51
|
Pacheva I, Ivanov I. Targeted Biomedical Treatment for Autism Spectrum Disorders. Curr Pharm Des 2020; 25:4430-4453. [PMID: 31801452 DOI: 10.2174/1381612825666191205091312] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 12/02/2019] [Indexed: 12/29/2022]
Abstract
BACKGROUND A diagnosis of autism spectrum disorders (ASD) represents presentations with impairment in communication and behaviour that vary considerably in their clinical manifestations and etiology as well as in their likely pathophysiology. A growing body of data indicates that the deleterious effect of oxidative stress, mitochondrial dysfunction, immune dysregulation and neuroinflammation, as well as their interconnections are important aspects of the pathophysiology of ASD. Glutathione deficiency decreases the mitochondrial protection against oxidants and tumor necrosis factor (TNF)-α; immune dysregulation and inflammation inhibit mitochondrial function through TNF-α; autoantibodies against the folate receptors underpin cerebral folate deficiency, resulting in disturbed methylation, and mitochondrial dysfunction. Such pathophysiological processes can arise from environmental and epigenetic factors as well as their combined interactions, such as environmental toxicant exposures in individuals with (epi)genetically impaired detoxification. The emerging evidence on biochemical alterations in ASD is forming the basis for treatments aimed to target its biological underpinnings, which is of some importance, given the uncertain and slow effects of the various educational interventions most commonly used. METHODS Literature-based review of the biomedical treatment options for ASD that are derived from established pathophysiological processes. RESULTS Most proposed biomedical treatments show significant clinical utility only in ASD subgroups, with specified pre-treatment biomarkers that are ameliorated by the specified treatment. For example, folinic acid supplementation has positive effects in ASD patients with identified folate receptor autoantibodies, whilst the clinical utility of methylcobalamine is apparent in ASD patients with impaired methylation capacity. Mitochondrial modulating cofactors should be considered when mitochondrial dysfunction is evident, although further research is required to identify the most appropriate single or combined treatment. Multivitamins/multiminerals formulas, as well as biotin, seem appropriate following the identification of metabolic abnormalities, with doses tapered to individual requirements. A promising area, requiring further investigations, is the utilization of antipurinergic therapies, such as low dose suramin. CONCLUSION The assessment and identification of relevant physiological alterations and targeted intervention are more likely to produce positive treatment outcomes. As such, current evidence indicates the utility of an approach based on personalized and evidence-based medicine, rather than treatment targeted to all that may not always be beneficial (primum non nocere).
Collapse
Affiliation(s)
- Iliyana Pacheva
- Department of Pediatrics and Medical Genetics, Medical University - Plovdiv, Plovdiv 4002, Bulgaria
| | - Ivan Ivanov
- Department of Pediatrics and Medical Genetics, Medical University - Plovdiv, Plovdiv 4002, Bulgaria
| |
Collapse
|
52
|
Maternal Immune Activation in Mice Only Partially Recapitulates the Autism Spectrum Disorders Symptomatology. Neuroscience 2020; 445:109-119. [PMID: 32445939 DOI: 10.1016/j.neuroscience.2020.05.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 04/03/2020] [Accepted: 05/05/2020] [Indexed: 02/06/2023]
Abstract
Prenatal viral/bacterial infections are considered risk factors for autism spectrum disorders (ASD) and rodent models of maternal immune activation (MIA) have been developed and extensively used in preclinical studies. Poly inosinic-cytidylic acid (Poly I:C) was injected in C57BL6/J dams to mimic a viral infection on gestational day 12.5; the experimental design includes 10/12 litters in each treatment group and data were analysed always considering the litter-effect; neonatal (spontaneous motor behaviour and ultrasonic vocalizations) and adult [open field, marble burying, social approach, fear conditioning, prepulse inhibition (PPI)] offspring of both sexes were tested. In vivo magnetic resonance imaging/spectroscopy (MRI-MRS) and high-performance liquid chromatography (HPLC) to quantify both aminoacid and/or neurotransmitter concentration in cortical and striatal regions were also carried out. In both sexes high levels of repetitive motor responses and sensory gating deficits in PPI were the more striking effects of Poly I:C, whereas no alteration of social responses were evidenced. Poly I:C treatment did not affect mean values, but, intriguingly, increased variability in the levels of four aminoacids (aspartate glycine and GABA) selectively in males. As a whole prenatal Poly I:C induced relevant long-term alterations in explorative-stereotyped motor responses and in sensory gating, sparing cognitive and social competences. When systematically assessing differences between male and female siblings within each litter, no significant sex differences were evident except for increased variability of four aminoacid levels in male brains. As a whole, prenatal Poly I:C paradigms appear to be a useful tool to investigate the profound and translationally-relevant effects of developmental immune activation on brain and behavioural development, not necessarily recapitulating the full ASD symptomatology.
Collapse
|
53
|
Haddad FL, Patel SV, Schmid S. Maternal Immune Activation by Poly I:C as a preclinical Model for Neurodevelopmental Disorders: A focus on Autism and Schizophrenia. Neurosci Biobehav Rev 2020; 113:546-567. [PMID: 32320814 DOI: 10.1016/j.neubiorev.2020.04.012] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 01/28/2020] [Accepted: 04/09/2020] [Indexed: 12/18/2022]
Abstract
Maternal immune activation (MIA) in response to a viral infection during early and mid-gestation has been linked through various epidemiological studies to a higher risk for the child to develop autism or schizophrenia-related symptoms.. This has led to the establishment of the pathogen-free poly I:C-induced MIA animal model for neurodevelopmental disorders, which shows relatively high construct and face validity. Depending on the experimental variables, particularly the timing of poly I:C administration, different behavioural and molecular phenotypes have been described that relate to specific symptoms of neurodevelopmental disorders such as autism spectrum disorder and/or schizophrenia. We here review and summarize epidemiological evidence for the effects of maternal infection and immune activation, as well as major findings in different poly I:C MIA models with a focus on poly I:C exposure timing, behavioural and molecular changes in the offspring, and characteristics of the model that relate it to autism spectrum disorder and schizophrenia.
Collapse
Affiliation(s)
- Faraj L Haddad
- Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada.
| | - Salonee V Patel
- Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada.
| | - Susanne Schmid
- Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada.
| |
Collapse
|
54
|
Bauman MD, Van de Water J. Translational opportunities in the prenatal immune environment: Promises and limitations of the maternal immune activation model. Neurobiol Dis 2020; 141:104864. [PMID: 32278881 DOI: 10.1016/j.nbd.2020.104864] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 03/03/2020] [Accepted: 04/02/2020] [Indexed: 12/15/2022] Open
Abstract
The prenatal environment, and in particular, the maternal-fetal immune environment, has emerged as a targeted area of research for central nervous system (CNS) diseases with neurodevelopmental origins. Converging evidence from both clinical and preclinical research indicates that changes in the maternal gestational immune environment can alter fetal brain development and increase the risk for certain neurodevelopmental disorders. Here we focus on the translational potential of one prenatal animal model - the maternal immune activation (MIA) model. This model stems from the observation that a subset of pregnant women who are exposed to infection during pregnancy have an increased risk of giving birth to a child who will later be diagnosed with a neurodevelopmental disorder, such as autism spectrum disorder (ASD) or schizophrenia (SZ). The preclinical MIA model provides a system in which to explore causal relationships, identify underlying neurobiological mechanisms, and, ultimately, develop novel therapeutic interventions and preventative strategies. In this review, we will highlight converging evidence from clinical and preclinical research that links changes in the maternal-fetal immune environment with lasting changes in offspring brain and behavioral development. We will then explore the promises and limitations of the MIA model as a translational tool to develop novel therapeutic interventions. As the translational potential of the MIA model has been the focus of several excellent review articles, here we will focus on what is perhaps the least well developed area of MIA model research - novel preventative strategies and therapeutic interventions.
Collapse
Affiliation(s)
- Melissa D Bauman
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, United States of America; California National Primate Research Center, University of California, Davis, United States of America; The MIND Institute, University of California, Davis, United States of America.
| | - Judy Van de Water
- The MIND Institute, University of California, Davis, United States of America; Rheumatology/Allergy and Clinical Immunology, University of California, Davis, United States of America
| |
Collapse
|
55
|
Abstract
While there are numerous medical comorbidities associated with ASD, gastrointestinal (GI) issues have a significant impact on quality of life for these individuals. Recent findings continue to support the relationship between the gut microbiome and both GI symptoms and behavior, but the heterogeneity within the autism spectrum requires in-depth clinical characterization of these clinical cohorts. Large, diverse, well-controlled studies in this area of research are still needed. Although there is still much to discover about the brain-gut-microbiome axis in ASD, microbially mediated therapies, specifically probiotics and fecal microbiota transplantation have shown promise in the treatment of GI symptoms in ASD, with potential benefit to the core behavioral symptoms of ASD as well. Future research and clinical trials must increasingly consider complex phenotypes in ASD in stratification of large datasets as well as in design of inclusion criteria for individual therapeutic interventions.
Collapse
Affiliation(s)
- Virginia Saurman
- Department of Pediatrics, Columbia University Medical Center, 620 West 168th Street, New York, NY 10032, USA
| | - Kara G. Margolis
- Department of Pediatrics, Columbia University Medical Center, 620 West 168th Street, New York, NY 10032, USA
| | - Ruth Ann Luna
- Department of Pathology and Immunology, Texas Children’s Microbiome Center, Baylor College of Medicine, Texas Children’s Hospital, Feigin Tower, 1102 Bates Avenue, Suite 955, Houston, TX 77030, USA
| |
Collapse
|
56
|
Abstract
Suramin is 100 years old and is still being used to treat the first stage of acute human sleeping sickness, caused by Trypanosoma brucei rhodesiense Suramin is a multifunctional molecule with a wide array of potential applications, from parasitic and viral diseases to cancer, snakebite, and autism. Suramin is also an enigmatic molecule: What are its targets? How does it get into cells in the first place? Here, we provide an overview of the many different candidate targets of suramin and discuss its modes of action and routes of cellular uptake. We reason that, once the polypharmacology of suramin is understood at the molecular level, new, more specific, and less toxic molecules can be identified for the numerous potential applications of suramin.
Collapse
|
57
|
Matsuo K, Yabuki Y, Fukunaga K. 5-aminolevulinic acid inhibits oxidative stress and ameliorates autistic-like behaviors in prenatal valproic acid-exposed rats. Neuropharmacology 2020; 168:107975. [PMID: 31991146 DOI: 10.1016/j.neuropharm.2020.107975] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 01/22/2020] [Accepted: 01/23/2020] [Indexed: 02/07/2023]
Abstract
Autism spectrum disorders (ASDs) constitute a neurodevelopmental disorder characterized by social deficits, repetitive behaviors, and learning disability. Oxidative stress and mitochondrial dysfunction are associated with ASD brain pathology. Here, we used oxidative stress in prenatal valproic acid (VPA)-exposed rats as an ASD model. After maternal VPA exposure (600 mg/kg, p.o.) on embryonic day (E) 12.5, temporal analyses of oxidative stress in the brain using an anti-4-hydroxy-2-nonenal antibody revealed that oxidative stress was increased in the hippocampus after birth. This was accompanied by aberrant enzymatic activity in the mitochondrial electron transport chain and reduced adenosine triphosphate (ATP) levels in the hippocampus. VPA-exposed rats exhibited impaired spatial reference and object recognition memory alongside impaired social behaviors and repetitive behaviors. ASD-like behaviors including learning and memory were rescued by chronic oral administration of 5-aminolevulinic acid (5-ALA; 30 mg/kg/day) and intranasal administration of oxytocin (OXT; 12 μg/kg/day), a neuropeptide that improves social behavior in ASD patients. 5-ALA but not OXT treatment ameliorated oxidative stress and mitochondrial dysfunction in the hippocampus of VPA-exposed rats. Fewer parvalbumin-positive interneurons were observed in VPA-exposed rats. Both 5-ALA and OXT treatments augmented the number of parvalbumin-positive interneurons. Collectively, our results indicate that oral 5-ALA administration ameliorated oxidative stress and mitochondrial dysfunction, suggesting that 5-ALA administration improves ASD-like neuropathology and behaviors via mechanisms different to those of OXT.
Collapse
Affiliation(s)
- Kazuya Matsuo
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Yasushi Yabuki
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan.
| |
Collapse
|
58
|
Hirsch MM, Deckmann I, Santos-Terra J, Staevie GZ, Fontes-Dutra M, Carello-Collar G, Körbes-Rockenbach M, Brum Schwingel G, Bauer-Negrini G, Rabelo B, Gonçalves MCB, Corrêa-Velloso J, Naaldijk Y, Castillo ARG, Schneider T, Bambini-Junior V, Ulrich H, Gottfried C. Effects of single-dose antipurinergic therapy on behavioral and molecular alterations in the valproic acid-induced animal model of autism. Neuropharmacology 2020; 167:107930. [PMID: 31904357 DOI: 10.1016/j.neuropharm.2019.107930] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 12/23/2019] [Accepted: 12/26/2019] [Indexed: 02/08/2023]
Abstract
Autism spectrum disorder (ASD) is characterized by deficits in communication and social interaction, restricted interests, and stereotyped behavior. Environmental factors, such as prenatal exposure to valproic acid (VPA), may contribute to the increased risk of ASD. Since disturbed functioning of the purinergic signaling system has been associated with the onset of ASD and used as a potential therapeutic target for ASD in both clinical and preclinical studies, we analyzed the effects of suramin, a non-selective purinergic antagonist, on behavioral, molecular and immunological in an animal model of autism induced by prenatal exposure to VPA. Treatment with suramin (20 mg/kg, intraperitoneal) restored sociability in the three-chamber apparatus and decreased anxiety measured by elevated plus maze apparatus, but had no impact on decreased reciprocal social interactions or higher nociceptive threshold in VPA rats. Suramin treatment did not affect VPA-induced upregulation of P2X4 and P2Y2 receptor expression in the hippocampus, and P2X4 receptor expression in the medial prefrontal cortex, but normalized an increased level of interleukin 6 (IL-6). Our results suggest an important role of purinergic signaling modulation in behavioral, molecular, and immunological aberrations described in VPA model, and indicate that the purinergic signaling system might be a potential target for pharmacotherapy in preclinical studies of ASD.
