DOI: 10.3389/fnins.2022.1031016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 10/31/2022] [Indexed: 07/27/2023] Open
Abstract
Altering the diet to treat disease dates to c. 400 BC when starvation was used to reduce seizures in persons with epilepsy. The current diversity of symptomology and mechanisms underlying autism spectrum disorders (ASDs) and a corresponding lack of disorder-specific effective treatments prompts an evaluation of diet as a therapeutic approach to improve symptoms of ASDs. In this review article, we summarize the main findings of nutritional studies in ASDs, with an emphasis on the most common monogenic cause of autism, Fragile X Syndrome (FXS), and the most studied dietary intervention, the ketogenic diet as well as other dietary interventions. We also discuss the gut microbiota in relation to pre- and probiotic therapies and provide insight into future directions that could aid in understanding the mechanism(s) underlying dietary efficacy.
Collapse
Affiliation(s)
- Sabiha Alam
- Department of Integrative Biology, Oklahoma State University, Stillwater, OK, United States
| | - Cara J. Westmark
- Department of Neurology, University of Wisconsin, Madison, WI, United States
- Molecular and Environmental Toxicology Center, University of Wisconsin, Madison, WI, United States
| | - Elizabeth A. McCullagh
- Department of Integrative Biology, Oklahoma State University, Stillwater, OK, United States
| |
Collapse
|
12
|
Dietary fish oil improves autistic behaviors and gut homeostasis by altering the gut microbial composition in a mouse model of fragile X syndrome. Brain Behav Immun 2023; 110:140-151. [PMID: 36858183 DOI: 10.1016/j.bbi.2023.02.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 02/11/2023] [Accepted: 02/25/2023] [Indexed: 03/03/2023] Open
Abstract
Fragile X syndrome (FXS) is the most common inherited intellectual disability, caused by a lack of the fragile X mental retardation protein (FMRP). Individuals with neurodevelopmental disorders frequently experience gastrointestinal problems that are primarily linked to gut microbial dysbiosis, inflammation, and increased intestinal permeability. Omega-3 polyunsaturated fatty acids (omega-3 PUFAs) are non-pharmacological agents that exert potential therapeutic effects against neurological disorders. However, it is unclear whether omega-3 PUFAs improve autistic behaviors in fragile X syndrome (FXS) by altering the gut microbial composition. Here, we describe gastrointestinal problems in Fmr1 knockout (KO) mice. FMRP deficiency causes intestinal homeostasis dysfunction in mice. Fish oil (FO) as a source of omega-3 PUFAs reduces intestinal inflammation but increases the mRNA and protein levels of TJP3 in the colon of juvenile Fmr1 KO mice. Fecal microbiota transplantation from FO-fed Fmr1 KO mice increased the gut abundance of Akkermansia and Gordonibacter in recipient Fmr1 KO mice and improved gut homeostasis and autistic behaviors. Our findings demonstrate that omega-3 PUFAs improve autistic behaviors and gut homeostasis in FMRP-deficient mice by suppressing gut microbiota dysbiosis, thereby presenting a novel therapeutic approach for juvenile FXS treatment.
Collapse
|
13
|
Abstract
The fragile X-related disorders are an important group of hereditary disorders that are caused by expanded CGG repeats in the 5' untranslated region of the FMR1 gene or by mutations in the coding sequence of this gene. Two categories of pathological CGG repeats are associated with these disorders, full mutation alleles and shorter premutation alleles. Individuals with full mutation alleles develop fragile X syndrome, which causes autism and intellectual disability, whereas those with premutation alleles, which have shorter CGG expansions, can develop fragile X-associated tremor/ataxia syndrome, a progressive neurodegenerative disease. Thus, fragile X-related disorders can manifest as neurodegenerative or neurodevelopmental disorders, depending on the size of the repeat expansion. Here, we review mouse models of fragile X-related disorders and discuss how they have informed our understanding of neurodegenerative and neurodevelopmental disorders. We also assess the translational value of these models for developing rational targeted therapies for intellectual disability and autism disorders.
Collapse
Affiliation(s)
- Rob Willemsen
- Department of Clinical Genetics, Erasmus University Medical Center, 3015 CN Rotterdam, the Netherlands. Department of Medical Genetics, University of Antwerp, 2000 Antwerp, Belgium
| | - R. Frank Kooy
- Department of Clinical Genetics, Erasmus University Medical Center, 3015 CN Rotterdam, the Netherlands. Department of Medical Genetics, University of Antwerp, 2000 Antwerp, Belgium
| |
Collapse
|
14
|
Petroni V, Subashi E, Premoli M, Memo M, Lemaire V, Pietropaolo S. Long-term behavioral effects of prenatal stress in the Fmr1-knock-out mouse model for fragile X syndrome. Front Cell Neurosci 2022; 16:917183. [PMID: 36385949 PMCID: PMC9647640 DOI: 10.3389/fncel.2022.917183] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 10/13/2022] [Indexed: 11/25/2022] Open
Abstract
Fragile X syndrome (FXS) is a major neurodevelopmental disorder and the most common monogenic cause of autism spectrum disorder (ASD). FXS is caused by a mutation in the X-linked FMR1 gene leading to the absence of the FMRP protein, inducing several behavioral deficits, including motor, emotional, cognitive, and social abnormalities. Beside its clear genetic origins, FXS can be modulated by environmental factors, e.g., stress exposure: indeed the behavioral phenotype of FXS, as well as of ASD patients can be exacerbated by the repeated experience of stressful events, especially early in life. Here we investigated the long-term effects of prenatal exposure to unpredictable chronic stress on the behavioral phenotype of the Fmr1-knock-out (KO) mouse model for FXS and ASD. Mice were tested for FXS- and ASD-relevant behaviors first at adulthood (3 months) and then at aging (18 months), in order to assess the persistence and the potential time-related progression of the stress effects. Stress induced the selective emergence of behavioral deficits in Fmr1-KO mice that were evident in spatial memory only at aging. Stress also exerted several age-specific behavioral effects in mice of both genotypes: at adulthood it enhanced anxiety levels and reduced social interaction, while at aging it enhanced locomotor activity and reduced the complexity of ultrasonic calls. Our findings underline the relevance of gene-environment interactions in mouse models of neurodevelopmental syndromes and highlight the long-term behavioral impact of prenatal stress in laboratory mice.
Collapse
Affiliation(s)
- Valeria Petroni
- Univ. Bordeaux, CNRS, INCIA, UMR 5287, F-33000 Bordeaux, France
| | - Enejda Subashi
- Univ. Bordeaux, CNRS, INCIA, UMR 5287, F-33000 Bordeaux, France
| | - Marika Premoli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Maurizio Memo
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Valerie Lemaire
- Univ. Bordeaux, CNRS, INCIA, UMR 5287, F-33000 Bordeaux, France
| | - Susanna Pietropaolo
- Univ. Bordeaux, CNRS, INCIA, UMR 5287, F-33000 Bordeaux, France
- *Correspondence: Susanna Pietropaolo,
| |
Collapse
|
15
|
Abolghasemi A, Carullo MP, Aguilera EC, Laroui A, Plantefeve R, Rojas D, Benachenhou S, Ramírez MV, Proteau-Lemieux M, Lepage JF, Corbin F, Plourde M, Farez M, Cogram P, Çaku A. Alteration of Fatty Acid Profile in Fragile X Syndrome. Int J Mol Sci 2022; 23:ijms231810815. [PMID: 36142726 PMCID: PMC9502195 DOI: 10.3390/ijms231810815] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 08/28/2022] [Accepted: 09/12/2022] [Indexed: 11/16/2022] Open
Abstract
Fragile X Syndrome (FXS) is the most prevalent monogenic cause of Autism Spectrum Disorders (ASDs). Despite a common genetic etiology, the affected individuals display heterogenous metabolic abnormalities including hypocholesterolemia. Although changes in the metabolism of fatty acids (FAs) have been reported in various neuropsychiatric disorders, it has not been explored in humans with FXS. In this study, we investigated the FA profiles of two different groups: (1) an Argentinian group, including FXS individuals and age- and sex-matched controls, and (2) a French-Canadian group, including FXS individuals and their age- and sex-matched controls. Since phospholipid FAs are an indicator of medium-term diet and endogenous metabolism, we quantified the FA profile in plasma phospholipids using gas chromatography. Our results showed significantly lower levels in various plasma FAs including saturated, monosaturated, ω-6 polyunsaturated, and ω-3 polyunsaturated FAs in FXS individuals compared to the controls. A decrease in the EPA/ALA (eicosapentaenoic acid/alpha linoleic acid) ratio and an increase in the DPA/EPA (docosapentaenoic acid/eicosapentaenoic acid) ratio suggest an alteration associated with desaturase and elongase activity, respectively. We conclude that FXS individuals present an abnormal profile of FAs, specifically FAs belonging to the ω-3 family, that might open new avenues of treatment to improve core symptoms of the disorder.
