1
|
Tang X, Ran X, Liang Z, Zhuang H, Yan X, Feng C, Qureshi A, Gao Y, Shen L. Screening biomarkers for autism spectrum disorder using plasma proteomics combined with machine learning methods. Clin Chim Acta 2025; 565:120018. [PMID: 39481511 DOI: 10.1016/j.cca.2024.120018] [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: 05/20/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 11/02/2024]
Abstract
BACKGROUND AND AIMS Autism spectrum disorder (ASD) is a common neurodevelopmental disorder in children. Early intervention is effective. Investigation of novel blood biomarkers of ASD facilitates early detection and intervention. MATERIALS AND METHODS Sequential window acquisition of all theoretical spectra-mass spectrometry (SWATH-MS)-based proteomics technology and 30 DSM-V defined ASD cases versus age- and sex-matched controls were initially evaluated, and candidate biomarkers were screened using machine learning methods. Candidate biomarkers were validated by targeted proteomics multiple reaction monitoring (MRM) analysis using an independent group of 30 ASD cases vs. controls. RESULTS Fifty-one differentially expressed proteins (DEPs) were identified by SWATH analysis. They were associated with the immune response, complements and coagulation cascade pathways, and apolipoprotein-related metabolic pathways. Machine learning analysis screened 10 proteins as biomarker combinations (TFRC, PPBP, APCS, ALDH1A1, CD5L, SPARC, FGG, SHBG, S100A9, and PF4V1). In the MRM analysis, four proteins (PPBP, APCS, FGG, and PF4V1) were significantly different between the groups, and their combination as a screening indicator showed high potential (AUC = 0.8087, 95 % confidence interval 0.6904-0.9252, p < 0.0001). CONCLUSIONS Our study provides data that suggests that a few plasma proteins have potential use in screening for ASD.
Collapse
Affiliation(s)
- Xiaoxiao Tang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518071, PR China
| | - Xiaoqian Ran
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518071, PR China
| | - Zhiyuan Liang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518071, PR China
| | - Hongbin Zhuang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518071, PR China
| | - Xi Yan
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518071, PR China
| | - Chengyun Feng
- Maternal and Child Health Hospital of Baoan, Shenzhen 518100, PR China
| | - Ayesha Qureshi
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518071, PR China
| | - Yan Gao
- Maternal and Child Health Hospital of Baoan, Shenzhen 518100, PR China
| | - Liming Shen
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518071, PR China; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research, Institutions, Shenzhen 518055, PR China.
| |
Collapse
|
2
|
Mukhtar I. Unravelling the critical role of neuroinflammation in epilepsy-associated neuropsychiatric comorbidities: A review. Prog Neuropsychopharmacol Biol Psychiatry 2025; 136:111135. [PMID: 39237022 DOI: 10.1016/j.pnpbp.2024.111135] [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: 01/01/2024] [Revised: 09/01/2024] [Accepted: 09/01/2024] [Indexed: 09/07/2024]
Abstract
Epilepsy is a complex neurological disorder characterized not only by seizures but also by significant neuropsychiatric comorbidities, affecting approximately one-third of those diagnosed. This review explores the intricate relationship between epilepsy and its associated psychiatric and cognitive disturbances, with a focus on the role of inflammation. Recent definitions of epilepsy emphasize its multifaceted nature, linking it to neurobiological, psychiatric, cognitive, and social deficits. Inflammation has emerged as a critical factor influencing both seizure activity and neuropsychiatric outcomes in epilepsy patients. This paper critically examines how dysregulated inflammatory pathways disrupt neurotransmitter transmission and contribute to depression, mood disorders, and anxiety prevalent among individuals with epilepsy. It also evaluates current therapeutic approaches and underscores the potential of anti-inflammatory therapies in managing epilepsy and related neuropsychiatric conditions. Additionally, the review highlights the importance of the anti-inflammatory effects of anti-seizure medications, antidepressants, and antipsychotics and their therapeutic implications for mood disorders. Also, the role of ketogenic diet in managing epilepsy and its psychiatric comorbidities is briefly presented. Furthermore, it briefly discusses the role of the gut-brain axis in maintaining neurological health and how its dysregulation is associated with epilepsy. The review concludes that inflammation plays a pivotal role in linking epilepsy with its neuropsychiatric comorbidities, suggesting that targeted anti-inflammatory interventions may offer promising therapeutic strategies. Future research should focus on longitudinal studies comparing outcomes between epileptic patients with and without neuropsychiatric comorbidities, the development of diagnostic tools, and the exploration of novel anti-inflammatory treatments to better manage these complex interactions.
Collapse
Affiliation(s)
- Iqra Mukhtar
- Faculty of Pharmacy, Iqra University, Karachi, Pakistan.
| |
Collapse
|
3
|
Gong H, Lu Y, Deng SL, Lv KY, Luo J, Luo Y, Du ZL, Wu LF, Liu TY, Wang XQ, Zhao JH, Wang L, Xia ML, Zhu DM, Wang LW, Fan XT. Targeting S100A9 attenuates social dysfunction by modulating neuroinflammation and myelination in a mouse model of autism. Pharmacol Res 2025; 211:107568. [PMID: 39733843 DOI: 10.1016/j.phrs.2024.107568] [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: 10/05/2024] [Revised: 12/15/2024] [Accepted: 12/25/2024] [Indexed: 12/31/2024]
Abstract
Growing evidence supports a role for dysregulated neuroinflammation in autism. However, the underlying mechanisms of microglia-evoked neuroinflammation in the development of autistic phenotypes have not been elucidated. This study aimed to investigate the role and underlying mechanisms of microglial S100 calcium-binding protein A9 (S100A9) in autistic phenotypes. We utilized the BTBR T + tf/J (BTBR) mouse, a reliable preclinical model for autism that displays core behavioral features of autism as well as persistent immune dysregulation. A combination of behavioral, pharmacological, immunological, genetic, molecular, and transcriptomics approaches were used to uncover the potential role of S100A9 in autism. Significant overexpression of microglial S100A9 was observed in the hippocampus of BTBR mice. BTBR mice displayed decreased social communication and increased repetitive behaviors compared to C57BL/6 mice. Interestingly, the above social dysfunction was attenuated by a pharmacological inhibitor of S100A9, accompanied by a significant reduction in the activated microglia morphological phenotype, inflammatory receptors, and proinflammatory cytokines. Notably, S100A9 inhibition decreased c-Fos+ cells and promoted myelination in the cornu ammonis 3 of BTBR mice. Furthermore, the promyelinating compound administration ameliorated the autism-relevant behaviors in BTBR mice. Our findings indicate that microglia-derived S100A9 triggers the neuroinflammation cascade, myelination deficits, and social dysfunction. Targeting S100A9 could, therefore, be a promising therapeutic strategy for neuroinflammation-related neurodevelopmental disorders.
Collapse
Affiliation(s)
- Hong Gong
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing 40038, China
| | - Yao Lu
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing 40038, China; Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 22100, China
| | - Shi-Long Deng
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing 40038, China; Nursing Department, The Affiliated Hospital of Southwest Medical University, Sichuan Province, Luzhou 646000, China
| | - Ke-Yi Lv
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing 40038, China
| | - Jing Luo
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing 40038, China
| | - Yi Luo
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing 40038, China
| | - Zhu-Lin Du
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing 40038, China
| | - Ling-Feng Wu
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing 40038, China; Battalion 7 of the Cadet Brigade, Third Military Medical University (Army Medical University), Chongqing 40038, China
| | - Tian-Yao Liu
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing 40038, China
| | - Xia-Qing Wang
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing 40038, China
| | - Jing-Hui Zhao
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing 40038, China
| | - Lian Wang
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing 40038, China
| | - Mei-Ling Xia
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing 40038, China
| | - Dong-Mei Zhu
- Department of Hospital Infection Control, Chongqing Health Center for Women and Children, Chongqing 401147, China; Department of Hospital Infection Control, Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, China
| | - Li-Wei Wang
- Department of Anesthesiology, Xuzhou Central Hospital, Xuzhou 221009, China; Department of Anesthesiology, Xuzhou Clinical College of Xuzhou Medical University, Xuzhou 221009, China.
| | - Xiao-Tang Fan
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing 40038, China.
| |
Collapse
|
4
|
Usui N, Kobayashi H, Shimada S. Neuroinflammation and Oxidative Stress in the Pathogenesis of Autism Spectrum Disorder. Int J Mol Sci 2023; 24:ijms24065487. [PMID: 36982559 PMCID: PMC10049423 DOI: 10.3390/ijms24065487] [Citation(s) in RCA: 69] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 03/08/2023] [Accepted: 03/09/2023] [Indexed: 03/16/2023] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder (NDD) characterized by impairments in social communication, repetitive behaviors, restricted interests, and hyperesthesia/hypesthesia caused by genetic and/or environmental factors. In recent years, inflammation and oxidative stress have been implicated in the pathogenesis of ASD. In this review, we discuss the inflammation and oxidative stress in the pathophysiology of ASD, particularly focusing on maternal immune activation (MIA). MIA is a one of the common environmental risk factors for the onset of ASD during pregnancy. It induces an immune reaction in the pregnant mother’s body, resulting in further inflammation and oxidative stress in the placenta and fetal brain. These negative factors cause neurodevelopmental impairments in the developing fetal brain and subsequently cause behavioral symptoms in the offspring. In addition, we also discuss the effects of anti-inflammatory drugs and antioxidants in basic studies on animals and clinical studies of ASD. Our review provides the latest findings and new insights into the involvements of inflammation and oxidative stress in the pathogenesis of ASD.