Collapse
Affiliation(s)
- Mauro Mozael Hirsch
- Translational Research Group in Autism Spectrum Disorders-GETTEA, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600, 90035-003, Porto Alegre, RS, Brazil; Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, 90035-003, Porto Alegre, RS, Brazil; National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Avenida Brasil, 4365, Manguinhos, 21045-900, Rio de Janeiro, RJ, Brazil.
| | - Iohanna Deckmann
- Translational Research Group in Autism Spectrum Disorders-GETTEA, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600, 90035-003, Porto Alegre, RS, Brazil; Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, 90035-003, Porto Alegre, RS, Brazil; National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Avenida Brasil, 4365, Manguinhos, 21045-900, Rio de Janeiro, RJ, Brazil
| | - Júlio Santos-Terra
- Translational Research Group in Autism Spectrum Disorders-GETTEA, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600, 90035-003, Porto Alegre, RS, Brazil; Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, 90035-003, Porto Alegre, RS, Brazil; National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Avenida Brasil, 4365, Manguinhos, 21045-900, Rio de Janeiro, RJ, Brazil
| | - Gabriela Zanotto Staevie
- Translational Research Group in Autism Spectrum Disorders-GETTEA, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600, 90035-003, Porto Alegre, RS, Brazil; Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, 90035-003, Porto Alegre, RS, Brazil; National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Avenida Brasil, 4365, Manguinhos, 21045-900, Rio de Janeiro, RJ, Brazil
| | - Mellanie Fontes-Dutra
- Translational Research Group in Autism Spectrum Disorders-GETTEA, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600, 90035-003, Porto Alegre, RS, Brazil; Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, 90035-003, Porto Alegre, RS, Brazil; National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Avenida Brasil, 4365, Manguinhos, 21045-900, Rio de Janeiro, RJ, Brazil
| | - Giovanna Carello-Collar
- Translational Research Group in Autism Spectrum Disorders-GETTEA, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600, 90035-003, Porto Alegre, RS, Brazil; Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, 90035-003, Porto Alegre, RS, Brazil; National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Avenida Brasil, 4365, Manguinhos, 21045-900, Rio de Janeiro, RJ, Brazil
| | - Marília Körbes-Rockenbach
- Translational Research Group in Autism Spectrum Disorders-GETTEA, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600, 90035-003, Porto Alegre, RS, Brazil; Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, 90035-003, Porto Alegre, RS, Brazil; National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Avenida Brasil, 4365, Manguinhos, 21045-900, Rio de Janeiro, RJ, Brazil
| | - Gustavo Brum Schwingel
- Translational Research Group in Autism Spectrum Disorders-GETTEA, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600, 90035-003, Porto Alegre, RS, Brazil; Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, 90035-003, Porto Alegre, RS, Brazil; National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Avenida Brasil, 4365, Manguinhos, 21045-900, Rio de Janeiro, RJ, Brazil
| | - Guilherme Bauer-Negrini
- Translational Research Group in Autism Spectrum Disorders-GETTEA, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600, 90035-003, Porto Alegre, RS, Brazil; Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, 90035-003, Porto Alegre, RS, Brazil; National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Avenida Brasil, 4365, Manguinhos, 21045-900, Rio de Janeiro, RJ, Brazil
| | - Bruna Rabelo
- Translational Research Group in Autism Spectrum Disorders-GETTEA, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600, 90035-003, Porto Alegre, RS, Brazil; Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, 90035-003, Porto Alegre, RS, Brazil; National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Avenida Brasil, 4365, Manguinhos, 21045-900, Rio de Janeiro, RJ, Brazil
| | - Maria Carolina Bittencourt Gonçalves
- Department of Biochemistry, Institute of Chemistry, Universidade de São Paulo (USP), Avenida Professor Lineu Prestes , 748, 05508-000, Vila Universitãria, São Paulo, SP, Brazil
| | - Juliana Corrêa-Velloso
- Department of Biochemistry, Institute of Chemistry, Universidade de São Paulo (USP), Avenida Professor Lineu Prestes , 748, 05508-000, Vila Universitãria, São Paulo, SP, Brazil
| | - Yahaira Naaldijk
- Department of Biochemistry, Institute of Chemistry, Universidade de São Paulo (USP), Avenida Professor Lineu Prestes , 748, 05508-000, Vila Universitãria, São Paulo, SP, Brazil
| | - Ana Regina Geciauskas Castillo
- Department of Biochemistry, Institute of Chemistry, Universidade de São Paulo (USP), Avenida Professor Lineu Prestes , 748, 05508-000, Vila Universitãria, São Paulo, SP, Brazil
| | - Tomasz Schneider
- School of Medicine, Pharmacy and Health, Durham University, Durham, DH1, UK
| | - Victorio Bambini-Junior
- Translational Research Group in Autism Spectrum Disorders-GETTEA, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600, 90035-003, Porto Alegre, RS, Brazil; National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Avenida Brasil, 4365, Manguinhos, 21045-900, Rio de Janeiro, RJ, Brazil; School of Pharmacy and Biomedical Sciences, University of Central Lancashire, PR1 2HE, Lancashire, Preston, England, UK; Autism Wellbeing And Research Development (AWARD) Institute, University of Central Lancashire, PR1 2HE, Lancashire, Preston, England, UK
| | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, Universidade de São Paulo (USP), Avenida Professor Lineu Prestes , 748, 05508-000, Vila Universitãria, São Paulo, SP, Brazil; Autism Wellbeing And Research Development (AWARD) Institute, University of Central Lancashire, PR1 2HE, Lancashire, Preston, England, UK
| | - Carmem Gottfried
- Translational Research Group in Autism Spectrum Disorders-GETTEA, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600, 90035-003, Porto Alegre, RS, Brazil; Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, 90035-003, Porto Alegre, RS, Brazil; National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Avenida Brasil, 4365, Manguinhos, 21045-900, Rio de Janeiro, RJ, Brazil; Autism Wellbeing And Research Development (AWARD) Institute, University of Central Lancashire, PR1 2HE, Lancashire, Preston, England, UK.
| |
Collapse
|
59
|
Rodrigues RJ, Marques JM, Cunha RA. Purinergic signalling and brain development. Semin Cell Dev Biol 2019; 95:34-41. [DOI: 10.1016/j.semcdb.2018.12.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 12/01/2018] [Accepted: 12/01/2018] [Indexed: 11/27/2022]
|
60
|
Kowash HM, Potter HG, Edye ME, Prinssen EP, Bandinelli S, Neill JC, Hager R, Glazier JD. Poly(I:C) source, molecular weight and endotoxin contamination affect dam and prenatal outcomes, implications for models of maternal immune activation. Brain Behav Immun 2019; 82:160-166. [PMID: 31415868 DOI: 10.1016/j.bbi.2019.08.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/10/2019] [Accepted: 08/10/2019] [Indexed: 01/08/2023] Open
Abstract
The viral mimetic polyinosinic:polycytidylic acid (poly(I:C)) is increasingly used to induce maternal immune activation (mIA) to model neurodevelopmental disorders (NDDs). Robust and reproducible phenotypes across studies are essential for the generation of models that will enhance our understanding of NDDs and enable the development of improved therapeutic strategies. However, differences in mIA-induced phenotypes using poly(I:C) have been widely observed, and this has prompted the reporting of useful and much needed methodological guidelines. Here, we perform a detailed investigation of molecular weight and endotoxin variations in poly(I:C) procured from two of the most commonly used suppliers, Sigma and InvivoGen. We demonstrate that endotoxin contamination and molecular weight differences in poly(I:C) composition lead to considerable variability in maternal IL-6 response in rats treated on gestational day (GD)15 and impact on fetal outcomes. Specifically, both endotoxin contamination and molecular weight predicted reductions in litter size on GD21. Further, molecular weight predicted a reduction in placental weight at GD21. While fetal body weight at GD21 was not affected by poly(I:C) treatment, male fetal brain weight was significantly reduced by poly(I:C), dependent on supplier. Our data are in agreement with recent reports of the importance of poly(I:C) molecular weight, and extend this work to demonstrate a key role of endotoxin on relevant phenotypic outcomes. We recommend that the source and batch numbers of poly(I:C) used should always be stated and that molecular weight variability and endotoxin contamination should be minimised for more robust mIA modelling.
Collapse
Affiliation(s)
- H M Kowash
- Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9WL, UK
| | - H G Potter
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK
| | - M E Edye
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Medicine, Biology and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK
| | - E P Prinssen
- Roche Innovation Centre, Basel, 124 Grenzacherstrasse, Basel, CH 4070, Switzerland
| | - S Bandinelli
- Roche Innovation Centre, Basel, 124 Grenzacherstrasse, Basel, CH 4070, Switzerland
| | - J C Neill
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Medicine, Biology and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK. http://www.b-neuro.com
| | - R Hager
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK
| | - J D Glazier
- Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9WL, UK; Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK
| |
Collapse
|
61
|
Johansson KE, Ståhl AL, Arvidsson I, Loos S, Tontanahal A, Rebetz J, Chromek M, Kristoffersson AC, Johannes L, Karpman D. Shiga toxin signals via ATP and its effect is blocked by purinergic receptor antagonism. Sci Rep 2019; 9:14362. [PMID: 31591425 PMCID: PMC6779916 DOI: 10.1038/s41598-019-50692-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 09/17/2019] [Indexed: 12/26/2022] Open
Abstract
Shiga toxin (Stx) is the main virulence factor of enterohemorrhagic Escherichia coli (EHEC), that cause gastrointestinal infection leading to hemolytic uremic syndrome. The aim of this study was to investigate if Stx signals via ATP and if blockade of purinergic receptors could be protective. Stx induced ATP release from HeLa cells and in a mouse model. Toxin induced rapid calcium influx into HeLa cells, as well as platelets, and a P2X1 receptor antagonist, NF449, abolished this effect. Likewise, the P2X antagonist suramin blocked calcium influx in Hela cells. NF449 did not affect toxin intracellular retrograde transport, however, cells pre-treated with NF449 exhibited significantly higher viability after exposure to Stx for 24 hours, compared to untreated cells. NF449 protected HeLa cells from protein synthesis inhibition and from Stx-induced apoptosis, assayed by caspase 3/7 activity. The latter effect was confirmed by P2X1 receptor silencing. Stx induced the release of toxin-positive HeLa cell- and platelet-derived microvesicles, detected by flow cytometry, an effect significantly reduced by NF449 or suramin. Suramin decreased microvesicle levels in mice injected with Stx or inoculated with Stx-producing EHEC. Taken together, we describe a novel mechanism of Stx-mediated cellular injury associated with ATP signaling and inhibited by P2X receptor blockade.
Collapse
Affiliation(s)
- Karl E Johansson
- Department of Pediatrics, Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Anne-Lie Ståhl
- Department of Pediatrics, Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Ida Arvidsson
- Department of Pediatrics, Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Sebastian Loos
- Department of Pediatrics, Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Ashmita Tontanahal
- Department of Pediatrics, Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Johan Rebetz
- Department of Pediatrics, Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Milan Chromek
- Department of Pediatrics, Clinical Sciences Lund, Lund University, Lund, Sweden
| | | | - Ludger Johannes
- Institut Curie, U1143 INSERM, UMR3666 CNRS, Cellular and Chemical Biology unit, Paris, France
| | - Diana Karpman
- Department of Pediatrics, Clinical Sciences Lund, Lund University, Lund, Sweden.
| |
Collapse
|
62
|
Hong MP, Erickson CA. Investigational drugs in early-stage clinical trials for autism spectrum disorder. Expert Opin Investig Drugs 2019; 28:709-718. [PMID: 31352835 DOI: 10.1080/13543784.2019.1649656] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Introduction: Pharmacologic interventions in Autism Spectrum Disorder (ASD) have historically focused on symptom-based approaches. However, a treatment for the core social deficits has remained unidentified. While a definitive theory for the cause of ASD is not yet known, recent advances in our understanding of ASD pathophysiology have opened the door for research on new pharmaceutical methods to target core symptomology. Areas covered: Herein, we review the novel pharmacologic therapies undergoing early-stage clinical trials for the treatment of the social symptoms associated with ASD. Specifically, these strategies center on altering neurologic excitatory and inhibitory imbalance, neuropeptide abnormalities, immunologic dysfunction, and biochemical deficiencies in ASD. Expert opinion: Utilizing the growing field of knowledge regarding the pathological mechanisms and altered neurobiology of individuals with ASD has led to the development of many innovative pharmaceutical interventions. Clinical trials for neurobiologic and immunologic targets show promise in impacting the social behavior and processing deficits in ASD but need evaluation in larger clinical trials and continued biomarker development to more effectively and consistently assess pharmacologic effects. Additionally, evaluating patient-specific drug responsivity and integrating behavioral intervention in conjunction with pharmacologic treatment is crucial to developing a successful approach to ASD treatment.
Collapse
Affiliation(s)
- Michael P Hong
- a Division of Psychiatry, Cincinnati Children's Hospital Medical Center , Cincinnati , OH , USA.,b College of Medicine, University of Cincinnati , Cincinnati , Oh , USA
| | - Craig A Erickson
- a Division of Psychiatry, Cincinnati Children's Hospital Medical Center , Cincinnati , OH , USA.,b College of Medicine, University of Cincinnati , Cincinnati , Oh , USA
| |
Collapse
|
63
|
Amodeo DA, Pahua AE, Zarate M, Taylor JA, Peterson S, Posadas R, Oliver BL, Amodeo LR. Differences in the expression of restricted repetitive behaviors in female and male BTBR T + tf/J mice. Behav Brain Res 2019; 372:112028. [PMID: 31212059 DOI: 10.1016/j.bbr.2019.112028] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 03/04/2019] [Accepted: 06/08/2019] [Indexed: 01/28/2023]
Abstract
Autism spectrum disorder (ASD) is characterized by the expression of restricted repetitive behaviors (RRBs) and impairments in social recognition and communication. Epidemiological studies demonstrate males are three times more likely than females to be affected. Although this is the case, more recent studies suggest females may be underrepresented in these numbers due to standard clinical measures of RRBs and social behaviors. In addition, many studies examining mouse models of ASD exclude females due to the sex disparity in diagnoses. The present study examined how female and male BTBR T + Itpr3tf /J (BTBR) compare to control C57BL/6J mice on tests of RRBs (probabilistic reversal learning, repetitive grooming, spontaneous alternation, and marble burying) and social behaviors (three chambered social approach task). Utilizing a spatial reversal learning test with 80/20 probabilistic feedback, in which ASD individuals have exhibited deficits, we find that female BTBR mice do not show the same impairment found in male BTBR mice. Interestingly, control female C57BL/6J mice required more trials to reach criterion. Female BTBR mice expressed comparable rates of repetitive grooming, marble burying and spontaneous alternation compared to female C57BL/6J mice. Male BTBR mice expressed higher rates of grooming behavior and locomotor activity compared to male C57BL/6J mice, as found in previous studies. Similarly, male BTBR mice showed a reduction in both measures of social approach compared to controls. Both male and female BTBR mice showed a reduction in sniff time for the stranger mouse compared to controls. Together these findings demonstrate how female BTBR mice do not display the RRB profile expressed by male BTBR mice. Testing of repetitive behaviors in ASD needs to better reflect the sex differences in how RRBs manifest in females compared to their extensively researched male counterparts.