Collapse
Affiliation(s)
- Armita Abolghasemi
- Centre de Recherche du CHUS, Department of Biochemistry, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Maria Paulina Carullo
- Department of Child Neurology, Raúl Carrea Institute for Neurological Research (FLENI), Buenos Aires C1428AQK, Argentina
| | - Ester Cisneros Aguilera
- Centre de Recherche sur le Vieillissement, Departments of Medicine, University of Sherbrooke, Sherbrooke, QC J1H 4C4, Canada
| | - Asma Laroui
- Centre de Recherche du CHUS, Department of Biochemistry, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Rosalie Plantefeve
- Centre de Recherche du CHUS, Department of Biochemistry, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Daniela Rojas
- Centre de Recherche du CHUS, Department of Biochemistry, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Serine Benachenhou
- Centre de Recherche du CHUS, Department of Biochemistry, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - María Victoria Ramírez
- Department of Child Neurology, Raúl Carrea Institute for Neurological Research (FLENI), Buenos Aires C1428AQK, Argentina
| | - Mélodie Proteau-Lemieux
- Department of Pediatrics and Centre de Recherche du CHUS, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Jean-François Lepage
- Department of Pediatrics and Centre de Recherche du CHUS, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - François Corbin
- Centre de Recherche du CHUS, Department of Biochemistry, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Mélanie Plourde
- Centre de Recherche sur le Vieillissement, Departments of Medicine, University of Sherbrooke, Sherbrooke, QC J1H 4C4, Canada
| | - Mauricio Farez
- Department of Child Neurology, Raúl Carrea Institute for Neurological Research (FLENI), Buenos Aires C1428AQK, Argentina
| | - Patricia Cogram
- Biomedicine Division, Centre for Systems Biotechnology, Fraunhofer Chile Research Foundation, Santiago 7500588, Chile
| | - Artuela Çaku
- Centre de Recherche du CHUS, Department of Biochemistry, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
- Correspondence:
| |
Collapse
|
16
|
Premoli M, Petroni V, Bulthuis R, Bonini SA, Pietropaolo S. Ultrasonic Vocalizations in Adult C57BL/6J Mice: The Role of Sex Differences and Repeated Testing. Front Behav Neurosci 2022; 16:883353. [PMID: 35910678 PMCID: PMC9330122 DOI: 10.3389/fnbeh.2022.883353] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 06/20/2022] [Indexed: 11/15/2022] Open
Abstract
Ultrasonic vocalizations (USVs) are a major tool for assessing social communication in laboratory mice during their entire lifespan. At adulthood, male mice preferentially emit USVs toward a female conspecific, while females mostly produce ultrasonic calls when facing an adult intruder of the same sex. Recent studies have developed several sophisticated tools to analyze adult mouse USVs, especially in males, because of the increasing relevance of adult communication for behavioral phenotyping of mouse models of autism spectrum disorder (ASD). Little attention has been instead devoted to adult female USVs and impact of sex differences on the quantitative and qualitative characteristics of mouse USVs. Most of the studies have also focused on a single testing session, often without concomitant assessment of other social behaviors (e.g., sniffing), so little is still known about the link between USVs and other aspects of social interaction and their stability/variations across multiple encounters. Here, we evaluated the USVs emitted by adult male and female mice during 3 repeated encounters with an unfamiliar female, with equal or different pre-testing isolation periods between sexes. We demonstrated clear sex differences in several USVs' characteristics and other social behaviors, and these were mostly stable across the encounters and independent of pre-testing isolation. The estrous cycle of the tested females exerted quantitative effects on their vocal and non-vocal behaviors, although it did not affect the qualitative composition of ultrasonic calls. Our findings obtained in B6 mice, i.e., the strain most widely used for engineering of transgenic mouse lines, contribute to provide new guidelines for assessing ultrasonic communication in male and female adult mice.
Collapse
Affiliation(s)
- Marika Premoli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | | | | | - Sara Anna Bonini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | | |
Collapse
|
17
|
Nolan SO, Hodges SL, Binder MS, Smith GD, Okoh JT, Jefferson TS, Escobar B, Lugo JN. Dietary rescue of adult behavioral deficits in the Fmr1 knockout mouse. PLoS One 2022; 17:e0262916. [PMID: 35089938 PMCID: PMC8797197 DOI: 10.1371/journal.pone.0262916] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 01/09/2022] [Indexed: 11/21/2022] Open
Abstract
The current study aimed to further address important questions regarding the therapeutic efficacy of omega-3 fatty acids for various behavioral and neuroimmune aspects of the Fmr1 phenotype. To address these questions, our experimental design utilized two different omega-3 fatty acid administration timepoints, compared to both standard laboratory chow controls ("Standard") and a diet controlling for the increase in fat content ("Control Fat"). In the first paradigm, post-weaning supplementation (after postnatal day 21) with the omega-3 fatty acid diet ("Omega-3") reversed deficits in startle threshold, but not deficits in prepulse inhibition, and the effect on startle threshold was not specific to the Omega-3 diet. However, post-weaning supplementation with both experimental diets also impaired acquisition of a fear response, recall of the fear memory and contextual fear conditioning compared to the Standard diet. The post-weaning Omega-3 diet reduced hippocampal expression of IL-6 and this reduction of IL-6 was significantly associated with diminished performance in the fear conditioning task. In the perinatal experimental paradigm, the Omega-3 diet attenuated hyperactivity and acquisition of a fear response. Additionally, perinatal exposure to the Control Fat diet (similar to a "Western" diet) further diminished nonsocial anxiety in the Fmr1 knockout. This study provides significant evidence that dietary fatty acids throughout the lifespan can significantly impact the behavioral and neuroimmune phenotype of the Fmr1 knockout model.
Collapse
Affiliation(s)
- Suzanne O. Nolan
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, United States of America
| | - Samantha L. Hodges
- Institute of Biomedical Studies, Baylor University, Waco, Texas, United States of America
| | - Matthew S. Binder
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, United States of America
| | - Gregory D. Smith
- Institute of Biomedical Studies, Baylor University, Waco, Texas, United States of America
| | - James T. Okoh
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, United States of America
| | - Taylor S. Jefferson
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, United States of America
| | - Brianna Escobar
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, United States of America
| | - Joaquin N. Lugo
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, United States of America
- Institute of Biomedical Studies, Baylor University, Waco, Texas, United States of America
| |
Collapse
|
18
|
Parental Reports on Early Autism Behaviors in Their Children with Fragile X Syndrome as a Function of Infant Feeding. Nutrients 2021; 13:nu13082888. [PMID: 34445048 PMCID: PMC8401950 DOI: 10.3390/nu13082888] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 08/13/2021] [Accepted: 08/20/2021] [Indexed: 11/16/2022] Open
Abstract
This study evaluates the prevalence of autistic behaviors in fragile X syndrome as a function of infant diet. Retrospective survey data from the Fragile X Syndrome Nutrition Study, which included data on infant feeding and caregiver-reported developmental milestones for 190 children with fragile X syndrome enrolled in the Fragile X Online Registry with Accessible Database (FORWARD), were analyzed. Exploratory, sex-specific associations were found linking the use of soy-based infant formula with worse autistic behaviors related to language in females and self-injurious behavior in males. These findings prompt prospective evaluation of the effects of soy-based infant formula on disease comorbidities in fragile X syndrome, a rare disorder for which newborn screening could be implemented if there was an intervention. Gastrointestinal problems were the most common reason cited for switching to soy-based infant formula. Thus, these findings also support the study of early gastrointestinal problems in fragile X syndrome, which may underly the development and severity of disease comorbidities. In conjunction with comorbidity data from the previous analyses of the Fragile X Syndrome Nutrition Study, the findings indicate that premutation fragile X mothers should be encouraged to breastfeed.
Collapse
|
19
|
Fyke W, Premoli M, Echeverry Alzate V, López-Moreno JA, Lemaire-Mayo V, Crusio WE, Marsicano G, Wöhr M, Pietropaolo S. Communication and social interaction in the cannabinoid-type 1 receptor null mouse: Implications for autism spectrum disorder. Autism Res 2021; 14:1854-1872. [PMID: 34173729 DOI: 10.1002/aur.2562] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 05/04/2021] [Accepted: 05/28/2021] [Indexed: 12/17/2022]
Abstract
Clinical and preclinical findings have suggested a role of the endocannabinoid system (ECS) in the etiopathology of autism spectrum disorder (ASD). Previous mouse studies have investigated the role of ECS in several behavioral domains; however, none of them has performed an extensive assessment of social and communication behaviors, that is, the main core features of ASD. This study employed a mouse line lacking the primary endocannabinoid receptor (CB1r) and characterized ultrasonic communication and social interaction in CB1-/- , CB1+/- , and CB1+/+ males and females. Quantitative and qualitative alterations in ultrasonic vocalizations (USVs) were observed in CB1 null mice both during early development (i.e., between postnatal days 4 and 10), and at adulthood (i.e., at 3 months of age). Adult mutants also showed marked deficits in social interest in the three-chamber test and social investigation in the direct social interaction test. These behavioral alterations were mostly observed in both sexes and appeared more marked in CB1-/- than CB1+/- mutant mice. Importantly, the adult USV alterations could not be attributed to differences in anxiety or sensorimotor abilities, as assessed by the elevated plus maze and auditory startle tests. Our findings demonstrate the role of CB1r in social communication and behavior, supporting the use of the CB1 full knockout mouse in preclinical research on these ASD-relevant core domains. LAY SUMMARY: The endocannabinoid system (ECS) is important for brain development and neural function and is therefore likely to be involved in neurodevelopmental disorders such as Autism Spectrum Disorder (ASD). Here we investigated changes in social behavior and communication, which are core features of ASD, in male and female mice lacking the chief receptor of this system. Our results show that loss of this receptor results in several changes in social behavior and communication both during early development and in adulthood, thus supporting the role of the ECS in these ASD-core behavioral domains.