Collapse
Affiliation(s)
- Noriyoshi Usui
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan
- United Graduate School of Child Development, Osaka University, Suita 565-0871, Japan
- Global Center for Medical Engineering and Informatics, Osaka University, Suita 565-0871, Japan
- Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka 541-8567, Japan
- Correspondence: ; Tel.: +81-668-79-3124
| | - Hikaru Kobayashi
- SANKEN (Institute of Scientific and Industrial Research), Osaka University, Suita 567-0047, Japan
| | - Shoichi Shimada
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan
- United Graduate School of Child Development, Osaka University, Suita 565-0871, Japan
- Global Center for Medical Engineering and Informatics, Osaka University, Suita 565-0871, Japan
- Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka 541-8567, Japan
| |
Collapse
|
5
|
Shah F, Dwivedi M. Pathophysiological Role of Gut Microbiota Affecting Gut–Brain Axis and Intervention of Probiotics and Prebiotics in Autism Spectrum Disorder. PROBIOTIC RESEARCH IN THERAPEUTICS 2022:69-115. [DOI: 10.1007/978-981-16-6760-2_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
6
|
Chen HJ, Liu YW. The Impacts of Probiotics on Microbiota in Patients With Autism Spectrum Disorder. COMPREHENSIVE GUT MICROBIOTA 2022:296-319. [DOI: 10.1016/b978-0-12-819265-8.00101-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
7
|
Davison LM, Alberto AA, Dand HA, Keller EJ, Patt M, Khan A, Dvorina N, White A, Sakurai N, Liegl LN, Vogl T, Jorgensen TN. S100a9 Protects Male Lupus-Prone NZBWF1 Mice From Disease Development. Front Immunol 2021; 12:681503. [PMID: 34220829 PMCID: PMC8248531 DOI: 10.3389/fimmu.2021.681503] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 06/01/2021] [Indexed: 11/13/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disorder disproportionally affecting women. A similar sex difference exists in the murine New Zealand Black/White hybrid model (NZBWF1) of SLE with all females, but only 30-40% of males, developing disease within the first year of life. Myeloid-derived suppressor cells (MDSCs) are prominent in NZBWF1 males and while depletion of these cells in males, but not females, promotes disease development, the mechanism of suppression remains unknown. S100a9, expressed by neutrophils and MDSCs, has previously been shown to exert immunosuppressive functions in cancer and inflammation. Here we investigated if S100a9 exerts immunosuppressive functions in NZBWF1 male and female mice. S100a9+/+, S100a9+/- and S100a9-/- NZBWF1 mice were followed for disease development for up to 8 months of age. Serum autoantibody levels, splenomegaly, lymphocyte activation, glomerulonephritis and proteinuria were measured longitudinally or at the time of harvest. In accordance with an immunosuppressive function of MDSCs in male mice, S100a9-deficient male NZBWF1 mice developed accelerated autoimmunity as indicated by increased numbers of differentiated effector B and T cells, elevated serum autoantibody levels, increased immune-complex deposition and renal inflammation, and accelerated development of proteinuria. In contrast, female mice showed either no response to S100a9-deficiency or even a slight reduction in disease symptoms. Furthermore, male, but not female, S100a9-/- NZBWF1 mice displayed an elevated type I interferon-induced gene signature, suggesting that S100a9 may dampen a pathogenic type I interferon signal in male mice. Taken together, S100a9 exerts an immunosuppressive function in male NZBWF1 mice effectively moderating lupus-like disease development via inhibition of type I interferon production, lymphocyte activation, autoantibody production and the development of renal disease.
Collapse
Affiliation(s)
- Laura M Davison
- Cleveland Clinic Lerner College of Medicine, Department of Molecular Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Andres A Alberto
- Department of Inflammation and Immunity, Lerner Research Institute, NE40, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Hardik A Dand
- Department of Inflammation and Immunity, Lerner Research Institute, NE40, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Emma J Keller
- Cleveland Clinic Lerner College of Medicine, Department of Molecular Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Madeline Patt
- Department of Inflammation and Immunity, Lerner Research Institute, NE40, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Ayesha Khan
- Department of Inflammation and Immunity, Lerner Research Institute, NE40, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Nina Dvorina
- Department of Inflammation and Immunity, Lerner Research Institute, NE40, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Alexandra White
- Cleveland Clinic Lerner College of Medicine at Case Western Reserve University, Cleveland, OH, United States
| | - Nodoka Sakurai
- Department of Inflammation and Immunity, Lerner Research Institute, NE40, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Lauren N Liegl
- Department of Inflammation and Immunity, Lerner Research Institute, NE40, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Thomas Vogl
- Institute of Immunology, University of Muenster, Muenster, Germany
| | - Trine N Jorgensen
- Department of Inflammation and Immunity, Lerner Research Institute, NE40, Cleveland Clinic Foundation, Cleveland, OH, United States
| |
Collapse
|
8
|
Increased Serum Concentrations of High Mobility Group Box 1 (HMGB1) Protein in Children with Autism Spectrum Disorder. CHILDREN-BASEL 2021; 8:children8060478. [PMID: 34198762 PMCID: PMC8228126 DOI: 10.3390/children8060478] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/01/2021] [Accepted: 06/03/2021] [Indexed: 12/19/2022]
Abstract
High mobility group box 1 protein (HMGB1) has been suggested to be involved in the immune dysfunction and inflammation reported in autism spectrum disorder (ASD). We aimed to assess HMGB1 serum concentrations (SCs) in high-functioning ASD children compared to typically developing (TD) controls and to explore their associations with the autism spectrum quotient (AQ), the empathy quotient (EQ), and the systemizing quotient (SQ). The study involved 42 ASD children and 38 TD children, all-male, aged between 6.1 and 13.3 years old. HMGB1 SCs were measured by enzyme-linked immunosorbent assay (ELISA). Groups were comparable regarding age, general IQ, birth weight, and maternal age at birth. ASD children showed significantly higher HMGB1 SCs compared to TD children (1.25 ± 0.84 ng/mL versus 1.13 ± 0.79 ng/mL, respectively, p = 0.039). The Spearman’s rho revealed that HMGB1 SCs were positively correlated with the AQ attention to detail subscale (rs = 0.46, p = 0.045) and with the SQ total score (rs = 0.42, p = 0.04) in the ASD group. These results show that HMGB1 serum concentrations are altered in ASD children, and suggest that inflammatory processes mediated by HMGB1 may be associated with specific cognitive features observed in ASD.
Collapse
|
9
|
Puricelli C, Rolla R, Gigliotti L, Boggio E, Beltrami E, Dianzani U, Keller R. The Gut-Brain-Immune Axis in Autism Spectrum Disorders: A State-of-Art Report. Front Psychiatry 2021; 12:755171. [PMID: 35185631 PMCID: PMC8850385 DOI: 10.3389/fpsyt.2021.755171] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 12/29/2021] [Indexed: 12/20/2022] Open
Abstract
The interest elicited by the large microbial population colonizing the human gut has ancient origins and has gone through a long evolution during history. However, it is only in the last decades that the introduction of high-throughput technologies has allowed to broaden this research field and to disentangle the numerous implications that gut microbiota has in health and disease. This comprehensive ecosystem, constituted mainly by bacteria but also by fungi, parasites, and viruses, is proven to be involved in several physiological and pathological processes that transcend the intestinal homeostasis and are deeply intertwined with apparently unrelated body systems, such as the immune and the nervous ones. In this regard, a novel speculation is the relationship between the intestinal microbial flora and the pathogenesis of some neurological and neurodevelopmental disorders, including the clinical entities defined under the umbrella term of autism spectrum disorders. The bidirectional interplay has led researchers to coin the term gut-brain-immune system axis, subverting the theory of the brain as an immune-privileged site and underscoring the importance of this reciprocal influence already from fetal life and especially during the pre- and post-natal neurodevelopmental process. This revolutionary theory has also unveiled the possibility to modify the gut microbiota as a way to treat and even to prevent different kinds of pathologies. In this sense, some attempts have been made, ranging from probiotic administration to fecal microbiota transplantation, with promising results that need further elaboration. This state-of-art report will describe the main aspects regarding the human gut microbiome and its specific role in the pathogenesis of autism and its related disorders, with a final discussion on the therapeutic and preventive strategies aiming at creating a healthy intestinal microbial environment, as well as their safety and ethical implications.
