1
|
Gotfryd L, Fesser E, Cambiasso MY, Stinson MG, Birolo S, Nemirovsky SI, Cánepa ET, Calvo JC, Fontana VA. Paternal ethanol exposure alters offspring motor skills and behavior in a sex-dependent manner and modifies early growth response 1 expression in the medial prefrontal cortex. J Affect Disord 2025; 381:388-400. [PMID: 40189063 DOI: 10.1016/j.jad.2025.04.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 04/02/2025] [Accepted: 04/03/2025] [Indexed: 04/13/2025]
Abstract
BACKGROUND Alcohol consumption is linked to various health issues exerting direct effects on the consumer and indirectly on offspring through both maternal and paternal transmission pathways. Our recent studies highlight the importance of paternal health before conception, showing that male ethanol consumption can alter epigenetic sperm marks and DNA integrity and testicular organization which led to adverse effects on embryonic development and induced alterations in testicular and sperm characteristics in the offspring. METHODS Based on these findings, this study explores the effects of paternal ethanol (15 % v/v) consumption for 12 days on motor development in mice offspring. We also analyzed different behavioral parameters and evaluated the expression of immediate early genes from the medial prefrontal cortex in the progeny during adulthood. RESULTS Paternal alcohol intake negatively affects the offspring, showing a delay in the acquisition of motor developmental skills at an early age and some modifications of behavior in a sex-dependent manner in adulthood. Furthermore, this consumption shows an increase in the expression of the Early Growth Response 1 gene in both males and females in the medial prefrontal cortex. LIMITATIONS In situ expression of the early growth response 1 gene was not measured. Hormonal fluctuations during the estrous cycle of the female offspring were not considered, these changes could interact with the observed outcomes. CONCLUSIONS This gene plays a key role in regulating cognition, emotion, and behavior. These findings highlight the importance of considering paternal health and alcohol consumption when assessing the risks to future generations.
Collapse
Affiliation(s)
- Lucila Gotfryd
- Instituto de Biología Y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Estefanía Fesser
- Laboratorio de Neuroepigenética, CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales - CONICET (IQUIBICEN), Buenos Aires, Argentina
| | - Maite Yael Cambiasso
- Instituto de Biología Y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Marcelo Gabriel Stinson
- Universidad de Buenos Aires, Departamento de Química Biológica de la Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina
| | - Sol Birolo
- Universidad de Buenos Aires, Departamento de Química Biológica de la Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina
| | - Sergio Iván Nemirovsky
- Universidad de Buenos Aires, Departamento de Química Biológica de la Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina
| | - Eduardo Tomás Cánepa
- Laboratorio de Neuroepigenética, CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales - CONICET (IQUIBICEN), Buenos Aires, Argentina
| | - Juan Carlos Calvo
- Instituto de Biología Y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Vanina Andrea Fontana
- Instituto de Biología Y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina; Universidad de Buenos Aires, Departamento de Química Biológica de la Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina.
| |
Collapse
|
2
|
Kurita H, Ohuchi K, Inden M. Effects of Environmental Non-Essential Toxic Heavy Metals on Epigenetics During Development. TOXICS 2025; 13:167. [PMID: 40137494 PMCID: PMC11946632 DOI: 10.3390/toxics13030167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 02/23/2025] [Accepted: 02/25/2025] [Indexed: 03/29/2025]
Abstract
We are exposed to a variety of environmental chemicals in our daily lives. It is possible that the effects of this daily chemical exposure could accumulate in the organism in some form and influence health and disease development. The exposure effects extend throughout the human lifetime, not only after birth, but also during the embryonic period. Epigenetics is an important target for the molecular mechanisms of daily environmental chemical effects. Epigenetics is a mechanism of gene transcription regulation that does not involve changes in DNA sequence. The Developmental Origins of Health and Disease (DOHaD) theory has also been proposed, in which effects such as exposure to environmental chemicals during embryonic period are mediated by epigenetic changes, which may lead to risk for disease development and adverse health effects after maturity. This review summarizes the association between embryonic exposure and the epigenetics of well-known non-essential toxic heavy metals (methylmercury, cadmium, arsenic, and lead), a representative group of environmental chemicals. In the future, it will be important to predict the epigenetic mechanisms of unknown chemical and combined exposures. In addition, further experimental investigations using experimental animals and the accumulation of knowledge are needed to study the transgenerational effects of environmental chemicals in the future.
Collapse
Affiliation(s)
- Hisaka Kurita
- Laboratory of Medical Therapeutics and Molecular Therapeutics, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu 501-1196, Japan; (K.O.); (M.I.)
| | | | | |
Collapse
|
3
|
Gelmez P, Karakoc TE, Ulucan O. Autism Spectrum Disorder and Atypical Brain Connectivity: Novel Insights from Brain Connectivity-Associated Genes by Combining Random Forest and Support Vector Machine Algorithm. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2024; 28:563-572. [PMID: 39417279 DOI: 10.1089/omi.2024.0167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
It is estimated that approximately one in every 100 children is diagnosed with autism spectrum disorder (ASD) around the globe. Currently, there are no curative pharmacological treatments for ASD. Discoveries on key molecular mechanisms of ASD are essential for precision medicine strategies. Considering that atypical brain connectivity patterns have been observed in individuals with ASD, this study examined the brain connectivity-associated genes and their putatively distinct expression patterns in brain samples from individuals diagnosed with ASD and using an iterative strategy based on random forest and support vector machine algorithms. We discovered a potential gene signature capable of differentiating ASD from control samples with a 92% accuracy. This gene signature comprised 14 brain connectivity-associated genes exhibiting enrichment in synapse-related terms. Of these genes, 11 were previously associated with ASD in varying degrees of evidence. Notably, NFKBIA, WNT10B, and IFT22 genes were identified as ASD-related for the first time in this study. Subsequent clustering analysis revealed the existence of two distinct ASD subtypes based on our gene signature. The expression levels of signature genes have the potential to influence brain connectivity patterns, potentially contributing to the manifestation of ASD. Further studies on the omics of ASD are called for so as to elucidate the molecular basis of ASD and for diagnostic and therapeutic innovations. Finally, we underscore that advances in ASD research can benefit from integrative bioinformatics and data science approaches.
Collapse
Affiliation(s)
- Pelin Gelmez
- Department of Genetics and Bioengineering, Istanbul Bilgi University, Eyupsultan, Turkey
| | - Talha Emir Karakoc
- Department of Genetics and Bioengineering, Istanbul Bilgi University, Eyupsultan, Turkey
| | - Ozlem Ulucan
- Department of Genetics and Bioengineering, Istanbul Bilgi University, Eyupsultan, Turkey
| |
Collapse
|
4
|
Varma M, Bhandari R, Sarkar A, Jain M, Paliwal JK, Medhi B, Kuhad A. Exploring Astrocytes Involvement and Glutamate Induced Neuroinflammation in Chlorpyrifos-Induced Paradigm Of Autism Spectrum Disorders (ASD). Neurochem Res 2024; 49:2573-2599. [PMID: 38896196 DOI: 10.1007/s11064-024-04191-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 05/19/2024] [Accepted: 06/03/2024] [Indexed: 06/21/2024]
Abstract
Autism spectrum disorders (ASD) are neurodevelopmental disorders manifested mainly in children, with symptoms ranging from social/communication deficits and stereotypies to associated behavioral anomalies like anxiety, depression, and ADHD. While the patho-mechanism is not well understood, the role of neuroinflammation has been suggested. Nevertheless, the triggers giving rise to this neuroinflammation have not previously been explored in detail, so the present study was aimed at exploring the role of glutamate on these processes, potentially carried out through increased activity of inflammatory cells like astrocytes, and a decline in neuronal health. A novel chlorpyrifos-induced paradigm of ASD in rat pups was used for the present study. The animals were subjected to tests assessing their neonatal development and adolescent behaviors (social skills, stereotypies, sensorimotor deficits, anxiety, depression, olfactory, and pain perception). Markers for inflammation and the levels of molecules involved in glutamate excitotoxicity, and neuroinflammation were also measured. Additionally, the expression of reactive oxygen species and markers of neuronal inflammation (GFAP) and function (c-Fos) were evaluated, along with an assessment of histopathological alterations. Based on these evaluations, it was found that postnatal administration of CPF had a negative impact on neurobehavior during both the neonatal and adolescent phases, especially on developmental markers, and brought about the generation of ASD-like symptoms. This was further corroborated by elevations in the expression of glutamate and downstream calcium, as well as certain cytokines and neuroinflammatory markers, and validated through histopathological and immunohistochemical results showing a decline in neuronal health in an astrocyte-mediated cytokine-dependent fashion. Through our findings, conclusive evidence regarding the involvement of glutamate in neuroinflammatory pathways implicated in the development of ASD-like symptoms, as well as its ability to activate further downstream processes linked to neuronal damage has been obtained. The role of astrocytes and the detrimental effect on neuronal health are also concluded. The significance of our study and its findings lies in the evaluation of the involvement of chlorpyrifos-induced neurotoxicity in the development of ASD, particularly in relation to glutamatergic dysfunction and neuronal damage.
Collapse
Affiliation(s)
- Manasi Varma
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC- Centre of Advanced Study, Panjab University, Chandigarh, 160 014, India
| | - Ranjana Bhandari
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC- Centre of Advanced Study, Panjab University, Chandigarh, 160 014, India.
- Division of Pharmaceutics, University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University, Chandigarh, 160 014, India.
- Director, AKB INNOVANT HEALHCARE PVT. LTD., Chandigarh, India.
| | - Ankan Sarkar
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC- Centre of Advanced Study, Panjab University, Chandigarh, 160 014, India
| | - Manish Jain
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC- Centre of Advanced Study, Panjab University, Chandigarh, 160 014, India
| | - Jyoti K Paliwal
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC- Centre of Advanced Study, Panjab University, Chandigarh, 160 014, India
| | - Bikash Medhi
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC- Centre of Advanced Study, Panjab University, Chandigarh, 160 014, India
- Institute of Medical Education & Research (PGIMER), Chandigarh, India
| | - Anurag Kuhad
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC- Centre of Advanced Study, Panjab University, Chandigarh, 160 014, India.
| |
Collapse
|
5
|
Tartaglione AM, Camoni L, Calamandrei G, Chiarotti F, Venerosi A. The contribution of environmental pollutants to the risk of autism and other neurodevelopmental disorders: A systematic review of case-control studies. Neurosci Biobehav Rev 2024; 164:105815. [PMID: 39053787 DOI: 10.1016/j.neubiorev.2024.105815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/09/2024] [Accepted: 07/16/2024] [Indexed: 07/27/2024]
Abstract
Exposure to environmental pollutants, such as metals, pesticides, and air pollutants during early life, is a risk factor for neurodevelopmental disorders (NDDs), including Autism Spectrum Disorder (ASD). Our systematic review aimed to select and summarize more recent case-control studies that examined the association between prenatal and early postnatal exposure to environmental pollutants and NDDs. We searched five databases (Web of Science, PubMed, Embase, Scopus, Ovid), screened 2261 records, and included 24 eligible case-control studies. Meta-analyses were conducted on subgroups of at least three studies that shared both the outcome and the exposure. A noteworthy discovery from this literature review is the existence of non-linear or non-monotonic dose-response relationships between the exposure to certain metals and the risk of ASD. The meta-analysis revealed a significant association between exposure to particular matter (PM)10 during the first year of life and the risk of ASD. Overall, studies included in our systematic review indicate that exposure to several pollutants within the first three years of life was significantly associated with the risk of NDDs.