Collapse
Affiliation(s)
- William Fyke
- University of Bordeaux, CNRS, EPHE, INCIA, UMR 5287, Bordeaux, France.,Graduate Program in Neural and Behavioral Science, SUNY Downstate Medical Center, Brooklyn, New York, USA
| | - Marika Premoli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Victor Echeverry Alzate
- Department of Psychobiology and Methodology on Behavioral Sciences, Faculty of Psychology, Madrid Complutense University, Spain.,Unidad Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Malaga University, Spain
| | - José A López-Moreno
- Department of Psychobiology and Methodology on Behavioral Sciences, Faculty of Psychology, Madrid Complutense University, Spain
| | | | - Wim E Crusio
- University of Bordeaux, CNRS, EPHE, INCIA, UMR 5287, Bordeaux, France
| | - Giovanni Marsicano
- University of Bordeaux, INSERM, U862 NeuroCentre Magendie, Group Endocannabinoids and Neuroadaptation, Bordeaux, France
| | - Markus Wöhr
- KU Leuven, Faculty of Psychology and Educational Sciences, Research Unit Brain and Cognition, Laboratory of Biological Psychology, Social and Affective Neuroscience Research Group, Leuven, Belgium.,KU Leuven, Leuven Brain Institute, Leuven, Belgium.,Faculty of Psychology, Experimental and Biological Psychology, Behavioral Neuroscience, Philipps-University of Marburg, Marburg, Germany.,Center for Mind, Brain and Behavior, Philipps-University of Marburg, Marburg, Germany
| | | |
Collapse
|
20
|
Martinat M, Rossitto M, Di Miceli M, Layé S. Perinatal Dietary Polyunsaturated Fatty Acids in Brain Development, Role in Neurodevelopmental Disorders. Nutrients 2021; 13:1185. [PMID: 33918517 PMCID: PMC8065891 DOI: 10.3390/nu13041185] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 03/30/2021] [Accepted: 03/30/2021] [Indexed: 12/26/2022] Open
Abstract
n-3 and n-6 polyunsaturated fatty acids (PUFAs) are essential fatty acids that are provided by dietary intake. Growing evidence suggests that n-3 and n-6 PUFAs are paramount for brain functions. They constitute crucial elements of cellular membranes, especially in the brain. They are the precursors of several metabolites with different effects on inflammation and neuron outgrowth. Overall, long-chain PUFAs accumulate in the offspring brain during the embryonic and post-natal periods. In this review, we discuss how they accumulate in the developing brain, considering the maternal dietary supply, the polymorphisms of genes involved in their metabolism, and the differences linked to gender. We also report the mechanisms linking their bioavailability in the developing brain, their transfer from the mother to the embryo through the placenta, and their role in brain development. In addition, data on the potential role of altered bioavailability of long-chain n-3 PUFAs in the etiologies of neurodevelopmental diseases, such as autism, attention deficit and hyperactivity disorder, and schizophrenia, are reviewed.
Collapse
|
21
|
Endocannabinoids and aging-Inflammation, neuroplasticity, mood and pain. VITAMINS AND HORMONES 2021; 115:129-172. [PMID: 33706946 DOI: 10.1016/bs.vh.2020.12.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Aging is associated with changes in hormones, slowing of metabolism, diminished physiological processes, chronic inflammation and high exposure to oxidative stress factors, generally described as the biological cost of living. Lifestyle interventions of diet and exercise can improve the quality of life during aging and lower diet-related chronic disease. The endocannabinoid system (ECS) has important effects on systemic metabolism and physiological systems, including the central and peripheral nervous systems. Exercise can reduce the loss of muscle mass and improve strength, and increase the levels of endocannabinoids (eCB) in brain and blood. Although the ECS exerts controls on multiple systems throughout life it affords benefits to natural aging. The eCB are synthesized from polyunsaturated fatty acids (PUFA) and the primary ones are produced from arachidonic acid (n-6 PUFA) and others from the n-3 PUFA, namely eicosapentaenoic and docosahexaenoic acids. The eCB ligands bind to their receptors, CB1 and CB2, with effects on appetite stimulation, metabolism, immune functions, and brain physiology and neuroplasticity. Dietary families of PUFA are a primary factor that can influence the types and levels of eCB and as a consequence, the downstream actions when the ligands bind to their receptors. Furthermore, the association of eCB with the synthesis of oxylipins (OxL) is a connection between the physiological actions of eCB and the lipid derived immunological OxL mediators of inflammation. OxL are ubiquitous and influence neuroinflammation and inflammatory processes. The emerging actions of eCB on neuroplasticity, well-being and pain are important to aging. Herein, we present information about the ECS and its components, how exercise and diet affects specific eCB, their role in neuroplasticity, neuroinflammation, pain, mood, and relationship to OxL. Poor nutrition status and low nutrient intakes observed with many elderly are reasons to examine the role of dietary PUFA actions on the ECS to improve health.
Collapse
|
22
|
Nolan SO, Hodges SL, Okoh JT, Binder MS, Lugo JN. Prenatal High-Fat Diet Rescues Communication Deficits in Fmr1 Mutant Mice in a Sex-Specific Manner. Dev Neurosci 2021; 42:94-104. [PMID: 33395685 PMCID: PMC7864857 DOI: 10.1159/000509797] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 06/25/2020] [Indexed: 12/22/2022] Open
Abstract
Using high-throughput analysis methods, the present study sought to determine the impact of prenatal high-fat dietary manipulations on isolation-induced ultrasonic vocalization production in both male and female Fmr1mutants on postnatal day 9. Prior to breeding, male FVB/129 Fmr1 wildtype and female Fmr1 heterozygous breeding pairs were assigned to 1 of 3 diet conditions: standard lab chow, omega-3 fatty acid-enriched chow, and a diet controlling for the fat increase. Prenatal exposure to omega-3 fatty acids improved reductions in the number of calls produced by Fmr1heterozygotes females. Moreover, diminished spectral purity in the female Fmr1homozygous mouse was rescued by exposure to both high-fat diets, although these effects were not seen in the male Fmr1knockout. Prenatal dietary fat manipulation also influenced several other aspects of vocalization production, such as the number of calls produced and their fundamental frequency, aside from effects due to loss of Fmr1.Specifically, in males, regardless of genotype, prenatal exposure to high omega-3s increased the average fundamental frequency of calls. These data support the need for future preclinical and clinical work elucidating the full potential of prenatal high-fat diets as a novel therapeutic alternative forFragile X syndrome.
Collapse
Affiliation(s)
- Suzanne O Nolan
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, USA
| | - Samantha L Hodges
- Institute of Biomedical Studies, Baylor University, Waco, Texas, USA
| | - James T Okoh
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, USA
| | - Matthew S Binder
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, USA
| | - Joaquin N Lugo
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, USA,
- Institute of Biomedical Studies, Baylor University, Waco, Texas, USA,
- Department of Biology, Baylor University, Waco, Texas, USA,
| |
Collapse
|
23
|
Schiavi S, Carbone E, Melancia F, Buzzelli V, Manduca A, Campolongo P, Pallottini V, Trezza V. Perinatal supplementation with omega-3 fatty acids corrects the aberrant social and cognitive traits observed in a genetic model of autism based on FMR1 deletion in rats. Nutr Neurosci 2020; 25:898-911. [PMID: 32912100 DOI: 10.1080/1028415x.2020.1819107] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background and objective: Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder for which no treatments exist. Fragile X syndrome (FXS) is the most common form of inherited mental retardation and the most frequent monogenic cause of ASD. Given the lack of pharmacological treatments for ASD, increasing interest is devoted to non-pharmacological approaches, including dietary interventions. Omega-3 polyunsaturated fatty acids (PUFAs) are critical for neurobehavioraldevelopment. This study had two aims: 1. To validatethe recently developed Fmr1-Δexon 8 rat model of FXS; 2. To assess the impact of omega-3 PUFAs dietary supplementation during pregnancy and lactation on the altered behavior displayed by Fmr1-Δexon 8 rats.Methods: Female Fmr1-Δexon 8 and wild-type Sprague-Dawley rats were fed with either an omega-3 PUFAs enriched diet or with an isocaloric control diet during pregnancy and lactation. Behavioral experiments were carried out on the infant (Postnatal days (PNDs) 9 and 13), juvenile (PND 35) and adult (PND 90) male offspring.Results: Fmr1-Δexon 8 pups showed hypolocomotion, reduced ultrasonic vocalizations (USVs) emission and impaired social discrimination compared to wild-type controls. Juvenile and adult Fmr1-Δexon 8 rats showed deficits in the social and cognitive domains, that were counteracted by perinatal omega-3 PUFAs supplementation.Conclusion: Our results support the validity of the Fmr1-Δexon 8 rat model to mimic key autistic-like features and support an important role of omega-3 PUFAs during of neurodevelopment. Although the mechanisms underlying the beneficial effects of omega-3 PUFAs supplementation in ASD needs to be clarified, this dietary intervention holds promise to mitigate core and comorbid autistic features.
Collapse
Affiliation(s)
- Sara Schiavi
- Department of Science, University 'Roma Tre', Rome Italy
| | - Emilia Carbone
- Department of Science, University 'Roma Tre', Rome Italy
| | | | | | | | - Patrizia Campolongo
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy.,Neurobiology of Behavior Laboratory, Santa Lucia Foundation, Rome, Italy
| | | | - Viviana Trezza
- Department of Science, University 'Roma Tre', Rome Italy
| |
Collapse
|
24
|
Inflammation (IL-1β) Modifies the Effect of Vitamin D and Omega-3 Long Chain Polyunsaturated Fatty Acids on Core Symptoms of Autism Spectrum Disorder-An Exploratory Pilot Study ‡. Nutrients 2020; 12:nu12030661. [PMID: 32121236 PMCID: PMC7146497 DOI: 10.3390/nu12030661] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 02/26/2020] [Accepted: 02/27/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The role of vitamin D and omega-3 long chain polyunsaturated fatty acids (omega-3 LCPUFA) in improving core symptoms of autism spectrum disorder (ASD) in children has been investigated by a few randomised controlled trials and the results are mixed and inconclusive. The response to treatment with these nutrients is heterogenous and may be influenced by inflammatory state. As an exploratory analysis, we investigated whether inflammatory state would modulate the effect of these nutrients on core symptoms of ASD. Methods: Seventy-three New Zealand children with ASD (2.5-8.0 years) completed a 12-month randomised, double-blind, placebo-controlled trial of vitamin D (VID, 2000 IU/day), omega-3 LCPUFA; (OM, 722 mg/day docosahexaenoic acid), or both (VIDOM). Non-fasting baseline plasma interleukin-1β (IL-1β) was available for 67 children (VID = 15, OM = 21, VIDOM = 15, placebo = 16). Children were categorised as having undetectable/normal IL-1β (<3.2 pg/ml, n=15) or elevated IL-1β (≥3.2 pg/mL, n = 52). The Social Responsiveness Scale (SRS) questionnaire was used to assess core symptoms of ASD (baseline, 12-month). Mixed model repeated measure analyses (including all children or only children with elevated IL-1β) were used. RESULTS We found evidence for an interaction between baseline IL-1β and treatment response for SRS-total, SRS-social communicative functioning, SRS-awareness and SRS-communication (all Pinteraction < 0.10). When all children were included in the analysis, two outcome comparisons (treatments vs. placebo) showed greater improvements: VID, no effect (all P > 0.10); OM and VIDOM (P = 0.01) for SRS-awareness. When only children with elevated IL-1β were included, five outcomes showed greater improvements: OM (P = 0.01) for SRS-total; OM (P = 0.03) for SRS-social communicative functioning; VID (P = 0.01), OM (P = 0.003) and VIDOM (P = 0.01) for SRS-awareness. CONCLUSION Inflammatory state may have modulated responses to vitamin D and omega-3 LCPUFA intervention in children with ASD, suggesting children with elevated inflammation may benefit more from daily vitamin D and omega-3 LCPUFA supplementation.