Collapse
Affiliation(s)
- Chiara Puricelli
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy.,Clinical Biochemistry Laboratory, Ospedale Maggiore della Carità, Novara, Italy
| | - Roberta Rolla
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy.,Clinical Biochemistry Laboratory, Ospedale Maggiore della Carità, Novara, Italy
| | - Luca Gigliotti
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Elena Boggio
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Eleonora Beltrami
- Clinical Biochemistry Laboratory, Ospedale Maggiore della Carità, Novara, Italy
| | - Umberto Dianzani
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy.,Clinical Biochemistry Laboratory, Ospedale Maggiore della Carità, Novara, Italy
| | - Roberto Keller
- Mental Health Department, Adult Autism Center, ASL Città di Torino, Turin, Italy
| |
Collapse
|
10
|
Ansari F, Pourjafar H, Tabrizi A, Homayouni A. The Effects of Probiotics and Prebiotics on Mental Disorders: A Review on Depression, Anxiety, Alzheimer, and Autism Spectrum Disorders. Curr Pharm Biotechnol 2020; 21:555-565. [PMID: 31914909 DOI: 10.2174/1389201021666200107113812] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 08/05/2019] [Accepted: 11/22/2019] [Indexed: 12/26/2022]
Abstract
BACKGROUND Probiotics and their nutrient sources (prebiotics) have been shown to have positive effects on different organs of the host. The idea of their potential benefits on Central Nervous Systems (CNS) and the incidence of Anxiety, Schizophrenia, Alzheimer, Depression, Autism, and other mental disorders has proposed a new category of medicines called "psychobiotic" which is hoped to be of low-side effect anti-inflammatory, antidepressant, and anti-anxiety constitutes. OBJECTIVE In the current review, we present valuable insights into the complicated interactions between the GI microbiota (especially in the colon), brain, immune and central nervous systems and provide a summary of the main findings of the effects of pro- and prebiotics on important mental disorders from the potential mechanisms of action to their application in clinical practice. METHODS Google Scholar, Pub Med, Scopus, and Science Direct databases were searched using following key words: "probiotics", "prebiotics", "mental disorders", "psychological disorders", "depression", "anxiety", "stress", "Alzheimer" and "autism spectrum". The full text of potentially eligible studies was retrieved and assessed in detail by the reviewers. Data were extracted and then summarized from the selected papers. RESULTS The results of the provided evidence suggest that probiotic and prebiotics might improve mental function via several mechanisms. The beneficial effects of their application in Depression, Anxiety, Alzheimer and autism spectrum diseases have also been supported in clinical studies. CONCLUSION Pro and prebiotics can improve mental health and psychological function and can be offered as new medicines for common mental disorders, however, more clinical studies are necessary to conduct regarding the clinical significance of the effects and their bioequivalence or superiority against current treatments.
Collapse
Affiliation(s)
- Fereshteh Ansari
- Research Center for Evidence-Based Medicine, Health Management and Safety Promotion Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Iranian EBM Centre: A Joanna Briggs Institute Affiliated Group, Tabriz, Iran.,Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Tehran, Iran
| | - Hadi Pourjafar
- Department of Food Sciences and Nutrition, Maragheh University of Medical Sciences, Maragheh, Iran.,Alborz University of Medical Sciences, Dietary supplements and Probiotic Research Center, Karaj, Iran
| | - Aydin Tabrizi
- Pediatrics Neurology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Aziz Homayouni
- Department of Food Science and Technology, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
11
|
Abdellatif B, McVeigh C, Bendriss G, Chaari A. The Promising Role of Probiotics in Managing the Altered Gut in Autism Spectrum Disorders. Int J Mol Sci 2020; 21:E4159. [PMID: 32532137 PMCID: PMC7312735 DOI: 10.3390/ijms21114159] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/12/2020] [Accepted: 05/14/2020] [Indexed: 02/06/2023] Open
Abstract
Gastrointestinal symptoms (GIS) have been reported repeatedly in people with autism spectrum disorder (ASD) and studies have reported interesting correlations between severity of behavioral and gastrointestinal symptoms. Growing evidence indicates that the gut microbiota in ASD is altered with various shifts described at different taxonomic levels, pointing to the importance of considering the gut-brain axis in treatment of these disorders. Probiotics are live beneficial bacteria that are ingested as food or customized pills. These beneficial bacteria, when added in sufficient amounts, can correct the dysbiosis. Because probiotics have shown success in treating irritable bowel syndrome (IBS), it is plausible to investigate whether they can induce alleviation of behavioral symptoms as well. Probiotics show, in some clinical studies, their potential benefits (1) in improving gastrointestinal dysfunction, (2) in correcting dysbiosis, (3) in consequently reducing the severity of ASD symptoms. This review compiles data from selected studies that investigate these benefits and the mechanisms that mediate these effects, which include the production of metabolites, hormones, and neurotransmitters and the regulation of pro-inflammatory and regulatory cytokines. Future research based on more randomized, controlled studies with a larger population size and standardized use of strains, concentration of probiotics, duration of treatments, and methods of DNA extraction is still needed in this area, which may lead to more robust results.
Collapse
Affiliation(s)
| | | | | | - Ali Chaari
- Premedical Department, Weill Cornell Medicine, Qatar Foundation, Education City, Doha, P.O. Box 24144, Qatar; (B.A.); (C.M.); (G.B.)
| |
Collapse
|
12
|
Fattorusso A, Di Genova L, Dell'Isola GB, Mencaroni E, Esposito S. Autism Spectrum Disorders and the Gut Microbiota. Nutrients 2019; 11:521. [PMID: 30823414 PMCID: PMC6471505 DOI: 10.3390/nu11030521] [Citation(s) in RCA: 259] [Impact Index Per Article: 43.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 02/22/2019] [Accepted: 02/25/2019] [Indexed: 02/08/2023] Open
Abstract
In recent years, there has been an emerging interest in the possible role of the gut microbiota as a co-factor in the development of autism spectrum disorders (ASDs), as many studies have highlighted the bidirectional communication between the gut and brain (the so-called "gut-brain axis"). Accumulating evidence has shown a link between alterations in the composition of the gut microbiota and both gastrointestinal and neurobehavioural symptoms in children with ASD. The aim of this narrative review was to analyse the current knowledge about dysbiosis and gastrointestinal (GI) disorders in ASD and assess the current evidence for the role of probiotics and other non-pharmacological approaches in the treatment of children with ASD. Analysis of the literature showed that gut dysbiosis in ASD has been widely demonstrated; however, there is no single distinctive profile of the composition of the microbiota in people with ASD. Gut dysbiosis could contribute to the low-grade systemic inflammatory state reported in patients with GI comorbidities. The administration of probiotics (mostly a mixture of Bifidobacteria, Streptococci and Lactobacilli) is the most promising treatment for neurobehavioural symptoms and bowel dysfunction, but clinical trials are still limited and heterogeneous. Well-designed, randomized, placebo-controlled clinical trials are required to validate the effectiveness of probiotics in the treatment of ASD and to identify the appropriate strains, dose, and timing of treatment.