Collapse
Affiliation(s)
- Dionisio A Amodeo
- Department of Psychology, California State University San Bernardino, 5500 University Parkway, San Bernardino, CA, 92407, United States.
| | - Alma E Pahua
- Department of Psychology, California State University San Bernardino, 5500 University Parkway, San Bernardino, CA, 92407, United States
| | - Marta Zarate
- Department of Psychology, California State University San Bernardino, 5500 University Parkway, San Bernardino, CA, 92407, United States
| | - Jordan A Taylor
- Department of Psychology, California State University San Bernardino, 5500 University Parkway, San Bernardino, CA, 92407, United States
| | - Sophie Peterson
- Department of Psychology, California State University San Bernardino, 5500 University Parkway, San Bernardino, CA, 92407, United States
| | - Rebekah Posadas
- Department of Psychology, California State University San Bernardino, 5500 University Parkway, San Bernardino, CA, 92407, United States
| | - Brandon L Oliver
- Department of Psychology, California State University San Bernardino, 5500 University Parkway, San Bernardino, CA, 92407, United States
| | - Leslie R Amodeo
- Department of Psychology, California State University San Bernardino, 5500 University Parkway, San Bernardino, CA, 92407, United States
| |
Collapse
|
64
|
Sex-Dependent Effects of Perinatal Inflammation on the Brain: Implication for Neuro-Psychiatric Disorders. Int J Mol Sci 2019; 20:ijms20092270. [PMID: 31071949 PMCID: PMC6539135 DOI: 10.3390/ijms20092270] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 05/03/2019] [Accepted: 05/06/2019] [Indexed: 12/12/2022] Open
Abstract
Individuals born preterm have higher rates of neurodevelopmental disorders such as schizophrenia, autistic spectrum, and attention deficit/hyperactivity disorders. These conditions are often sexually dimorphic and with different developmental trajectories. The etiology is likely multifactorial, however, infections both during pregnancy and in childhood have emerged as important risk factors. The association between sex- and age-dependent vulnerability to neuropsychiatric disorders has been suggested to relate to immune activation in the brain, including complex interactions between sex hormones, brain transcriptome, activation of glia cells, and cytokine production. Here, we will review sex-dependent effects on brain development, including glia cells, both under normal physiological conditions and following perinatal inflammation. Emphasis will be given to sex-dependent effects on brain regions which play a role in neuropsychiatric disorders and inflammatory reactions that may underlie early-life programming of neurobehavioral disturbances later in life.
Collapse
|
65
|
Haida O, Al Sagheer T, Balbous A, Francheteau M, Matas E, Soria F, Fernagut PO, Jaber M. Sex-dependent behavioral deficits and neuropathology in a maternal immune activation model of autism. Transl Psychiatry 2019; 9:124. [PMID: 30923308 PMCID: PMC6438965 DOI: 10.1038/s41398-019-0457-y] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 02/14/2019] [Accepted: 03/12/2019] [Indexed: 01/29/2023] Open
Abstract
Infections during gestation and the consequent maternal immune activation (MIA) increase the risk of developing neuropsychiatric disorders in infants and throughout life, including autism spectrum disorders (ASD). ASD is a neurodevelopmental disorder that affects three times more males than females and is mainly characterized by deficits in social communication and restricted interests. Consistent findings also indicate that ASD patients suffer from movement disorders, although these symptoms are not yet considered as diagnosis criteria. Here we used the double-stranded RNA analog polyinosinic:polycytidylic acid (poly I:C) MIA animal model of ASD in mice and explored its effects in males and females on social and motor behavior. We then investigated brain areas implicated in controlling and coordinating movements, namely the nigro-striatal pathway, motor cortex and cerebellum. We show that male mice are more affected by this treatment than females as they show reduced social interactions as well as motor development and coordination deficits. Reduced numbers of Purkinje cells in the cerebellum was found more widespread and within distinct lobules in males than in females. Moreover, a reduced number of neurons was found in the motor cortex of males only. These results suggest that females are better protected against developmental insults leading to ASD symptoms in mice. They also point to brain areas that may be targeted to better manage social and motor consequences of ASD.
Collapse
Affiliation(s)
- Obelia Haida
- 0000 0001 2160 6368grid.11166.31Université de Poitiers, INSERM, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, France
| | - Tareq Al Sagheer
- 0000 0001 2160 6368grid.11166.31Université de Poitiers, INSERM, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, France
| | - Anais Balbous
- 0000 0001 2160 6368grid.11166.31Université de Poitiers, INSERM, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, France ,0000 0000 9336 4276grid.411162.1CHU Poitiers, Poitiers, France
| | - Maureen Francheteau
- 0000 0001 2160 6368grid.11166.31Université de Poitiers, INSERM, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, France
| | - Emmanuel Matas
- 0000 0001 2160 6368grid.11166.31Université de Poitiers, INSERM, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, France
| | - Federico Soria
- grid.462010.1Université de Bordeaux, CNRS, Institut des Maladies Neurodégénératives, Bordeaux, France
| | - Pierre Olivier Fernagut
- 0000 0001 2160 6368grid.11166.31Université de Poitiers, INSERM, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, France ,grid.462010.1Université de Bordeaux, CNRS, Institut des Maladies Neurodégénératives, Bordeaux, France
| | - Mohamed Jaber
- Université de Poitiers, INSERM, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, France. .,CHU Poitiers, Poitiers, France.
| |
Collapse
|
66
|
Huang L, Otrokocsi L, Sperlágh B. Role of P2 receptors in normal brain development and in neurodevelopmental psychiatric disorders. Brain Res Bull 2019; 151:55-64. [PMID: 30721770 DOI: 10.1016/j.brainresbull.2019.01.030] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 01/17/2019] [Accepted: 01/19/2019] [Indexed: 12/19/2022]
Abstract
The purinergic signaling system, including P2 receptors, plays an important role in various central nervous system (CNS) disorders. Over the last few decades, a substantial amount of accumulated data suggest that most P2 receptor subtypes (P2X1, 2, 3, 4, 6, and 7, and P2Y1, 2, 6, 12, and 13) regulate neuronal/neuroglial developmental processes, such as proliferation, differentiation, migration of neuronal precursors, and neurite outgrowth. However, only a few of these subtypes (P2X2, P2X3, P2X4, P2X7, P2Y1, and P2Y2) have been investigated in the context of neurodevelopmental psychiatric disorders. The activation of these potential target receptors and their underlying mechanisms mainly influence the process of neuroinflammation. In particular, P2 receptor-mediated inflammatory cytokine release has been indicated to contribute to the complex mechanisms of a variety of CNS disorders. The released inflammatory cytokines could be utilized as biomarkers for neurodevelopmental and psychiatric disorders to improve the early diagnosis intervention, and prognosis. The population changes in gut microbiota after birth are closely linked to neurodevelopmental/neuropsychiatric disorders in later life; thus, the dynamic expression and function of P2 receptors on gut epithelial cells during disease processes indicate a novel avenue for the evaluation of disease progression and for the discovery of related therapeutic compounds.
Collapse
Affiliation(s)
- Lumei Huang
- Laboratory of Molecular Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary; János Szentágothai School of Neurosciences, Semmelweis University School of PhD Studies, Budapest, Hungary
| | - Lilla Otrokocsi
- Laboratory of Molecular Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Beáta Sperlágh
- Laboratory of Molecular Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary.
| |
Collapse
|
67
|
Amodeo DA, Lai CY, Hassan O, Mukamel EA, Behrens MM, Powell SB. Maternal immune activation impairs cognitive flexibility and alters transcription in frontal cortex. Neurobiol Dis 2019; 125:211-218. [PMID: 30716470 DOI: 10.1016/j.nbd.2019.01.025] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 12/20/2018] [Accepted: 01/17/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Epidemiological studies suggest that the risk of neurodevelopmental disorders such as autism spectrum disorder (ASD) and schizophrenia is increased by prenatal exposure to viral or bacterial infection during pregnancy. It is still unclear how activation of the maternal immune response interacts with underlying genetic factors to influence observed ASD phenotypes. METHODS The current study investigated how maternal immune activation (MIA) in mice impacts gene expression in the frontal cortex in adulthood, and how these molecular changes relate to deficits in cognitive flexibility and social behavior, and increases in repetitive behavior that are prevalent in ASD. Poly(I:C) (20 mg/kg) was administered to dams on E12.5 and offspring were tested for social approach behavior, repetitive grooming, and probabilistic reversal learning in adulthood (n = 8 vehicle; n = 9 Poly(I:C)). We employed next-generation high-throughput mRNA sequencing (RNA-seq) to comprehensively investigate the transcriptome profile in frontal cortex of adult offspring of Poly(I:C)-exposed dams. RESULTS Exposure to poly(I:C) during gestation impaired probabilistic reversal learning and decreased social approach in MIA offspring compared to controls. We found long-term effects of MIA on expression of 24 genes, including genes involved in glutamatergic neurotransmission, mTOR signaling and potassium ion channel activity. Correlations between gene expression and specific behavioral measures provided insight into genes that may be responsible for ASD-like behavioral alterations. CONCLUSIONS These findings suggest that MIA can lead to impairments in cognitive flexibility in mice similar to those exhibited in ASD individuals, and that these impairments are associated with altered gene expression in frontal cortex.
Collapse
Affiliation(s)
- Dionisio A Amodeo
- Department of Psychiatry, University of California San Diego, CA 9500 Gilman Drive, La Jolla, CA 92093, United States; Department of Psychology, California State University San Bernardino, 5500 University Parkway, San Bernardino, CA 92407, United States
| | - Chi-Yu Lai
- Computational Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92093, United States
| | - Omron Hassan
- Department of Psychiatry, University of California San Diego, CA 9500 Gilman Drive, La Jolla, CA 92093, United States
| | - Eran A Mukamel
- Department of Cognitive Science, University of California San Diego, CA 9500 Gilman Drive, La Jolla, CA 92093, United States
| | - M Margarita Behrens
- Computational Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92093, United States; Department of Psychiatry, University of California San Diego, CA 9500 Gilman Drive, La Jolla, CA 92093, United States.
| | - Susan B Powell
- Department of Psychiatry, University of California San Diego, CA 9500 Gilman Drive, La Jolla, CA 92093, United States; VISN-22 Mental Illness Research, Education and Clinical Center (MIRECC), VA San Diego Healthcare System, La Jolla, CA, United States.
| |
Collapse
|
68
|
P2X7 Receptors Drive Poly(I:C) Induced Autism-like Behavior in Mice. J Neurosci 2019; 39:2542-2561. [PMID: 30683682 DOI: 10.1523/jneurosci.1895-18.2019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 01/09/2019] [Accepted: 01/12/2019] [Indexed: 02/08/2023] Open
Abstract
Maternal immune activation (MIA) is a principal environmental risk factor contributing to autism spectrum disorder (ASD), which compromises fetal brain development at critical periods of pregnancy and might be causally linked to ASD symptoms. We report that endogenous activation of the purinergic ion channel P2X7 (P2rx7) is necessary and sufficient to transduce MIA to autistic phenotype in male offspring. MIA induced by poly(I:C) injections to P2rx7 WT mouse dams elicited an autism-like phenotype in their offspring, and these alterations were not observed in P2rx7-deficient mice, or following maternal treatment with a specific P2rx7 antagonist, JNJ47965567. Genetic deletion and pharmacological inhibition of maternal P2rx7s also counteracted the induction of IL-6 in the maternal plasma and fetal brain, and disrupted brain development, whereas postnatal P2rx7 inhibition alleviated behavioral and morphological alterations in the offspring. Administration of ATP to P2rx7 WT dams also evoked autistic phenotype, but not in KO dams, implying that P2rx7 activation by ATP is sufficient to induce autism-like features in offspring. Our results point to maternal and offspring P2rx7s as potential therapeutic targets for the early prevention and treatment of ASD.SIGNIFICANCE STATEMENT Autism spectrum disorder (ASD) is a neurodevelopmental psychiatric disorder caused by genetic and environmental factors. Recent studies highlighted the importance of perinatal risks, in particular, maternal immune activation (MIA), showing strong association with the later emergence of ASD in the affected children. MIA could be mimicked in animal models via injection of a nonpathogenic agent poly(I:C) during pregnancy. This is the first report showing the key role of a ligand gated ion channel, the purinergic P2X7 receptor in MIA-induced autism-like behavioral and biochemical features. We show that genetic or pharmacological inhibition of both maternal and offspring P2X7 receptors could reverse the compromised brain development and autistic phenotype pointing to new possibilities for prevention and treatment of ASD.
Collapse
|
69
|
Beamer E, Conte G, Engel T. ATP release during seizures - A critical evaluation of the evidence. Brain Res Bull 2019; 151:65-73. [PMID: 30660718 DOI: 10.1016/j.brainresbull.2018.12.021] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 12/06/2018] [Accepted: 12/20/2018] [Indexed: 01/17/2023]
Abstract
That adenosine 5' triphosphate (ATP) functions as an extracellular signaling molecule has been established since the 1970s. Ubiquitous throughout the body as the principal molecular store of intracellular energy, ATP has a short extracellular half-life and is difficult to measure directly. Extracellular ATP concentrations are dependent both on the rate of cellular release and of enzymatic degradation. Some findings from in vitro studies suggest that extracellular ATP concentrations increase during high levels of neuronal activity and seizure-like events in hippocampal slices. Pharmacological studies suggest that antagonism of ATP-sensitive purinergic receptors can suppress the severity of seizures and block epileptogenesis. Directly measuring extracellular ATP concentrations in the brain, however, has a number of specific challenges, notably, the rapid hydrolysis of ATP and huge gradient between intracellular and extracellular compartments. Two studies using microdialysis found no change in extracellular ATP in the hippocampus of rats during experimentally-induced status epilepticus. One of which demonstrated that ATP increased measurably, only in the presence of ectoATPase inhibitors, with the other study demonstrating increases only during later spontaneous seizures. Current evidence is mixed and seems highly dependent on the model used and method of detection. More sensitive methods of detection with higher spatial resolution, which induce less tissue disruption will be necessary to provide evidence for or against the hypothesis of seizure-induced elevations in extracellular ATP. Here we describe the current hypothesis for ATP release during seizures and its role in epileptogenesis, describe the technical challenges involved and critically examine the current evidence.