Collapse
|
25
|
Hodges SL, Nolan SO, Tomac LA, Muhammad IDA, Binder MS, Taube JH, Lugo JN. Lipopolysaccharide-induced inflammation leads to acute elevations in pro-inflammatory cytokine expression in a mouse model of Fragile X syndrome. Physiol Behav 2019; 215:112776. [PMID: 31838149 DOI: 10.1016/j.physbeh.2019.112776] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/11/2019] [Accepted: 12/11/2019] [Indexed: 02/07/2023]
Abstract
Fragile X syndrome (FXS) is a neurodevelopmental disorder caused by a single genetic mutation in the Fmr1 gene, serving as the largest genetic cause of intellectual disability. Trinucleotide expansion mutations in Fmr1 result in silencing and hypermethylation of the gene, preventing synthesis of the RNA binding protein Fragile X mental retardation protein which functions as a translational repressor. Abnormal immune responses have been demonstrated to play a role in FXS pathophysiology, however, whether these alterations impact how those with FXS respond to an immune insult behaviorally is not entirely known. In the current study, we examine how Fmr1 knockout (KO) and wild type (WT) mice respond to the innate immune stimulus lipopolysaccharide (LPS), both on a molecular and behavioral level, to determine if Fmr1 mutations impact the normal physiological response to an immune insult. In response to LPS, Fmr1 KO mice had elevated hippocampal IL-1β and IL-6 mRNA levels 4 h post-treatment compared to WT mice, with no differences detected in any cytokines at baseline or between genotypes 24 h post-LPS administration. Fmr1 KO mice also had upregulated hippocampal BDNF gene expression 4 h post-treatment compared to WT mice, which was not dependent on LPS administration. There were no differences in hippocampal protein expression between genotypes in microglia (Iba1) or astrocyte (GFAP) reactivity. Further, both genotypes displayed the typical sickness response following LPS stimulation, demonstrated by a significant reduction in food burrowed by LPS-treated mice in a burrowing task. Additional investigation is critical to determine if the transient increases in cytokine expression could lead to long-term changes in downstream molecular signaling in FXS.
Collapse
Affiliation(s)
- Samantha L Hodges
- Institute of Biomedical Studies, Baylor University, Waco, TX 76798, USA
| | - Suzanne O Nolan
- Department of Psychology and Neuroscience, Baylor University, One Bear Place # 97334, Waco, TX 76798, USA
| | - Lindsay A Tomac
- Department of Psychology and Neuroscience, Baylor University, One Bear Place # 97334, Waco, TX 76798, USA
| | - Ilyasah D A Muhammad
- Department of Psychology and Neuroscience, Baylor University, One Bear Place # 97334, Waco, TX 76798, USA
| | - Matthew S Binder
- Department of Psychology and Neuroscience, Baylor University, One Bear Place # 97334, Waco, TX 76798, USA
| | - Joseph H Taube
- Institute of Biomedical Studies, Baylor University, Waco, TX 76798, USA; Department of Biology, Baylor University, Waco, TX 76798, USA
| | - Joaquin N Lugo
- Institute of Biomedical Studies, Baylor University, Waco, TX 76798, USA; Department of Psychology and Neuroscience, Baylor University, One Bear Place # 97334, Waco, TX 76798, USA; Department of Biology, Baylor University, Waco, TX 76798, USA.
| |
Collapse
|
26
|
Joffre C, Rey C, Layé S. N-3 Polyunsaturated Fatty Acids and the Resolution of Neuroinflammation. Front Pharmacol 2019; 10:1022. [PMID: 31607902 PMCID: PMC6755339 DOI: 10.3389/fphar.2019.01022] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 08/12/2019] [Indexed: 12/22/2022] Open
Abstract
In the past few decades, as a result of their anti-inflammatory properties, n-3 long chain polyunsaturated fatty acids (n-3 LC-PUFAs), have gained greater importance in the regulation of inflammation, especially in the central nervous system (in this case known as neuroinflammation). If sustained, neuroinflammation is a common denominator of neurological disorders, including Alzheimer’s disease and major depression, and of aging. Hence, limiting neuroinflammation is a real strategy for neuroinflammatory disease therapy and treatment. Recent data show that n-3 LC-PUFAs exert anti-inflammatory properties in part through the synthesis of specialized pro-resolving mediators (SPMs) such as resolvins, maresins and protectins. These SPMs are crucially involved in the resolution of inflammation. They could be good candidates to resolve brain inflammation and to contribute to neuroprotective functions and could lead to novel therapeutics for brain inflammatory diseases. This review presents an overview 1) of brain n-3 LC-PUFAs as precursors of SPMs with an emphasis on the effect of n-3 PUFAs on neuroinflammation, 2) of the formation and action of SPMs in the brain and their biological roles, and the possible regulation of their synthesis by environmental factors such as inflammation and nutrition and, in particular, PUFA consumption.
Collapse
Affiliation(s)
- Corinne Joffre
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, Bordeaux, France.,Université de Bordeaux 2, Bordeaux, France
| | - Charlotte Rey
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, Bordeaux, France.,Université de Bordeaux 2, Bordeaux, France.,ITERG, Nutrition Health and Lipid Biochemistry Department, Canéjan, France
| | - Sophie Layé
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, Bordeaux, France.,Université de Bordeaux 2, Bordeaux, France
| |
Collapse
|
27
|
Ornoy A, Weinstein-Fudim L, Ergaz Z. Prevention or Amelioration of Autism-Like Symptoms in Animal Models: Will it Bring Us Closer to Treating Human ASD? Int J Mol Sci 2019; 20:ijms20051074. [PMID: 30832249 PMCID: PMC6429371 DOI: 10.3390/ijms20051074] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 02/23/2019] [Indexed: 11/17/2022] Open
Abstract
Since the first animal model of valproic acid (VPA) induced autistic-like behavior, many genetic and non-genetic experimental animal models for Autism Spectrum Disorder (ASD) have been described. The more common non-genetic animal models induce ASD in rats and mice by infection/inflammation or the prenatal or early postnatal administration of VPA. Through the establishment of these models, attempts have been made to ameliorate or even prevent ASD-like symptoms. Some of the genetic models have been successfully treated by genetic manipulations or the manipulation of neurotransmission. Different antioxidants have been used (i.e., astaxanthin, green tea, piperine) to reduce brain oxidative stress in VPA-induced ASD models. Agents affecting brain neurotransmitters (donepezil, agmatine, agomelatine, memantine, oxytocin) also successfully reduced ASD-like symptoms. However, complete prevention of the development of symptoms was achieved only rarely. In our recent study, we treated mouse offspring exposed on postnatal day four to VPA with S-adenosine methionine (SAM) for three days, and prevented ASD-like behavior, brain oxidative stress, and the changes in gene expression induced by VPA. In this review, we describe, in addition to our data, the existing literature on the prevention/amelioration of ASD-like symptoms. We also discuss the possible mechanisms underlying some of these phenomena. Finally, we describe some of the clinical trials in children with ASD that were carried out as a result of data from animal studies, especially those with polyunsaturated fatty acids (PUFAs).
Collapse
Affiliation(s)
- Asher Ornoy
- Laboratory of Teratology, Department of Medical Neurobiology, Hebrew University Hadassah Medical School, Jerusalem 9112001, Israel.
| | - Liza Weinstein-Fudim
- Laboratory of Teratology, Department of Medical Neurobiology, Hebrew University Hadassah Medical School, Jerusalem 9112001, Israel.
| | - Zivanit Ergaz
- Laboratory of Teratology, Department of Medical Neurobiology, Hebrew University Hadassah Medical School, Jerusalem 9112001, Israel.
- Neonatology Department, Hadassah Hebrew University Medical Center, Jerusalem 9112001, Israel.
| |
Collapse
|
28
|
Kosel F, Torres Munoz P, Yang JR, Wong AA, Franklin TB. Age-related changes in social behaviours in the 5xFAD mouse model of Alzheimer's disease. Behav Brain Res 2019; 362:160-172. [PMID: 30659846 DOI: 10.1016/j.bbr.2019.01.029] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 01/09/2019] [Accepted: 01/15/2019] [Indexed: 12/16/2022]
Abstract
In addition to memory impairments, patients with Alzheimer's disease (AD) exhibit a number of behavioural and psychological symptoms that can affect social interactions over the course of the disease. While altered social interactions have been demonstrated in a number of mouse models of AD, many models only recapitulate the initial stages of the disease, and these behavioural changes have yet to be examined over the course of disease progression. By performing a longitudinal study using the 5xFAD mouse model, we have demonstrated that transgenic females exhibit progressive alterations in social investigation compared to wild-type controls. Transgenic females exhibited an age-related reduction in interest for social odours, as well as reduced investigative behaviours towards novel conspecifics in a novel environment. However, transgenic mice exhibited no obvious olfactory deficits, nor any changes in scent-marking behaviour compared to wild-type controls, indicating that changes in investigative behaviour were due to motivation to engage with a social stimulus. This evidence suggests that transgenic 5xFAD females exhibit increased social anxiety in novel environments compared to wild-type controls. Overall, transgenic 5xFAD female mice mimic some features of social withdrawal observed in human AD patients suggesting this strain may be suitable for modelling aspects of the social dysfunction observed in human patients.