Collapse
Affiliation(s)
- A M Tartaglione
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy.
| | - L Camoni
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| | - G Calamandrei
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| | - F Chiarotti
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| | - A Venerosi
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
6
|
Ma Q, Yang Z, Yang C, Lin M, Gong M, Deng P, He M, Lu Y, Zhang K, Pi H, Qu M, Yu Z, Zhou Z, Chen C. A single-cell transcriptomic landscape of cadmium-hindered brain development in mice. Commun Biol 2024; 7:997. [PMID: 39147853 PMCID: PMC11327346 DOI: 10.1038/s42003-024-06685-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 08/06/2024] [Indexed: 08/17/2024] Open
Abstract
The effects of neurotoxicant cadmium (Cd) exposure on brain development have not been well elucidated. To investigate this, we have herein subjected pregnant mice to low-dose Cd throughout gestation. Using single-cell RNA sequencing (scRNA-seq), we explored the cellular responses in the embryonic brain to Cd exposure, and identified 18 distinct cell subpopulations that exhibited varied responses to Cd. Typically, Cd exposure impeded the development and maturation of cells in the brain, especially progenitor cells such as neural progenitor cells (NPCs) and oligodendrocyte progenitor cells (OPCs). It also caused significant cell subpopulation shifts in almost all the types of cells in the brain. Additionally, Cd exposure reduced the dendritic sophistication of cortical neurons in the offspring. Importantly, these changes led to aberrant Ca2+ activity in the cortex and neural behavior changes in mature offspring. These data contribute to our understanding of the effects and mechanisms of Cd exposure on brain development and highlight the importance of controlling environmental neurotoxicant exposure at the population level.
Collapse
Affiliation(s)
- Qinlong Ma
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhiqi Yang
- Brain Research Center, Army Medical University (Third Military Medical University), Chongqing, China
| | - Chuanyan Yang
- Brain Research Center, Army Medical University (Third Military Medical University), Chongqing, China
| | - Min Lin
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing, China
| | - Mingyue Gong
- Brain Research Center, Army Medical University (Third Military Medical University), Chongqing, China
| | - Ping Deng
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing, China
| | - Mindi He
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing, China
| | - Yonghui Lu
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing, China
| | - Kuan Zhang
- Brain Research Center, Army Medical University (Third Military Medical University), Chongqing, China
| | - Huifeng Pi
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing, China
| | - Mingyue Qu
- The PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Zhengping Yu
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhou Zhou
- Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing, China.
| | - Chunhai Chen
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing, China.
| |
Collapse
|
7
|
Khattab NA, El-Kadem AH, Elblehi SS, Ahmed El-Mahdy N, El-Shitany NA. The emerging role of α7nAChRs/caspase-3/Nrf-2 signaling pathway in citicoline improved autistic behavior induced by thimerosal in mice. Int Immunopharmacol 2024; 130:111736. [PMID: 38401462 DOI: 10.1016/j.intimp.2024.111736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 02/13/2024] [Accepted: 02/19/2024] [Indexed: 02/26/2024]
Abstract
AIMS Autism spectrum disorder (ASD) is a global concern,affecting around 75 million individuals.Various factors contribute to ASD,including mercury-containing preservatives like thimerosal (Thim) found in some vaccines.This study explored whether citicoline could be a therapeutic option for Thim-induced neuronal damage in a mouse model of ASD.Additionally,the study investigated the effects of citicoline on the α7nAChRs/Akt/Nrf2/caspase-3 pathway,which may be involved in the development of ASD. MATERIALS AND METHODS The study separated newborn mice into four groups.The control group received saline injections,while the Thim group received intramuscular injections of 3000 μg Hg/kg Thim on days 7,9,11,and 15 after birth.The two citicoline groups were administered Thim followed by intraperitoneal injections of 250 mg/kg or 500 mg/kg citicoline for three weeks.Afterward,various parameters were assessed, including growth,behavior,brain histopathology,oxidative stress,apoptotic,and inflammatory markers. KEY FINDINGS Untreated Thim-exposed mice exhibited significant brain damage,which was substantially alleviated by citicoline treatment.This beneficial effect was associated with increased expressions and concentrations of brain α7nAChRs and Akt, increased brain content of Nrf2, and the hippocampus contents of acetylcholine. Citicoline treatment decreased the brain levels of oxidative stress markers (MDA and NO),the apoptotic marker caspase-3,and pro-inflammatory markers (NF-κB,TNF-α,and IL-1β). The drug also increased the brain GPx activity. SIGNIFICANCE Based on the results of this study,the α7nAChRs pathway appears to be essential for the therapeutic effectiveness of citicoline in treating Thim-induced ASD in mice.
Collapse
Affiliation(s)
- Nourhan A Khattab
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt.
| | - Aya H El-Kadem
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| | - Samar S Elblehi
- Department of Pathology, Faculty of Veterinary Medicine, Alexandria University, Alexandria 22758, Egypt
| | - Nageh Ahmed El-Mahdy
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| | - Nagla A El-Shitany
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| |
Collapse
|
8
|
Gonçalves CL, Doifode T, Rezende VL, Costa MA, Rhoads JM, Soutullo CA. The many faces of microbiota-gut-brain axis in autism spectrum disorder. Life Sci 2024; 337:122357. [PMID: 38123016 DOI: 10.1016/j.lfs.2023.122357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/02/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023]
Abstract
The gut-brain axis is gaining more attention in neurodevelopmental disorders, especially autism spectrum disorder (ASD). Many factors can influence microbiota in early life, including host genetics and perinatal events (infections, mode of birth/delivery, medications, nutritional supply, and environmental stressors). The gut microbiome can influence blood-brain barrier (BBB) permeability, drug bioavailability, and social behaviors. Developing microbiota-based interventions such as probiotics, gastrointestinal (GI) microbiota transplantation, or metabolite supplementation may offer an exciting approach to treating ASD. This review highlights that RNA sequencing, metabolomics, and transcriptomics data are needed to understand how microbial modulators can influence ASD pathophysiology. Due to the substantial clinical heterogeneity of ASD, medical caretakers may be unlikely to develop a broad and effective general gut microbiota modulator. However, dietary modulation followed by administration of microbiota modulators is a promising option for treating ASD-related behavioral and gastrointestinal symptoms. Future work should focus on the accuracy of biomarker tests and developing specific psychobiotic agents tailored towards the gut microbiota seen in ASD patients, which may include developing individualized treatment options.
Collapse
Affiliation(s)
- Cinara L Gonçalves
- Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil.
| | - Tejaswini Doifode
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health (UTHealth), Houston, TX, USA
| | - Victoria L Rezende
- Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Maiara A Costa
- Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - J Marc Rhoads
- Department of Pediatrics, Division of Pediatric Gastroenterology, McGovern Medical School, The University of Texas Health (UTHealth), Houston, TX, USA
| | - Cesar A Soutullo
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health (UTHealth), Houston, TX, USA
| |
Collapse
|
9
|
EFSA Panel on Nutrition, Novel Foods and Food Allergens (NDA), Turck D, Bohn T, Castenmiller J, de Henauw S, Hirsch‐Ernst K, Knutsen HK, Maciuk A, Mangelsdorf I, McArdle HJ, Pentieva K, Siani A, Thies F, Tsabouri S, Vinceti M, Bornhorst J, Cubadda F, Dopter A, FitzGerald R, de Sesmaisons Lecarré A, das Neves Ferreira P, Fabiani L, Horvath Z, Matijević L, Naska A. Scientific opinion on the tolerable upper intake level for manganese. EFSA J 2023; 21:e8413. [PMID: 38075631 PMCID: PMC10704406 DOI: 10.2903/j.efsa.2023.8413] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2024] Open
Abstract
Following a request from the European Commission (EC), the EFSA Panel on Nutrition, Novel Foods and Food Allergens (NDA) was asked to deliver a scientific opinion on the tolerable upper intake level (UL) for manganese. Systematic reviews of the literature of human and animal data were conducted to assess evidence regarding excess manganese intake (including authorised manganese salts) and the priority adverse health effect, i.e. manganese-induced neurotoxicity. Available human and animal studies support neurotoxicity as a critical effect, however, data are not sufficient and suitable to characterise a dose-response relationship and identify a reference point for manganese-induced neurotoxicity. In the absence of adequate data to establish an UL, estimated background dietary intakes (i.e. manganese intakes from natural dietary sources only) observed among high consumers (95th percentile) were used to provide an indication of the highest level of intake where there is reasonable confidence on the absence of adverse effects. A safe level of intake of 8 mg/day was established for adults ≥ 18 years (including pregnant and lactating women) and ranged between 2 and 7 mg/day for other population groups. The application of the safe level of intake is more limited than an UL because the intake level at which the risk of adverse effects starts to increase is not defined.
Collapse
|
10
|
Posar A, Visconti P. Autism Spectrum Disorder in 2023: A Challenge Still Open. Turk Arch Pediatr 2023; 58:566-571. [PMID: 37850666 PMCID: PMC10724724 DOI: 10.5152/turkarchpediatr.2023.23194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 09/05/2023] [Indexed: 10/19/2023]
Abstract
In this paper, we provide an update on autism spectrum disorder (ASD), including epidemiology, etiopathogenesis, clinical presentation, instrumental investigations, early signs, onset patterns, neuropsychological hypotheses, treatments, and long-term outcome. The prevalence of this condition has increased enormously over the last few decades. This increase prompted a search for possible environmental factors whose effects would add up to a genetic predisposition leading to the development of autism. But the genetic and environmental variables involved are extremely numerous, and conclusive data regarding the etiopathogenesis are still far away. Assuming that a well-defined etiology is still found today only in a minority of cases, numerous pathogenetic mechanisms have been hypothesized. Among these, we mention oxidative stress, mitochondrial dysfunction, alteration of the intestinal microbiota, immune dysregulation, and neuroinflammation. These pathogenetic mechanisms could alter epigenetic status and gene expression, finally leading to ASD. Inherent in the term spectrum is the great clinical heterogeneity of this condition, mainly due to the frequent comorbidity that characterizes it. The earlier the diagnosis is made and the earlier psychoeducational treatment begins, the better the prognosis. In this sense, the role of pediatricians can be decisive in making children with signs suggestive of autism undergo a specialist diagnostic course. The development of increasingly advanced cognitive-behavioral educational techniques has considerably improved the prognosis of affected individuals, even though only a small minority of them come off the autistic spectrum. Pharmacological therapies are used to treat comorbidities. During childhood, the most important prognostic factor for long-term outcome seems to be intellectual functioning.
Collapse
Affiliation(s)
- Annio Posar
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOSI Disturbi dello Spettro Autistico, Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, Bologna University, Bologna, Italy
| | - Paola Visconti
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOSI Disturbi dello Spettro Autistico, Bologna, Italy
| |
Collapse
|
11
|
Mei Z, Liu G, Zhao B, He Z, Gu S. Emerging roles of epigenetics in lead-induced neurotoxicity. ENVIRONMENT INTERNATIONAL 2023; 181:108253. [PMID: 37864902 DOI: 10.1016/j.envint.2023.108253] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 09/19/2023] [Accepted: 10/06/2023] [Indexed: 10/23/2023]
Abstract
Lead is a common environmental heavy metal contaminant. Humans are highly susceptible to lead accumulation in the body, which causes nervous system damage and leads to a variety of nervous system diseases, such as Alzheimer's disease, Parkinson's disease, and autism spectrum disorder. Recent research has focused on the mechanisms of lead-induced neurotoxicity at multiple levels, including DNA methylation, histone modifications, and non-coding RNAs, which are involved in various lead-induced nervous system diseases. We reviewed the latest articles and summarised the emerging roles of DNA methylation, histone modification, and non-coding RNAs in lead-induced neurotoxicity. Our summary provides a theoretical basis and directions for future research on the prevention, diagnosis, and treatment of lead-induced neurological diseases.