Collapse
Affiliation(s)
- Edward Beamer
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, D02YN77, Dublin, Ireland.
| | - Giorgia Conte
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, D02YN77, Dublin, Ireland
| | - Tobias Engel
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, D02YN77, Dublin, Ireland
| |
Collapse
|
70
|
Jyonouchi H, Geng L, Rose S, Bennuri SC, Frye RE. Variations in Mitochondrial Respiration Differ in IL-1ß/IL-10 Ratio Based Subgroups in Autism Spectrum Disorders. Front Psychiatry 2019; 10:71. [PMID: 30842746 PMCID: PMC6391925 DOI: 10.3389/fpsyt.2019.00071] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 01/30/2019] [Indexed: 12/31/2022] Open
Abstract
Autism spectrum disorder (ASD)7 is associated with multiple physiological abnormalities, including immune dysregulation, and mitochondrial dysfunction. However, an association between these two commonly reported abnormalities in ASD has not been studied in depth. This study assessed the association between previously identified alterations in cytokine profiles by ASD peripheral blood monocytes (PBMo) and mitochondrial dysfunction. In 112 ASD and 38 non-ASD subjects, cytokine production was assessed by culturing purified PBMo overnight with stimuli of innate immunity. Parameters of mitochondrial respiration including proton-leak respiration (PLR), ATP-linked respiration (ALR), maximal respiratory capacity (MRC), and reserve capacity (RC) were measured in peripheral blood mononuclear cells (PBMCs). The ASD samples were analyzed by subgrouping them into high, normal, and low IL-1ß/IL-10 ratio groups, which was previously shown to be associated with changes in behaviors and PBMo miRNA expression. MRC, RC, and RC/PLR, a marker of electron transport chain (ETC) efficiency, were higher in ASD PBMCs than controls. The expected positive associations between PLR and ALR were found in control non-ASD PBMCs, but not in ASD PBMCs. Higher MRC, RC, RC/PLR in ASD PBMCs were secondary to higher levels of these parameters in the high and normal IL-1ß/IL-10 ratio ASD subgroups than controls. Associations between mitochondrial parameters and monocyte cytokine profiles differed markedly across the IL-1ß/IL-10 ratio based ASD subgroups, rendering such associations less evident when ASD samples as a whole were compared to non-ASD controls. Our results indicate for the first time, an association between PBMC mitochondrial function and PBMo cytokine profiles in ASD subjects. This relationship differs across the IL-1ß/IL-10 ratio based ASD subgroups. Changes in mitochondrial function are likely due to adaptive changes or mitochondrial dysfunction, resulting from chronic oxidative stress. These results may indicate alteration in molecular pathways affecting both the immune system and mitochondrial function in some ASD subjects.
Collapse
Affiliation(s)
- Harumi Jyonouchi
- Department of Pediatrics, Saint Peter's University Hospital, New Brunswick, NJ, United States.,Robert Wood Johnson Medical School-Rutgers, New Brunswick, NJ, United States
| | - Lee Geng
- Department of Pediatrics, Saint Peter's University Hospital, New Brunswick, NJ, United States
| | - Shannon Rose
- Arkansas Children's Research Institute, Little Rock, AR, United States.,Department of Pediatrics, University of Arkansas of Medical Sciences, Little Rock, AR, United States
| | - Sirish C Bennuri
- Arkansas Children's Research Institute, Little Rock, AR, United States.,Department of Pediatrics, University of Arkansas of Medical Sciences, Little Rock, AR, United States
| | - Richard E Frye
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States.,Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States
| |
Collapse
|
71
|
Grković I, Drakulić D, Martinović J, Mitrović N. Role of Ectonucleotidases in Synapse Formation During Brain Development: Physiological and Pathological Implications. Curr Neuropharmacol 2019; 17:84-98. [PMID: 28521702 PMCID: PMC6341498 DOI: 10.2174/1570159x15666170518151541] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 04/19/2017] [Accepted: 05/16/2017] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Extracellular adenine nucleotides and nucleosides, such as ATP and adenosine, are among the most recently identified and least investigated diffusible signaling factors that contribute to the structural and functional remodeling of the brain, both during embryonic and postnatal development. Their levels in the extracellular milieu are tightly controlled by various ectonucleotidases: ecto-nucleotide pyrophosphatase/phosphodiesterases (E-NPP), alkaline phosphatases (AP), ecto-nucleoside triphosphate diphosphohydrolases (E-NTPDases) and ecto-5'- nucleotidase (eN). METHODS Studies related to the expression patterns of ectonucleotidases and their known features during brain development are reviewed, highlighting involvement of these enzymes in synapse formation and maturation in physiological as well as in pathological states. RESULTS During brain development and in adulthood all ectonucleotidases have diverse expression pattern, cell specific localization and function. NPPs are expressed at early embryonic days, but the expression of NPP3 is reduced and restricted to ependymal area in adult brain. NTPDase2 is dominant ectonucleotidase existing in the progenitor cells as well as main astrocytic NTPDase in the adult brain, while NTPDase3 is fully expressed after third postnatal week, almost exclusively on varicose fibers. Specific brain AP is functionally associated with synapse formation and this enzyme is sufficient for adenosine production during neurite growth and peak of synaptogenesis. eN is transiently associated with synapses during synaptogenesis, however in adult brain it is more glial than neuronal enzyme. CONCLUSION Control of extracellular adenine nucleotide levels by ectonucleotidases are important for understanding the role of purinergic signaling in developing tissues and potential targets in developmental disorders such as autism.
Collapse
Affiliation(s)
- Ivana Grković
- Department of Molecular Biology and Endocrinology, VINČA Institute of Nuclear Sciences, University of Belgrade, Mike Petrovića Alasa 12-14, 11001 Belgrade, Serbia
| | - Dunja Drakulić
- Department of Molecular Biology and Endocrinology, VINČA Institute of Nuclear Sciences, University of Belgrade, Mike Petrovića Alasa 12-14, 11001 Belgrade, Serbia
| | - Jelena Martinović
- Department of Molecular Biology and Endocrinology, VINČA Institute of Nuclear Sciences, University of Belgrade, Mike Petrovića Alasa 12-14, 11001 Belgrade, Serbia
| | - Nataša Mitrović
- Department of Molecular Biology and Endocrinology, VINČA Institute of Nuclear Sciences, University of Belgrade, Mike Petrovića Alasa 12-14, 11001 Belgrade, Serbia
| |
Collapse
|
72
|
Bennuri SC, Rose S, Frye RE. Mitochondrial Dysfunction Is Inducible in Lymphoblastoid Cell Lines From Children With Autism and May Involve the TORC1 Pathway. Front Psychiatry 2019; 10:269. [PMID: 31133888 PMCID: PMC6514096 DOI: 10.3389/fpsyt.2019.00269] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 04/09/2019] [Indexed: 01/25/2023] Open
Abstract
We previously developed a lymphoblastoid cell line (LCL) model of mitochondrial dysfunction in autism spectrum disorder (ASD); some individuals with ASD showed mitochondrial dysfunction (AD-A) while other individuals (AD-N) demonstrated mitochondrial respiration similar to controls (CNT). To test the hypothesis that mitochondrial dysfunction could be a consequence of environmental exposures through chronic elevations in reactive oxygen species (ROS), we exposed LCLs to prolonged ROS. We also examined expression of metabolic regulatory genes and the modulating effect of the mechanistic target of rapamycin (mTOR) pathway. Prolonged ROS exposure induced or worsened mitochondrial dysfunction in all LCL groups. Expression of genes associated with ROS protection was elevated in both AD-N and AD-A LCLs, but mitochondrial fission/fusion and mitoplasticity gene expression was only increased in AD-N LCLs. Partial least squares discriminant analysis showed that mTOR, UCP2 (uncoupling protein 2), SIRT1 (sirtuin 1), and MFN2 (mitofusin-2) gene expression differentiated LCL groups. Low-dose rapamycin (0.1 nM) normalized respiration with the magnitude of this normalization greater for AD-A LCLs, suggesting that the mammalian target of rapamycin complex 1 (mTORC1) pathway may have a different dynamic range for regulating mitochondrial activity in individuals with ASD with and without mitochondrial dysfunction, potentially related to S6K1 (S6 kinase beta-1) regulation. Understanding pathways that underlie mitochondrial dysfunction in ASD may lead to novel treatments.
Collapse
Affiliation(s)
- Sirish C Bennuri
- Arkansas Children's Research Institute and Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Shannon Rose
- Arkansas Children's Research Institute and Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Richard E Frye
- Barrow Neurologic Institute at Phoenix Children's Hospital, Phoenix, AZ, United States.,Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
| |
Collapse
|
73
|
Bergdolt L, Dunaevsky A. Brain changes in a maternal immune activation model of neurodevelopmental brain disorders. Prog Neurobiol 2018; 175:1-19. [PMID: 30590095 DOI: 10.1016/j.pneurobio.2018.12.002] [Citation(s) in RCA: 154] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 12/13/2018] [Accepted: 12/20/2018] [Indexed: 12/11/2022]
Abstract
The developing brain is sensitive to a variety of insults. Epidemiological studies have identified prenatal exposure to infection as a risk factor for a range of neurological disorders, including autism spectrum disorder and schizophrenia. Animal models corroborate this association and have been used to probe the contribution of gene-environment interactions to the etiology of neurodevelopmental disorders. Here we review the behavior and brain phenotypes that have been characterized in MIA offspring, including the studies that have looked at the interaction between maternal immune activation and genetic risk factors for autism spectrum disorder or schizophrenia. These phenotypes include behaviors relevant to autism, schizophrenia, and other neurological disorders, alterations in brain anatomy, and structural and functional neuronal impairments. The link between maternal infection and these phenotypic changes is not fully understood, but there is increasing evidence that maternal immune activation induces prolonged immune alterations in the offspring's brain which could underlie epigenetic alterations which in turn may mediate the behavior and brain changes. These concepts will be discussed followed by a summary of the pharmacological interventions that have been tested in the maternal immune activation model.
Collapse
Affiliation(s)
- Lara Bergdolt
- University of Nebraska Medical Center, Neurological Sciences, 985960 Nebraska Medical Center, 68105, Omaha, NE, United States
| | - Anna Dunaevsky
- University of Nebraska Medical Center, Neurological Sciences, 985960 Nebraska Medical Center, 68105, Omaha, NE, United States.
| |
Collapse
|
74
|
Waddington JL, O'Tuathaigh CM. Modelling the neuromotor abnormalities of psychotic illness: Putative mechanisms and systems dysfunction. Schizophr Res 2018; 200:12-19. [PMID: 28867516 DOI: 10.1016/j.schres.2017.08.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 08/11/2017] [Accepted: 08/16/2017] [Indexed: 12/20/2022]
Abstract
Limitations in access to antipsychotic-naïve patients and in the incisiveness of studies that can be conducted on them, together with the inevitability of subsequent antipsychotic treatment, indicate an enduring role for animal models that can inform on the pathobiology of neuromotor abnormalities in schizophrenia and related psychotic illness. This review focusses particularly on genetically modified mouse models that involve genes associated with risk for schizophrenia and with mechanisms implicated in the neuromotor abnormalities evident in psychotic patients, as well as developmental models that seek to mirror the trajectory, phenomenology and putative pathophysiology of psychotic illness. Such abnormalities are inconsistent and subtle in mice mutant for some schizophrenia risk genes but more evident for others. The phenotype of dopaminergic and glutamatergic mutants indicates the involvement of these mechanisms, informs on the roles of specific receptor subtypes, and implicates the interplay of cortical and subcortical processes. Developmental models suggest a criticality in the timing of early adversity for diversity in the relative emergence of psychological symptoms vis-à-vis neuromotor abnormalities in the overall psychosis phenotype. These findings elaborate current concepts of dysfunction in a neuronal network linking the cerebral cortex, basal ganglia, thalamus and cerebellum. Both findings in model systems and clinical evidence converge in indicating that any distinction between 'psychomotor' and 'neuromotor' abnormality is artificial and arbitrary due to a unitary origin in developmentally determined systems/network dysfunction that underlies the lifetime trajectory of psychotic illness.
Collapse
Affiliation(s)
- John L Waddington
- Molecular & Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin, Ireland; Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psychiatric-Diseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China.
| | | |
Collapse
|
75
|
Smolders S, Notter T, Smolders SMT, Rigo JM, Brône B. Controversies and prospects about microglia in maternal immune activation models for neurodevelopmental disorders. Brain Behav Immun 2018; 73:51-65. [PMID: 29870753 DOI: 10.1016/j.bbi.2018.06.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 05/26/2018] [Accepted: 06/01/2018] [Indexed: 12/16/2022] Open
Abstract
Activation of the maternal immune system during pregnancy is a well-established risk factor for neuropsychiatric disease in the offspring, yet, the underlying mechanisms leading to altered brain function remain largely undefined. Microglia, the resident immune cells of the brain, are key to adequate development of the central nervous system (CNS), and are prime candidates to mediate maternal immune activation (MIA)-induced brain abnormalities. As such, the effects of MIA on the immunological phenotype of microglia has been widely investigated. However, contradicting results due to differences in read-out and methodological approaches impede final conclusions on MIA-induced microglial alterations. The aim of this review is to critically discuss the evidence for an activated microglial phenotype upon MIA.
Collapse
Affiliation(s)
- Silke Smolders
- Uhasselt - BIOMED, Hasselt, Belgium; Laboratory of Neuronal Differentiation, VIB Center for the Biology of Disease, Leuven and Center for Human Genetics, KU Leuven Leuven, Belgium.
| | - Tina Notter
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland.
| | - Sophie M T Smolders
- Uhasselt - BIOMED, Hasselt, Belgium; INSERM, UMR S 1130, Université Pierre et Marie Curie Paris, France; CNRS, UMR 8246, Université Pierre et Marie Curie Paris, France; UM 119 NPS, Université Pierre et Marie Curie Paris, France.
| | | | | |
Collapse
|
76
|
Kim JW, Hong JY, Bae SM. Microglia and Autism Spectrum Disorder: Overview of Current Evidence and Novel Immunomodulatory Treatment Options. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE : THE OFFICIAL SCIENTIFIC JOURNAL OF THE KOREAN COLLEGE OF NEUROPSYCHOPHARMACOLOGY 2018; 16:246-252. [PMID: 30121973 PMCID: PMC6124874 DOI: 10.9758/cpn.2018.16.3.246] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 03/12/2018] [Accepted: 04/05/2018] [Indexed: 12/18/2022]
Abstract
Autism spectrum disorder is a rapidly increasing heterogeneous neurodevelopmental syndrome, remarked by persistent deficit in social communication, and restricted, repetitive patterns of behavior and interest. Lately, maternal immune activation and micgroglial dysfunction in the developing brain have been gaining mounting evidence and leading to studies of various novel agents as potential treatment options. A few immunomodulatory treatment options-luteolin, minocycline, suramin, vitamin D, gut microbiota-are discussed in the current article, regarding the current understanding of their mechanisms and evidence for potential clinical use. More studies are warranted to understand their exact mechanisms of action and to verify efficacy and safety in human subjects.