Collapse
Affiliation(s)
- Filip Kosel
- The Social Lab, Department of Psychology and Neuroscience, Dalhousie University, Halifax, B3H 4R2, Canada
| | - Paula Torres Munoz
- The Social Lab, Department of Psychology and Neuroscience, Dalhousie University, Halifax, B3H 4R2, Canada
| | - J Renee Yang
- The Social Lab, Department of Psychology and Neuroscience, Dalhousie University, Halifax, B3H 4R2, Canada
| | - Aimee A Wong
- The Social Lab, Department of Psychology and Neuroscience, Dalhousie University, Halifax, B3H 4R2, Canada
| | - Tamara B Franklin
- The Social Lab, Department of Psychology and Neuroscience, Dalhousie University, Halifax, B3H 4R2, Canada.
| |
Collapse
|
29
|
van Elst K, Brouwers JF, Merkens JE, Broekhoven MH, Birtoli B, Helms JB, Kas MJH. Chronic dietary changes in n-6/n-3 polyunsaturated fatty acid ratios cause developmental delay and reduce social interest in mice. Eur Neuropsychopharmacol 2019; 29:16-31. [PMID: 30563719 DOI: 10.1016/j.euroneuro.2018.11.1106] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 10/24/2018] [Accepted: 11/09/2018] [Indexed: 01/21/2023]
Abstract
Polyunsaturated fatty acids (PUFAs) are one of the main cellular building blocks, and dietary changes in PUFA composition are proposed as a potential route to influence brain development. For example, initial studies indicated that there is a relation between blood omega-6(n-6)/omega-3(n-3) PUFA ratios and neurodevelopmental disease diagnosis. To study the consequences of dietary n-6/n-3 PUFA ratio changes, we investigated the impact of a n-3 supplemented and n-3 deficient diet in developing BTBR T + Itpr3tf/J (BTBR) - a mouse inbred strain displaying Autism Spectrum Disorder (ASD)-like symptomatology - and control C57BL/6J mice. This study showed that pre- and postnatal changed dietary n-6/n-3 ratio intake has a major impact on blood and brain PUFA composition, and led to delayed physical development and puberty onset in both strains. The PUFA induced developmental delay did not impact adult cognitive performance, but resulted in reduced social interest, a main ASD behavioral feature. Thus, both chronic dietary n-3 PUFA supplementation and depletion may not be beneficial.
Collapse
Affiliation(s)
- Kim van Elst
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jos F Brouwers
- Department of Biochemistry and Biology, Faculty of Veterinary Medicine, Utrecht University, The Netherlands
| | - Jessica E Merkens
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Mark H Broekhoven
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - J Bernd Helms
- Department of Biochemistry and Biology, Faculty of Veterinary Medicine, Utrecht University, The Netherlands
| | - Martien J H Kas
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands; Groningen Institute for Evolutionary Life Sciences, University of Groningen, The Netherlands.
| |
Collapse
|
30
|
Zemdegs J, Rainer Q, Grossmann CP, Rousseau-Ralliard D, Grynberg A, Ribeiro E, Guiard BP. Anxiolytic- and Antidepressant-Like Effects of Fish Oil-Enriched Diet in Brain-Derived Neurotrophic Factor Deficient Mice. Front Neurosci 2018; 12:974. [PMID: 30622454 PMCID: PMC6308198 DOI: 10.3389/fnins.2018.00974] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 12/05/2018] [Indexed: 12/29/2022] Open
Abstract
Despite significant advances in the understanding of the therapeutic activity of antidepressant drugs, treatment-resistant depression is a public health issue prompting research to identify new therapeutic strategies. Evidence strongly suggests that nutrition might exert a significant impact on the onset, the duration and the severity of major depression. Accordingly, preclinical and clinical investigations demonstrated the beneficial effects of omega-3 fatty acids in anxiety and mood disorders. Although the neurobiological substrates of its action remain poorly documented, basic research has shown that omega-3 increases brain-derived neurotrophic factor (BDNF) levels in brain regions associated with depression, as antidepressant drugs do. In contrast, low BDNF levels and hippocampal atrophy were observed in animal models of depression. In this context, the present study compared the effects of long-lasting fish oil-enriched diet, an important source of omega-3 fatty acids, between heterozygous BDNF+/- mice and their wild-type littermates. Our results demonstrated lower activation of Erk in BDNF+/- mice whereas this deficit was rescued by fish oil-enriched diet. In parallel, BDNF+/- mice displayed elevated hippocampal extracellular 5-HT levels in relation with a local decreased serotonin transporter protein level. Fish oil-enriched diet restored normal serotonergic tone by increasing the protein levels of serotonin transporter. At the cellular level, fish oil-enriched diet increased the pool of immature neurons in the dentate gyrus of BDNF+/- mice and the latter observations coincide with its ability to promote anxiolytic- and antidepressant-like response in these mutants. Collectively, our results demonstrate that the beneficial effects of long-term exposure to fish oil-enriched diet in behavioral paradigms known to recapitulate diverse abnormalities related to the depressive state specifically in mice with a partial loss of BDNF. These findings contrast with the mechanism of action of currently available antidepressant drugs for which the full manifestation of their therapeutic activity depends on the enhancement of serotoninergic and BDNF signaling. Further studies are warranted to determine whether fish oil supplementation could be used as an add-on strategy to conventional pharmacological interventions in treatment-resistant patients and relevant animal models.
Collapse
Affiliation(s)
- Juliane Zemdegs
- Department of Physiology, Discipline of Nutrition Physiology, Universidade Federal de São Paulo, São Paulo, Brazil.,Faculté de Pharmacie, Université Paris Sud, Université Paris-Saclay, Chatenay-Malabry, France.,Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Centre National de la Recherche Scientifique, Université de Toulouse, Toulouse, France
| | - Quentin Rainer
- Faculté de Pharmacie, Université Paris Sud, Université Paris-Saclay, Chatenay-Malabry, France
| | - Cindy P Grossmann
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Centre National de la Recherche Scientifique, Université de Toulouse, Toulouse, France
| | - Delphine Rousseau-Ralliard
- INRA, Unité Mixte de Recherche BDR, ENVA, Université Paris Saclay, Jouy-en-Josas, France.,INRA, Unité Mixte de Recherche 1154, Laboratoire Lipides Membranaires et Régulations Fonctionnelles du Coeur et des Vaisseaux, Jouy-en-Josas, France
| | - Alain Grynberg
- INRA, Unité Mixte de Recherche 1154, Laboratoire Lipides Membranaires et Régulations Fonctionnelles du Coeur et des Vaisseaux, Jouy-en-Josas, France
| | - Eliane Ribeiro
- Department of Physiology, Discipline of Nutrition Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Bruno P Guiard
- Faculté de Pharmacie, Université Paris Sud, Université Paris-Saclay, Chatenay-Malabry, France.,Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Centre National de la Recherche Scientifique, Université de Toulouse, Toulouse, France
| |
Collapse
|
31
|
Sun GY, Simonyi A, Fritsche KL, Chuang DY, Hannink M, Gu Z, Greenlief CM, Yao JK, Lee JC, Beversdorf DQ. Docosahexaenoic acid (DHA): An essential nutrient and a nutraceutical for brain health and diseases. Prostaglandins Leukot Essent Fatty Acids 2018; 136:3-13. [PMID: 28314621 PMCID: PMC9087135 DOI: 10.1016/j.plefa.2017.03.006] [Citation(s) in RCA: 161] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 03/06/2017] [Accepted: 03/09/2017] [Indexed: 01/01/2023]
Abstract
Docosahexaenoic acid (DHA), a polyunsaturated fatty acid (PUFA) enriched in phospholipids in the brain and retina, is known to play multi-functional roles in brain health and diseases. While arachidonic acid (AA) is released from membrane phospholipids by cytosolic phospholipase A2 (cPLA2), DHA is linked to action of the Ca2+-independent iPLA2. DHA undergoes enzymatic conversion by 15-lipoxygenase (Alox 15) to form oxylipins including resolvins and neuroprotectins, which are powerful lipid mediators. DHA can also undergo non-enzymatic conversion by reacting with oxygen free radicals (ROS), which cause the production of 4-hydoxyhexenal (4-HHE), an aldehyde derivative which can form adducts with DNA, proteins and lipids. In studies with both animal models and humans, there is evidence that inadequate intake of maternal n-3 PUFA may lead to aberrant development and function of the central nervous system (CNS). What is less certain is whether consumption of n-3 PUFA is important in maintaining brain health throughout one's life span. Evidence mostly from non-human studies suggests that DHA intake above normal nutritional requirements might modify the risk/course of a number of diseases of the brain. This concept has fueled much of the present interest in DHA research, in particular, in attempts to delineate mechanisms whereby DHA may serve as a nutraceutical and confer neuroprotective effects. Current studies have revealed ability for the oxylipins to regulation of cell redox homeostasis through the Nuclear factor (erythroid-derived 2)-like 2/Antioxidant response element (Nrf2/ARE) anti-oxidant pathway, and impact signaling pathways associated with neurotransmitters, and modulation of neuronal functions involving brain-derived neurotropic factor (BDNF). This review is aimed at describing recent studies elaborating these mechanisms with special regard to aging and Alzheimer's disease, autism spectrum disorder, schizophrenia, traumatic brain injury, and stroke.