Collapse
Affiliation(s)
- Zongqin Mei
- Institute of Preventive Medicine, School of Public Health, Dali University, No. 22, Wanhua Road, Dali, Yunnan 671000, People's Republic of China
| | - Guofen Liu
- Institute of Preventive Medicine, School of Public Health, Dali University, No. 22, Wanhua Road, Dali, Yunnan 671000, People's Republic of China
| | - Bo Zhao
- Institute of Preventive Medicine, School of Public Health, Dali University, No. 22, Wanhua Road, Dali, Yunnan 671000, People's Republic of China
| | - Zuoshun He
- Institute of Preventive Medicine, School of Public Health, Dali University, No. 22, Wanhua Road, Dali, Yunnan 671000, People's Republic of China.
| | - Shiyan Gu
- Institute of Preventive Medicine, School of Public Health, Dali University, No. 22, Wanhua Road, Dali, Yunnan 671000, People's Republic of China.
| |
Collapse
|
12
|
Herrera K, Maldonado-Ruiz R, Camacho-Morales A, de la Garza AL, Castro H. Maternal methyl donor supplementation regulates the effects of cafeteria diet on behavioral changes and nutritional status in male offspring. Food Nutr Res 2023; 67:9828. [PMID: 37920679 PMCID: PMC10619398 DOI: 10.29219/fnr.v67.9828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/30/2023] [Accepted: 09/01/2023] [Indexed: 11/04/2023] Open
Abstract
Background Nutritional status and maternal feeding during the perinatal and postnatal periods can program the offspring to develop long-term health alterations. Epidemiologic studies have demonstrated an association between maternal obesity and intellectual disability/cognitive deficits like autism spectrum disorders (ASDs) in offspring. Experimental findings have consistently been indicating that maternal supplementation with methyl donors, attenuated the social alterations and repetitive behavior in offspring. Objective This study aims to analyze the effect of maternal cafeteria diet and methyl donor-supplemented diets on social, anxiety-like, and repetitive behavior in male offspring, besides evaluating weight gain and food intake in both dams and male offspring. Design C57BL/6 female mice were randomized into four dietary formulas: control Chow (CT), cafeteria (CAF), control + methyl donor (CT+M), and cafeteria + methyl donor (CAF+M) during the pre-gestational, gestational, and lactation period. Behavioral phenotyping in the offspring was performed by 2-month-old using Three-Chamber Test, Open Field Test, and Marble Burying Test. Results We found that offspring prenatally exposed to CAF diet displayed less social interaction index when compared with subjects exposed to Chow diet (CT group). Notably, offspring exposed to CAF+M diet recovered social interaction when compared to the CAF group. Discussion These findings suggest that maternal CAF diet is efficient in promoting reduced social interaction in murine models. In our study, we hypothesized that a maternal methyl donor supplementation could improve the behavioral alterations expected in maternal CAF diet offspring. Conclusions The CAF diet also contributed to a social deficit and anxiety-like behavior in the offspring. On the other hand, a maternal methyl donor-supplemented CAF diet normalized the social interaction in the offspring although it led to an increase in anxiety-like behaviors. These findings suggest that a methyl donor supplementation could protect against aberrant social behavior probably targeting key genes related to neurotransmitter pathways.
Collapse
Affiliation(s)
- Katya Herrera
- Universidad Autonoma de Nuevo León, Facultad de Salud Pública y Nutrición, Centro de Investigación en Nutrición y Salud Pública. Monterrey, Nuevo León, México
| | - Roger Maldonado-Ruiz
- Universidad Autonoma de Nuevo Leon, Unidad de Neurometabolismo, Centro de Investigación y Desarrollo en Ciencias de la Salud. Monterrey, Nuevo León, México
| | - Alberto Camacho-Morales
- Universidad Autonoma de Nuevo Leon, Unidad de Neurometabolismo, Centro de Investigación y Desarrollo en Ciencias de la Salud. Monterrey, Nuevo León, México
- Universidad Autonoma de Nuevo Leon, Facultad de Medicina, Departamento de Bioquímica. Monterrey, Nuevo León, México
| | - Ana Laura de la Garza
- Universidad Autonoma de Nuevo Leon, Unidad de Nutrición, Centro de Investigación y Desarrollo en Ciencias de la Salud. Monterrey, Nuevo León, México
| | - Heriberto Castro
- Universidad Autonoma de Nuevo León, Facultad de Salud Pública y Nutrición, Centro de Investigación en Nutrición y Salud Pública. Monterrey, Nuevo León, México
- Universidad Autonoma de Nuevo Leon, Unidad de Nutrición, Centro de Investigación y Desarrollo en Ciencias de la Salud. Monterrey, Nuevo León, México
| |
Collapse
|
13
|
Grgurevic N. Testing the extreme male hypothesis in the valproate mouse model; sex-specific effects on plasma testosterone levels and tyrosine hydroxylase expression in the anteroventral periventricular nucleus, but not on parental behavior. Front Behav Neurosci 2023; 17:1107226. [PMID: 36818606 PMCID: PMC9932272 DOI: 10.3389/fnbeh.2023.1107226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 01/16/2023] [Indexed: 02/05/2023] Open
Abstract
Introduction Autism is a neurodevelopmental disorder with a strong male bias in prevalence and severity. The extreme male hypothesis proposed that autism is a manifestation of extreme male traits as evidenced by increased masculine behaviors, hypermasculinization of some brain regions, and alterations in androgen metabolism. In the present study, the extreme male hypothesis was tested in the valproate (VPA) mouse model. Methods Females of the C57BL/6JOlaHsd mouse strain were treated with 500 mg/kg VPA on gestational day 12. Offspring of both sexes were tested at 3 to 4 months of age in the elevated plus maze (EPM), open field, sociability tests, and for parental behavior. After sacrifice at 5 to 6 months of age, plasma testosterone was measured in males, while the brains of both sexes were examined for tyrosine hydroxylase (TH) expression in the anteroventral periventricular nucleus (AVPV). Results VPA treatment significantly increased plasma testosterone levels and decreased AVPV TH expression in males, whereas the expression of TH in females remained at the same level. In parental behavior test none of the pup-oriented behavior was affected by VPA treatment in both sexes, the exception was nest quality which was lower after VPA exposure in males, but not in females. Discussion Our results suggest a hypermasculinizing effect of VPA that occurred specifically in males but not in females, and this effect could be related to changes in androgen physiology. Nevertheless, a generalized interpretation of the extreme male hypothesis on brain and behavior should be avoided due to the complex effects of VPA.
Collapse
|
14
|
Lin J, de Rezende VL, de Aguiar da Costa M, de Oliveira J, Gonçalves CL. Cholesterol metabolism pathway in autism spectrum disorder: From animal models to clinical observations. Pharmacol Biochem Behav 2023; 223:173522. [PMID: 36717034 DOI: 10.1016/j.pbb.2023.173522] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/18/2022] [Accepted: 01/24/2023] [Indexed: 01/29/2023]
Abstract
Autism Spectrum Disorder (ASD) is a neurodevelopmental disorder characterized by a persistent impairment of social skills, including aspects of perception, interpretation, and response, combined with restricted and repetitive behavior. ASD is a complex and multifactorial condition, and its etiology could be attributed to genetic and environmental factors. Despite numerous clinical and experimental studies, no etiological factor, biomarker, and specific model of transmission have been consistently associated with ASD. However, an imbalance in cholesterol levels has been observed in many patients, more specifically, a condition of hypocholesterolemia, which seems to be shared between ASD and ASD-related genetic syndromes such as fragile X syndrome (FXS), Rett syndrome (RS), and Smith- Lemli-Opitz (SLO). Furthermore, it is known that alterations in cholesterol levels lead to neuroinflammation, oxidative stress, impaired myelination and synaptogenesis. Thus, the aim of this review is to discuss the cholesterol metabolic pathways in the ASD context, as well as in genetic syndromes related to ASD, through clinical observations and animal models. In fact, SLO, FXS, and RS patients display early behavioral markers of ASD followed by cholesterol disturbances. Several studies have demonstrated the role of cholesterol in psychiatric conditions and how its levels modulate brain neurodevelopment. This review suggests an important relationship between ASD pathology and cholesterol metabolism impairment; thus, some strategies could be raised - at clinical and pre-clinical levels - to explore whether cholesterol metabolism disturbance has a generally adverse effect in exacerbating the symptoms of ASD patients.
Collapse
Affiliation(s)
- Jaime Lin
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Victória Linden de Rezende
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Maiara de Aguiar da Costa
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Jade de Oliveira
- Laboratory for Research in Metabolic Disorders and Neurodegenerative Diseases, Graduate Program in Health Sciences, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Cinara Ludvig Gonçalves
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil.
| |
Collapse
|
15
|
Majerczyk D, Ayad E, Brewton K, Saing P, Hart P. Systemic maternal inflammation promotes ASD via IL-6 and IFN-γ. Biosci Rep 2022; 42:BSR20220713. [PMID: 36300375 PMCID: PMC9670245 DOI: 10.1042/bsr20220713] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 09/30/2022] [Accepted: 10/26/2022] [Indexed: 07/25/2023] Open
Abstract
Autism spectrum disorder (ASD) is a neurological disorder that manifests during early development, impacting individuals through their ways of communicating, social behaviors, and their ability to perform day-to-day activities. There have been different proposed mechanisms on how ASD precipitates within a patient, one of which being the impact cytokines have on fetal development once a mother's immune system has been activated (referred to as maternal immune activation, MIA). The occurrence of ASD has long been associated with elevated levels of several cytokines, including interleukin-6 (IL-6) and interferon gamma (IFN-γ). These proinflammatory cytokines can achieve high systemic levels in response to immune activating pathogens from various extrinsic sources. Transfer of cytokines such as IL-6 across the placental barrier allows accumulation in the fetus, potentially inducing neuroinflammation and consequently altering neurodevelopmental processes. Individuals who have been later diagnosed with ASD have been observed to have elevated levels of IL-6 and other proinflammatory cytokines during gestation. Moreover, the outcome of MIA has been associated with neurological effects such as impaired social interaction and an increase in repetitive behavior in animal models, supporting a mechanistic link between gestational inflammation and development of ASD-like characteristics. The present review attempts to provide a concise overview of the available preclinical and clinical data that suggest cross-talk between IL-6 and IFN-γ through both extrinsic and intrinsic factors as a central mechanism of MIA that may promote the development of ASD.
Collapse
Affiliation(s)
- Daniel Majerczyk
- College of Science, Health and Pharmacy, Roosevelt University, Illinois 60173, U.S.A
- Loyola Medicine, Berwyn, Illinois 60402, U.S.A
| | - Elizabeth G. Ayad
- College of Science, Health and Pharmacy, Roosevelt University, Illinois 60173, U.S.A
| | - Kari L. Brewton
- College of Science, Health and Pharmacy, Roosevelt University, Illinois 60173, U.S.A
| | - Pichrasmei Saing
- College of Science, Health and Pharmacy, Roosevelt University, Illinois 60173, U.S.A
| | - Peter C. Hart
- College of Science, Health and Pharmacy, Roosevelt University, Illinois 60173, U.S.A
| |
Collapse
|
16
|
Rahdar M, Hajisoltani R, Davoudi S, Karimi SA, Borjkhani M, Khatibi VA, Hosseinmardi N, Behzadi G, Janahmadi M. Alterations in the intrinsic discharge activity of CA1 pyramidal neurons associated with possible changes in the NADPH diaphorase activity in a rat model of autism induced by prenatal exposure to valproic acid. Brain Res 2022; 1792:148013. [PMID: 35841982 DOI: 10.1016/j.brainres.2022.148013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/24/2022] [Accepted: 07/10/2022] [Indexed: 11/02/2022]
Abstract
Autism spectrum disorder is a neurodevelopmental disorder characterized by sensory abnormalities, social skills impairment and cognitive deficits. Although recent evidence indicated that induction of autism-like behavior in animal models causes abnormal neuronal excitability, the impact of autism on neuronal properties is still an important issue. Thus, new findings at the cellular level may shed light on the pathophysiology of autism and may help to find effective treatment strategies. Here, we investigated the behavioral, electrophysiological and histochemical impacts of prenatal exposure to valproic acid (VPA) in rats. Findings revealed that VPA exposure caused a significant increase in the hot plate response latency. The novel object recognition ability was also impaired in VPA-exposed rats. Along with these behavioral alterations, neurons from VPA-exposed animals exhibited altered excitability features in response to depolarizing current injections relative to control neurons. In the VPA-exposed group, these changes consisted of a significant increase in the amplitude, evoked firing frequency and the steady-state standard deviation of spike timing of action potentials (APs). Moreover, the half-width, the AHP amplitude and the decay time constant of APs were significantly decreased in this group. These changes in the evoked electrophysiological properties were accompanied by intrinsic hyperexcitability and lower spike-frequency adaptation and also a significant increase in the number of NADPH-diaphorase stained neurons in the hippocampal CA1 area of the VPA-exposed rats. Taken together, findings demonstrate that abnormal nociception and recognition memory is associated with alterations in the neuronal responsiveness and nitrergic system in a rat model of autism-like.