Collapse
Affiliation(s)
- Jung Won Kim
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL,
USA
| | - Ji Yeon Hong
- Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul,
Korea
| | - Seung Min Bae
- Department of Psychiatry, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon,
Korea
| |
Collapse
|
77
|
Sakaguchi T, Iwasaki S, Okada M, Okamoto K, Ikegaya Y. Ethanol facilitates socially evoked memory recall in mice by recruiting pain-sensitive anterior cingulate cortical neurons. Nat Commun 2018; 9:3526. [PMID: 30166546 PMCID: PMC6117351 DOI: 10.1038/s41467-018-05894-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 07/09/2018] [Indexed: 11/09/2022] Open
Abstract
Alcohol is a traditional social-bonding reinforcer; however, the neural mechanism underlying ethanol-driven social behaviors remains elusive. Here, we report that ethanol facilitates observational fear response. Observer mice exhibited stronger defensive immobility while observing cagemates that received repetitive foot shocks if the observer mice had experienced a brief priming foot shock. This enhancement was associated with an observation-induced recruitment of subsets of anterior cingulate cortex (ACC) neurons in the observer mouse that were responsive to its own pain. The vicariously activated ACC neurons projected their axons preferentially to the basolateral amygdala. Ethanol shifted the ACC neuronal balance toward inhibition, facilitated the preferential ACC neuronal recruitment during observation, and enhanced observational fear response, independent of an oxytocin signaling pathway. Furthermore, ethanol enhanced socially evoked fear response in autism model mice.
Collapse
Affiliation(s)
- Tetsuya Sakaguchi
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Satoshi Iwasaki
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Mami Okada
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Kazuki Okamoto
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Yuji Ikegaya
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan. .,Center for Information and Neural Networks, National Institute of Information and Communications Technology, Suita City, Osaka, 565-0871, Japan.
| |
Collapse
|
78
|
Naviaux RK. Metabolic features and regulation of the healing cycle-A new model for chronic disease pathogenesis and treatment. Mitochondrion 2018; 46:278-297. [PMID: 30099222 DOI: 10.1016/j.mito.2018.08.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 08/02/2018] [Indexed: 02/07/2023]
Abstract
Without healing, multicellular life on Earth would not exist. Without healing, one injury predisposes to another, leading to disability, chronic disease, accelerated aging, and death. Over 60% of adults and 30% of children and teens in the United States now live with a chronic illness. Advances in mass spectrometry and metabolomics have given scientists a new lens for studying health and disease. This study defines the healing cycle in metabolic terms and reframes the pathophysiology of chronic illness as the result of metabolic signaling abnormalities that block healing and cause the normal stages of the cell danger response (CDR) to persist abnormally. Once an injury occurs, active progress through the stages of healing is driven by sequential changes in cellular bioenergetics and the disposition of oxygen and carbon skeletons used for fuel, signaling, defense, repair, and recovery. >100 chronic illnesses can be organized into three persistent stages of the CDR. One hundred and two targetable chemosensory G-protein coupled and ionotropic receptors are presented that regulate the CDR and healing. Metabokines are signaling molecules derived from metabolism that regulate these receptors. Reframing the pathogenesis of chronic illness in this way, as a systems problem that maintains disease, rather than focusing on remote trigger(s) that caused the initial injury, permits new research to focus on novel signaling therapies to unblock the healing cycle, and restore health when other approaches have failed.
Collapse
Affiliation(s)
- Robert K Naviaux
- The Mitochondrial and Metabolic Disease Center, Departments of Medicine, Pediatrics, and Pathology, University of California, San Diego School of Medicine, 214 Dickinson St., Bldg CTF, Rm C102, MC#8467, San Diego, CA 92103, United States.
| |
Collapse
|
79
|
Rogozin IB, Gertz EM, Baranov PV, Poliakov E, Schaffer AA. Genome-Wide Changes in Protein Translation Efficiency Are Associated with Autism. Genome Biol Evol 2018; 10:1902-1919. [PMID: 29986017 PMCID: PMC6086092 DOI: 10.1093/gbe/evy146] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/06/2018] [Indexed: 01/05/2023] Open
Abstract
We previously proposed that changes in the efficiency of protein translation are associated with autism spectrum disorders (ASDs). This hypothesis connects environmental factors and genetic factors because each can alter translation efficiency. For genetic factors, we previously tested our hypothesis using a small set of ASD-associated genes, a small set of ASD-associated variants, and a statistic to quantify by how much a single nucleotide variant (SNV) in a protein coding region changes translation speed. In this study, we confirm and extend our hypothesis using a published set of 1,800 autism quartets (parents, one affected child and one unaffected child) and genome-wide variants. Then, we extend the test statistic to combine translation efficiency with other possibly relevant variables: ribosome profiling data, presence/absence of CpG dinucleotides, and phylogenetic conservation. The inclusion of ribosome profiling abundances strengthens our results for male-male sibling pairs. The inclusion of CpG information strengthens our results for female-female pairs, giving an insight into the significant gender differences in autism incidence. By combining the single-variant test statistic for all variants in a gene, we obtain a single gene score to evaluate how well a gene distinguishes between affected and unaffected siblings. Using statistical methods, we compute gene sets that have some power to distinguish between affected and unaffected siblings by translation efficiency of gene variants. Pathway and enrichment analysis of those gene sets suggest the importance of Wnt signaling pathways, some other pathways related to cancer, ATP binding, and ATP-ase pathways in the etiology of ASDs.
Collapse
Affiliation(s)
- Igor B Rogozin
- National Center for Biotechnology Information, National Library of Medicine, NIH, Bethesda, Maryland
| | - E Michael Gertz
- National Center for Biotechnology Information, National Library of Medicine, NIH, Bethesda, Maryland
| | - Pasha V Baranov
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Eugenia Poliakov
- National Eye Institute, NIH, Laboratory of Retinal Cell and Molecular Biology, Bethesda, Maryland
| | - Alejandro A Schaffer
- National Center for Biotechnology Information, National Library of Medicine, NIH, Bethesda, Maryland
| |
Collapse
|
80
|
Cattane N, Richetto J, Cattaneo A. Prenatal exposure to environmental insults and enhanced risk of developing Schizophrenia and Autism Spectrum Disorder: focus on biological pathways and epigenetic mechanisms. Neurosci Biobehav Rev 2018; 117:253-278. [PMID: 29981347 DOI: 10.1016/j.neubiorev.2018.07.001] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 06/11/2018] [Accepted: 07/01/2018] [Indexed: 12/15/2022]
Abstract
When considering neurodevelopmental disorders (NDDs), Schizophrenia (SZ) and Autism Spectrum Disorder (ASD) are considered to be among the most severe in term of prevalence, morbidity and impact on the society. Similar features and overlapping symptoms have been observed at multiple levels, suggesting common pathophysiological bases. Indeed, recent genome-wide association studies (GWAS) and epidemiological data report shared vulnerability genes and environmental triggers across the two disorders. In this review, we will discuss the possible biological mechanisms, including glutamatergic and GABAergic neurotransmissions, inflammatory signals and oxidative stress related systems, which are targeted by adverse environmental exposures and that have been associated with the development of SZ and ASD. We will also discuss the emerging role of the gut microbiome as possible interplay between environment, immune system and brain development. Finally, we will describe the involvement of epigenetic mechanisms in the maintenance of long-lasting effects of adverse environments early in life. This will allow us to better understand the pathophysiology of these NDDs, and also to identify novel targets for future treatment strategies.
Collapse
Affiliation(s)
- Nadia Cattane
- Biological Psychiatry Unit, IRCCS Fatebenefratelli San Giovanni di Dio, via Pilastroni 4, Brescia, Italy
| | - Juliet Richetto
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
| | - Annamaria Cattaneo
- Biological Psychiatry Unit, IRCCS Fatebenefratelli San Giovanni di Dio, via Pilastroni 4, Brescia, Italy; Stress, Psychiatry and Immunology Laboratory, Department of Psychological Medicine, Institute of Psychiatry, King's College London, London, 125 Coldharbour Lane, SE5 9NU, London, UK.
| |
Collapse
|
81
|
GABAergic malfunction in the anterior cingulate cortex underlying maternal immune activation-induced social deficits. J Neuroimmunol 2018; 321:92-96. [PMID: 29957393 DOI: 10.1016/j.jneuroim.2018.06.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 06/06/2018] [Accepted: 06/06/2018] [Indexed: 12/15/2022]
Abstract
Social deficits are one of the major symptoms of psychiatric disorders, including autism spectrum disorders (ASDs) and schizophrenia. However, the underlying mechanism remains ill-defined. Here, we focused on the anterior cingulate cortex (ACC), a brain region that is related to social behaviors, of mice that received poly(I:C)-induced maternal immune activation. Offspring born from poly(I:C)-treated dams exhibited social deficits in a three-chamber task at juvenile stages. Using whole-cell patch clamp recordings, we found that layer 2/3 pyramidal cells were hyperactive in acute ACC slices prepared from poly(I:C)-treated mice compared to those from saline-treated mice. The hyperexcitation was associated with a reduction in inhibitory synapse activity. Local injection of the GABAA receptor enhancer clonazepam into the ACC of poly(I:C)-treated mice restored the social behaviors of the mice. These results suggest that the balanced excitability of ACC neurons is essential for social ability.
Collapse
|
82
|
Lombardo MV, Moon HM, Su J, Palmer TD, Courchesne E, Pramparo T. Maternal immune activation dysregulation of the fetal brain transcriptome and relevance to the pathophysiology of autism spectrum disorder. Mol Psychiatry 2018; 23:1001-1013. [PMID: 28322282 PMCID: PMC5608645 DOI: 10.1038/mp.2017.15] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 12/31/2016] [Accepted: 01/09/2017] [Indexed: 12/12/2022]
Abstract
Maternal immune activation (MIA) via infection during pregnancy is known to increase risk for autism spectrum disorder (ASD). However, it is unclear how MIA disrupts fetal brain gene expression in ways that may explain this increased risk. Here we examine how MIA dysregulates rat fetal brain gene expression (at a time point analogous to the end of the first trimester of human gestation) in ways relevant to ASD-associated pathophysiology. MIA downregulates expression of ASD-associated genes, with the largest enrichments in genes known to harbor rare highly penetrant mutations. MIA also downregulates expression of many genes also known to be persistently downregulated in the ASD cortex later in life and which are canonically known for roles in affecting prenatally late developmental processes at the synapse. Transcriptional and translational programs that are downstream targets of highly ASD-penetrant FMR1 and CHD8 genes are also heavily affected by MIA. MIA strongly upregulates expression of a large number of genes involved in translation initiation, cell cycle, DNA damage and proteolysis processes that affect multiple key neural developmental functions. Upregulation of translation initiation is common to and preserved in gene network structure with the ASD cortical transcriptome throughout life and has downstream impact on cell cycle processes. The cap-dependent translation initiation gene, EIF4E, is one of the most MIA-dysregulated of all ASD-associated genes and targeted network analyses demonstrate prominent MIA-induced transcriptional dysregulation of mTOR and EIF4E-dependent signaling. This dysregulation of translation initiation via alteration of the Tsc2-mTor-Eif4e axis was further validated across MIA rodent models. MIA may confer increased risk for ASD by dysregulating key aspects of fetal brain gene expression that are highly relevant to pathophysiology affecting ASD.
Collapse
Affiliation(s)
- M V Lombardo
- Center for Applied Neuroscience, Department of Psychology, University of Cyprus, Nicosia, Cyprus,Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, UK,Neuroscience University of California, San Diego, 8110 La Jolla Shores Drive Suite 201, La Jolla, CA 92093, USA. E-mail: or
| | - H M Moon
- Department of Neurosurgery, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
| | - J Su
- Department of Neurosurgery, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
| | - T D Palmer
- Department of Neurosurgery, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
| | - E Courchesne
- Department of Neuroscience, University of California, San Diego, San Diego, CA, USA
| | - T Pramparo
- Department of Neuroscience, University of California, San Diego, San Diego, CA, USA,Neuroscience University of California, San Diego, 8110 La Jolla Shores Drive Suite 201, La Jolla, CA 92093, USA. E-mail: or
| |
Collapse
|
83
|
Robicsek O, Ene HM, Karry R, Ytzhaki O, Asor E, McPhie D, Cohen BM, Ben-Yehuda R, Weiner I, Ben-Shachar D. Isolated Mitochondria Transfer Improves Neuronal Differentiation of Schizophrenia-Derived Induced Pluripotent Stem Cells and Rescues Deficits in a Rat Model of the Disorder. Schizophr Bull 2018; 44:432-442. [PMID: 28586483 PMCID: PMC5814822 DOI: 10.1093/schbul/sbx077] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Dysfunction of mitochondria, key players in various essential cell processes, has been repeatedly reported in schizophrenia (SZ). Recently, several studies have reported functional recovery and cellular viability following mitochondrial transplantation, mostly in ischemia experimental models. Here, we aimed to demonstrate beneficial effects of isolated active normal mitochondria (IAN-MIT) transfer in vitro and in vivo, using SZ-derived induced pluripotent stem cells (iPSCs) differentiating into glutamatergic neuron, as well as a rodent model of SZ. First, we show that IAN-MIT enter various cell types without manipulation. Next, we show that IAN-MIT transfer into SZ-derived lymphoblasts induces long-lasting improvement in various mitochondrial functions including cellular oxygen consumption and mitochondrial membrane potential (Δ ψ m). We also demonstrate improved differentiation of SZ-derived iPSCs into neurons, by increased expression of neuronal and glutamatergic markers β3-tubulin, synapsin1, and Tbr1 and by an activation of the glutamate-glutamine cycle. In the animal model, we show that intra-prefrontal cortex injection of IAN-MIT in adolescent rats exposed prenatally to a viral mimic prevents mitochondrial Δ ψ m and attentional deficit at adulthood. Our results provide evidence for a direct link between mitochondrial function and SZ-related deficits both in vitro and in vivo and suggest a therapeutic potential for IAN-MIT transfer in diseases with bioenergetic and neurodevelopmental abnormalities such as SZ.