Collapse
Affiliation(s)
- Grace Y Sun
- Biochemistry Department, University of Missouri, Columbia, MO, United States
| | - Agnes Simonyi
- Biochemistry Department, University of Missouri, Columbia, MO, United States
| | - Kevin L Fritsche
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | - Dennis Y Chuang
- Department of Neurology, University Hospitals Cleveland Medical Center and Case Western Reserve University, Cleveland, OH, United States
| | - Mark Hannink
- Biochemistry Department, University of Missouri, Columbia, MO, United States
| | - Zezong Gu
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO, United States
| | | | - Jeffrey K Yao
- Medical Research Service, VA Pittsburgh Healthcare System, and Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - James C Lee
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, United States
| | - David Q Beversdorf
- Department of Radiology, Neurology, and Psychological Sciences, and the Thompson Center, William and Nancy Thompson Endowed Chair in Radiology, University of Missouri School of Medicine, Columbia, MO, United States
| |
Collapse
|
32
|
Brief Report: Implementation of a Specific Carbohydrate Diet for a Child with Autism Spectrum Disorder and Fragile X Syndrome. J Autism Dev Disord 2018; 50:1800-1808. [DOI: 10.1007/s10803-018-3704-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
33
|
Adult Fmr1 knockout mice present with deficiencies in hippocampal interleukin-6 and tumor necrosis factor-α expression. Neuroreport 2018; 28:1246-1249. [PMID: 28915148 DOI: 10.1097/wnr.0000000000000905] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Fragile X syndrome (FXS) is a neurodevelopmental disorder caused by a single genetic mutation in the FMR1 gene. Mutations in the FMR1 gene are the largest monogenic cause of autism spectrum disorder (ASD), and thus both disorders share many of the same cognitive and behavioral impairments. There is increasing evidence suggesting that dysregulated immune responses play a role in the pathophysiology of ASD; however, the association between FXS and altered immunity requires further investigation. This study examined whether Fmr1 knockout (KO) and wild-type mice on a FVB/NJ background strain had altered cytokine expression at baseline levels in the hippocampus. Results showed Fmr1 KO mice to have decreased proinflammatory cytokine hippocampal mRNA expression, specifically interleukin (IL)-6 and tumor necrosis factor-α, compared with wild-type mice. However, no differences were detected in the expression levels of IL-1β, MCP-1, interferon-γ, or IL-10. Despite the high comorbidity between FXS and ASD, these results suggest that the Fmr1 KO mouse does not mimic the increased proinflammatory cytokine expression commonly found in ASD mouse models and patients. Further investigation of the immune profile of the Fmr1 KO mouse is critical to understand whether this deficiency of cytokines in the hippocampus is indicative of a broader immunologic deficit associated with FXS.
Collapse
|
34
|
Dahlhaus R. Of Men and Mice: Modeling the Fragile X Syndrome. Front Mol Neurosci 2018; 11:41. [PMID: 29599705 PMCID: PMC5862809 DOI: 10.3389/fnmol.2018.00041] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 01/31/2018] [Indexed: 12/26/2022] Open
Abstract
The Fragile X Syndrome (FXS) is one of the most common forms of inherited intellectual disability in all human societies. Caused by the transcriptional silencing of a single gene, the fragile x mental retardation gene FMR1, FXS is characterized by a variety of symptoms, which range from mental disabilities to autism and epilepsy. More than 20 years ago, a first animal model was described, the Fmr1 knock-out mouse. Several other models have been developed since then, including conditional knock-out mice, knock-out rats, a zebrafish and a drosophila model. Using these model systems, various targets for potential pharmaceutical treatments have been identified and many treatments have been shown to be efficient in preclinical studies. However, all attempts to turn these findings into a therapy for patients have failed thus far. In this review, I will discuss underlying difficulties and address potential alternatives for our future research.
Collapse
Affiliation(s)
- Regina Dahlhaus
- Institute for Biochemistry, Emil-Fischer Centre, University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
35
|
Matsui F, Hecht P, Yoshimoto K, Watanabe Y, Morimoto M, Fritsche K, Will M, Beversdorf D. DHA Mitigates Autistic Behaviors Accompanied by Dopaminergic Change in a Gene/Prenatal Stress Mouse Model. Neuroscience 2017; 371:407-419. [PMID: 29288796 DOI: 10.1016/j.neuroscience.2017.12.029] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 12/18/2017] [Accepted: 12/20/2017] [Indexed: 12/20/2022]
Abstract
Autism Spectrum Disorder (ASD) is characterized by impairments in social interaction, social communication, and repetitive and stereotyped behaviors. Recent work has begun to explore gene × environmental interactions in the etiology of ASD. We previously reported that prenatal stress exposure in stress-susceptible heterozygous serotonin transporter (SERT) KO pregnant dams in a mouse model resulted in autism-like behavior in the offspring (SERT/S mice). The association between prenatal stress and ASD appears to be affected by maternal SERT genotype in clinical populations as well. Using the mouse model, we examined autistic-like behaviors in greater detail, and additionally explored whether diet supplementation with docosahexaenoic acid (DHA) may mitigate the behavioral changes. Only male SERT/S mice showed social impairment and stereotyped behavior, and DHA supplementation ameliorated some of these behaviors. We also measured monoamine levels in the SERT/S mice after three treatment paradigms: DHA-rich diet continuously from breeding (DHA diet), DHA-rich diet only after weaning (CTL/DHA diet) and control diet only (CTL diet). The dopamine (DA) content in the striatum was significantly increased in the SERT/S mice compared with wild-type (WT) mice, whereas no difference was observed with noradrenaline and serotonin content. Moreover, DA content in the striatum was significantly reduced in the SERT/S mice with the DHA-rich diet provided continuously from breeding. The results indicate that autism-associated behaviors and changes in the dopaminergic system in this setting can be mitigated with DHA supplementation.
Collapse
Affiliation(s)
- Fumihiro Matsui
- Department of Radiology, University of Missouri, Columbia, MO, USA; Department of Pediatrics, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Patrick Hecht
- Interdisciplinary Neuroscience Program, University of Missouri, Columbia, MO, USA
| | - Kanji Yoshimoto
- Department of Food Science and Biotechnology, Hiroshima Institute of Technology, Hiroshima, Japan
| | - Yoshihisa Watanabe
- Department of Basic Geriatrics, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masafumi Morimoto
- Department of Pediatrics, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kevin Fritsche
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, USA
| | - Matthew Will
- Department of Psychological Sciences, University of Missouri, Columbia, MO, USA
| | - David Beversdorf
- Department of Radiology, University of Missouri, Columbia, MO, USA; Interdisciplinary Neuroscience Program, University of Missouri, Columbia, MO, USA; Department of Psychological Sciences, University of Missouri, Columbia, MO, USA; Departments of Radiology and Neurology, William and Nancy Thompson Chair in Radiology, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
36
|
Park HJ, Lee HS. Monocyte chemoattractant protein-1 polymorphism interaction with spirulina immunomodulatory effects in healthy Korean elderly: A 16 week, double-blind randomized clinical trial. Nutr Res Pract 2017; 11:290-299. [PMID: 28765775 PMCID: PMC5537538 DOI: 10.4162/nrp.2017.11.4.290] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 03/25/2017] [Accepted: 04/11/2017] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND/OBJECTIVES Spirulina is a known a functional food related to lipid profiles, immune functions, and antioxidant capacity. Circulating monocyte chemoattractant protein-1 (MCP-1) level is associated with inflammation markers. Single nucleotide polymorphism in the MCP-1 promoter region -2518 have been identified and shown to affect gene transcription. Gene variation may also impact functional food supplementary effects. The current study investigated the interaction of MCP-1 -2518 polymorphism with spirulina supplements on anti-inflammatory capacity in Korean elderly. SUBJECTS/METHODS After genotyping, healthy elderly subjects (n = 78) were included in a randomized, double blind, and placebo controlled study. Baseline characteristic, body composition, and dietary intake were measured twice (baseline vs. week 16). For 16 weeks, subjects consumed 8 g either spirulina or placebo daily. Plasma MCP-1, interleukin (IL) -2, IL-6, tumor necrosis factor (TNF)-α, complement (C) 3, immunoglobulin (Ig) G, and Ig A concentrations and lymphocyte proliferation rate (LPR) were analyzed as inflammatory markers. RESULTS In the placebo group with A/A genotype, MCP-1 level was significantly increased, but the spirulina group with A/A genotype was unchanged. IL-2 was significantly increased only in subjects with spirulina supplementation. TNF-α was significantly reduced in subjects with the G carrier. C3 was significantly increased in the placebo group, particularly when A/A increased more than G, but not when spirulina was ingested. LPR was significantly different only in subjects with A/A genotype; there was a significant increase in phytohemagglutinin and lipopolysaccharide induced LPR in the spirulina group. CONCLUSION In healthy Korean elderly, spirulina supplementation may influence different inflammatory markers by the MCP-1 genotype. These results may be useful for customized dietary guidelines to improve immune function in Koreans.