Collapse
Affiliation(s)
- Mona Rahdar
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Razieh Hajisoltani
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Shima Davoudi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Asaad Karimi
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Tehran, Iran
| | - Mehdi Borjkhani
- Department of Electrical Engineering, Urmia University of Technology, Urmia, Iran
| | - Vahid Ahli Khatibi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Narges Hosseinmardi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Gila Behzadi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahyar Janahmadi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
17
|
Cuomo D, Foster MJ, Threadgill D. Systemic review of genetic and epigenetic factors underlying differential toxicity to environmental lead (Pb) exposure. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:35583-35598. [PMID: 35244845 PMCID: PMC9893814 DOI: 10.1007/s11356-022-19333-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 02/17/2022] [Indexed: 05/03/2023]
Abstract
Lead (Pb) poisoning is a major public health concern in environmental justice communities of the USA and in many developing countries. There is no identified safety threshold for lead in blood, as low-level Pb exposures can lead to severe toxicity in highly susceptible individuals and late onset of diseases from early-life exposure. However, identifying "susceptibility genes" or "early exposure biomarkers" remains challenging in human populations. There is a considerable variation in susceptibility to harmful effects from Pb exposure in the general population, likely due to the complex interplay of genetic and/or epigenetic factors. This systematic review summarizes current state of knowledge on the role of genetic and epigenetic factors in determining individual susceptibility in response to environmental Pb exposure in humans and rodents. Although a number of common genetic and epigenetic factors have been identified, the reviewed studies, which link these factors to various adverse health outcomes following Pb exposure, have provided somewhat inconsistent evidence of main health effects. Acknowledging the compelling need for new approaches could guide us to better characterize individual responses, predict potential adverse outcomes, and identify accurate and usable biomarkers for Pb exposure to improve mitigation therapies to reduce future adverse health outcomes of Pb exposure.
Collapse
Affiliation(s)
- Danila Cuomo
- Department of Molecular and Cellular Medicine, Texas A&M University, College Station, TX, USA.
| | - Margaret J Foster
- Medical Sciences Library, Texas A&M University, College Station, TX, USA
| | - David Threadgill
- Department of Molecular and Cellular Medicine and Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
18
|
Alymov AA, Kapitsa IG, Voronina TA. Behavioral Effects of Afobazole in an Experimental Model of Autism Spectrum Disorders. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022010185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
19
|
Varela RB, Cararo JH, Tye SJ, Carvalho AF, Valvassori SS, Fries GR, Quevedo J. Contributions of epigenetic inheritance to the predisposition of major psychiatric disorders: theoretical framework, evidence, and implications. Neurosci Biobehav Rev 2022; 135:104579. [DOI: 10.1016/j.neubiorev.2022.104579] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 01/10/2022] [Accepted: 02/11/2022] [Indexed: 02/08/2023]
|
20
|
Tasin FR, Ahmed A, Halder D, Mandal C. On-going consequences of in utero exposure of Pb: An epigenetic perspective. J Appl Toxicol 2022; 42:1553-1569. [PMID: 35023172 DOI: 10.1002/jat.4287] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 12/23/2021] [Accepted: 01/01/2022] [Indexed: 11/08/2022]
Abstract
Epigenetic modifications by toxic heavy metals are one of the intensively investigated fields of modern genomic research. Among a diverse group of heavy metals, lead (Pb) is an extensively distributed toxicant causing an immense number of abnormalities in the developing fetus via a wide variety of epigenetic changes. As a divalent cation, Pb can readily cross the placental membrane and the fetal blood brain barrier leading to far-reaching alterations in DNA methylation patterns, histone protein modifications and micro-RNA expression. Over recent years, several human cohorts and animal model studies have documented hyper- and hypo-methylation of developmental genes along with altered DNA methyl-transferase expression by in utero Pb exposure in a dose-, duration- and sex-dependent manner. Modifications in the expression of specific histone acetyltransferase enzymes along with histone acetylation and methylation levels have been reported in rodent and murine models. Apart from these, down-regulation and up-regulation of certain microRNAs crucial for fetal development have been shown to be associated with in utero Pb exposure in human placenta samples. All these modifications in the developing fetus during the prenatal and perinatal stages reportedly caused severe abnormalities in early or adult age, such as - impaired growth, obesity, autism, diabetes, cardiovascular diseases, risks of cancer development and Alzheimer's disease. In this review, currently available information on Pb-mediated alterations in the fetal epigenome is summarized. Further research on Pb-induced epigenome modification will help to understand the mechanisms in detail and will enable us to formulate safety guidelines for pregnant women and developing children.
Collapse
Affiliation(s)
- Fahim Rejanur Tasin
- Biotechnology and Genetic Engineering Discipline, Khulna University, Khulna, Bangladesh
| | - Asif Ahmed
- Biotechnology and Genetic Engineering Discipline, Khulna University, Khulna, Bangladesh
| | - Debasish Halder
- Rare Disease research center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Chanchal Mandal
- Biotechnology and Genetic Engineering Discipline, Khulna University, Khulna, Bangladesh
| |
Collapse
|
21
|
Abstract
PURPOSE OF REVIEW At elevated levels, the essential element manganese (Mn) is neurotoxic and increasing evidence indicates that environmental Mn exposure early in life negatively affects neurodevelopment. In this review, we describe how underlying genetics may confer susceptibility to elevated Mn concentrations and how the epigenetic effects of Mn may explain the association between Mn exposure early in life and its toxic effects later in life. RECENT FINDINGS Common polymorphisms in the Mn transporter genes SLC30A10 and SLC39A8 seem to have a large impact on intracellular Mn levels and, in turn, neurotoxicity. Genetic variation in iron regulatory genes may to lesser extent also influence Mn levels and toxicity. Recent studies on Mn and epigenetic mechanisms indicate that Mn-related changes in DNA methylation occur early in life. One human and two animal studies found persistent changes from in utero exposure to Mn but whether these changes have functional effects remains unknown. Genetics seems to play a major role in susceptibility to Mn toxicity and should therefore be considered in risk assessment. Mn appears to interfere with epigenetic processes, potentially leading to persistent changes in developmental programming, which warrants further study.
Collapse
|
22
|
Pugsley K, Scherer SW, Bellgrove MA, Hawi Z. Environmental exposures associated with elevated risk for autism spectrum disorder may augment the burden of deleterious de novo mutations among probands. Mol Psychiatry 2022; 27:710-730. [PMID: 34002022 PMCID: PMC8960415 DOI: 10.1038/s41380-021-01142-w] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 04/16/2021] [Accepted: 04/21/2021] [Indexed: 12/11/2022]
Abstract
Although the full aetiology of autism spectrum disorder (ASD) is unknown, familial and twin studies demonstrate high heritability of 60-90%, indicating a predominant role of genetics in the development of the disorder. The genetic architecture of ASD consists of a complex array of rare and common variants of all classes of genetic variation usually acting additively to augment individual risk. The relative contribution of heredity in ASD persists despite selective pressures against the classic autistic phenotype; a phenomenon thought to be explained, in part, by the incidence of spontaneous (or de novo) mutations. Notably, environmental exposures attributed as salient risk factors for ASD may play a causal role in the emergence of deleterious de novo variations, with several ASD-associated agents having significant mutagenic potential. To explore this hypothesis, this review article assesses published epidemiological data with evidence derived from assays of mutagenicity, both in vivo and in vitro, to determine the likely role such agents may play in augmenting the genetic liability in ASD. Broadly, these exposures were observed to elicit genomic alterations through one or a combination of: (1) direct interaction with genetic material; (2) impaired DNA repair; or (3) oxidative DNA damage. However, the direct contribution of these factors to the ASD phenotype cannot be determined without further analysis. The development of comprehensive prospective birth cohorts in combination with genome sequencing is essential to forming a causal, mechanistic account of de novo mutations in ASD that links exposure, genotypic alterations, and phenotypic consequences.
Collapse
Affiliation(s)
- Kealan Pugsley
- grid.1002.30000 0004 1936 7857Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Melbourne, VIC Australia
| | - Stephen W. Scherer
- grid.42327.300000 0004 0473 9646The Centre for Applied Genomics and Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938McLaughlin Centre and Department of Molecular Genetics, University of Toronto, Toronto, ON Canada
| | - Mark A. Bellgrove
- grid.1002.30000 0004 1936 7857Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Melbourne, VIC Australia
| | - Ziarih Hawi
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
23
|
Amadi CN, Orish CN, Frazzoli C, Orisakwe OE. Association of autism with toxic metals: A systematic review of case-control studies. Pharmacol Biochem Behav 2021; 212:173313. [PMID: 34896416 DOI: 10.1016/j.pbb.2021.173313] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 12/05/2021] [Accepted: 12/06/2021] [Indexed: 02/07/2023]
Abstract
Environmental factors have been associated with the etiology of autism spectrum disorder ASD in recent times. The involvement of toxic metals in the generation of reactive oxygen species and their epigenetics effects have been implicated in ASD. This systemic review examines the association of toxic metals with autism in children. A systematic literature search was performed in scientific databases such as PubMed, Google scholar, and Scopus. Case-control studies evaluating toxic metal levels in different tissues of ASD children and comparing them to healthy children (control group) were identified. The Newcastle-Ottawa Scale was used to evaluate the risk of bias of the included studies. Six case-control studies with 425 study subjects met our inclusion criteria. A total of four studies indicated higher levels of As, Pb, Hg, Cd, Al, Sn, Sb, Ba, TI, W, and Zr in whole blood, RBC, in whole blood, RBC, and hair samples of children with autism compared with control suggestive of a greater toxic metal exposure (immediate and long-term). Three studies identified significantly higher concentrations of Cd, Pb and Hg in urine and hair samples of autistic children compared to control suggesting decreased excretion and possible high body burden of these metals. The findings from this review demonstrate that high levels of toxic metals are associated with ASD, therefore, critical care is necessary to reduce body burden of these metals in children with ASD as a major therapeutic strategy.
Collapse
Affiliation(s)
- Cecilia N Amadi
- Department of Experimental Pharmacology & Toxicology, Faculty of Pharmacy, University of Port Harcourt, PMB, 5323 Port Harcourt, Choba, Nigeria
| | - Chinna N Orish
- Department of Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, University of Port Harcourt, PMB, 5323 Port Harcourt, Choba, Nigeria
| | - Chiara Frazzoli
- Department for Cardiovascular, Dysmetabolic and Aging Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Orish E Orisakwe
- Department of Experimental Pharmacology & Toxicology, Faculty of Pharmacy, University of Port Harcourt, PMB, 5323 Port Harcourt, Choba, Nigeria; African Centre of Excellence for Public Health and Toxicological Research (ACE-PUTOR), University of Port Harcourt, PMB, 5323 Port Harcourt, Choba, Nigeria.
| |
Collapse
|
24
|
Adjimann TS, Argañaraz CV, Soiza-Reilly M. Serotonin-related rodent models of early-life exposure relevant for neurodevelopmental vulnerability to psychiatric disorders. Transl Psychiatry 2021; 11:280. [PMID: 33976122 PMCID: PMC8113523 DOI: 10.1038/s41398-021-01388-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 04/16/2021] [Accepted: 04/21/2021] [Indexed: 01/22/2023] Open
Abstract
Mental disorders including depression and anxiety are continuously rising their prevalence across the globe. Early-life experience of individuals emerges as a main risk factor contributing to the developmental vulnerability to psychiatric disorders. That is, perturbing environmental conditions during neurodevelopmental stages can have detrimental effects on adult mood and emotional responses. However, the possible maladaptive neural mechanisms contributing to such psychopathological phenomenon still remain poorly understood. In this review, we explore preclinical rodent models of developmental vulnerability to psychiatric disorders, focusing on the impact of early-life environmental perturbations on behavioral aspects relevant to stress-related and psychiatric disorders. We limit our analysis to well-established models in which alterations in the serotonin (5-HT) system appear to have a crucial role in the pathophysiological mechanisms. We analyze long-term behavioral outcomes produced by early-life exposures to stress and psychotropic drugs such as the selective 5-HT reuptake inhibitor (SSRI) antidepressants or the anticonvulsant valproic acid (VPA). We perform a comparative analysis, identifying differences and commonalities in the behavioral effects produced in these models. Furthermore, this review discusses recent advances on neurodevelopmental substrates engaged in these behavioral effects, emphasizing the possible existence of maladaptive mechanisms that could be shared by the different models.