Collapse
Affiliation(s)
- Antonella Fattorusso
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, 06132 Perugia, Italy.
| | - Lorenza Di Genova
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, 06132 Perugia, Italy.
| | - Giovanni Battista Dell'Isola
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, 06132 Perugia, Italy.
| | - Elisabetta Mencaroni
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, 06132 Perugia, Italy.
| | - Susanna Esposito
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, 06132 Perugia, Italy.
| |
Collapse
|
13
|
Di Salvo E, Casciaro M, Quartuccio S, Genovese L, Gangemi S. Do Alarmins Have a Potential Role in Autism Spectrum Disorders Pathogenesis and Progression? Biomolecules 2018; 9:E2. [PMID: 30577568 PMCID: PMC6358895 DOI: 10.3390/biom9010002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 12/09/2018] [Accepted: 12/17/2018] [Indexed: 02/07/2023] Open
Abstract
Autism spectrum disorders (ASDs) represent a disabling condition in early childhood. A number of risk factors were proposed in order to explain their pathogenesis. A multifactorial model was proposed, and data supported the implication of genetic and environmental factors. One of the most accepted speculations is the existence of an imbalance of the immune system. Altered levels of cytokines, chemokines and immunoglobulins were demonstrated in patients with ASDs; in particular, proinflammatory mediators were significantly increased. Alarmins are a multifunctional heterogeneous group of proteins, structurally belonging to specific cells or incorporated by them. They are released in the surrounding tissues as a consequence of cell damage or inflammation. Their functions are multiple as they could activate innate immunity or recruit and activate antigen-presenting cells stimulating an adaptive response. Alarmins are interesting both for understanding the inflammatory process and for diagnostic purposes as biomarkers. Moreover, recent studies, separately, showed that alarmins like interleukin (IL)-33, high-mobility group box 1 (HMGB1), heat-shock protein (HSP) and S100 protein (S100) could play a relevant role in the pathogenesis of ASDs. According to the literature, some of these alarmins could be suitable as biomarkers of inflammation in ASD. Other alarmins, by interfering with the immune system blocking pro-inflammatory mediators, could be the key for ameliorating symptoms and behaviours in autistic disorders.
Collapse
Affiliation(s)
- Eleonora Di Salvo
- National Research Council of Italy (CNR), Institute of Biological Resources and Marine Biotechnologies (IRBIM), Messina 98122, Italy.
- National Research Council of Italy (CNR), Institute of Applied Science and Intelligent System (ISASI), Messina 98164, Italy.
| | - Marco Casciaro
- School and Operative Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, Messina 98125, Italy.
| | | | - Lucrezia Genovese
- National Research Council of Italy (CNR), Institute of Biological Resources and Marine Biotechnologies (IRBIM), Messina 98122, Italy.
| | - Sebastiano Gangemi
- School and Operative Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, Messina 98125, Italy.
| |
Collapse
|
14
|
Abstract
Supplemental Digital Content is available in the text. Background: Previous autism spectrum disorder (ASD) and air pollution studies focused on pregnancy exposures, but another vulnerable period is immediate postnatally. Here, we examined early life exposures to air pollution from the pre- to the postnatal period and ASD/ASD subtypes in the Danish population. Methods: With Danish registers, we conducted a nationwide case–control study of 15,387 children with ASD born 1989–2013 and 68,139 population controls matched by birth year and sex identified from the birth registry. We generated air dispersion geographic information system (AirGIS) model estimates for nitrogen dioxide (NO2), sulfur dioxide (SO2), particulate matter 2.5 (PM2.5), and particulate matter 10 (PM10) at mothers’ home from 9 months before to 9 months after pregnancy and calculated odds ratios (ORs) and 95% confidence intervals (CIs), adjusting for parental age, neighborhood socioeconomic indicators, and maternal smoking using conditional logistic regression. Results: In models that included all exposure periods, we estimated adjusted ORs for ASD per interquartile range (IQR) increase for 9 months after pregnancy with NO2 of 1.08 (95% CI = 1.01, 1.15) and with PM2.5 of 1.06 (95% CI = 1.01, 1.11); associations were smaller for PM10 (1.04; 95% CI = 1.00, 1.09) and strongest for SO2 (1.21; 95% CI = 1.13, 1.29). Also, associations for pollutants were stronger in more recent years (2000–2013) and in larger cities compared with provincial towns/rural counties. For particles and NO2, associations were only specific to autism and Asperger diagnoses. Conclusions: Our data suggest that air pollutant exposure in early infancy but not during pregnancy increases the risk of being diagnosed with autism and Asperger among children born in Denmark.
Collapse
|
15
|
Yenkoyan K, Harutyunyan H, Harutyunyan A. A certain role of SOD/CAT imbalance in pathogenesis of autism spectrum disorders. Free Radic Biol Med 2018; 123:85-95. [PMID: 29782990 DOI: 10.1016/j.freeradbiomed.2018.05.070] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 05/16/2018] [Accepted: 05/17/2018] [Indexed: 12/16/2022]
Abstract
The real impact of reactive oxygen species, antioxidant enzymes, mitochondrial dysfunction and chronic inflammation on the development of autism spectrum disorders (ASD) remains unclear, and even controversial. In this study we compared the plasma levels of antioxidant enzymes and their cofactors, markers of oxidative damage, and the respiratory burst in peripheral blood polymorphonuclear leucocytes (PMNL) as surrogate marker of chronic inflammation obtained from 10 children (4-10 year old) who met DSM-5 criteria and their siblings. We demonstrated diminished superoxide dismutase (SOD) and enhanced catalase (CAT) activities resulting in a markedly decreased SOD/CAT ratio and enhanced carbonyl content in the plasma of ASD patients. A strong correlation was present between SOD and CAT activities in the control group, which was not noted in ASD patients. Moreover, in autistic patients, we observed negative correlation between SOD activity on one side, and carbonyl content in plasma, 8-Hydroxy-2-deoxyguanosin content in urine, and respiratory burst intensity in PMNL on the other side. At the same time, low SOD level in autistic children was positively correlated with the magnesium content in the packed RBCs, which might indicate the involvement of the mitochondrial MnSOD in ASD pathogenesis, and therefore the consequent partaking of mitochondrial dysfunction in the development of ASD. Altogether, these results indicate that decreased antioxidant capacity and increased oxidative stress in ASD patients may have functional consequence in terms of increased superoxide leakage, oxidative protein damage, chronic inflammatory response, and, finally, neuronal cell abnormal functioning or death.
Collapse
Affiliation(s)
- Konstantin Yenkoyan
- Department of Biochemistry, Yerevan State Medical University after Mkhitar Heratsi, Yerevan, Armenia; Laboratory of Biochemical and Biophysical Investigations, Scientific-Research Centre, Yerevan State Medical University after Mkhitar Heratsi, Yerevan, Armenia.
| | - Hayk Harutyunyan
- Laboratory of Biochemical and Biophysical Investigations, Scientific-Research Centre, Yerevan State Medical University after Mkhitar Heratsi, Yerevan, Armenia
| | - Aida Harutyunyan
- Department of Biochemistry, Yerevan State Medical University after Mkhitar Heratsi, Yerevan, Armenia
| |
Collapse
|
16
|
Vezzani A, Dingledine R, Rossetti AO. Immunity and inflammation in status epilepticus and its sequelae: possibilities for therapeutic application. Expert Rev Neurother 2018; 15:1081-92. [PMID: 26312647 DOI: 10.1586/14737175.2015.1079130] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Status epilepticus (SE) is a life-threatening neurological emergency often refractory to available treatment options. It is a very heterogeneous condition in terms of clinical presentation and causes, which besides genetic, vascular and other structural causes also include CNS or severe systemic infections, sudden withdrawal from benzodiazepines or anticonvulsants and rare autoimmune etiologies. Treatment of SE is essentially based on expert opinions and antiepileptic drug treatment per se seems to have no major impact on prognosis. There is, therefore, urgent need of novel therapies that rely upon a better understanding of the basic mechanisms underlying this clinical condition. Accumulating evidence in animal models highlights that inflammation ensuing in the brain during SE may play a determinant role in ongoing seizures and their long-term detrimental consequences, independent of an infection or auto-immune cause; this evidence encourages reconsideration of the treatment flow in SE patients.
Collapse
Affiliation(s)
- Annamaria Vezzani
- a 1 Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Milano, Italy
| | | | | |
Collapse
|
17
|
Parker W, Hornik CD, Bilbo S, Holzknecht ZE, Gentry L, Rao R, Lin SS, Herbert MR, Nevison CD. The role of oxidative stress, inflammation and acetaminophen exposure from birth to early childhood in the induction of autism. J Int Med Res 2017; 45:407-438. [PMID: 28415925 PMCID: PMC5536672 DOI: 10.1177/0300060517693423] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The wide range of factors associated with the induction of autism is invariably linked with either inflammation or oxidative stress, and sometimes both. The use of acetaminophen in babies and young children may be much more strongly associated with autism than its use during pregnancy, perhaps because of well-known deficiencies in the metabolic breakdown of pharmaceuticals during early development. Thus, one explanation for the increased prevalence of autism is that increased exposure to acetaminophen, exacerbated by inflammation and oxidative stress, is neurotoxic in babies and small children. This view mandates extreme urgency in probing the long-term effects of acetaminophen use in babies and the possibility that many cases of infantile autism may actually be induced by acetaminophen exposure shortly after birth.