Collapse
Affiliation(s)
- Hee Jung Park
- Department of Foods and Nutrition, Kookmin University, Seoul 02707, Korea
| | - Hyun Sook Lee
- Department of Food Science and Nutrition, Dongseo University, 47 Jurye-ro, Sasang-gu, Busan 47011, Korea
| |
Collapse
|
37
|
Gaudissard J, Ginger M, Premoli M, Memo M, Frick A, Pietropaolo S. Behavioral abnormalities in the Fmr1-KO2 mouse model of fragile X syndrome: The relevance of early life phases. Autism Res 2017; 10:1584-1596. [DOI: 10.1002/aur.1814] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 04/07/2017] [Accepted: 05/02/2017] [Indexed: 11/12/2022]
Affiliation(s)
- Julie Gaudissard
- University of Bordeaux, INCIA; Pessac France
- CNRS, INCIA, UMR 5287; Pessac France
| | - Melanie Ginger
- INSERM, Neurocentre Magendie, U1215; Bordeaux France
- University of Bordeaux, Neurocentre Magendie, U1215; Bordeaux France
| | - Marika Premoli
- Department of Molecular and Translational Medicine; University of Brescia; Brescia Italy
| | - Maurizio Memo
- Department of Molecular and Translational Medicine; University of Brescia; Brescia Italy
| | - Andreas Frick
- INSERM, Neurocentre Magendie, U1215; Bordeaux France
- University of Bordeaux, Neurocentre Magendie, U1215; Bordeaux France
| | - Susanna Pietropaolo
- University of Bordeaux, INCIA; Pessac France
- CNRS, INCIA, UMR 5287; Pessac France
| |
Collapse
|
38
|
Gauducheau M, Lemaire-Mayo V, D'Amato FR, Oddi D, Crusio WE, Pietropaolo S. Age-specific autistic-like behaviors in heterozygous Fmr1-KO female mice. Autism Res 2017; 10:1067-1078. [PMID: 28301083 DOI: 10.1002/aur.1743] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 12/13/2016] [Accepted: 12/21/2016] [Indexed: 12/16/2022]
Abstract
Fragile X syndrome (FXS) is a major developmental disorder and the most frequent monogenic cause of autism. Surprisingly, most existing studies on the Fmr1-KO mouse model for FXS have focused on males, although FX women, who are mostly heterozygous for the Fmr1 mutation, are known to exhibit several behavioral deficits, including autistic-like features. Furthermore, most animal research has been carried out on adults only; so that little is known about the age progression of the behavioral phenotype of Fmr1 mutants, which is a crucial issue to optimize the impact of therapeutic interventions. Here, we performed an extensive analysis of autistic-like social behaviors in heterozygous (HET) Fmr1-KO females and their WT littermates at different ages. No behavioral difference between HET and WT mice was observed at infancy, but some abnormalities in social interaction and communication were first detected at juvenile age. At adulthood some of these alterations disappeared, but avoidance of social novelty appeared, together with other FXS-relevant behavioral deficits, such as hyperactivity and reduced contextual fear response. Our data provide for the first time evidence for the presence of autistic-relevant behavioral abnormalities in Fmr1-HET female mice, demonstrating the utility of this mouse line to model autistic-like behaviors in both sexes. These results also highlight the importance of taking into account age differences when using the Fmr1-KO mouse model, suggesting that the early post-natal phases are the most promising target for preventive interventions and the adult age is the most appropriate to investigate the behavioral impact of potential therapies. Autism Res 2017. © 2017 International Society for Autism Research, Wiley Periodicals, Inc. Autism Res 2017, 10: 1067-1078. © 2017 International Society for Autism Research, Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Manon Gauducheau
- Univ. Bordeaux, INCIA, Pessac cedex, France.,CNRS, INCIA, UMR 5287, Pessac cedex, France
| | - Valerie Lemaire-Mayo
- Univ. Bordeaux, INCIA, Pessac cedex, France.,CNRS, INCIA, UMR 5287, Pessac cedex, France
| | - Francesca R D'Amato
- CNR, Cell Biology and Neurobiology Institute, IRCCS, Santa Lucia Foundation, Rome, Italy
| | - Diego Oddi
- CNR, Cell Biology and Neurobiology Institute, IRCCS, Santa Lucia Foundation, Rome, Italy
| | - Wim E Crusio
- Univ. Bordeaux, INCIA, Pessac cedex, France.,CNRS, INCIA, UMR 5287, Pessac cedex, France
| | - Susanna Pietropaolo
- Univ. Bordeaux, INCIA, Pessac cedex, France.,CNRS, INCIA, UMR 5287, Pessac cedex, France
| |
Collapse
|
39
|
Behavioral effects of chronic stress in the Fmr1 mouse model for fragile X syndrome. Behav Brain Res 2017; 320:128-135. [PMID: 27939692 DOI: 10.1016/j.bbr.2016.11.051] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 11/24/2016] [Accepted: 11/30/2016] [Indexed: 11/21/2022]
Abstract
Fragile X Syndrome (FXS) is a pervasive developmental disorder due to a mutation in the FMR1 X-linked gene. Despite its clear genetic cause, the expression of FXS symptoms is known to be modulated by environmental factors, including stress. Furthermore, several studies have shown disturbances in stress regulatory systems in FXS patients and Fmr1 mice. These studies have mostly focused on the hormonal responses to stress, using the acute exposure to a single type of stressor. Hence, little is known about the behavioral effects of stress in FXS, and the importance of the nature of the stressing procedure, especially in the context of a repeated exposure that more closely resembles real life conditions. Here we evaluated the effects of chronic exposure to different types of stress (i.e., either repeated restraint or unpredictable stress) on the behavioral phenotype of adult Fmr1 mice. Our results demonstrated that chronic stress induced deficits in social interaction and working memory only in WT mice and the impact of stress depended on the type of stressors and the specific behavior tested. Our data suggest that the behavioral sensitivity to stress is dramatically reduced in FXS, opening new views on the impact of gene-environment interactions in this pathology.
Collapse
|
40
|
Neurotrophic Factors in Mouse Models of Autism Spectrum Disorder: Focus on BDNF and IGF-1. TRANSLATIONAL ANATOMY AND CELL BIOLOGY OF AUTISM SPECTRUM DISORDER 2017; 224:121-134. [DOI: 10.1007/978-3-319-52498-6_7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
|
41
|
Shmakova OP. [The clinical dynamics of mental retardation and social adaptation of patients as they get older]. Zh Nevrol Psikhiatr Im S S Korsakova 2016; 116:16-23. [PMID: 27845310 DOI: 10.17116/jnevro201611610116-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
AIM To establish the patterns of social adaptation of patients with mental retardation (MR) as they get older in relation to the clinical dynamics of MR, to analyze the association between the formation of MR and factors and circumstances that determine the adaptive capacity of patients. MATERIAL AND METHODS One hundred and eighty-seven patients with MR were followed-up from childhood to young adulthood (23-25 years old). The average duration of follow-up was 17.7±3.7 years. Clinical-psychopathological method was the main method of research. To assess social functioning of patients, the registration card of social skills in adolescents with chronic mental disorders and the questionnaire for assessment of social functioning and quality of life of mentally ill people were used. RESULTS AND CONCLUSION About 90% of young adults with mild MR got a professional education, but only about 40% had permanent jobs, 35% had temporary income. 80% of patients with moderate mental retardation did not work, less than 10% had temporary jobs. The status of the disabled person received in childhood, has been preserved in most patients. Several early risk factors of predisposition to MR were identified. It has been established that the level of social adaptation of patients is correlated with the severity of intellectual deficit and depends on some external factors.
Collapse
|
42
|
Neuroinflammation in Autism: Plausible Role of Maternal Inflammation, Dietary Omega 3, and Microbiota. Neural Plast 2016; 2016:3597209. [PMID: 27840741 PMCID: PMC5093279 DOI: 10.1155/2016/3597209] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 08/24/2016] [Accepted: 09/27/2016] [Indexed: 02/06/2023] Open
Abstract
Several genetic causes of autism spectrum disorder (ASD) have been identified. However, more recent work has highlighted that certain environmental exposures early in life may also account for some cases of autism. Environmental insults during pregnancy, such as infection or malnutrition, seem to dramatically impact brain development. Maternal viral or bacterial infections have been characterized as disruptors of brain shaping, even if their underlying mechanisms are not yet fully understood. Poor nutritional diversity, as well as nutrient deficiency, is strongly associated with neurodevelopmental disorders in children. For instance, imbalanced levels of essential fatty acids, and especially polyunsaturated fatty acids (PUFAs), are observed in patients with ASD and other neurodevelopmental disorders (e.g., attention deficit hyperactivity disorder (ADHD) and schizophrenia). Interestingly, PUFAs, and specifically n-3 PUFAs, are powerful immunomodulators that exert anti-inflammatory properties. These prenatal dietary and immunologic factors not only impact the fetal brain, but also affect the microbiota. Recent work suggests that the microbiota could be the missing link between environmental insults in prenatal life and future neurodevelopmental disorders. As both nutrition and inflammation can massively affect the microbiota, we discuss here how understanding the crosstalk between these three actors could provide a promising framework to better elucidate ASD etiology.
Collapse
|
43
|
Lozano R, Azarang A, Wilaisakditipakorn T, Hagerman RJ. Fragile X syndrome: A review of clinical management. Intractable Rare Dis Res 2016; 5:145-57. [PMID: 27672537 PMCID: PMC4995426 DOI: 10.5582/irdr.2016.01048] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The fragile X mental retardation 1 gene, which codes for the fragile X mental retardation 1 protein, usually has 5 to 40 CGG repeats in the 5' untranslated promoter. The full mutation is the almost always the cause of fragile X syndrome (FXS). The prevalence of FXS is about 1 in 4,000 to 1 in 7,000 in the general population although the prevalence varies in different regions of the world. FXS is the most common inherited cause of intellectual disability and autism. The understanding of the neurobiology of FXS has led to many targeted treatments, but none have cured this disorder. The treatment of the medical problems and associated behaviors remain the most useful intervention for children with FXS. In this review, we focus on the non-pharmacological and pharmacological management of medical and behavioral problems associated with FXS as well as current recommendations for follow-up and surveillance.
Collapse
Affiliation(s)
- Reymundo Lozano
- Medical Investigation of Neurodevelopmental Disorders MIND Institute, UC Davis, CA, USA
- Department of Pediatrics, UC Davis, Sacramento, CA, USA
- Address correspondence to: Dr. Reymundo Lozano, Medical Investigation of Neurodevelopmental Disorders MIND Institute, UC Davis, CA, USA; Department of Pediatrics, UC Davis, Sacramento, CA, USA. E-mail:
| | - Atoosa Azarang
- Medical Investigation of Neurodevelopmental Disorders MIND Institute, UC Davis, CA, USA
- Department of Pediatrics, UC Davis, Sacramento, CA, USA
| | - Tanaporn Wilaisakditipakorn
- Medical Investigation of Neurodevelopmental Disorders MIND Institute, UC Davis, CA, USA
- Department of Pediatrics, UC Davis, Sacramento, CA, USA
| | - Randi J Hagerman
- Medical Investigation of Neurodevelopmental Disorders MIND Institute, UC Davis, CA, USA
- Department of Pediatrics, UC Davis, Sacramento, CA, USA
| |
Collapse
|
44
|
Nutritional Omega-3 Deficiency Alters Glucocorticoid Receptor-Signaling Pathway and Neuronal Morphology in Regionally Distinct Brain Structures Associated with Emotional Deficits. Neural Plast 2015; 2016:8574830. [PMID: 27057368 PMCID: PMC4710953 DOI: 10.1155/2016/8574830] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 10/07/2015] [Accepted: 10/19/2015] [Indexed: 12/31/2022] Open
Abstract
Extensive evidence suggests that long term dietary n-3 polyunsaturated fatty acids (PUFAs) deficiency results in altered emotional behaviour. We have recently demonstrated that n-3 PUFAs deficiency induces emotional alterations through abnormal corticosterone secretion which leads to altered dendritic arborisation in the prefrontal cortex (PFC). Here we show that hypothalamic-pituitary-adrenal (HPA) axis feedback inhibition was not compromised in n-3 deficient mice. Rather, glucocorticoid receptor (GR) signaling pathway was inactivated in the PFC but not in the hippocampus of n-3 deficient mice. Consequently, only dendritic arborisation in PFC was affected by dietary n-3 PUFAs deficiency. In addition, occlusion experiment with GR blockade altered GR signaling in the PFC of control mice, with no further alterations in n-3 deficient mice. In conclusion, n-3 PUFAs deficiency compromised PFC, leading to dendritic atrophy, but did not change hippocampal GR function and dendritic arborisation. We argue that this GR sensitivity contributes to n-3 PUFAs deficiency-related emotional behaviour deficits.