Collapse
Affiliation(s)
- Tamara S. Adjimann
- grid.7345.50000 0001 0056 1981Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Carla V. Argañaraz
- grid.7345.50000 0001 0056 1981Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mariano Soiza-Reilly
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
25
|
Cowell W, Colicino E, Levin-Schwartz Y, Enlow MB, Amarasiriwardena C, Andra SS, Gennings C, Wright RO, Wright RJ. Prenatal metal mixtures and sex-specific infant negative affectivity. Environ Epidemiol 2021; 5:e147. [PMID: 33870019 PMCID: PMC8043734 DOI: 10.1097/ee9.0000000000000147] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 03/04/2021] [Indexed: 11/27/2022] Open
Abstract
Prenatal exposure to metals has been associated with a range of adverse neurocognitive outcomes; however, associations with early behavioral development are less well understood. We examined joint exposure to multiple co-occurring metals in relation to infant negative affect, a stable temperamental trait linked to psychopathology among children and adults. METHODS Analyses included 308 mother-infant pairs enrolled in the PRISM pregnancy cohort. We measured As, Ba, Cd, Cs, Cr, Pb, and Sb in urine, collected on average during late pregnancy, by ICP-MS. At age 6 months, we assessed negative affect using the Infant Behavior Questionnaire-Revised. We used Weighted Quantile Sum (WQS) regression with repeated holdout validation to estimate the joint association between the metals and global negative affectivity, as well as four subdomains (Fear, Sadness, Distress to Limitations, and Falling Reactivity). We also tested for a sex interaction with estimated stratified weights. RESULTS In adjusted models, urinary metals were associated with higher scores on the Fear scale (βWQS = 0.20, 95% confidence interval [CI]: 0.09, 0.30), which captures behavioral inhibition, characterized by startle or distress to sudden changes in the environment and inhibited approach to novelty. We observed a significant sex interaction (95% CI for the cross-product term: -0.19, -0.01), and stratified weights showed girls (61.6%) contributed substantially more to the mixture effect compared with boys (38.4%). Overall, Ba contributed the greatest mixture weight (22.5%), followed by Cs (14.9%) and As (14.6%). CONCLUSIONS Prenatal exposure to metals was associated with increased infant scores on the temperamental domain of fear, with girls showing particular sensitivity.Key words: Prenatal; Metals; Mixtures; Temperament; Infancy; Negative affect.
Collapse
Affiliation(s)
- Whitney Cowell
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Elena Colicino
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Yuri Levin-Schwartz
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Michelle Bosquet Enlow
- Department of Psychiatry, Boston Children’s Hospital, Boston, MA
- Department of Psychiatry, Harvard Medical School, Boston, MA
| | - Chitra Amarasiriwardena
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Syam S. Andra
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Chris Gennings
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY
- Institute for Exposomic Research, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Robert O. Wright
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY
- Institute for Exposomic Research, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Rosalind J. Wright
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY
- Institute for Exposomic Research, Icahn School of Medicine at Mount Sinai, New York, NY
- Kravis Children’s Hospital, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
26
|
Shabani S. A mechanistic view on the neurotoxic effects of air pollution on central nervous system: risk for autism and neurodegenerative diseases. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:6349-6373. [PMID: 33398761 DOI: 10.1007/s11356-020-11620-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Accepted: 11/09/2020] [Indexed: 06/12/2023]
Abstract
Many reports have shown a strong association between exposure to neurotoxic air pollutants like heavy metal and particulate matter (PM) as an active participant and neurological disorders. While the effects of these toxic pollutants on cardiopulmonary morbidity have principally been studied, growing evidence has shown that exposure to polluted air is associated with memory impairment, communication deficits, and anxiety/depression among all ages. So, these toxic pollutants in the environment increase the risk of neurodegenerative disease, ischemia, and autism spectrum disorders (ASD). The precise mechanisms in which air pollutants lead to communicative inability, social inability, and declined cognition have remained unknown. Various animal model studies show that amyloid precursor protein (APP), processing, oxidant/antioxidant balance, and inflammation pathways change following the exposure to constituents of polluted air. In the present review study, we collect the probable molecular mechanisms of deleterious CNS effects in response to various air pollutants.
Collapse
Affiliation(s)
- Sahreh Shabani
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| |
Collapse
|
27
|
Zhou H, Lin Y, Zhao W, Teng Y, Cui Y, Wang T, Li C, Jiang YH, Zhang JJ, Wang Y. The role of Hipk2-p53 pathways in arsenic-induced autistic behaviors: A translational study from rats to humans. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 267:115568. [PMID: 33254717 DOI: 10.1016/j.envpol.2020.115568] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/25/2020] [Accepted: 08/27/2020] [Indexed: 06/12/2023]
Abstract
Previous studies have associated the risk of autism spectrum disorder (ASD) with increased exposures to metals and metalloids such as arsenic. In this study, we used an animal-to-human translational strategy to identify key molecular changes that potentially mediated the effects of arsenic exposures on ASD development. In a previously established rat model, we have induced autistic behaviors in rat pups with gestational arsenic exposures (10 and 45 μg/L As2O3 in drinking water). Neuronal apoptosis and the associated epigenetic dysregulations in frontal cortex were assayed to screen potential mediating pathways, which were subsequently validated with qPCR, western blotting, and immunohistochemistry analyses. Furthermore, the identified pathway, along with serum levels of 26 elements including arsenic, were characterized in a case-control study with 21 ASD children and 21 age-matched healthy controls. In animals, we found that arsenic exposures caused difficulties of social interaction and increased stereotypic behaviors in a dose-dependent manner, accompanied by increased neuronal apoptosis and upregulation of Hipk2-p53 pathway in the frontal cortex. In humans, we found that serum levels of Hipk2 and p53 were 24.7 (95%CI: 8.5 to 43.4) % and 23.7 (95%CI: 10.5 to 38.5) % higher in ASD children than in healthy controls. ASD children had significantly higher serum levels of 15 elements, among which arsenic, silicon, strontium, and vanadium were positively associated with both Hipk2 and p53. Results from both the rat arsenic exposure and human case-control studies suggest a likely role of Hipk2-p53 pathway in ASD development induced by exposures to environmental pollutants such as arsenic.
Collapse
Affiliation(s)
- Hao Zhou
- Department of Pediatrics, Guizhou Provincial People's Hospital, Medical College of Guizhou University, Guiyang, 550002, China; Department of Neurology, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Yan Lin
- Global Health Institute & Nicholas School of the Environment, Duke University, Durham, NC, 27705, USA
| | - Weiqing Zhao
- Department of Pediatrics, Guizhou Provincial People's Hospital, Medical College of Guizhou University, Guiyang, 550002, China
| | - Yanbo Teng
- Global Health Research Center, Duke Kunshan University, Jiangsu Province, China
| | - Yuxia Cui
- Department of Pediatrics, Guizhou Provincial People's Hospital, Medical College of Guizhou University, Guiyang, 550002, China
| | - Tianqi Wang
- Department of Neurology, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Chunpei Li
- Department of Neurology, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Yong-Hui Jiang
- Department of Genetics and Pediatrics, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Junfeng Jim Zhang
- Global Health Institute & Nicholas School of the Environment, Duke University, Durham, NC, 27705, USA; Global Health Research Center, Duke Kunshan University, Jiangsu Province, China.
| | - Yi Wang
- Department of Neurology, Children's Hospital of Fudan University, Shanghai, 201102, China.
| |
Collapse
|
28
|
Chaliha D, Albrecht M, Vaccarezza M, Takechi R, Lam V, Al-Salami H, Mamo J. A Systematic Review of the Valproic-Acid-Induced Rodent Model of Autism. Dev Neurosci 2020; 42:12-48. [DOI: 10.1159/000509109] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 06/02/2020] [Indexed: 11/19/2022] Open
|
29
|
Keever MR, Zhang P, Bolt CR, Antonson AM, Rymut HE, Caputo MP, Houser AK, Hernandez AG, Southey BR, Rund LA, Johnson RW, Rodriguez-Zas SL. Lasting and Sex-Dependent Impact of Maternal Immune Activation on Molecular Pathways of the Amygdala. Front Neurosci 2020; 14:774. [PMID: 32848554 PMCID: PMC7431923 DOI: 10.3389/fnins.2020.00774] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 07/01/2020] [Indexed: 12/23/2022] Open
Abstract
The prolonged and sex-dependent impact of maternal immune activation (MIA) during gestation on the molecular pathways of the amygdala, a brain region that influences social, emotional, and other behaviors, is only partially understood. To address this gap, we investigated the effects of viral-elicited MIA during gestation on the amygdala transcriptome of pigs, a species of high molecular and developmental homology to humans. Gene expression levels were measured using RNA-Seq on the amygdala for 3-week-old female and male offspring from MIA and control groups. Among the 403 genes that exhibited significant MIA effect, a prevalence of differentially expressed genes annotated to the neuroactive ligand-receptor pathway, glutamatergic functions, neuropeptide systems, and cilium morphogenesis were uncovered. Genes in these categories included corticotropin-releasing hormone receptor 2, glutamate metabotropic receptor 4, glycoprotein hormones, alpha polypeptide, parathyroid hormone 1 receptor, vasointestinal peptide receptor 2, neurotensin, proenkephalin, and gastrin-releasing peptide. These categories and genes have been associated with the MIA-related human neurodevelopmental disorders, including schizophrenia and autism spectrum disorders. Gene network reconstruction highlighted differential vulnerability to MIA effects between sexes. Our results advance the understanding necessary for the development of multifactorial therapies targeting immune modulation and neurochemical dysfunction that can ameliorate the effects of MIA on offspring behavior later in life.
Collapse
Affiliation(s)
- Marissa R. Keever
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Pan Zhang
- Illinois Informatics Institute, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Courtni R. Bolt
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Adrienne M. Antonson
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Haley E. Rymut
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Megan P. Caputo
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Alexandra K. Houser
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Alvaro G. Hernandez
- High-throughput Sequencing and Genotyping Unit, Roy J. Carver Biotechnology Center, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Bruce R. Southey
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Laurie A. Rund
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Rodney W. Johnson
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Sandra L. Rodriguez-Zas
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- Illinois Informatics Institute, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- Department of Statistics, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| |
Collapse
|
30
|
Hsu CC, Madsen TE, O'Gorman E, Gourley SL, Rainnie DG. Reward-related dynamical coupling between basolateral amygdala and nucleus accumbens. Brain Struct Funct 2020; 225:1873-1888. [PMID: 32556583 PMCID: PMC7405940 DOI: 10.1007/s00429-020-02099-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 06/06/2020] [Indexed: 12/28/2022]
Abstract
Recognizing reward-related stimuli is crucial for survival. Neuronal projections from the basolateral amygdala (BLA) to the nucleus accumbens (NAc) play an important role in processing reward-related cues. Previous studies revealed synchronization between distant brain regions in reward-sensitive neurocircuits; however, whether the NAc synchronizes with the BLA is unknown. Here, we recorded local field potentials simultaneously from the BLA and NAc of rats during social preference tests and an appetitive conditioning test in which explicit stimuli were associated with food. BLA-NAc coherence in the theta band (5-8 Hz) increased in response to food-associated cues. Meanwhile, the modulatory strength of theta-high gamma (50-110 Hz) phase-amplitude cross-frequency coupling (PAC) in the NAc decreased. Importantly, both of these neuromodulations disappeared upon extinction. In contrast, both theta and gamma power oscillations in each region increased in the presence of social conspecifics or contexts associated with conspecifics, but coherence did not change. To potentially disrupt behavior and associated neural activity, a subgroup of rats was exposed prenatally to valproic acid (VPA), which has been shown to disrupt transcriptome and excitatory/inhibitory balance in the amygdala. VPA-exposed rats demonstrated impulsive-like behavior, but VPA did not affect BLA-NAc coherence. These findings reveal changes in BLA-NAc coherence in response to select reward-related stimuli (i.e., food-predictive cues); the differences between the tasks used here could shed light onto the functional nature of BLA-NAc coherence and are discussed.
Collapse
Affiliation(s)
- Chia-Chun Hsu
- Yerkes National Primate Research Center, Emory University, Atlanta, USA
- Neuroscience Graduate Program, Emory University, Atlanta, USA
- Department of Pediatrics, Emory University, Atlanta, USA
| | - Teresa E Madsen
- Yerkes National Primate Research Center, Emory University, Atlanta, USA
| | | | - Shannon L Gourley
- Yerkes National Primate Research Center, Emory University, Atlanta, USA.
- Neuroscience Graduate Program, Emory University, Atlanta, USA.