Collapse
Affiliation(s)
- Odile Robicsek
- Laboratory of Psychobiology, Department of Psychiatry, Rambam Health Care Campus, B. Rappaport Faculty of Medicine and Rappaport Family Institute for Research in Medical Sciences, Technion IIT, Haifa, Israel
| | - Hila M Ene
- Laboratory of Psychobiology, Department of Psychiatry, Rambam Health Care Campus, B. Rappaport Faculty of Medicine and Rappaport Family Institute for Research in Medical Sciences, Technion IIT, Haifa, Israel
| | - Rachel Karry
- Laboratory of Psychobiology, Department of Psychiatry, Rambam Health Care Campus, B. Rappaport Faculty of Medicine and Rappaport Family Institute for Research in Medical Sciences, Technion IIT, Haifa, Israel
| | - Ofer Ytzhaki
- Laboratory of Psychobiology, Department of Psychiatry, Rambam Health Care Campus, B. Rappaport Faculty of Medicine and Rappaport Family Institute for Research in Medical Sciences, Technion IIT, Haifa, Israel
| | - Eyal Asor
- Laboratory of Psychobiology, Department of Psychiatry, Rambam Health Care Campus, B. Rappaport Faculty of Medicine and Rappaport Family Institute for Research in Medical Sciences, Technion IIT, Haifa, Israel
| | - Donna McPhie
- Department of Psychiatry, Harvard Medical School, Boston, McLean Hospital, Belmont, MA
| | - Bruce M Cohen
- Department of Psychiatry, Harvard Medical School, Boston, McLean Hospital, Belmont, MA
| | - Rotem Ben-Yehuda
- School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ina Weiner
- School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Dorit Ben-Shachar
- Laboratory of Psychobiology, Department of Psychiatry, Rambam Health Care Campus, B. Rappaport Faculty of Medicine and Rappaport Family Institute for Research in Medical Sciences, Technion IIT, Haifa, Israel,To whom correspondence should be addressed; Laboratory of Psychobiology, Department of Psychiatry, Rambam Health Care Campus and B. Rappaport Faculty of Medicine, Technion ITT, POB 9649, Haifa 31096, Israel; tel: +972-4-8295224, fax: +972-4-8295220, e-mail:
| |
Collapse
|
84
|
Microglia and Neonatal Brain Injury. Neuroscience 2018; 405:68-76. [PMID: 29352997 DOI: 10.1016/j.neuroscience.2018.01.023] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 12/21/2017] [Accepted: 01/09/2018] [Indexed: 12/16/2022]
Abstract
Microglial cells are now recognized as the "gate-keepers" of healthy brain microenvironment with their disrupted functions adversely affecting neurovascular integrity, neuronal homeostasis, and network connectivity. The perception that these cells are purely toxic under neurodegenerative conditions has been challenged by a continuously increasing understanding of their complexity, the existence of a broad array of microglial phenotypes, and their ability to rapidly change in a context-dependent manner to attenuate or exacerbate injuries of different nature. Recent studies have demonstrated that microglial cells exert crucial physiological functions during embryonic and postnatal brain development, some of these functions being unique to particular stages of development, and extending far beyond sensing dangerous signals and serving as antigen presenting cells. In this focused review we cover the roles of microglial cells in regulating embryonic vasculogenesis, neurogenesis, and establishing network connectivity during postnatal brain development. We further discuss context-dependent microglial contribution to neonatal brain injuries associated with prenatal and postnatal infection and inflammation, in relation to neurodevelopmental disorders, as well as perinatal hypoxia-ischemia and arterial focal stroke. We also emphasize microglial phenotypic diversity, notably at the ultrastructural level, and their sex-dependent influence on the pathophysiology of neurodevelopmental disorders.
Collapse
|
85
|
Purinergic system in psychiatric diseases. Mol Psychiatry 2018; 23:94-106. [PMID: 28948971 DOI: 10.1038/mp.2017.188] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 07/15/2017] [Accepted: 07/20/2017] [Indexed: 12/14/2022]
Abstract
Psychiatric disorders are debilitating diseases, affecting >80 million people worldwide. There are no causal cures for psychiatric disorders and available therapies only treat the symptoms. The etiology of psychiatric disorders is unknown, although it has been speculated to be a combination of environmental, stress and genetic factors. One of the neurotransmitter systems implicated in the biology of psychiatric disorders is the purinergic system. In this review, we performed a comprehensive search of the literature about the role and function of the purinergic system in the development and predisposition to psychiatric disorders, with a focus on depression, schizophrenia, bipolar disorder, autism, anxiety and attention deficit/hyperactivity disorder. We also describe how therapeutics used for psychiatric disorders act on the purinergic system.
Collapse
|
86
|
Fumagalli M, Lecca D, Abbracchio MP, Ceruti S. Pathophysiological Role of Purines and Pyrimidines in Neurodevelopment: Unveiling New Pharmacological Approaches to Congenital Brain Diseases. Front Pharmacol 2017; 8:941. [PMID: 29375373 PMCID: PMC5770749 DOI: 10.3389/fphar.2017.00941] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 12/11/2017] [Indexed: 12/17/2022] Open
Abstract
In recent years, a substantial body of evidence has emerged demonstrating that purine and pyrimidine synthesis and metabolism play major roles in controlling embryonic and fetal development and organogenesis. Dynamic and time-dependent changes in the expression of purine metabolizing enzymes (such as ectonucleotidases and adenosine deaminase) represent a key checkpoint for the correct sequential generation of the different signaling molecules, that in turn activate their specific membrane receptors. In neurodevelopment, Ca2+ release from radial glia mediated by P2Y1 purinergic receptors is fundamental to allow neuroblast migration along radial glia processes, and their correct positioning in the different layers of the developing neocortex. Moreover, ATP is involved in the development of synaptic transmission and contributes to the establishment of functional neuronal networks in the developing brain. Additionally, several purinergic receptors (spanning from adenosine to P2X and P2Y receptor subtypes) are differentially expressed by neural stem cells, depending on their maturation stage, and their activation tightly regulates cell proliferation and differentiation to either neurons or glial cells, as well as their correct colonization of the developing telencephalon. The purinergic control of neurodevelopment is not limited to prenatal life, but is maintained in postnatal life, when it plays fundamental roles in controlling oligodendrocyte maturation from precursors and their terminal differentiation to fully myelinating cells. Based on the above-mentioned and other literature evidence, it is now increasingly clear that any defect altering the tight regulation of purinergic transmission and of purine and pyrimidine metabolism during pre- and post-natal brain development may translate into functional deficits, which could be at the basis of severe pathologies characterized by mental retardation or other disturbances. This can occur either at the level of the recruitment and/or signaling of specific nucleotide or nucleoside receptors or through genetic alterations in key steps of the purine salvage pathway. In this review, we have provided a critical analysis of what is currently known on the pathophysiological role of purines and pyrimidines during brain development with the aim of unveiling new future strategies for pharmacological intervention in different neurodevelopmental disorders.
Collapse
Affiliation(s)
- Marta Fumagalli
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Davide Lecca
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Maria P Abbracchio
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Stefania Ceruti
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
87
|
Antipurinergic therapy for autism-An in-depth review. Mitochondrion 2017; 43:1-15. [PMID: 29253638 DOI: 10.1016/j.mito.2017.12.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 12/11/2017] [Accepted: 12/14/2017] [Indexed: 12/13/2022]
Abstract
Are the symptoms of autism caused by a treatable metabolic syndrome that traces to the abnormal persistence of a normal, alternative functional state of mitochondria? A small clinical trial published in 2017 suggests this is possible. Based on a new unifying theory of pathogenesis for autism called the cell danger response (CDR) hypothesis, this study of 10 boys, ages 5-14years, showed that all 5 boys who received antipurinergic therapy (APT) with a single intravenous dose of suramin experienced improvements in all the core symptoms of autism that lasted for 5-8weeks. Language, social interaction, restricted interests, and repetitive movements all improved. Two children who were non-verbal spoke their first sentences. None of these improvements were observed in the placebo group. Larger and longer studies are needed to confirm this promising discovery. This review introduces the concept of M2 (anti-inflammatory) and M1 (pro-inflammatory) mitochondria that are polarized along a functional continuum according to cell stress. The pathophysiology of the CDR, the complementary functions of M1 and M2 mitochondria, relevant gene-environment interactions, and the metabolic underpinnings of behavior are discussed as foundation stones for understanding the improvements in ASD behaviors produced by antipurinergic therapy in this small clinical trial.
Collapse
|
88
|
Kennedy T, Broadie K. Fragile X Mental Retardation Protein Restricts Small Dye Iontophoresis Entry into Central Neurons. J Neurosci 2017; 37:9844-9858. [PMID: 28887386 PMCID: PMC5637114 DOI: 10.1523/jneurosci.0723-17.2017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 07/27/2017] [Accepted: 08/29/2017] [Indexed: 01/29/2023] Open
Abstract
Fragile X mental retardation protein (FMRP) loss causes Fragile X syndrome (FXS), a major disorder characterized by autism, intellectual disability, hyperactivity, and seizures. FMRP is both an RNA- and channel-binding regulator, with critical roles in neural circuit formation and function. However, it remains unclear how these FMRP activities relate to each other and how dysfunction in their absence underlies FXS neurological symptoms. In testing circuit level defects in the Drosophila FXS model, we discovered a completely unexpected and highly robust neuronal dye iontophoresis phenotype in the well mapped giant fiber (GF) circuit. Controlled dye injection into the GF interneuron results in a dramatic increase in dye uptake in neurons lacking FMRP. Transgenic wild-type FMRP reintroduction rescues the mutant defect, demonstrating a specific FMRP requirement. This phenotype affects only small dyes, but is independent of dye charge polarity. Surprisingly, the elevated dye iontophoresis persists in shaking B mutants that eliminate gap junctions and dye coupling among GF circuit neurons. We therefore used a wide range of manipulations to investigate the dye uptake defect, including timed injection series, pharmacology and ion replacement, and optogenetic activity studies. The results show that FMRP strongly limits the rate of dye entry via a cytosolic mechanism. This study reveals an unexpected new phenotype in a physical property of central neurons lacking FMRP that could underlie aspects of FXS disruption of neural function.SIGNIFICANCE STATEMENT FXS is a leading heritable cause of intellectual disability and autism spectrum disorders. Although researchers established the causal link with FMRP loss >;25 years ago, studies continue to reveal diverse FMRP functions. The Drosophila FXS model is key to discovering new FMRP roles, because of its genetic malleability and individually identified neuron maps. Taking advantage of a well characterized Drosophila neural circuit, we discovered that neurons lacking FMRP take up dramatically more current-injected small dye. After examining many neuronal properties, we determined that this dye defect is cytoplasmic and occurs due to a highly elevated dye iontophoresis rate. We also report several new factors affecting neuron dye uptake. Understanding how FMRP regulates iontophoresis should reveal new molecular factors underpinning FXS dysfunction.
Collapse
Affiliation(s)
| | - Kendal Broadie
- Department of Biological Sciences,
- Department of Cell and Developmental Biology, and
- Vanderbilt Brain Institute, Vanderbilt University and Medical Center, Nashville, Tennessee 37235
| |
Collapse
|
89
|
Chadman KK. Animal models for autism in 2017 and the consequential implications to drug discovery. Expert Opin Drug Discov 2017; 12:1187-1194. [PMID: 28971687 DOI: 10.1080/17460441.2017.1383982] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Autism spectrum disorder (ASD) is characterized by deficits in social communication and restricted interests/repetitive behaviors, for which there are currently no approved drug treatments. The core symptoms of ASD vary widely in severity and are often accompanied by other neuropsychiatric disorders. Drug discovery has been challenging because of the lack of understanding of the underlying pathophysiology of ASD as well as the heterogeneity of symptoms and symptom severity. Areas covered: In this review, the author discusses animal models of ASD used as targets for drug discovery, focusing primarily on non-syndromic models, primarily rodents. They highlight the wide range of drug targets examined in animal models. While very little of this work has resulted in drug therapy for the behavioral symptoms of ASD yet, it has increased our knowledge of the biology of ASD that is critical for driving drug discovery and has already provided many new drug targets for investigation. Expert opinion: The information gathered from the animal models of ASD is increasing our understanding of the underlying pathophysiology for ASD and is leading to better therapeutic targets. However, the issue of small sample size, heterogeneity within clinical samples, and a lack of replicable outcome measures must be addressed to move forward.
Collapse
Affiliation(s)
- Kathryn K Chadman
- a Behavioral Pharmacology Laboratory , NYS Office for People with Developmental Disabilities, Institute for Basic Research in Developmental Disabilities , Staten Island , NY , USA
| |
Collapse
|
90
|
Naviaux RK, Curtis B, Li K, Naviaux JC, Bright AT, Reiner GE, Westerfield M, Goh S, Alaynick WA, Wang L, Capparelli EV, Adams C, Sun J, Jain S, He F, Arellano DA, Mash LE, Chukoskie L, Lincoln A, Townsend J. Low-dose suramin in autism spectrum disorder: a small, phase I/II, randomized clinical trial. Ann Clin Transl Neurol 2017; 4:491-505. [PMID: 28695149 PMCID: PMC5497533 DOI: 10.1002/acn3.424] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Revised: 04/18/2017] [Accepted: 04/20/2017] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE No drug is yet approved to treat the core symptoms of autism spectrum disorder (ASD). Low-dose suramin was effective in the maternal immune activation and Fragile X mouse models of ASD. The Suramin Autism Treatment-1 (SAT-1) trial was a double-blind, placebo-controlled, translational pilot study to examine the safety and activity of low-dose suramin in children with ASD. METHODS Ten male subjects with ASD, ages 5-14 years, were matched by age, IQ, and autism severity into five pairs, then randomized to receive a single, intravenous infusion of suramin (20 mg/kg) or saline. The primary outcomes were ADOS-2 comparison scores and Expressive One-Word Picture Vocabulary Test (EOWPVT). Secondary outcomes were the aberrant behavior checklist, autism treatment evaluation checklist, repetitive behavior questionnaire, and clinical global impression questionnaire. RESULTS Blood levels of suramin were 12 ± 1.5 μmol/L (mean ± SD) at 2 days and 1.5 ± 0.5 μmol/L after 6 weeks. The terminal half-life was 14.7 ± 0.7 days. A self-limited, asymptomatic rash was seen, but there were no serious adverse events. ADOS-2 comparison scores improved by -1.6 ± 0.55 points (n = 5; 95% CI = -2.3 to -0.9; Cohen's d = 2.9; P = 0.0028) in the suramin group and did not change in the placebo group. EOWPVT scores did not change. Secondary outcomes also showed improvements in language, social interaction, and decreased restricted or repetitive behaviors. INTERPRETATION The safety and activity of low-dose suramin showed promise as a novel approach to treatment of ASD in this small study.