Collapse
|
45
|
Essential role of docosahexaenoic acid towards development of a smarter brain. Neurochem Int 2015; 89:51-62. [DOI: 10.1016/j.neuint.2015.08.014] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 08/18/2015] [Accepted: 08/26/2015] [Indexed: 01/25/2023]
|
46
|
Abstract
PURPOSE OF REVIEW This work reviews recent research regarding treatment of fragile X syndrome (FXS), the most common inherited cause of intellectual disability and autism spectrum disorder. The phenotype includes anxiety linked to sensory hyperarousal, hyperactivity, and attentional problems consistent with attention deficit hyperactivity disorder and social deficits leading to autism spectrum disorder in 60% of boys and 25% of girls with FXS. RECENT FINDINGS Multiple targeted treatments for FXS have rescued the phenotype of the fmr1 knockout mouse, but few have been beneficial to patients with FXS. The failure of the metabotropic glutamate receptor 5 antagonists falls on the heels of the failure of Arbaclofen's efficacy in children and adults with autism or FXS. In contrast, efficacy has been demonstrated in a controlled trial of minocycline in children with FXS. Minocycline lowers the abnormally elevated levels of matrix metalloproteinase 9 in FXS. Acamprosate and lovastatin have been beneficial in open-label trials in FXS. The first 5 years of life may be the most efficacious time for intervention when combined with behavioral and/or educational interventions. SUMMARY Minocycline, acamprosate, lovastatin, and sertraline are treatments that can be currently prescribed and have shown benefit in children with FXS. Use of combined medical and behavioral interventions will likely be most efficacious for the treatment of FXS.
Collapse
Affiliation(s)
- Randi J. Hagerman
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis Health System, Sacramento, California, USA
- Department of Pediatrics, University of California Davis Health System, Sacramento, California, USA
| | - Jonathan Polussa
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis Health System, Sacramento, California, USA
- Department of Pediatrics, University of California Davis Health System, Sacramento, California, USA
| |
Collapse
|
47
|
Naviaux JC, Wang L, Li K, Bright AT, Alaynick WA, Williams KR, Powell SB, Naviaux RK. Antipurinergic therapy corrects the autism-like features in the Fragile X (Fmr1 knockout) mouse model. Mol Autism 2015; 6:1. [PMID: 25705365 PMCID: PMC4334917 DOI: 10.1186/2040-2392-6-1] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 12/16/2014] [Indexed: 02/07/2023] Open
Abstract
Background This study was designed to test a new approach to drug treatment of autism spectrum disorders (ASDs) in the Fragile X (Fmr1) knockout mouse model. Methods We used behavioral analysis, mass spectrometry, metabolomics, electron microscopy, and western analysis to test the hypothesis that the disturbances in social behavior, novelty preference, metabolism, and synapse structure are treatable with antipurinergic therapy (APT). Results Weekly treatment with the purinergic antagonist suramin (20 mg/kg intraperitoneally), started at 9 weeks of age, restored normal social behavior, and improved metabolism, and brain synaptosomal structure. Abnormalities in synaptosomal glutamate, endocannabinoid, purinergic, and IP3 receptor expression, complement C1q, TDP43, and amyloid β precursor protein (APP) were corrected. Comprehensive metabolomic analysis identified 20 biochemical pathways associated with symptom improvements. Seventeen pathways were shared with human ASD, and 11 were shared with the maternal immune activation (MIA) model of ASD. These metabolic pathways were previously identified as functionally related mediators of the evolutionarily conserved cell danger response (CDR). Conclusions The data show that antipurinergic therapy improves the multisystem, ASD-like features of both the environmental MIA, and the genetic Fragile X models. These abnormalities appeared to be traceable to mitochondria and regulated by purinergic signaling. Electronic supplementary material The online version of this article (doi:10.1186/2040-2392-6-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jane C Naviaux
- Department of Psychiatry, University of California, San Diego School of Medicine, 214 Dickinson St., Bldg CTF, Rm C102, San Diego, CA 92103-8467 USA
| | - Lin Wang
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, 214 Dickinson St., Bldg CTF, Rm C102, San Diego, CA 92103-8467 USA ; Department of Medicine, University of California, San Diego School of Medicine, 214 Dickinson St., Bldg CTF, Rm C102, San Diego, CA 92103-8467 USA
| | - Kefeng Li
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, 214 Dickinson St., Bldg CTF, Rm C102, San Diego, CA 92103-8467 USA ; Department of Medicine, University of California, San Diego School of Medicine, 214 Dickinson St., Bldg CTF, Rm C102, San Diego, CA 92103-8467 USA
| | - A Taylor Bright
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, 214 Dickinson St., Bldg CTF, Rm C102, San Diego, CA 92103-8467 USA ; Department of Medicine, University of California, San Diego School of Medicine, 214 Dickinson St., Bldg CTF, Rm C102, San Diego, CA 92103-8467 USA
| | - William A Alaynick
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, 214 Dickinson St., Bldg CTF, Rm C102, San Diego, CA 92103-8467 USA ; Department of Medicine, University of California, San Diego School of Medicine, 214 Dickinson St., Bldg CTF, Rm C102, San Diego, CA 92103-8467 USA
| | - Kenneth R Williams
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, 214 Dickinson St., Bldg CTF, Rm C102, San Diego, CA 92103-8467 USA ; Department of Medicine, University of California, San Diego School of Medicine, 214 Dickinson St., Bldg CTF, Rm C102, San Diego, CA 92103-8467 USA ; General Atomics, Inc, San Diego, CA USA
| | - Susan B Powell
- Department of Psychiatry, University of California, San Diego School of Medicine, 214 Dickinson St., Bldg CTF, Rm C102, San Diego, CA 92103-8467 USA ; Research Service, VA San Diego Healthcare System, La Jolla, CA USA
| | - Robert K Naviaux
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, 214 Dickinson St., Bldg CTF, Rm C102, San Diego, CA 92103-8467 USA ; Department of Medicine, University of California, San Diego School of Medicine, 214 Dickinson St., Bldg CTF, Rm C102, San Diego, CA 92103-8467 USA ; Department of Pediatrics, University of California, San Diego School of Medicine, 214 Dickinson St., Bldg CTF, Rm C102, San Diego, CA 92103-8467 USA ; Department of Pathology, University of California, San Diego School of Medicine, 214 Dickinson St., Bldg CTF, Rm C102, San Diego, CA 92103-8467 USA ; Veterans Affairs Center for Excellence in Stress and Mental Health (CESAMH), La Jolla, CA USA
| |
Collapse
|
48
|
Pietropaolo S, Crusio WE, D'amato FR. Treatment Approaches in Rodent Models for Autism Spectrum Disorder. Curr Top Behav Neurosci 2015; 30:325-340. [PMID: 26857461 DOI: 10.1007/7854_2015_433] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Recent years have seen an impressive amount of research devoted to the developing of therapies to treat autism spectrum disorder (ASD). This work has been largely based on rodent models, employing a multitude of genetic and environmental manipulations. Unfortunately, the task of identifying suitable treatments for ASD is extremely challenging, due to a variety of problems specific to the research in this field. Here, we first discuss these problems, including (I) the presence of a large variety of rodent models (often without universal consensus on their validity), (II) the difficulties in choosing the most appropriate behavioural markers to assess the efficacy of possible treatments, (III) the limited knowledge we still have of the neurobiological bases of ASD pathology and of its aetiology, and (IV) the complexity of ASD itself, including a highly heterogeneous group of disorders sometimes with markedly different symptoms (therefore unlikely to be treated with the same approaches). Second, we give a critical overview of the most relevant advances in designing treatments for ASD, focusing on the most commonly used animal model, the laboratory mouse. We include pharmacological and non-pharmacological approaches, underlining their specific advantages, but also their current limitations especially in relation to the problems discussed before. Finally, we highlight the theoretical (e.g. the combination of multiple rather than single treatments) and methodological (e.g. use of single-gene mouse models) approaches that seem more promising to us, suggesting various strategies that can be adopted to simplify the complex field of research on treatments for ASD.
Collapse
Affiliation(s)
- Susanna Pietropaolo
- INCIA, University of Bordeaux, Bat B2, Allée Geoffroy St. Hilaire, CS 50023, 33615, Pessac Cedex, France. .,INCIA, UMR 5287, CNRS, Bat B2, Allée Geoffroy St. Hilaire, CS 50023, 33615, Pessac Cedex, France.
| | - Wim E Crusio
- INCIA, University of Bordeaux, Bat B2, Allée Geoffroy St. Hilaire, CS 50023, 33615, Pessac Cedex, France.,INCIA, UMR 5287, CNRS, Bat B2, Allée Geoffroy St. Hilaire, CS 50023, 33615, Pessac Cedex, France
| | - Francesca R D'amato
- CNR, Cell Biology and Neurobiology Institute, IRCCS, Santa Lucia Foundation, Via del Fosso di Fiorano 64, 00143, Rome, Italy.,Department of Psychiatry and Neurosciences, Laval University, Québec City, Canada
| |
Collapse
|