Collapse
Affiliation(s)
- William Parker
- 1 Departments of Surgery, Duke University Medical Center, Durham, NC USA
| | - Chi Dang Hornik
- 2 Departments of Pediatrics, Duke University Medical Center, Durham, NC USA
| | - Staci Bilbo
- 3 Departments of Pediatrics, Harvard Medical School, Charlestown, MA, USA
| | - Zoie E Holzknecht
- 1 Departments of Surgery, Duke University Medical Center, Durham, NC USA
| | - Lauren Gentry
- 1 Departments of Surgery, Duke University Medical Center, Durham, NC USA
| | - Rasika Rao
- 1 Departments of Surgery, Duke University Medical Center, Durham, NC USA
| | - Shu S Lin
- 1 Departments of Surgery, Duke University Medical Center, Durham, NC USA
| | - Martha R Herbert
- 4 Departments of Neurology, Harvard Medical School, Charlestown, MA, USA
| | - Cynthia D Nevison
- 5 Institute for Arctic and Alpine Research, University of Colorado, Boulder, Boulder, CO, USA
| |
Collapse
|
18
|
Navarro F, Liu Y, Rhoads JM. Can probiotics benefit children with autism spectrum disorders? World J Gastroenterol 2016; 22:10093-10102. [PMID: 28028357 PMCID: PMC5155168 DOI: 10.3748/wjg.v22.i46.10093] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Revised: 10/05/2016] [Accepted: 11/12/2016] [Indexed: 02/06/2023] Open
Abstract
Children with autism are commonly affected by gastrointestinal problems such as abdominal pain, constipation and diarrhea. In recent years, there has been a growing interest in the use of probiotics in this population, as it hypothetically may help to improve bowel habits and the behavioral and social functioning of these individuals. The gut microbiome plays an important role in the pathophysiology of organic as well as functional gastrointestinal disorders. Microbial modification with the use of antibiotics, probiotics, and fecal transplantation have been effective in the treatment of conditions such as recurrent Clostridium difficile infection, pouchitis, and irritable bowel syndrome. The present review presents a number of reported clinical, immunological and microbiome-related changes seen in children with autism compared to normally developed children. It also discusses gut inflammation, permeability concerns, and absorption abnormalities that may contribute to these problems. Most importantly, it discusses evidence, from human and animal studies, of a potential role of probiotics in the treatment of gastrointestinal symptoms in children with autism.
Collapse
|
19
|
Miyashita M, Watanabe T, Ichikawa T, Toriumi K, Horiuchi Y, Kobori A, Kushima I, Hashimoto R, Fukumoto M, Koike S, Ujike H, Arinami T, Tatebayashi Y, Kasai K, Takeda M, Ozaki N, Okazaki Y, Yoshikawa T, Amano N, Washizuka S, Yamamoto H, Miyata T, Itokawa M, Yamamoto Y, Arai M. The regulation of soluble receptor for AGEs contributes to carbonyl stress in schizophrenia. Biochem Biophys Res Commun 2016; 479:447-452. [PMID: 27641663 DOI: 10.1016/j.bbrc.2016.09.074] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 09/15/2016] [Indexed: 11/28/2022]
Abstract
Our previous study showed that enhanced carbonyl stress is closely related to schizophrenia. The endogenous secretory receptor for advanced glycation end-products (esRAGE) is a splice variant of the AGER gene and is one of the soluble forms of RAGE. esRAGE is considered to be a key molecule for alleviating the burden of carbonyl stress by entrapping advanced glycation end-products (AGEs). In the current study, we conducted genetic association analyses focusing on AGER, in which we compared 212 schizophrenic patients to 214 control subjects. We also compared esRAGE levels among a subgroup of 104 patients and 89 controls and further carried out measurements of total circulating soluble RAGE (sRAGE) in 25 patients and 49 healthy subjects. Although the genetic association study yielded inconclusive results, multiple regression analysis indicated that a specific haplotype composed of rs17846798, rs2071288, and a 63 bp deletion, which were in perfect linkage disequilibrium (r2 = 1), and rs2070600 (Gly82Ser) were significantly associated with a marked decrease in serum esRAGE levels. Furthermore, compared to healthy subjects, schizophrenia showed significantly lower esRAGE (p = 0.007) and sRAGE (p = 0.03) levels, respectively. This is the first study to show that serum esRAGE levels are regulated by a newly identified specific haplotype in AGER and that a subpopulation of schizophrenic patients are more vulnerable to carbonyl stress.
Collapse
Affiliation(s)
- Mitsuhiro Miyashita
- Project for Schizophrenia Research, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan; Department of Psychiatry, Tokyo Metropolitan Matsuzawa Hospital, Tokyo, Japan; Department of Psychiatry, Shinshu University School of Medicine, Matsumoto, Japan.
| | - Takuo Watanabe
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Tomoe Ichikawa
- Project for Schizophrenia Research, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Kazuya Toriumi
- Project for Schizophrenia Research, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Yasue Horiuchi
- Project for Schizophrenia Research, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Akiko Kobori
- Project for Schizophrenia Research, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Itaru Kushima
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ryota Hashimoto
- Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Motoyuki Fukumoto
- Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shinsuke Koike
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | | | - Tadao Arinami
- Institute of Basic Medical Sciences, University of Tsukuba, Tsukuba, Japan
| | - Yoshitaka Tatebayashi
- Affective Disorder Research, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Kiyoto Kasai
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masatoshi Takeda
- Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Norio Ozaki
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuji Okazaki
- Department of Psychiatry, Tokyo Metropolitan Matsuzawa Hospital, Tokyo, Japan
| | - Takeo Yoshikawa
- Laboratory for Molecular Psychiatry, RIKEN Brain Science Institute, Wako, Japan
| | - Naoji Amano
- Department of Psychiatry, Shinshu University School of Medicine, Matsumoto, Japan
| | - Shinsuke Washizuka
- Department of Psychiatry, Shinshu University School of Medicine, Matsumoto, Japan
| | - Hiroshi Yamamoto
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Toshio Miyata
- Molecular Medicine and Therapy, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masanari Itokawa
- Project for Schizophrenia Research, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan; Department of Psychiatry, Tokyo Metropolitan Matsuzawa Hospital, Tokyo, Japan
| | - Yasuhiko Yamamoto
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Makoto Arai
- Project for Schizophrenia Research, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| |
Collapse
|
20
|
Gabriele S, Lombardi F, Sacco R, Napolioni V, Altieri L, Tirindelli MC, Gregorj C, Bravaccio C, Rousseau F, Persico AM. The GLO1 C332 (Ala111) allele confers autism vulnerability: family-based genetic association and functional correlates. J Psychiatr Res 2014; 59:108-16. [PMID: 25201284 DOI: 10.1016/j.jpsychires.2014.07.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 07/08/2014] [Accepted: 07/25/2014] [Indexed: 11/16/2022]
Abstract
Glyoxalase I (GLO1) is a homodimeric Zn(2+)-dependent isomerase involved in the detoxification of methylglyoxal and in limiting the formation of advanced glycation end-products (AGE). We previously found the rs4746 A332 (Glu111) allele of the GLO1 gene, which encodes for glyoxalase I, associated with "unaffected sibling" status in families with one or more children affected by Autism Spectrum Disorder (ASD). To identify and characterize this protective allele, we sequenced GLO1 exons and exon-intron junctions, detecting two additional SNPs (rs1049346, rs1130534) in linkage disequilibrium with rs4746. A family-based association study involving 385 simplex and 20 multiplex Italian families yielded a significant association with autism driven only by the rs4746 C332 (Ala111) allele itself (P < 0.05 and P < 0.001 under additive and dominant/recessive models, respectively). Glyoxalase enzymatic activity was significantly reduced both in leukocytes and in post-mortem temporocortical tissue (N = 38 and 13, respectively) of typically developing C332 allele carriers (P < 0.05 and <0.01), with no difference in Glo1 protein levels. Conversely, AGE amounts were significantly higher in the same C332 post-mortem brains (P = 0.001), with a strong negative correlation between glyoxalase activity and AGE levels (τ = -0.588, P < 0.01). Instead, 19 autistic brains show a dysregulation of the glyoxalase-AGE axis (τ = -0.209, P = 0.260), with significant blunting of glyoxalase activity and AGE amounts compared to controls (P < 0.05), and loss of rs4746 genotype effects. In summary, the GLO1 C332 (Ala111) allele confers autism vulnerability by reducing brain glyoxalase activity and enhancing AGE formation, but years after an autism diagnosis the glyoxalase-AGE axis appears profoundly disrupted, with loss of C332 allelic effects.