- Department of Pediatrics, Emory University, Atlanta, USA.
| | - Donald G Rainnie
- Yerkes National Primate Research Center, Emory University, Atlanta, USA.
| |
Collapse
|
31
|
Prenatal S-Adenosine Methionine (SAMe) Induces Changes in Gene Expression in the Brain of Newborn Mice That Are Prevented by Co-Administration of Valproic Acid (VPA). Int J Mol Sci 2020; 21:ijms21082834. [PMID: 32325788 PMCID: PMC7215397 DOI: 10.3390/ijms21082834] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/13/2020] [Accepted: 04/14/2020] [Indexed: 12/12/2022] Open
Abstract
In previous studies, we produced changes in gene expression in the brain of mice by early postnatal administration of valproic acid (VPA), with distinct differences between genders. The addition of S-adenosine methionine (SAMe) normalized the expression of most genes in both genders, while SAMe alone induced no changes. We treated pregnant dams with a single injection of VPA on day 12.5 of gestation, or with SAMe during gestational days 12–14, or by a combination of VPA and SAMe. In the frontal half of the brain, we studied the expression of 770 genes of the pathways involved in neurophysiology and neuropathology using the NanoString nCounter method. SAMe, but not VPA, induced statistically significant changes in the expression of many genes, with differences between genders. The expression of 112 genes was changed in both sexes, and another 170 genes were changed only in females and 31 only in males. About 30% of the genes were changed by more than 50%. One of the most important pathways changed by SAMe in both sexes was the VEGF (vascular endothelial growth factor) pathway. Pretreatment with VPA prevented almost all the changes in gene expression induced by SAMe. We conclude that large doses of SAMe, if administered prenatally, may induce significant epigenetic changes in the offspring. Hence, SAMe and possibly other methyl donors may be epigenetic teratogens.
Collapse
|
32
|
Gender Related Changes in Gene Expression Induced by Valproic Acid in A Mouse Model of Autism and the Correction by S-adenosyl Methionine. Does It Explain the Gender Differences in Autistic Like Behavior? Int J Mol Sci 2019; 20:ijms20215278. [PMID: 31652960 PMCID: PMC6862653 DOI: 10.3390/ijms20215278] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 10/21/2019] [Accepted: 10/22/2019] [Indexed: 12/14/2022] Open
Abstract
In previous studies we produced autism like behavioral changes in mice by Valproic acid (VPA) with significant differences between genders. S-adenosine methionine (SAM) prevented the autism like behavior in both genders. The expression of 770 genes of pathways involved in neurophysiology and neuropathology was studied in the prefrontal cortex of 60 days old male and female mice using the NanoString nCounter. In females, VPA induced statistically significant changes in the expression of 146 genes; 71 genes were upregulated and 75 downregulated. In males, VPA changed the expression of only 19 genes, 16 were upregulated and 3 downregulated. Eight genes were similarly changed in both genders. When considering only the genes that were changed by at least 50%, VPA changed the expression of 15 genes in females and 3 in males. Only Nts was similarly downregulated in both genders. SAM normalized the expression of most changed genes in both genders. We presume that genes that are involved in autism like behavior in our model were similarly changed in both genders and corrected by SAM. The behavioral and other differences between genders may be related to genes that were differently affected by VPA in males and females and/or differently affected by SAM.
Collapse
|
33
|
Zheng W, Hu Y, Chen D, Li Y, Wang S. [Improvement of a mouse model of valproic acid-induced autism]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2019; 39:718-723. [PMID: 31270052 DOI: 10.12122/j.issn.1673-4254.2019.06.14] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To establish an improved mouse model of valproic acid (VPA)-induced autism that better mimics human autism. METHODS We established mouse models of autism in female C57 mice by intraperitoneal injection of sodium valproate either at a single dose (600 mg/kg) on day 12.5 after conception (conventional group) or in two doses of 300 mg/kg each on days 10 and 12 after conception (modified group), and the control mice were injected with saline only on day 12.5. The responses of the mice to VPA injection, the uterus, mortality rate, and abortion rate were compared among the 3 groups. The morphology and development of the offspring mice were assessed, and their behavioral ontogeny was evaluated using 3- chambered social test, social test, juvenil play test, and open field test. RESULTS The mortality and abortion rates were significantly lower in the modified model group than in the conventional group (P < 0.01). Compared with those in the control group, the offspring mice in both the conventional group and the modified group showed developmental disorders (P < 0.05). The mortality rate of the newborn mice was significantly lower in the modified group than in the conventional group with a rate of curvy tail of up to 100% (P < 0.001). The offspring mice in both the modified group and conventional group exhibited autism-like behavioral abnormalities, including social disorder and repetitive stereotyped behavior (P < 0.05). CONCLUSIONS The mouse model of autism established using the modified method better mimics human autism with reduced mortality and abortion rates of the pregnant mice and also decreased mortality rate of the newborn mice.
Collapse
Affiliation(s)
- Wenxia Zheng
- College of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yuling Hu
- College of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Di Chen
- College of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yingbo Li
- College of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Shali Wang
- College of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
34
|
Khalid M, Abdollahi M. Epigenetic modifications associated with pathophysiological effects of lead exposure. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART C, ENVIRONMENTAL CARCINOGENESIS & ECOTOXICOLOGY REVIEWS 2019; 37:235-287. [PMID: 31402779 DOI: 10.1080/10590501.2019.1640581] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Lead (Pb) exposure during different stages of development has demonstrated dose, duration, sex, and tissue-specific pathophysiological outcomes due to altered epigenetic regulation via (a) DNA methylation, (b) histone modifications, (c) miRNAs, and (d) chromatin accessibility. Pb-induced alteration of epigenetic regulation causes neurotoxic and extra-neurotoxic pathophysiological outcomes. Neurotoxic effects of Pb include dysfunction of memory and learning, behavioral disorder, attention deficit hyperactivity disorder, autism spectrum disorder, aging, Alzheimer's disease, tauopathy, and neurodegeneration. Extra-neurotoxic effects of Pb include altered body weight, metabolic disorder, cardiovascular disorders, hematopoietic disorder, and reproductive impairment. Pb exposure either early in life or at any stage of development results in undesirable pathophysiological outcomes that tends to sustain and maintain for a lifetime.
Collapse
Affiliation(s)
- Madiha Khalid
- Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mohammad Abdollahi
- Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
35
|
Avram S, Udrea AM, Negrea A, Ciopec M, Duteanu N, Postolache C, Duda-Seiman C, Duda-Seiman D, Shaposhnikov S. Prevention of Deficit in Neuropsychiatric Disorders through Monitoring of Arsenic and Its Derivatives as Well as Through Bioinformatics and Cheminformatics. Int J Mol Sci 2019; 20:ijms20081804. [PMID: 31013686 PMCID: PMC6514589 DOI: 10.3390/ijms20081804] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 04/04/2019] [Accepted: 04/09/2019] [Indexed: 12/26/2022] Open
Abstract
Neuropsychiatric disorders are induced by various risk factors, including direct exposure to environmental chemicals. Arsenic exposure induces neurodegeneration and severe psychiatric disorders, but the molecular mechanisms by which brain damage is induced are not yet elucidated. Our aim is to better understand the molecular mechanisms of arsenic toxicity in the brain and to elucidate possible ways to prevent arsenic neurotoxicity, by reviewing significant experimental, bioinformatics, and cheminformatics studies. Brain damage induced by arsenic exposure is discussed taking in account: the correlation between neuropsychiatric disorders and the presence of arsenic and its derivatives in the brain; possible molecular mechanisms by which arsenic induces disturbances of cognitive and behavioral human functions; and arsenic influence during psychiatric treatments. Additionally, we present bioinformatics and cheminformatics tools used for studying brain toxicity of arsenic and its derivatives, new nanoparticles used as arsenic delivery systems into the human body, and experimental ways to prevent arsenic contamination by its removal from water. The main aim of the present paper is to correlate bioinformatics, cheminformatics, and experimental information on the molecular mechanism of cerebral damage induced by exposure to arsenic, and to elucidate more efficient methods used to reduce its toxicity in real groundwater.
Collapse
Affiliation(s)
- Speranta Avram
- Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, 050095 Bucharest, Romania.
| | - Ana Maria Udrea
- National Institute for Laser Plasma and Radiation Physics, Atomistilor Street 409, 077125 Magurele, Romania.
| | - Adina Negrea
- Politehnica University of Timisoara, Faculty of Industrial Chemistry and Environmental Engineering, Piata Victoriei, 2, 300006 Timisoara, Romania.
| | - Mihaela Ciopec
- Politehnica University of Timisoara, Faculty of Industrial Chemistry and Environmental Engineering, Piata Victoriei, 2, 300006 Timisoara, Romania.
| | - Narcis Duteanu
- Politehnica University of Timisoara, Faculty of Industrial Chemistry and Environmental Engineering, Piata Victoriei, 2, 300006 Timisoara, Romania.
| | - Carmen Postolache
- Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, 050095 Bucharest, Romania.
| | - Corina Duda-Seiman
- Faculty of Chemistry, Biology, Geography, West University of Timișoara, I.H.Pestalozzi 16, 300115 Timisoara, Romania.
| | - Daniel Duda-Seiman
- University of Medicine and Pharmacy "Victor Babes, Timişoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania.
| | | |
Collapse
|
36
|
Anacker AMJ, Moran JT, Santarelli S, Forsberg CG, Rogers TD, Stanwood GD, Hall BJ, Delpire E, Veenstra-VanderWeele J, Saxe MD. Enhanced Social Dominance and Altered Neuronal Excitability in the Prefrontal Cortex of Male KCC2b Mutant Mice. Autism Res 2019; 12:732-743. [PMID: 30977597 DOI: 10.1002/aur.2098] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 03/06/2019] [Accepted: 03/13/2019] [Indexed: 01/20/2023]
Abstract
The K-Cl cotransporter KCC2 is essential in the development of the "GABA switch" that produces a change in neuronal responses to GABA signaling from excitatory to inhibitory early in brain development, and alterations in this progression have previously been hypothesized to play a causal role in autism spectrum disorder (ASD). We investigated the KCC2b (Slc12a5) heterozygous knockout mouse using a battery of rodent behavioral tests relevant to core and comorbid ASD symptoms. Compared to wild-type littermates, KCC2+/- mice were normal in standard measures of locomotor activity, grooming and digging behaviors, and social, vocalization, and anxiety-like behaviors. However, KCC2+/- mice exhibited increased social dominance behaviors and increased amplitude of spontaneous postsynaptic currents in the medial prefrontal cortex (PFC) that were previously implicated in governing social hierarchy and dominance behaviors. Treatment of wild-type mouse brain slices with the KCC2 inhibitor VU0240511 increased the amplitude and frequency of excitatory postsynaptic currents, partially recapitulating the phenotype of KCC2+/- mice. These findings indicate that the activity of KCC2 plays a role in social dominance, in parallel with effects on PFC signaling, further suggesting that KCC2 function has some relevance to social behavior but without the breadth of impact on autism-like behavior suggested by previous studies. Further testing could assess whether KCC2 alters other circuits and whether additional factors such as environmental insults may precipitate autism-related behavioral phenotypes. Autism Research 2019, 12: 732-743. © 2019 International Society for Autism Research, Wiley Periodicals, Inc. LAY SUMMARY: A mouse model of altered chloride transporter expression was used to look for a role in behaviors and brain function relevant to autism. There was an imbalance in signaling in the prefrontal cortex, and increased social dominance behavior, although other autism-related behaviors were not changed. These findings indicate that altered chloride transporter function affects prefrontal cortex function and social dominance without a broader impact on autism-like behaviors.