Collapse
Affiliation(s)
- Robert K. Naviaux
- The Mitochondrial and Metabolic Disease CenterUniversity of CaliforniaSan Diego School of Medicine214 Dickinson St., Bldg CTF, Rm C102San Diego92103‐8467California
- Department of MedicineUniversity of CaliforniaSan Diego School of Medicine214 Dickinson St., Bldg CTF, Rm C102San Diego92103‐8467California
- Department of PediatricsUniversity of CaliforniaSan Diego School of Medicine214 Dickinson St., Bldg CTF, Rm C102San Diego92103‐8467California
- Department of PathologyUniversity of CaliforniaSan Diego School of Medicine214 Dickinson St., Bldg CTF, Rm C102San Diego92103‐8467California
| | - Brooke Curtis
- Alliant International University10455 Pomerado RoadSan DiegoCalifornia92131
| | - Kefeng Li
- The Mitochondrial and Metabolic Disease CenterUniversity of CaliforniaSan Diego School of Medicine214 Dickinson St., Bldg CTF, Rm C102San Diego92103‐8467California
- Department of MedicineUniversity of CaliforniaSan Diego School of Medicine214 Dickinson St., Bldg CTF, Rm C102San Diego92103‐8467California
| | - Jane C. Naviaux
- The Mitochondrial and Metabolic Disease CenterUniversity of CaliforniaSan Diego School of Medicine214 Dickinson St., Bldg CTF, Rm C102San Diego92103‐8467California
- Department of NeurosciencesUniversity of CaliforniaSan Diego School of Medicine9500 Gilman Drive.La JollaCA92093‐0662
| | - A. Taylor Bright
- The Mitochondrial and Metabolic Disease CenterUniversity of CaliforniaSan Diego School of Medicine214 Dickinson St., Bldg CTF, Rm C102San Diego92103‐8467California
- Department of MedicineUniversity of CaliforniaSan Diego School of Medicine214 Dickinson St., Bldg CTF, Rm C102San Diego92103‐8467California
| | - Gail E. Reiner
- The Mitochondrial and Metabolic Disease CenterUniversity of CaliforniaSan Diego School of Medicine214 Dickinson St., Bldg CTF, Rm C102San Diego92103‐8467California
- Department of NeurosciencesUniversity of CaliforniaSan Diego School of Medicine9500 Gilman Drive.La JollaCA92093‐0662
| | - Marissa Westerfield
- The Research in Autism and Development Laboratory (RAD Lab)University of California9500 Gilman DriveLa JollaCA92093‐0959
| | - Suzanne Goh
- Pediatric Neurology Therapeutics7090 Miratech DrSan DiegoCA92121
| | - William A. Alaynick
- The Mitochondrial and Metabolic Disease CenterUniversity of CaliforniaSan Diego School of Medicine214 Dickinson St., Bldg CTF, Rm C102San Diego92103‐8467California
- Department of MedicineUniversity of CaliforniaSan Diego School of Medicine214 Dickinson St., Bldg CTF, Rm C102San Diego92103‐8467California
| | - Lin Wang
- The Mitochondrial and Metabolic Disease CenterUniversity of CaliforniaSan Diego School of Medicine214 Dickinson St., Bldg CTF, Rm C102San Diego92103‐8467California
- Department of MedicineUniversity of CaliforniaSan Diego School of Medicine214 Dickinson St., Bldg CTF, Rm C102San Diego92103‐8467California
| | - Edmund V. Capparelli
- Department of Pediatricsand Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of CaliforniaSan Diego School of Medicine9500 Gilman DriveLa JollaCA92093‐0657
| | - Cynthia Adams
- Clinical and Translational Research Institute (CTRI)University of CaliforniaSan DiegoLa JollaCA92037
| | - Ji Sun
- Clinical and Translational Research Institute (CTRI)University of CaliforniaSan DiegoLa JollaCA92037
| | - Sonia Jain
- Department of Family Medicine and Public HealthUniversity of CaliforniaSan DiegoLa JollaCA92093
| | - Feng He
- Department of Family Medicine and Public HealthUniversity of CaliforniaSan DiegoLa JollaCA92093
| | - Deyna A. Arellano
- Clinical and Translational Research Institute (CTRI)University of CaliforniaSan DiegoLa JollaCA92037
| | - Lisa E. Mash
- The Research in Autism and Development Laboratory (RAD Lab)University of California9500 Gilman DriveLa JollaCA92093‐0959
- Department of PsychologySan Diego State University5500 Campanile DriveSan DiegoCA92182
| | - Leanne Chukoskie
- The Research in Autism and Development Laboratory (RAD Lab)University of California9500 Gilman DriveLa JollaCA92093‐0959
- Institute for Neural ComputationUniversity of California9500 Gilman DriveLa Jolla92093‐0523
| | - Alan Lincoln
- Alliant International University10455 Pomerado RoadSan DiegoCalifornia92131
| | - Jeanne Townsend
- Department of NeurosciencesUniversity of CaliforniaSan Diego School of Medicine9500 Gilman Drive.La JollaCA92093‐0662
- The Research in Autism and Development Laboratory (RAD Lab)University of California9500 Gilman DriveLa JollaCA92093‐0959
| |
Collapse
|
91
|
The placental interleukin-6 signaling controls fetal brain development and behavior. Brain Behav Immun 2017; 62:11-23. [PMID: 27838335 PMCID: PMC5373986 DOI: 10.1016/j.bbi.2016.11.007] [Citation(s) in RCA: 176] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 10/20/2016] [Accepted: 11/08/2016] [Indexed: 01/16/2023] Open
Abstract
Epidemiological studies show that maternal immune activation (MIA) during pregnancy is a risk factor for autism. However, mechanisms for how MIA affects brain development and behaviors in offspring remain poorly described. To determine whether placental interleukin-6 (IL-6) signaling is required for mediating MIA on the offspring, we generated mice with restricted deletion of the receptor for IL-6 (IL-6Rα) in placental trophoblasts (Cyp19-Cre+;Il6rafl/fl), and tested offspring of Cyp19-Cre+;Il6rafl/fl mothers for immunological, pathological and behavioral abnormalities following induction of MIA. We reveal that MIA results in acute inflammatory responses in the fetal brain. Lack of IL-6 signaling in trophoblasts effectively blocks MIA-induced inflammatory responses in the placenta and the fetal brain. Furthermore, behavioral abnormalities and cerebellar neuropathologies observed in MIA control offspring are prevented in Cyp19-Cre+;Il6rafl/fl offspring. Our results demonstrate that IL-6 activation in placenta is required for relaying inflammatory signals to the fetal brain and impacting behaviors and neuropathologies relevant to neurodevelopmental disease.
Collapse
|
92
|
Higa KK, Grim A, Kamenski ME, van Enkhuizen J, Zhou X, Li K, Naviaux JC, Wang L, Naviaux RK, Geyer MA, Markou A, Young JW. Nicotine withdrawal-induced inattention is absent in alpha7 nAChR knockout mice. Psychopharmacology (Berl) 2017; 234:1573-1586. [PMID: 28243714 PMCID: PMC5420484 DOI: 10.1007/s00213-017-4572-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 02/15/2017] [Indexed: 11/25/2022]
Abstract
RATIONALE Smoking is the leading cause of preventable death in the USA, but quit attempts result in withdrawal-induced cognitive dysfunction and predicts relapse. Greater understanding of the neural mechanism(s) underlying these cognitive deficits is required to develop targeted treatments to aid quit attempts. OBJECTIVES We examined nicotine withdrawal-induced inattention in mice lacking the α7 nicotinic acetylcholine receptor (nAChR) using the five-choice continuous performance test (5C-CPT). METHODS Mice were trained in the 5C-CPT prior to osmotic minipump implantation containing saline or nicotine. Experiment 1 used 40 mg kg-1 day-1 nicotine treatment and tested C57BL/6 mice 4, 28, and 52 h after pump removal. Experiment 2 used 14 and 40 mg kg-1 day-1 nicotine treatment in α7 nAChR knockout (KO) and wildtype (WT) littermates tested 4 h after pump removal. Subsets of WT mice were killed before and after pump removal to assess changes in receptor expression associated with nicotine administration and withdrawal. RESULTS Nicotine withdrawal impaired attention in the 5C-CPT, driven by response inhibition and target detection deficits. The overall attentional deficit was absent in α7 nAChR KO mice despite response disinhibition in these mice. Synaptosomal glutamate mGluR5 and dopamine D4 receptor expression were reduced during chronic nicotine but increased during withdrawal, potentially contributing to cognitive deficits. CONCLUSIONS The α7 nAChR may underlie nicotine withdrawal-induced deficits in target detection but is not required for response disinhibition deficits. Alterations to the glutamatergic and dopaminergic pathways may also contribute to withdrawal-induced attentional deficits, providing novel targets to alleviate the cognitive symptoms of withdrawal during quit attempts.
Collapse
Affiliation(s)
- K K Higa
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA, 92093-0804, USA
| | - A Grim
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA, 92093-0804, USA
| | - M E Kamenski
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA, 92093-0804, USA
| | - J van Enkhuizen
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA, 92093-0804, USA
| | - X Zhou
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA, 92093-0804, USA
- Research Service, VA San Diego Healthcare System, 3350 La Jolla Village Drive, San Diego, CA, 92037, USA
| | - K Li
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - J C Naviaux
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA, 92093-0804, USA
| | - L Wang
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - R K Naviaux
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - M A Geyer
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA, 92093-0804, USA
- Research Service, VA San Diego Healthcare System, 3350 La Jolla Village Drive, San Diego, CA, 92037, USA
| | - A Markou
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA, 92093-0804, USA
| | - J W Young
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA, 92093-0804, USA.
- Research Service, VA San Diego Healthcare System, 3350 La Jolla Village Drive, San Diego, CA, 92037, USA.
| |
Collapse
|
93
|
Careaga M, Murai T, Bauman MD. Maternal Immune Activation and Autism Spectrum Disorder: From Rodents to Nonhuman and Human Primates. Biol Psychiatry 2017; 81:391-401. [PMID: 28137374 PMCID: PMC5513502 DOI: 10.1016/j.biopsych.2016.10.020] [Citation(s) in RCA: 238] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 10/20/2016] [Accepted: 10/20/2016] [Indexed: 12/18/2022]
Abstract
A subset of women who are exposed to infection during pregnancy have an increased risk of giving birth to a child who will later be diagnosed with a neurodevelopmental or neuropsychiatric disorder. Although epidemiology studies have primarily focused on the association between maternal infection and an increased risk of offspring schizophrenia, mounting evidence indicates that maternal infection may also increase the risk of autism spectrum disorder. A number of factors, including genetic susceptibility, the intensity and timing of the infection, and exposure to additional aversive postnatal events, may influence the extent to which maternal infection alters fetal brain development and which disease phenotype (autism spectrum disorder, schizophrenia, other neurodevelopmental disorders) is expressed. Preclinical animal models provide a test bed to systematically evaluate the effects of maternal infection on fetal brain development, determine the relevance to human central nervous system disorders, and to evaluate novel preventive and therapeutic strategies. Maternal immune activation models in mice, rats, and nonhuman primates suggest that the maternal immune response is the critical link between exposure to infection during pregnancy and subsequent changes in brain and behavioral development of offspring. However, differences in the type, severity, and timing of prenatal immune challenge paired with inconsistencies in behavioral phenotyping approaches have hindered the translation of preclinical results to human studies. Here we highlight the promises and limitations of the maternal immune activation model as a preclinical tool to study prenatal risk factors for autism spectrum disorder, and suggest specific changes to improve reproducibility and maximize translational potential.
Collapse
Affiliation(s)
- Milo Careaga
- UC Davis MIND Institute, University of California, Davis, California; Department of Psychiatry and Behavioral Sciences, University of California, Davis, California
| | - Takeshi Murai
- UC Davis MIND Institute, University of California, Davis, California; California National Primate Research Center, University of California, Davis, California; Biomarker Group, Drug Development Research Laboratories, Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan
| | - Melissa D Bauman
- UC Davis MIND Institute, University of California, Davis, California; Department of Psychiatry and Behavioral Sciences, University of California, Davis, California; California National Primate Research Center, University of California, Davis, California.
| |
Collapse
|
94
|
Cheng N, Rho JM, Masino SA. Metabolic Dysfunction Underlying Autism Spectrum Disorder and Potential Treatment Approaches. Front Mol Neurosci 2017; 10:34. [PMID: 28270747 PMCID: PMC5318388 DOI: 10.3389/fnmol.2017.00034] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 01/30/2017] [Indexed: 12/14/2022] Open
Abstract
Autism spectrum disorder (ASD) is characterized by deficits in sociability and communication, and increased repetitive and/or restrictive behaviors. While the etio-pathogenesis of ASD is unknown, clinical manifestations are diverse and many possible genetic and environmental factors have been implicated. As such, it has been a great challenge to identify key neurobiological mechanisms and to develop effective treatments. Current therapies focus on co-morbid conditions (such as epileptic seizures and sleep disturbances) and there is no cure for the core symptoms. Recent studies have increasingly implicated mitochondrial dysfunction in ASD. The fact that mitochondria are an integral part of diverse cellular functions and are susceptible to many insults could explain how a wide range of factors can contribute to a consistent behavioral phenotype in ASD. Meanwhile, the high-fat, low-carbohydrate ketogenic diet (KD), used for nearly a century to treat medically intractable epilepsy, has been shown to enhance mitochondrial function through a multiplicity of mechanisms and affect additional molecular targets that may address symptoms and comorbidities of ASD. Here, we review the evidence for the use of metabolism-based therapies such as the KD in the treatment of ASD as well as emerging co-morbid models of epilepsy and autism. Future research directions aimed at validating such therapeutic approaches and identifying additional and novel mechanistic targets are also discussed.
Collapse
Affiliation(s)
- Ning Cheng
- Departments of Pediatrics, University of CalgaryCalgary, AB, Canada
| | - Jong M. Rho
- Departments of Pediatrics, University of CalgaryCalgary, AB, Canada
- Clinical Neurosciences, University of CalgaryCalgary, AB, Canada
- Physiology and Pharmacology, Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of CalgaryCalgary, AB, Canada
| | - Susan A. Masino
- Neuroscience Program, Department of Psychology, Trinity CollegeHartford, CT, USA
| |
Collapse
|
95
|
Ruskin DN, Murphy MI, Slade SL, Masino SA. Ketogenic diet improves behaviors in a maternal immune activation model of autism spectrum disorder. PLoS One 2017; 12:e0171643. [PMID: 28166277 PMCID: PMC5293204 DOI: 10.1371/journal.pone.0171643] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 01/24/2017] [Indexed: 01/31/2023] Open
Abstract
Prenatal factors influence autism spectrum disorder (ASD) incidence in children and can increase ASD symptoms in offspring of animal models. These may include maternal immune activation (MIA) due to viral or bacterial infection during the first trimesters. Unfortunately, regardless of ASD etiology, existing drugs are poorly effective against core symptoms. For nearly a century a ketogenic diet (KD) has been used to treat seizures, and recent insights into mechanisms of ASD and a growing recognition that immune/inflammatory conditions exacerbate ASD risk has increased interest in KD as a treatment for ASD. Here we studied the effects of KD on core ASD symptoms in offspring exposed to MIA. To produce MIA, pregnant C57Bl/6 mice were injected with the viral mimic polyinosinic-polycytidylic acid; after weaning offspring were fed KD or control diet for three weeks. Consistent with an ASD phenotype of a higher incidence in males, control diet-fed MIA male offspring were not social and exhibited high levels of repetitive self-directed behaviors; female offspring were unaffected. However, KD feeding partially or completely reversed all MIA-induced behavioral abnormalities in males; it had no effect on behavior in females. KD-induced metabolic changes of reduced blood glucose and elevated blood ketones were quantified in offspring of both sexes. Prior work from our laboratory and others demonstrate KDs improve relevant behaviors in several ASD models, and here we demonstrate clear benefits of KD in the MIA model of ASD. Together these studies suggest a broad utility for metabolic therapy in improving core ASD symptoms, and support further research to develop and apply ketogenic and/or metabolic strategies in patients with ASD.