Collapse
Affiliation(s)
- Stefano Gabriele
- Unit of Child and Adolescent NeuroPsychiatry, Laboratory of Molecular Psychiatry and Neurogenetics, University "Campus Bio-Medico", Rome, Italy; Department of Experimental Neurosciences, I.R.C.C.S. "Fondazione Santa Lucia", Rome, Italy
| | - Federica Lombardi
- Unit of Child and Adolescent NeuroPsychiatry, Laboratory of Molecular Psychiatry and Neurogenetics, University "Campus Bio-Medico", Rome, Italy; Department of Experimental Neurosciences, I.R.C.C.S. "Fondazione Santa Lucia", Rome, Italy
| | - Roberto Sacco
- Unit of Child and Adolescent NeuroPsychiatry, Laboratory of Molecular Psychiatry and Neurogenetics, University "Campus Bio-Medico", Rome, Italy; Department of Experimental Neurosciences, I.R.C.C.S. "Fondazione Santa Lucia", Rome, Italy
| | - Valerio Napolioni
- Unit of Child and Adolescent NeuroPsychiatry, Laboratory of Molecular Psychiatry and Neurogenetics, University "Campus Bio-Medico", Rome, Italy; Department of Experimental Neurosciences, I.R.C.C.S. "Fondazione Santa Lucia", Rome, Italy
| | - Laura Altieri
- Unit of Child and Adolescent NeuroPsychiatry, Laboratory of Molecular Psychiatry and Neurogenetics, University "Campus Bio-Medico", Rome, Italy; Department of Experimental Neurosciences, I.R.C.C.S. "Fondazione Santa Lucia", Rome, Italy
| | | | - Chiara Gregorj
- Hematology Transfusion Medicine, University "Campus Bio-Medico", Rome, Italy
| | - Carmela Bravaccio
- Department of Translational Medical Science, University "Federico II", Naples, Italy
| | | | - Antonio M Persico
- Unit of Child and Adolescent NeuroPsychiatry, Laboratory of Molecular Psychiatry and Neurogenetics, University "Campus Bio-Medico", Rome, Italy; Department of Experimental Neurosciences, I.R.C.C.S. "Fondazione Santa Lucia", Rome, Italy; Mafalda Luce Center for Pervasive Developmental Disorders, Milan, Italy.
| |
Collapse
|
21
|
Herbert MR, Sage C. Autism and EMF? Plausibility of a pathophysiological link - Part I. ACTA ACUST UNITED AC 2013; 20:191-209. [PMID: 24095003 DOI: 10.1016/j.pathophys.2013.08.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2013] [Revised: 05/06/2013] [Accepted: 07/15/2013] [Indexed: 01/02/2023]
Abstract
Although autism spectrum conditions (ASCs) are defined behaviorally, they also involve multileveled disturbances of underlying biology that find striking parallels in the physiological impacts of electromagnetic frequency and radiofrequency exposures (EMF/RFR). Part I of this paper will review the critical contributions pathophysiology may make to the etiology, pathogenesis and ongoing generation of core features of ASCs. We will review pathophysiological damage to core cellular processes that are associated both with ASCs and with biological effects of EMF/RFR exposures that contribute to chronically disrupted homeostasis. Many studies of people with ASCs have identified oxidative stress and evidence of free radical damage, cellular stress proteins, and deficiencies of antioxidants such as glutathione. Elevated intracellular calcium in ASCs may be due to genetics or may be downstream of inflammation or environmental exposures. Cell membrane lipids may be peroxidized, mitochondria may be dysfunctional, and various kinds of immune system disturbances are common. Brain oxidative stress and inflammation as well as measures consistent with blood-brain barrier and brain perfusion compromise have been documented. Part II of this paper will review how behaviors in ASCs may emerge from alterations of electrophysiological oscillatory synchronization, how EMF/RFR could contribute to these by de-tuning the organism, and policy implications of these vulnerabilities. Changes in brain and autonomic nervous system electrophysiological function and sensory processing predominate, seizures are common, and sleep disruption is close to universal. All of these phenomena also occur with EMF/RFR exposure that can add to system overload ('allostatic load') in ASCs by increasing risk, and worsening challenging biological problems and symptoms; conversely, reducing exposure might ameliorate symptoms of ASCs by reducing obstruction of physiological repair. Various vital but vulnerable mechanisms such as calcium channels may be disrupted by environmental agents, various genes associated with autism or the interaction of both. With dramatic increases in reported ASCs that are coincident in time with the deployment of wireless technologies, we need aggressive investigation of potential ASC - EMF/RFR links. The evidence is sufficient to warrant new public exposure standards benchmarked to low-intensity (non-thermal) exposure levels now known to be biologically disruptive, and strong, interim precautionary practices are advocated.
Collapse
Affiliation(s)
- Martha R Herbert
- TRANSCEND Research Program Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA.
| | | |
Collapse
|
22
|
Distler MG, Palmer AA. Role of Glyoxalase 1 (Glo1) and methylglyoxal (MG) in behavior: recent advances and mechanistic insights. Front Genet 2012. [PMID: 23181072 PMCID: PMC3500958 DOI: 10.3389/fgene.2012.00250] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Glyoxalase 1 (GLO1) is a ubiquitous cellular enzyme that participates in the detoxification of methylglyoxal (MG), a cytotoxic byproduct of glycolysis that induces protein modification (advanced glycation end-products, AGEs), oxidative stress, and apoptosis. The concentration of MG is elevated under high-glucose conditions, such as diabetes. As such, GLO1 and MG have been implicated in the pathogenesis of diabetic complications. Recently, findings have linked GLO1 to numerous behavioral phenotypes, including psychiatric diseases (anxiety, depression, schizophrenia, and autism) and pain. This review highlights GLO1's association with behavioral phenotypes, describes recent discoveries that have elucidated the underlying mechanisms, and identifies opportunities for future research.
Collapse
|
23
|
Rose S, Melnyk S, Pavliv O, Bai S, Nick TG, Frye RE, James SJ. Evidence of oxidative damage and inflammation associated with low glutathione redox status in the autism brain. Transl Psychiatry 2012; 2:e134. [PMID: 22781167 PMCID: PMC3410618 DOI: 10.1038/tp.2012.61] [Citation(s) in RCA: 326] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Despite increasing evidence of oxidative stress in the pathophysiology of autism, most studies have not evaluated biomarkers within specific brain regions, and the functional consequences of oxidative stress remain relatively understudied. We examined frozen samples from the cerebellum and temporal cortex (Brodmann area 22 (BA22)) from individuals with autism and unaffected controls (n=15 and n=12 per group, respectively). Biomarkers of oxidative stress, including reduced glutathione (GSH), oxidized glutathione (GSSG) and glutathione redox/antioxidant capacity (GSH/GSSG), were measured. Biomarkers of oxidative protein damage (3-nitrotyrosine; 3-NT) and oxidative DNA damage (8-oxo-deoxyguanosine; 8-oxo-dG) were also assessed. Functional indicators of oxidative stress included relative levels of 3-chlorotyrosine (3-CT), an established biomarker of a chronic inflammatory response, and aconitase activity, a biomarker of mitochondrial superoxide production. Consistent with previous studies on plasma and immune cells, GSH and GSH/GSSG were significantly decreased in both autism cerebellum (P<0.01) and BA22 (P<0.01). There was a significant increase in 3-NT in the autism cerebellum and BA22 (P<0.01). Similarly, 8-oxo-dG was significantly increased in autism cerebellum and BA22 (P<0.01 and P=0.01, respectively), and was inversely correlated with GSH/GSSG in the cerebellum (P<0.01). There was a significant increase in 3-CT levels in both brain regions (P<0.01), whereas aconitase activity was significantly decreased in autism cerebellum (P<0.01), and was negatively correlated with GSH/GSSG (P=0.01). Together, these results indicate that decreased GSH/GSSG redox/antioxidant capacity and increased oxidative stress in the autism brain may have functional consequence in terms of a chronic inflammatory response, increased mitochondrial superoxide production, and oxidative protein and DNA damage.