Collapse
Affiliation(s)
- Allison M J Anacker
- Division of Child & Adolescent Psychiatry, New York State Psychiatric Institute, Columbia University, New York, New York
| | - Jacqueline T Moran
- Roche Pharmaceutical Research and Early Development, Neuroscience, Ophthalmology and Rare Diseases, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland.,Tulane University Department of Cell and Molecular Biology and the Neuroscience Program, New Orleans, Louisiana
| | - Sara Santarelli
- Roche Pharmaceutical Research and Early Development, Neuroscience, Ophthalmology and Rare Diseases, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - C Gunnar Forsberg
- Departments of Psychiatry, Pediatrics, and Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Tiffany D Rogers
- Departments of Psychiatry, Pediatrics, and Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Gregg D Stanwood
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida
| | - Benjamin J Hall
- Roche Pharmaceutical Research and Early Development, Neuroscience, Ophthalmology and Rare Diseases, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland.,Tulane University Department of Cell and Molecular Biology and the Neuroscience Program, New Orleans, Louisiana
| | - Eric Delpire
- Departments of Psychiatry, Pediatrics, and Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Jeremy Veenstra-VanderWeele
- Division of Child & Adolescent Psychiatry, New York State Psychiatric Institute, Columbia University, New York, New York.,Departments of Psychiatry, Pediatrics, and Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Michael D Saxe
- Roche Pharmaceutical Research and Early Development, Neuroscience, Ophthalmology and Rare Diseases, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| |
Collapse
|
37
|
Zhou H, Zhao W, Ye L, Chen Z, Cui Y. Postnatal low-concentration arsenic exposure induces autism-like behavior and affects frontal cortex neurogenesis in rats. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2018; 62:188-198. [PMID: 30064059 DOI: 10.1016/j.etap.2018.07.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 07/19/2018] [Accepted: 07/23/2018] [Indexed: 06/08/2023]
Abstract
This study aimed to explore the effects of postnatal low-concentration arsenic exposure on learning, social skills and frontal cortex neurogenesis in rats. Water-based arsenic exposure rat models were established on postnatal days 4-10 (P4-P10). The experimental animals were divided into four groups: the control group, a 15 μg/L As2O3 water group, a 30 μg/L As2O3 water group, and a 45 μg/L As2O3 water group. Cognitive function was examined with the Morris water maze, anxiety-like behavior with the open field test and light-dark box test, and social skills with a social interaction test. The frontal cortices of pups from each experimental group were sectioned at various time points after arsenic exposure. The morphologies and neurogenesis of the neurons in the frontal cortices were observed by hematoxylin-eosin staining, Nissl staining, and doublecortin (DCX) immunostaining. Significant positive correlations between arsenic concentration and deficits in learning and social skills were found, and the arsenic exposure groups showed significant increases in anxiety-like behavior compared with the control group (all Ps<0.05). Abnormal morphologic changes in the external granular layer and external pyramidal layer were positively correlated with the water arsenic concentration in the acute phase of arsenic exposure. However, at five weeks after arsenic exposure, the frontal cortex morphology was restored. Moreover, immunohistochemistry revealed that compared to the control group, the groups that were exposed to arsenic exhibited significantly higher levels of DCX expression in the external granular and external pyramidal layers (all Ps<0.001). Furthermore, the 30 μg/L and 45 μg/L arsenic exposure groups still showed some DCX expression at five weeks after exposure. In conclusion, postnatal low-concentration arsenic exposure impaired learning and social skills and increased anxiety-like behaviors, and abnormal frontal cortex neurogenesis may be the mechanism underlying these effects.
Collapse
Affiliation(s)
- Hao Zhou
- Department of Pediatrics, Guizhou Provincial People's Hospital, Medical College of Guizhou University, Guiyang, 550002, China.
| | | | - Liu Ye
- Otolaryngological Department, Guizhou Provincial People's Hospital, Medical College of Guizhou University, Guiyang, 550002, China
| | - Zhihe Chen
- Department of Pediatrics, Guizhou Provincial People's Hospital, Medical College of Guizhou University, Guiyang, 550002, China
| | - Yuxia Cui
- Department of Pediatrics, Guizhou Provincial People's Hospital, Medical College of Guizhou University, Guiyang, 550002, China; Guizhou Medical University, 550004, China.
| |
Collapse
|
38
|
Kalashnikov V, Zajcev A, Atroshchenko M, Miroshnikov S, Frolov A, Zav'yalov O, Kalinkova L, Kalashnikova T. The content of essential and toxic elements in the hair of the mane of the trotter horses depending on their speed. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2018; 25:21961-21967. [PMID: 29797197 DOI: 10.1007/s11356-018-2334-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 05/15/2018] [Indexed: 06/08/2023]
Abstract
A study on the Russian trotting breeds was conducted to assess the impact of horses' sporting results and the degree of accumulation of chemical elements in the hair. In the first phase of the research, the elemental composition of the mane hair of trotter horses (n = 215) was studied. Based on these studies, percentile intervals for the distribution of concentrations of chemical elements in the hair have been established, and the values of 25 and 75 percentile adopted as a "physiological standard" have been defined. In the second stage of the research into clinically healthy Russian trotting breeds (n = 56), it was estimated that the sporting results were dependent on the elemental status defined by the hair. The elemental composition of the hair was defined by 25 chemical elements using atomic emission and mass spectrometry. It is established that the mane hair is closely related to the sporting results of trotter horses. Thus, in animal wool with the highest sporting achievements, there were reliably less I, Cr, Co, Li, V, Al, Pb, and Cd, and reliably more Si than the low ones. Differences in individual elements exceeded 200%. As sporting performance diminished, the number of elements within the standard increased. For example, for mares with average speed, there were deviations from the physiological standard by 6 elements (P, Fe, Mn, I, Co, Si), with the low one by 13 elements (P, Fe, Cu, Mn, I, Co, Si, K, Cr, Ni, V, Al, Pb). A comparative estimate of the mineralization of the horses' mane measured by the sum of the amount of substances showed that there was a negative correlation between the accumulation of toxic elements and the speed (r = - 0.59). On the basis of the above, a conclusion is reached on the future use of the mane hair to assess the speed qualities of trotter horses.
Collapse
Affiliation(s)
| | - Aleksandr Zajcev
- FSBSI All-Russian Research Institute of Horse Breeding, Ryazan, Russia
| | | | - Sergey Miroshnikov
- Federal Scientific Center of Biological Systems and Agrotechnologies of the Russian Academy of Sciences, Orenburg, Russia
- FSBEI HE Orenburg State University, Orenburg, Russia
| | - Alexey Frolov
- Federal Scientific Center of Biological Systems and Agrotechnologies of the Russian Academy of Sciences, Orenburg, Russia.
| | - Oleg Zav'yalov
- Federal Scientific Center of Biological Systems and Agrotechnologies of the Russian Academy of Sciences, Orenburg, Russia
| | - Liliya Kalinkova
- FSBSI All-Russian Research Institute of Horse Breeding, Ryazan, Russia
| | | |
Collapse
|
39
|
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: 1.9] [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
|
40
|
Gillet V, Hunting DJ, Takser L. Turing Revisited: Decoding the microRNA Messages in Brain Extracellular Vesicles for Early Detection of Neurodevelopmental Disorders. Curr Environ Health Rep 2018; 3:188-201. [PMID: 27301443 DOI: 10.1007/s40572-016-0093-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The prevention of neurodevelopmental disorders (NDD) of prenatal origin suffers from the lack of objective tools for early detection of susceptible individuals and the long time lag, usually in years, between the neurotoxic exposure and the diagnosis of mental dysfunction. Human data on the effects of alcohol, lead, and mercury and experimental data from animals on developmental neurotoxins and their long-term behavioral effects have achieved a critical mass, leading to the concept of the Developmental Origin of Health and Disease (DOHaD). However, there is currently no way to evaluate the degree of brain damage early after birth. We propose that extracellular vesicles (EVs) and particularly exosomes, released by brain cells into the fetal blood, may offer us a non-invasive means of assessing brain damage by neurotoxins. We are inspired by the strategy applied by Alan Turing (a cryptanalyst working for the British government), who created a first computer to decrypt German intelligence communications during World War II. Given the growing evidence that microRNAs (miRNAs), which are among the molecules carried by EVs, are involved in cell-cell communication, we propose that decrypting messages from EVs can allow us to detect damage thus offering an opportunity to cure, reverse, or prevent the development of NDD. This review summarizes recent findings on miRNAs associated with selected environmental toxicants known to be involved in the pathophysiology of NDD.
Collapse
Affiliation(s)
- Virginie Gillet
- Département Pédiatrie, Faculté de Médecine et Sciences de la Santé de l'Université de Sherbrooke, 3001, 12ème avenue Nord, Sherbrooke, Québec, Canada, J1H 5N4
| | - Darel John Hunting
- Département Radiobiologie, Faculté de Médecine et Sciences de la Santé de l'Université de Sherbrooke, 3001, 12ème avenue Nord, Sherbrooke, Québec, Canada, J1H 5N4
| | - Larissa Takser
- Département Pédiatrie, Faculté de Médecine et Sciences de la Santé de l'Université de Sherbrooke, 3001, 12ème avenue Nord, Sherbrooke, Québec, Canada, J1H 5N4.
| |
Collapse
|
41
|
Appleton AA, Jackson BP, Karagas M, Marsit CJ. Prenatal exposure to neurotoxic metals is associated with increased placental glucocorticoid receptor DNA methylation. Epigenetics 2017; 12:607-615. [PMID: 28548590 DOI: 10.1080/15592294.2017.1320637] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Epigenetic alterations related to prenatal neurotoxic metals exposure may be key in understanding the origins of cognitive and neurobehavioral problems in children. Placental glucocorticoid receptor (NR3C1) methylation has been linked to neurobehavioral risk in early life, but has not been examined in response to neurotoxic metals exposure despite parallel lines of research showing metals exposure and NR3C1 methylation each contribute to a similar set of neurobehavioral phenotypes. Thus, we conducted a study of prenatal neurotoxic metals exposure and placental NR3C1 methylation in a cohort of healthy term singleton pregnancies from Rhode Island, USA (n = 222). Concentrations of arsenic (As; median 0.02 ug/g), cadmium (Cd; median 0.03 μg/g), lead (Pb; median 0.40 μg/g), manganese (Mn; median 0.56 μg/g), mercury (Hg; median 0.02 μg/g), and zinc (Zn; 145.18 μg/g) measured in infant toenails were categorized as tertiles. Multivariable linear regression models tested the independent associations for each metal with NR3C1 methylation, as well as the cumulative risk of exposure to multiple metals simultaneously. Compared to the lowest exposure tertiles, higher levels of As, Cd, Pb, Mn, and Hg were each associated with increased placental NR3C1 methylation (all P<0.02). Coefficients for these associations corresponded with a 0.71-1.41 percent increase in NR3C1 methylation per tertile increase in metals concentrations. For Zn, the lowest exposure tertile compared with the highest tertile was associated with 1.26 percent increase in NR3C1 methylation (P=0.01). Higher cumulative metal risk scores were marginally associated with greater NR3C1 methylation. Taken together, these results indicate that prenatal exposure to neurotoxic metals may affect the offspring's NR3C1 activity, which may help explain cognitive and neurodevelopmental risk later in life.
Collapse
Affiliation(s)
- Allison A Appleton
- a Department of Epidemiology and Biostatistics , University at Albany School of Public Health , Rensselaer , NY , USA
| | - Brian P Jackson
- b Department of Earth Sciences, Dartmouth College , Hanover , NH , USA
| | - Margaret Karagas
- c Department of Epidemiology , Geisel School of Medicine at Dartmouth , One Medical Center Drive, Lebanon , NH , USA
| | - Carmen J Marsit
- d Department of Environmental Health, Rollins School of Public Health , Emory University , Atlanta , GA , USA
| |
Collapse
|
42
|
Posar A, Visconti P. Autism in 2016: the need for answers. J Pediatr (Rio J) 2017; 93:111-119. [PMID: 27837654 DOI: 10.1016/j.jped.2016.09.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 09/01/2016] [Accepted: 09/13/2016] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE Autism spectrum disorders are lifelong and often devastating conditions that severely affect social functioning and self-sufficiency. The etiopathogenesis is presumably multifactorial, resulting from a very complex interaction between genetic and environmental factors. The dramatic increase in autism spectrum disorder prevalence observed during the last decades has led to placing more emphasis on the role of environmental factors in the etiopathogenesis. The objective of this narrative biomedical review was to summarize and discuss the results of the most recent and relevant studies about the environmental factors hypothetically involved in autism spectrum disorder etiopathogenesis. SOURCES A search was performed in PubMed (United States National Library of Medicine) about the environmental factors hypothetically involved in the non-syndromic autism spectrum disorder etiopathogenesis, including: air pollutants, pesticides and other endocrine-disrupting chemicals, electromagnetic pollution, vaccinations, and diet modifications. SUMMARY OF THE FINDINGS While the association between air pollutants, pesticides and other endocrine-disrupting chemicals, and risk for autism spectrum disorder is receiving increasing confirmation, the hypothesis of a real causal relation between them needs further data. The possible pathogenic mechanisms by which environmental factors can lead to autism spectrum disorder in genetically predisposed individuals were summarized, giving particular emphasis to the increasingly important role of epigenetics. CONCLUSIONS Future research should investigate whether there is a significant difference in the prevalence of autism spectrum disorder among nations with high and low levels of the various types of pollution. A very important goal of the research concerning the interactions between genetic and environmental factors in autism spectrum disorder etiopathogenesis is the identification of vulnerable populations, also in view of proper prevention.