Collapse
Affiliation(s)
- David N. Ruskin
- Department of Psychology, Neuroscience Program, Trinity College, Hartford, CT, United States of America
- * E-mail:
| | - Michelle I. Murphy
- Department of Psychology, Neuroscience Program, Trinity College, Hartford, CT, United States of America
| | - Sierra L. Slade
- Department of Psychology, Neuroscience Program, Trinity College, Hartford, CT, United States of America
| | - Susan A. Masino
- Department of Psychology, Neuroscience Program, Trinity College, Hartford, CT, United States of America
| |
Collapse
|
96
|
Barańska J, Czajkowski R, Pomorski P. P2Y 1 Receptors - Properties and Functional Activities. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017. [PMID: 28639247 DOI: 10.1007/5584_2017_57] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In this chapter we try to show a comprehensive image of current knowledge of structure, activity and physiological role of the P2Y1 purinergic receptor. The structure, distribution and changes in the expression of this receptor are summarized, as well as the mechanism of its signaling activity by the intracellular calcium mobilization. We try to show the connection between the components of its G protein activation and cellular or physiological effects, starting from changes in protein phosphorylation patterns and ending with such remote effects as receptor-mediated apoptosis. The special emphasis is put on the role of the P2Y1 receptor in cancer cells and neuronal plasticity. We concentrate on the P2Y1 receptor, it is though impossible to completely abstract from other aspects of nucleotide signaling and cross-talk with other nucleotide receptors is here discussed. Especially, the balance between P2Y1 and P2Y12 receptors, sharing the same ligand but signaling through different pathways, is presented.
Collapse
Affiliation(s)
- Jolanta Barańska
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur St., PL 02-093, Warsaw, Poland
| | - Rafał Czajkowski
- Laboratory of Spatial Memory, Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur St., PL 02-093, Warsaw, Poland
| | - Paweł Pomorski
- Laboratory of Molecular Basis of Cell Motility, Department of Cell Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur St., PL 02-093, Warsaw, Poland.
| |
Collapse
|
97
|
Gevi F, Zolla L, Gabriele S, Persico AM. Urinary metabolomics of young Italian autistic children supports abnormal tryptophan and purine metabolism. Mol Autism 2016; 7:47. [PMID: 27904735 PMCID: PMC5121959 DOI: 10.1186/s13229-016-0109-5] [Citation(s) in RCA: 156] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2015] [Accepted: 11/11/2016] [Indexed: 12/13/2022] Open
Abstract
Background Autism spectrum disorder (ASD) is still diagnosed through behavioral observation, due to a lack of laboratory biomarkers, which could greatly aid clinicians in providing earlier and more reliable diagnoses. Metabolomics on human biofluids provides a sensitive tool to identify metabolite profiles potentially usable as biomarkers for ASD. Initial metabolomic studies, analyzing urines and plasma of ASD and control individuals, suggested that autistic patients may share some metabolic abnormalities, despite several inconsistencies stemming from differences in technology, ethnicity, age range, and definition of “control” status. Methods ASD-specific urinary metabolomic patterns were explored at an early age in 30 ASD children and 30 matched controls (age range 2–7, M:F = 22:8) using hydrophilic interaction chromatography (HILIC)-UHPLC and mass spectrometry, a highly sensitive, accurate, and unbiased approach. Metabolites were then subjected to multivariate statistical analysis and grouped by metabolic pathway. Results Urinary metabolites displaying the largest differences between young ASD and control children belonged to the tryptophan and purine metabolic pathways. Also, vitamin B6, riboflavin, phenylalanine-tyrosine-tryptophan biosynthesis, pantothenate and CoA, and pyrimidine metabolism differed significantly. ASD children preferentially transform tryptophan into xanthurenic acid and quinolinic acid (two catabolites of the kynurenine pathway), at the expense of kynurenic acid and especially of melatonin. Also, the gut microbiome contributes to altered tryptophan metabolism, yielding increased levels of indolyl 3-acetic acid and indolyl lactate. Conclusions The metabolic pathways most distinctive of young Italian autistic children largely overlap with those found in rodent models of ASD following maternal immune activation or genetic manipulations. These results are consistent with the proposal of a purine-driven cell danger response, accompanied by overproduction of epileptogenic and excitotoxic quinolinic acid, large reductions in melatonin synthesis, and gut dysbiosis. These metabolic abnormalities could underlie several comorbidities frequently associated to ASD, such as seizures, sleep disorders, and gastrointestinal symptoms, and could contribute to autism severity. Their diagnostic sensitivity, disease-specificity, and interethnic variability will merit further investigation. Electronic supplementary material The online version of this article (doi:10.1186/s13229-016-0109-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Federica Gevi
- Department of Ecological and Biological Sciences, University of Tuscia, Viterbo, Italy
| | - Lello Zolla
- Department of Ecological and Biological Sciences, University of Tuscia, Viterbo, Italy
| | - Stefano Gabriele
- Unit of Child and Adolescent Neuropsychiatry, Laboratory of Molecular Psychiatry and Neurogenetics, University Campus Bio-Medico, Rome, Italy
| | - Antonio M Persico
- Unit of Child and Adolescent Neuropsychiatry, Interdepartmental Program "Autism 0-90", "Gaetano Martino" University Hospital, University of Messina, Messina, Italy ; Mafalda Luce Center for Pervasive Developmental Disorders, Milan, Italy
| |
Collapse
|
98
|
Féron F, Gepner B, Lacassagne E, Stephan D, Mesnage B, Blanchard MP, Boulanger N, Tardif C, Devèze A, Rousseau S, Suzuki K, Izpisua Belmonte JC, Khrestchatisky M, Nivet E, Erard-Garcia M. Olfactory stem cells reveal MOCOS as a new player in autism spectrum disorders. Mol Psychiatry 2016; 21:1215-24. [PMID: 26239292 PMCID: PMC4995547 DOI: 10.1038/mp.2015.106] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 05/06/2015] [Accepted: 06/15/2015] [Indexed: 12/27/2022]
Abstract
With an onset under the age of 3 years, autism spectrum disorders (ASDs) are now understood as diseases arising from pre- and/or early postnatal brain developmental anomalies and/or early brain insults. To unveil the molecular mechanisms taking place during the misshaping of the developing brain, we chose to study cells that are representative of the very early stages of ontogenesis, namely stem cells. Here we report on MOlybdenum COfactor Sulfurase (MOCOS), an enzyme involved in purine metabolism, as a newly identified player in ASD. We found in adult nasal olfactory stem cells of 11 adults with ASD that MOCOS is downregulated in most of them when compared with 11 age- and gender-matched control adults without any neuropsychiatric disorders. Genetic approaches using in vivo and in vitro engineered models converge to indicate that altered expression of MOCOS results in neurotransmission and synaptic defects. Furthermore, we found that MOCOS misexpression induces increased oxidative-stress sensitivity. Our results demonstrate that altered MOCOS expression is likely to have an impact on neurodevelopment and neurotransmission, and may explain comorbid conditions, including gastrointestinal disorders. We anticipate our discovery to be a fresh starting point for the study on the roles of MOCOS in brain development and its functional implications in ASD clinical symptoms. Moreover, our study suggests the possible development of new diagnostic tests based on MOCOS expression, and paves the way for drug screening targeting MOCOS and/or the purine metabolism to ultimately develop novel treatments in ASD.
Collapse
Affiliation(s)
- F Féron
- Inserm CBT 1409, Centre d'Investigations Cliniques en Biothérapie, Marseille, France
- Aix Marseille Université, CNRS, NICN UMR 7259, Marseille, France
| | - B Gepner
- Aix Marseille Université, CNRS, NICN UMR 7259, Marseille, France
| | - E Lacassagne
- Aix Marseille Université, CNRS, NICN UMR 7259, Marseille, France
| | - D Stephan
- Aix Marseille Université, CNRS, NICN UMR 7259, Marseille, France
| | - B Mesnage
- Aix Marseille Université, CNRS, NICN UMR 7259, Marseille, France
| | - M-P Blanchard
- Aix Marseille Université, CNRS, CRN2M UMR 6231, Marseille, France
| | - N Boulanger
- Aix Marseille Université, TAGC UMR 1090, Marseille, France
| | - C Tardif
- Aix Marseille Université, PsyCLE, EA 3273, Aix en Provence, France
| | - A Devèze
- AP-HM, Département ORL, Marseille, France
| | - S Rousseau
- AP-HM, Département Anesthésie, Marseille, France
| | - K Suzuki
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - J C Izpisua Belmonte
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - M Khrestchatisky
- Aix Marseille Université, CNRS, NICN UMR 7259, Marseille, France
| | - E Nivet
- Aix Marseille Université, CNRS, NICN UMR 7259, Marseille, France
| | - M Erard-Garcia
- Aix Marseille Université, CNRS, NICN UMR 7259, Marseille, France
| |
Collapse
|
99
|
Kim KC, Gonzales EL, Lázaro MT, Choi CS, Bahn GH, Yoo HJ, Shin CY. Clinical and Neurobiological Relevance of Current Animal Models of Autism Spectrum Disorders. Biomol Ther (Seoul) 2016; 24:207-43. [PMID: 27133257 PMCID: PMC4859786 DOI: 10.4062/biomolther.2016.061] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 04/05/2016] [Indexed: 12/24/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by social and communication impairments, as well as repetitive and restrictive behaviors. The phenotypic heterogeneity of ASD has made it overwhelmingly difficult to determine the exact etiology and pathophysiology underlying the core symptoms, which are often accompanied by comorbidities such as hyperactivity, seizures, and sensorimotor abnormalities. To our benefit, the advent of animal models has allowed us to assess and test diverse risk factors of ASD, both genetic and environmental, and measure their contribution to the manifestation of autistic symptoms. At a broader scale, rodent models have helped consolidate molecular pathways and unify the neurophysiological mechanisms underlying each one of the various etiologies. This approach will potentially enable the stratification of ASD into clinical, molecular, and neurophenotypic subgroups, further proving their translational utility. It is henceforth paramount to establish a common ground of mechanistic theories from complementing results in preclinical research. In this review, we cluster the ASD animal models into lesion and genetic models and further classify them based on the corresponding environmental, epigenetic and genetic factors. Finally, we summarize the symptoms and neuropathological highlights for each model and make critical comparisons that elucidate their clinical and neurobiological relevance.
Collapse
Affiliation(s)
- Ki Chan Kim
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.,Center for Neuroscience Research, SMART Institute of Advanced Biomedical Sciences, Konkuk University, Seoul 05029, Republic of Korea
| | - Edson Luck Gonzales
- Center for Neuroscience Research, SMART Institute of Advanced Biomedical Sciences, Konkuk University, Seoul 05029, Republic of Korea.,School of Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - María T Lázaro
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Chang Soon Choi
- Center for Neuroscience Research, SMART Institute of Advanced Biomedical Sciences, Konkuk University, Seoul 05029, Republic of Korea.,School of Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Geon Ho Bahn
- Department of Neuropsychiatry, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Hee Jeong Yoo
- Department of Neuropsychiatry, Seoul National University Bungdang Hospital, Seongnam 13620, Republic of Korea
| | - Chan Young Shin
- Center for Neuroscience Research, SMART Institute of Advanced Biomedical Sciences, Konkuk University, Seoul 05029, Republic of Korea.,School of Medicine, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
100
|
Monpays C, Deslauriers J, Sarret P, Grignon S. Mitochondrial Dysfunction in Schizophrenia: Determination of Mitochondrial Respiratory Activity in a Two-Hit Mouse Model. J Mol Neurosci 2016; 59:440-51. [PMID: 27034067 DOI: 10.1007/s12031-016-0746-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 03/22/2016] [Indexed: 10/22/2022]
Abstract
Schizophrenia is a chronic mental illness in which mitochondrial dysfunction has been suggested. Our laboratory recently developed a juvenile murine two-hit model (THM) of schizophrenia based on the combination of gestational inflammation, followed by juvenile restraint stress. We previously reported that relevant behaviors and neurochemical disturbances, including oxidative stress, were reversed by the antioxidant lipoic acid (LA), thereby pointing to the central role played by oxidative abnormalities and prompting us to investigate mitochondrial function. Mitochondrial activity was determined with the MitoXpress® commercial kit in two schizophrenia-relevant regions (prefrontal cortex (PFC) and striatum). Measurements were performed in state 3, with substrates for complex I- and complex II-induced respiratory activity (IRA). We observed an increase in complex I IRA in the PFC and striatum in both sexes but an increase in complex II activity only in males. LA treatment prevented this increase only in complex II IRA in males. Expression levels of the different respiratory chain complexes, as well as fission/fusion proteins and protein carbonylation, were unchanged. In conclusion, our juvenile schizophrenia THM shows an increase in mitochondrial activity reversed by LA, specifically in complex II IRA in males. Further investigations are required to determine the mechanisms of these modifications.
Collapse
Affiliation(s)
- Cécile Monpays
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 12e avenue Nord, Sherbrooke, QC, J1H 5N4, Canada
| | - Jessica Deslauriers
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 12e avenue Nord, Sherbrooke, QC, J1H 5N4, Canada
| | - Philippe Sarret
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 12e avenue Nord, Sherbrooke, QC, J1H 5N4, Canada
| | - Sylvain Grignon
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 12e avenue Nord, Sherbrooke, QC, J1H 5N4, Canada. .,Department of Psychiatry, Centre Hospitalier Universitaire de Sherbrooke, 580 Bowen Sud, Sherbrooke, QC, J1G 2E8, Canada.
| |
Collapse
|