Collapse
Affiliation(s)
- S Rose
- Department of Pediatrics, University of Arkansas for Medical Sciences, Arkansas Children's Hospital Research Institute, Little Rock, AR 72202, USA.
| | - S Melnyk
- Department of Pediatrics, University of Arkansas for Medical Sciences, Arkansas Children's Hospital Research Institute, Little Rock, AR, USA
| | - O Pavliv
- Department of Pediatrics, University of Arkansas for Medical Sciences, Arkansas Children's Hospital Research Institute, Little Rock, AR, USA
| | - S Bai
- Department of Pediatrics, University of Arkansas for Medical Sciences, Arkansas Children's Hospital Research Institute, Little Rock, AR, USA
| | - T G Nick
- Department of Pediatrics, University of Arkansas for Medical Sciences, Arkansas Children's Hospital Research Institute, Little Rock, AR, USA
| | - R E Frye
- Department of Pediatrics, University of Arkansas for Medical Sciences, Arkansas Children's Hospital Research Institute, Little Rock, AR, USA
| | - S J James
- Department of Pediatrics, University of Arkansas for Medical Sciences, Arkansas Children's Hospital Research Institute, Little Rock, AR, USA
| |
Collapse
|
24
|
Intracellular and extracellular redox status and free radical generation in primary immune cells from children with autism. AUTISM RESEARCH AND TREATMENT 2011; 2012:986519. [PMID: 22928106 PMCID: PMC3420377 DOI: 10.1155/2012/986519] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2011] [Revised: 08/12/2011] [Accepted: 09/12/2011] [Indexed: 12/01/2022]
Abstract
The modulation of the redox microenvironment is an important regulator of immune cell activation and proliferation. To investigate immune cell redox status in autism we quantified the intracellular glutathione redox couple (GSH/GSSG) in resting peripheral blood mononuclear cells (PBMCs), activated monocytes and CD4 T cells and the extracellular cysteine/cystine redox couple in the plasma from 43 children with autism and 41 age-matched control children. Resting PBMCs and activated monocytes from children with autism exhibited significantly higher oxidized glutathione (GSSG) and percent oxidized glutathione equivalents and decreased glutathione redox status (GSH/GSSG). In activated CD4 T cells from children with autism, the percent oxidized glutathione equivalents were similarly increased, and GSH and GSH/GSSG were decreased. In the plasma, both glutathione and cysteine redox ratios were decreased in autistic compared to control children. Consistent with decreased intracellular and extracellular redox status, generation of free radicals was significantly elevated in lymphocytes from the autistic children. These data indicate primary immune cells from autistic children have a more oxidized intracellular and extracellular microenvironment and a deficit in glutathione-mediated redox/antioxidant capacity compared to control children. These results suggest that the loss of glutathione redox homeostasis and chronic oxidative stress may contribute to immune dysregulation in autism.
Collapse
|
25
|
Vezzani A, Aronica E, Mazarati A, Pittman QJ. Epilepsy and brain inflammation. Exp Neurol 2011; 244:11-21. [PMID: 21985866 DOI: 10.1016/j.expneurol.2011.09.033] [Citation(s) in RCA: 421] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2011] [Revised: 09/15/2011] [Accepted: 09/26/2011] [Indexed: 02/06/2023]
Abstract
During the last decade, experimental research has demonstrated a prominent role of glial cells, activated in brain by various injuries, in the mechanisms of seizure precipitation and recurrence. In particular, alterations in the phenotype and function of activated astrocytes and microglial cells have been described in experimental and human epileptic tissue, including modifications in potassium and water channels, alterations of glutamine/glutamate cycle, changes in glutamate receptor expression and transporters, release of neuromodulatory molecules (e.g. gliotransmitters, neurotrophic factors), and induction of molecules involved in inflammatory processes (e.g. cytokines, chemokines, prostaglandins, complement factors, cell adhesion molecules) (Seifert et al., 2006; Vezzani et al., 2011; Wetherington et al., 2008). In particular, brain injury or proconvulsant events can activate microglia and astrocytes to release a number of proinflammatory mediators, thus initiating a cascade of inflammatory processes in brain tissue. Proinflammatory molecules can alter neuronal excitability and affect the physiological functions of glia by paracrine or autocrine actions, thus perturbing the glioneuronal communications. In experimental models, these changes contribute to decreasing the threshold to seizures and may compromise neuronal survival (Riazi et al., 2010; Vezzani et al., 2008). In this context, understanding which are the soluble mediators and the molecular mechanisms crucially involved in glio-neuronal interactions is instrumental to shed light on how brain inflammation may contribute to neuronal hyperexcitability in epilepsy. This review will report the clinical observations in drug-resistant human epilepsies and the experimental findings in adult and immature rodents linking brain inflammation to the epileptic process in a causal and reciprocal manner. By confronting the clinical evidence with the experimental findings, we will discuss the role of specific soluble inflammatory mediators in the etiopathogenesis of seizures, reporting evidence for both their acute and long term effects on seizure threshold. The possible contribution of these mediators to co-morbidities often described in epilepsy patients will be also discussed. Finally, we will report on the anti-inflammatory treatments with anticonvulsant actions in experimental models highlighting possible therapeutic options for treating drug-resistant seizures and for prevention of epileptogenesis.
Collapse
Affiliation(s)
- Annamaria Vezzani
- Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Via G. La Masa 19, 20156 Milano, Italy.
| | | | | | | |
Collapse
|
26
|
Meguid NA, Dardir AA, Abdel-Raouf ER, Hashish A. Evaluation of oxidative stress in autism: defective antioxidant enzymes and increased lipid peroxidation. Biol Trace Elem Res 2011; 143:58-65. [PMID: 20845086 DOI: 10.1007/s12011-010-8840-9] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2010] [Accepted: 09/02/2010] [Indexed: 10/19/2022]
Abstract
Autism is a neurodevelopmental disorder of childhood with poorly understood etiology and pathology. This pilot study aims to evaluate the levels of antioxidant enzymes, superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px), and levels of malondialdehyde (MDA), a marker of lipid peroxidation, in Egyptian autistic children. Autism is a neurodevelopmental disorder of childhood with poorly understood etiology and pathology. The present study included 20 children with autism diagnosed by DSM-IV-TR criteria and Childhood Autism Rating Scale. Controls included 25 age-matched healthy children. Cases were referred to Outpatient Clinic of Children with Special Needs Department, National Research Center, Cairo, Egypt. We compared levels of SOD, GSH-Px, and MDA in children with autism and controls. In children less than 6 years of age, levels of SOD, and GSH-Px were significantly lower in autistic children compared with their controls, while MDA was significantly higher among patients than controls. In children older than 6 years, there was no significant difference in any of these values between cases and controls. We concluded that children with autism are more vulnerable to oxidative stress in the form of increased lipid peroxidation and deficient antioxidant defense mechanism especially at younger children. We highlight that autistic children might benefit from antioxidants supplementation coupled with polyunsaturated fatty acids. Moreover, early assessment of antioxidant status would have better prognosis as it may decrease the oxidative stress before inducing more irreversible brain damage.
Collapse
Affiliation(s)
- Nagwa A Meguid
- Department of Research on Children with Special Needs, National Research Center, Tahrir St, Cairo, Egypt.
| | | | | | | |
Collapse
|
27
|
Volk HE, Hertz-Picciotto I, Delwiche L, Lurmann F, McConnell R. Residential proximity to freeways and autism in the CHARGE study. ENVIRONMENTAL HEALTH PERSPECTIVES 2011; 119:873-7. [PMID: 21156395 PMCID: PMC3114825 DOI: 10.1289/ehp.1002835] [Citation(s) in RCA: 236] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2010] [Accepted: 12/13/2010] [Indexed: 05/17/2023]
Abstract
BACKGROUND Little is known about environmental causes and contributing factors for autism. Basic science and epidemiologic research suggest that oxidative stress and inflammation may play a role in disease development. Traffic-related air pollution, a common exposure with established effects on these pathways, contains substances found to have adverse prenatal effects. OBJECTIVES We examined the association between autism and proximity of residence to freeways and major roadways during pregnancy and near the time of delivery, as a surrogate for air pollution exposure. METHODS Data were from 304 autism cases and 259 typically developing controls enrolled in the Childhood Autism Risks from Genetics and the Environment (CHARGE) study. The mother's address recorded on the birth certificate and trimester-specific addresses derived from a residential history obtained by questionnaire were geocoded, and measures of distance to freeways and major roads were calculated using ArcGIS software. Logistic regression models compared residential proximity to freeways and major roads for autism cases and typically developing controls. RESULTS Adjusting for sociodemographic factors and maternal smoking, maternal residence at the time of delivery was more likely be near a freeway (≤ 309 m) for cases than for controls [odds ratio (OR)=1.86; 95% confidence interval (CI), 1.04-3.45]. Autism was also associated with residential proximity to a freeway during the third trimester (OR=2.22; CI, 1.16-4.42). After adjustment for socioeconomic and sociodemographic characteristics, these associations were unchanged. Living near other major roads at birth was not associated with autism. CONCLUSIONS Living near a freeway was associated with autism. Examination of associations with measured air pollutants is needed.