Collapse
Affiliation(s)
- Annio Posar
- IRCCS Institute of Neurological Sciences of Bologna, Child Neurology and Psychiatry Unit, Bologna, Italy; University of Bologna, Department of Biomedical and Neuromotor Sciences, Bologna, Italy.
| | - Paola Visconti
- IRCCS Institute of Neurological Sciences of Bologna, Child Neurology and Psychiatry Unit, Bologna, Italy
| |
Collapse
|
43
|
Sheida E, Sipailova O, Miroshnikov S, Sizova E, Lebedev S, Rusakova E, Notova S. The effect of iron nanoparticles on performance of cognitive tasks in rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2017; 24:8700-8710. [PMID: 28210948 DOI: 10.1007/s11356-017-8531-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 01/31/2017] [Indexed: 06/06/2023]
Abstract
To assess the influence of 62.5 ± 0.6 nm iron nanoparticles on the status of central nervous system, a study was conducted on Wistar rats, which were subjected to abdominal injection of the studied nanoparticles at doses of 2 and 14 mg/kg. Based on the analysis of the structural and functional status of the cerebral cortex of rats, behavioral reactions of animals, and the elemental composition of the cerebral cortex, we investigated the nanoparticles' neurotoxic effect, whose degree and nature varied depending on the dosage and the time elapsed after the injection. We identified pathological changes in motor and somatosensory areas of the rats' cerebral cortex and established pronounced changes in the elemental homeostasis of the animals' cerebral cortex in experimental groups. Identified structural changes were accompanied by an increase in exploratory activity, locomotor activity, and emotional status of the animals. At that, these activities were more pronounced in rats, which were administered iron nanoparticles at a dose of 14 mg/kg. By the end of the experiment, the excitation processes prevailed over the inhibition processes that have led to the inhibition of central nervous system activity in experimental animals against the adaptation to stress in rats of the control group.
Collapse
Affiliation(s)
| | | | - Sergei Miroshnikov
- Federal State Budgetary Scientific Institution "All-Russian Research Institute of Beef Cattle Breeding", Orenburg, Russia
| | - Elena Sizova
- Orenburg State University, Orenburg, Russia
- Federal State Budgetary Scientific Institution "All-Russian Research Institute of Beef Cattle Breeding", Orenburg, Russia
| | | | | | | |
Collapse
|
44
|
|
45
|
Galvez-Contreras AY, Campos-Ordonez T, Gonzalez-Castaneda RE, Gonzalez-Perez O. Alterations of Growth Factors in Autism and Attention-Deficit/Hyperactivity Disorder. Front Psychiatry 2017; 8:126. [PMID: 28751869 PMCID: PMC5507945 DOI: 10.3389/fpsyt.2017.00126] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 06/29/2017] [Indexed: 12/27/2022] Open
Abstract
Growth factors (GFs) are cytokines that regulate the neural development. Recent evidence indicates that alterations in the expression level of GFs during embryogenesis are linked to the pathophysiology and clinical manifestations of attention-deficit/hyperactivity disorder (ADHD) and autism spectrum disorders (ASD). In this concise review, we summarize the current evidence that supports the role of brain-derived neurotrophic factor, insulin-like growth factor 2, hepatocyte growth factor (HGF), glial-derived neurotrophic factor, nerve growth factor, neurotrophins 3 and 4, and epidermal growth factor in the pathogenesis of ADHD and ASD. We also highlight the potential use of these GFs as clinical markers for diagnosis and prognosis of these neurodevelopmental disorders.
Collapse
Affiliation(s)
- Alma Y Galvez-Contreras
- Department of Neuroscience, Institute of Translational Neuroscience, Centro Universitario de Ciencias de la Salud, University of Guadalajara, Guadalajara, Mexico.,Unidad de Atencion en Neurosciencias, Department of Neuroscience, Centro Universitario de Ciencias de la Salud, University of Guadalajara, Guadalajara, Mexico
| | - Tania Campos-Ordonez
- Laboratory of Neuroscience, School of Psychology, University of Colima, Colima, Mexico.,Medical Science PhD Program, School of Medicine, University of Colima, Colima, Mexico
| | - Rocio E Gonzalez-Castaneda
- Department of Neuroscience, Institute of Translational Neuroscience, Centro Universitario de Ciencias de la Salud, University of Guadalajara, Guadalajara, Mexico
| | - Oscar Gonzalez-Perez
- Laboratory of Neuroscience, School of Psychology, University of Colima, Colima, Mexico.,El Colegio de Colima, Colima, Mexico
| |
Collapse
|
46
|
Carter CJ, Blizard RA. Autism genes are selectively targeted by environmental pollutants including pesticides, heavy metals, bisphenol A, phthalates and many others in food, cosmetics or household products. Neurochem Int 2016; 101:S0197-0186(16)30197-8. [PMID: 27984170 DOI: 10.1016/j.neuint.2016.10.011] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 10/18/2016] [Accepted: 10/26/2016] [Indexed: 11/21/2022]
Abstract
The increasing incidence of autism suggests a major environmental influence. Epidemiology has implicated many candidates and genetics many susceptibility genes. Gene/environment interactions in autism were analysed using 206 autism susceptibility genes (ASG's) from the Autworks database to interrogate ∼1 million chemical/gene interactions in the comparative toxicogenomics database. Any bias towards ASG's was statistically determined for each chemical. Many suspect compounds identified in epidemiology, including tetrachlorodibenzodioxin, pesticides, particulate matter, benzo(a)pyrene, heavy metals, valproate, acetaminophen, SSRI's, cocaine, bisphenol A, phthalates, polyhalogenated biphenyls, flame retardants, diesel constituents, terbutaline and oxytocin, inter alia showed a significant degree of bias towards ASG's, as did relevant endogenous agents (retinoids, sex steroids, thyroxine, melatonin, folate, dopamine, serotonin). Numerous other suspected endocrine disruptors (over 100) selectively targeted ASG's including paraquat, atrazine and other pesticides not yet studied in autism and many compounds used in food, cosmetics or household products, including tretinoin, soy phytoestrogens, aspartame, titanium dioxide and sodium fluoride. Autism polymorphisms influence the sensitivity to some of these chemicals and these same genes play an important role in barrier function and control of respiratory cilia sweeping particulate matter from the airways. Pesticides, heavy metals and pollutants also disrupt barrier and/or ciliary function, which is regulated by sex steroids and by bitter/sweet taste receptors. Further epidemiological studies and neurodevelopmental and behavioural research is warranted to determine the relevance of large number of suspect candidates whose addition to the environment, household, food and cosmetics might be fuelling the autism epidemic in a gene-dependent manner.
Collapse
Affiliation(s)
- C J Carter
- PolygenicPathways, Flat 2, 40 Baldslow Road, Hastings, East Sussex, TN34 2EY, UK.
| | - R A Blizard
- Molecular Psychiatry Laboratory, Mental Health Sciences Unit, University College, London, UK
| |
Collapse
|
47
|
Benítez-Burraco A, Lattanzi W, Murphy E. Language Impairments in ASD Resulting from a Failed Domestication of the Human Brain. Front Neurosci 2016; 10:373. [PMID: 27621700 PMCID: PMC5002430 DOI: 10.3389/fnins.2016.00373] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 08/02/2016] [Indexed: 11/16/2022] Open
Abstract
Autism spectrum disorders (ASD) are pervasive neurodevelopmental disorders entailing social and cognitive deficits, including marked problems with language. Numerous genes have been associated with ASD, but it is unclear how language deficits arise from gene mutation or dysregulation. It is also unclear why ASD shows such high prevalence within human populations. Interestingly, the emergence of a modern faculty of language has been hypothesized to be linked to changes in the human brain/skull, but also to the process of self-domestication of the human species. It is our intention to show that people with ASD exhibit less marked domesticated traits at the morphological, physiological, and behavioral levels. We also discuss many ASD candidates represented among the genes known to be involved in the “domestication syndrome” (the constellation of traits exhibited by domesticated mammals, which seemingly results from the hypofunction of the neural crest) and among the set of genes involved in language function closely connected to them. Moreover, many of these genes show altered expression profiles in the brain of autists. In addition, some candidates for domestication and language-readiness show the same expression profile in people with ASD and chimps in different brain areas involved in language processing. Similarities regarding the brain oscillatory behavior of these areas can be expected too. We conclude that ASD may represent an abnormal ontogenetic itinerary for the human faculty of language resulting in part from changes in genes important for the “domestication syndrome” and, ultimately, from the normal functioning of the neural crest.
Collapse
Affiliation(s)
| | - Wanda Lattanzi
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore Rome, Italy
| | - Elliot Murphy
- Division of Psychology and Language Sciences, University College London London, UK
| |
Collapse
|
48
|
Yang Y, Ibrahim AA, Hashemi P, Stockdill JL. Real-Time, Selective Detection of Copper(II) Using Ionophore-Grafted Carbon-Fiber Microelectrodes. Anal Chem 2016; 88:6962-6. [DOI: 10.1021/acs.analchem.6b00825] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Yuanyuan Yang
- Department
of Chemistry, Wayne State University, Detroit, Michigan 48202, United States
| | - Ahmad A. Ibrahim
- Department
of Chemistry, Wayne State University, Detroit, Michigan 48202, United States
| | - Parastoo Hashemi
- Department
of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Jennifer L. Stockdill
- Department
of Chemistry, Wayne State University, Detroit, Michigan 48202, United States
| |
Collapse
|
49
|
Cartocci V, Segatto M, Di Tunno I, Leone S, Pfrieger FW, Pallottini V. Modulation of the Isoprenoid/Cholesterol Biosynthetic Pathway During Neuronal Differentiation In Vitro. J Cell Biochem 2016; 117:2036-44. [PMID: 27392312 DOI: 10.1002/jcb.25500] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 01/25/2016] [Indexed: 12/27/2022]
Abstract
During differentiation, neurons acquire their typical shape and functional properties. At present, it is unclear, whether this important developmental step involves metabolic changes. Here, we studied the contribution of the mevalonate (MVA) pathway to neuronal differentiation using the mouse neuroblastoma cell line N1E-115 as experimental model. Our results show that during differentiation, the activity of 3-hydroxy 3-methylglutaryl Coenzyme A reductase (HMGR), a key enzyme of MVA pathway, and the level of Low Density Lipoprotein receptor (LDLr) decrease, whereas the level of LDLr-related protein-1 (LRP1) and the dimerization of Scavanger Receptor B1 (SRB-1) rise. Pharmacologic inhibition of HMGR by simvastatin accelerated neuronal differentiation by modulating geranylated proteins. Collectively, our data suggest that during neuronal differentiation, the activity of the MVA pathway decreases and we postulate that any interference with this process impacts neuronal morphology and function. Therefore, the MVA pathway appears as an attractive pharmacological target to modulate neurological and metabolic symptoms of developmental neuropathologies. J. Cell. Biochem. 117: 2036-2044, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Veronica Cartocci
- Department of Science, Biomedical and Technology Science Section, University Roma Tre, Viale Marconi, 446, 00146, Rome, Italy
| | - Marco Segatto
- Department of Biosciences, University of Milan, Via Giovanni Celoria, 26, 20133, Milan, Italy
| | - Ilenia Di Tunno
- Department of Science, Biomedical and Technology Science Section, University Roma Tre, Viale Marconi, 446, 00146, Rome, Italy
| | - Stefano Leone
- Department of Science, Biomedical and Technology Science Section, University Roma Tre, Viale Marconi, 446, 00146, Rome, Italy
| | - Frank W Pfrieger
- Institute of Cellular and Integrative Neurosciences (INCI) CNRS UPR 3212, University of Strasbourg, 5 rue Blaise Pascal, 67084, Strasbourg, France
| | - Valentina Pallottini
- Department of Science, Biomedical and Technology Science Section, University Roma Tre, Viale Marconi, 446, 00146, Rome, Italy
| |
Collapse
|