Collapse
Affiliation(s)
- Heather E Volk
- Department of Preventive Medicine, Zilkha Neurogenetic Institute, Keck School of Medicine, Children's Hospital Los Angeles, University of Southern California, Los Angeles, California 90033, USA.
| | | | | | | | | |
Collapse
|
28
|
Barua M, Jenkins EC, Chen W, Kuizon S, Pullarkat RK, Junaid MA. Glyoxalase I polymorphism rs2736654 causing the Ala111Glu substitution modulates enzyme activity--implications for autism. Autism Res 2011; 4:262-70. [PMID: 21491613 DOI: 10.1002/aur.197] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2009] [Accepted: 03/12/2011] [Indexed: 11/08/2022]
Abstract
Autism is a pervasive, heterogeneous, neurodevelopmental disability characterized by impairments in verbal communications, reciprocal social interactions, and restricted repetitive stereotyped behaviors. Evidence suggests the involvement of multiple genetic factors in the etiology of autism, and extensive genome-wide association studies have revealed several candidate genes that bear single nucleotide polymorphisms (SNPs) in non-coding and coding regions. We have shown that a non-conservative, non-synonymous SNP in the glyoxalase I gene, GLOI, may be an autism susceptibility factor. The GLOI rs2736654 SNP is a C→A change that causes an Ala111Glu change in the Glo1 enzyme. To identify the significance of the SNP, we have conducted functional assays for Glo1. We now present evidence that the presence of the A-allele at rs2736654 results in reduced enzyme activity. Glo1 activity is decreased in lymphoblastoid cells that are homozygous for the A allele. The Glu-isoform of Glo1 in these cells is hyperphosphorylated. Direct HPLC measurements of the glyoxalase I substrate, methylglyoxal (MG), show an increase in MG in these cells. Western blot analysis revealed elevated levels of the receptor for advanced glycation end products (RAGEs). We also show that MG is toxic to the developing neuronal cells. We suggest that accumulation of MG results in the formation of AGEs, which induce expression of the RAGE that during crucial neuronal development may be a factor in the pathology of autism.
Collapse
Affiliation(s)
- Madhabi Barua
- Department of Developmental Biochemistry, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, NY 10314, USA
| | | | | | | | | | | |
Collapse
|
29
|
Emanuele E, Boso M, Brondino N, Pietra S, Barale F, Ucelli di Nemi S, Politi P. Increased serum levels of high mobility group box 1 protein in patients with autistic disorder. Prog Neuropsychopharmacol Biol Psychiatry 2010; 34:681-3. [PMID: 20302902 DOI: 10.1016/j.pnpbp.2010.03.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2010] [Revised: 02/28/2010] [Accepted: 03/12/2010] [Indexed: 11/19/2022]
Abstract
BACKGROUND High mobility group box 1 (HMGB1) is a highly conserved, ubiquitous protein that functions as an activator for inducing the immune response and can be released from neurons after glutamate excitotoxicity. The objective of the present study was to measure serum levels of HMGB1 in patients with autistic disorder and to study their relationship with clinical characteristics. METHODS We enrolled 22 adult patients with autistic disorder (mean age: 28.1+/-7.7 years) and 28 age- and gender-matched healthy controls (mean age: 28.7+/-8.1 years). Serum levels of HMGB1 were measured by enzyme-linked immunosorbent assay (ELISA). RESULTS Compared with healthy subjects, serum levels of HMGB1 were significantly higher in patients with autistic disorder (10.8+/-2.6 ng/mL versus 5.6+/-2.5 ng/mL, respectively, P<0.001). After adjustment for potential confounders, serum HMGB1 levels were independently associated with their domain A scores in the Autism Diagnostic Interview-Revised, which reflects their impairments in social interaction. CONCLUSIONS These results suggest that HMGB1 levels may be affected in autistic disorder. Increased HMGB1 may be a biological correlate of the impaired reciprocal social interactions in this neurodevelopmental disorder.
Collapse
Affiliation(s)
- Enzo Emanuele
- Department of Health Sciences, Section of Psychiatry, University of Pavia, Via Bassi, 21, I-27100, Pavia, Italy.
| | | | | | | | | | | | | |
Collapse
|
30
|
Youn SI, Jin SH, Kim SH, Lim S. Porphyrinuria in Korean children with autism: correlation with oxidative stress. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2010; 73:701-710. [PMID: 20391113 DOI: 10.1080/15287391003614000] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder believed to be associated with heavy metal exposure, especially mercury (Hg), and is characterized by disturbances in metal elimination. Various studies correlated elevated heavy metal body burden with ASD diagnoses as evidenced by increased urinary porphyrin levels in patients. Urinary porphyrins were also determined in Korean patients diagnosed with ASD (n = 65) who visited AK Eastern Medicinal Clinic in Kangnam-gu, Seoul, from June 2007 to September 2008, compared to controls (n = 9) residing in the same area, by means of Metametrix (CLIA-approved) laboratory testing. Further, urinary organic acids as indicators of hepatic detoxification/oxidative stress were also analyzed among patients diagnosed with ASD. Significant increases were found in patients diagnosed with ASD for proporphyrins, pentacarboxyporphyrin, precoproporphyrin, coproporphyrins, and total porphyrins. Significant correlations were observed between hepatic detoxification/oxidative stress markers and urinary porphyrins. In agreement with published data, the present results demonstrated that measurement of porphyrins serves as a reliable tool for diagnosis of heavy metal involvement in ASD.
Collapse
Affiliation(s)
- Seung-Il Youn
- Department of Basic Eastern Medical Science, Graduate School, KyungHee University, Seoul, Republic of Korea
| | | | | | | |
Collapse
|
31
|
James SJ, Rose S, Melnyk S, Jernigan S, Blossom S, Pavliv O, Gaylor DW. Cellular and mitochondrial glutathione redox imbalance in lymphoblastoid cells derived from children with autism. FASEB J 2009; 23:2374-83. [PMID: 19307255 DOI: 10.1096/fj.08-128926] [Citation(s) in RCA: 168] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Research into the metabolic phenotype of autism has been relatively unexplored despite the fact that metabolic abnormalities have been implicated in the pathophysiology of several other neurobehavioral disorders. Plasma biomarkers of oxidative stress have been reported in autistic children; however, intracellular redox status has not yet been evaluated. Lymphoblastoid cells (LCLs) derived from autistic children and unaffected controls were used to assess relative concentrations of reduced glutathione (GSH) and oxidized disulfide glutathione (GSSG) in cell extracts and isolated mitochondria as a measure of intracellular redox capacity. The results indicated that the GSH/GSSG redox ratio was decreased and percentage oxidized glutathione increased in both cytosol and mitochondria in the autism LCLs. Exposure to oxidative stress via the sulfhydryl reagent thimerosal resulted in a greater decrease in the GSH/GSSG ratio and increase in free radical generation in autism compared to control cells. Acute exposure to physiological levels of nitric oxide decreased mitochondrial membrane potential to a greater extent in the autism LCLs, although GSH/GSSG and ATP concentrations were similarly decreased in both cell lines. These results suggest that the autism LCLs exhibit a reduced glutathione reserve capacity in both cytosol and mitochondria that may compromise antioxidant defense and detoxification capacity under prooxidant conditions.
Collapse
Affiliation(s)
- S Jill James
- Department of Pediatrics, University of Arkansas for Medical Sciences, Arkansas Children's Hospital Research Institute, 1120 Marshall St., Little Rock, AR 72202, USA.
| | | | | | | | | | | | | |
Collapse
|
32
|
Geier DA, Geier MR. Autism spectrum disorder-associated biomarkers for case evaluation and management by clinical geneticists. Expert Rev Mol Diagn 2009; 8:671-4. [PMID: 18999918 DOI: 10.1586/14737159.8.6.671] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
33
|
Abstract
Improving clinical tests are allowing us to more precisely classify autism spectrum disorders and diagnose them at earlier ages. This raises the possibility of earlier and potentially more effective therapeutic interventions. To fully capitalize on this opportunity, however, will require better understanding of the neurobiological changes underlying this devastating group of developmental disorders. It is becoming clear that the normal trajectory of neurodevelopment is altered in autism, with aberrations in brain growth, neuronal patterning and cortical connectivity. Changes to the structure and function of synapses and dendrites have also been strongly implicated in the pathology of autism by morphological, genetic and animal modeling studies. Finally, environmental factors are likely to interact with the underlying genetic profile, and foster the clinical heterogeneity seen in autism spectrum disorders. In this review we attempt to link the molecular pathways altered in autism to the neurodevelopmental and clinical changes that characterize the disease. We focus on signaling molecules such as neurotrophin, Reelin, PTEN and hepatocyte growth factor, neurotransmitters such as serotonin and glutamate, and synaptic proteins such as neurexin, SHANK and neuroligin. We also discuss evidence implicating oxidative stress, neuroglial activation and neuroimmunity in autism.
Collapse
Affiliation(s)
- Carlos A Pardo
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287. USA.
| | | |
Collapse
|