1
|
Davias A, Lyon-Caen S, Rolland M, Iszatt N, Thomsen C, Sabaredzovic A, Sakhi AK, Monot C, Rayah Y, Ilhan ZE, Philippat C, Eggesbø M, Lepage P, Slama R. ASSOCIATIONS BETWEEN PRE- AND POST-NATAL EXPOSURE TO PHTHALATE AND DINCH METABOLITES AND GUT MICROBIOTA IN ONE-YEAR OLD CHILDREN. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024:125204. [PMID: 39490662 DOI: 10.1016/j.envpol.2024.125204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 10/03/2024] [Accepted: 10/25/2024] [Indexed: 11/05/2024]
Abstract
The gut microbiota is a collection of symbiotic microorganisms in the gastrointestinal tract. Its sensitivity to chemicals with widespread exposure, such as phthalates, is little known. We aimed to investigate the impact of perinatal exposure to phthalates on the infant gut microbiota at 12 months of age. Within SEPAGES cohort (Suivi de l'Exposition à la Pollution Atmosphérique durant la Grossesse et Effet sur la Santé), we assessed 13 phthalate metabolites and 2 di(isononyl) cyclohexane-1,2-dicarboxylate (DINCH) metabolites in repeated urine samples collected in pregnant women and their offspring. We obtained stool samples from 356 children at 12 months of age and sequenced the V3-V4 region of the 16S rRNA gene, allowing gut bacterial profiling. We used single-chemical (linear regressions) and mixture (BKMR, Bayesian Kernel Machine Regression) models to examine associations of phthalates and DINCH metabolites, with gut microbiota indices of α-diversity (specific richness and Shannon diversity) and the relative abundances of the most abundant microbiota phyla and genera. After correction for multiple testing, di(2-ethylhexyl) phthalate (ΣDEHP), diethyl phthalate (DEP) and bis(2-propylheptyl) phthalate (DPHP) metabolites 12-month urinary concentrations were associated with higher Shannon α-diversity of the child gut microbiota in single-chemical models. The multiple-chemical model (BKMR) suggested higher α-diversity with exposure to the phthalate mixture at 12 months, driven by the same phthalates. There were no associations between phthalate and DINCH exposure biomarkers at other time points and α-diversity after correction for multiple testing. ΣDEHP metabolites concentration at 12 months was associated with higher Coprococcus genus. Finally, ΣDEHP exposure at 12 months tended to be associated with higher phylum Firmicutes, an association not maintained after correction for multiple testing. Infancy exposure to phthalate might disrupt children's gut microbiota. The observed associations were cross-sectional, so that reverse causality cannot be excluded.
Collapse
Affiliation(s)
- Aline Davias
- Environmental Epidemiology applied to Development and Respiratory Health team, Institute for Advanced Biosciences, Inserm U1209, CNRS UMR 5309, University Grenoble Alpes, 38000 Grenoble, France.
| | - Sarah Lyon-Caen
- Environmental Epidemiology applied to Development and Respiratory Health team, Institute for Advanced Biosciences, Inserm U1209, CNRS UMR 5309, University Grenoble Alpes, 38000 Grenoble, France
| | - Matthieu Rolland
- Environmental Epidemiology applied to Development and Respiratory Health team, Institute for Advanced Biosciences, Inserm U1209, CNRS UMR 5309, University Grenoble Alpes, 38000 Grenoble, France
| | - Nina Iszatt
- Division of Climate and Environmental Health, Norwegian Institute of Public Health (NIPH), 0213 Oslo, Norway
| | - Cathrine Thomsen
- Division of Climate and Environmental Health, Norwegian Institute of Public Health (NIPH), 0213 Oslo, Norway
| | - Azemira Sabaredzovic
- Division of Climate and Environmental Health, Norwegian Institute of Public Health (NIPH), 0213 Oslo, Norway
| | - Amrit Kaur Sakhi
- Division of Climate and Environmental Health, Norwegian Institute of Public Health (NIPH), 0213 Oslo, Norway
| | - Celine Monot
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, F-78350, Jouy-en-Josas, France
| | - Yamina Rayah
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, F-78350, Jouy-en-Josas, France
| | - Zehra Esra Ilhan
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, F-78350, Jouy-en-Josas, France
| | - Claire Philippat
- Environmental Epidemiology applied to Development and Respiratory Health team, Institute for Advanced Biosciences, Inserm U1209, CNRS UMR 5309, University Grenoble Alpes, 38000 Grenoble, France
| | - Merete Eggesbø
- Department of Clinical and Molecular Medicine, NTNU, Trondheim, Norway
| | - Patricia Lepage
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, F-78350, Jouy-en-Josas, France
| | - Rémy Slama
- Environmental Epidemiology applied to Development and Respiratory Health team, Institute for Advanced Biosciences, Inserm U1209, CNRS UMR 5309, University Grenoble Alpes, 38000 Grenoble, France
| |
Collapse
|
2
|
Kurokawa S, Nomura K, Sanada K, Miyaho K, Ishii C, Fukuda S, Iwamoto C, Naraoka M, Yoneda S, Imafuku M, Matsuzaki J, Saito Y, Mimura M, Kishimoto T. A comparative study on dietary diversity and gut microbial diversity in children with autism spectrum disorder, attention-deficit hyperactivity disorder, their neurotypical siblings, and non-related neurotypical volunteers: a cross-sectional study. J Child Psychol Psychiatry 2024; 65:1184-1195. [PMID: 38562118 DOI: 10.1111/jcpp.13962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/29/2023] [Indexed: 04/04/2024]
Abstract
BACKGROUND Previous research has shown a significant link between gut microbiota in children with autism spectrum disorder (ASD) and attention-deficit hyperactivity disorder (ADHD). However, much remains unknown because of the heterogeneity of disorders and the potential confounders such as dietary patterns and control group variations. METHODS Children aged 6-12 years who had been clinically diagnosed with ASD and/or ADHD, their unaffected neurotypical siblings, and non-related neurotypical volunteers were recruited cross-sectionally. The ASD diagnosis was confirmed using the Autism Diagnostic Observation Schedule-2 (ADOS-2) in all patients, including those with ADHD. Standardized DNA extraction and sequencing methods were used to compare gut microbial alpha-diversity among the groups. Dietary diversity was calculated from a standardized dietary questionnaire form. We compared the difference in gut microbiome between patients with ASD and/or ADHD with neurotypical siblings and non-related neurotypical controls. RESULTS Ninety-eight subjects were included in the study (18 with ASD, 19 with ADHD, 20 with both ASD and ADHD, 13 neurotypical siblings, and 28 non-related neurotypical controls). The alpha-diversity indices, such as Chao 1 and Shannon index, showed a significant difference between the groups in a Linear mixed-effect model (F(4, 93) = 4.539, p = .02), (F(4, 93) = 3.185, p = .017), respectively. In a post-hoc pairwise comparison, patients with ASD had lower alpha-diversity compared with non-related controls after Bonferroni correction. Dietary diversity shown in Shannon index did not differ among the groups (F(4, 84) = 1.494, p = .211). CONCLUSIONS Our study indicates disorder-specific microbiome differences in patients with ASD. In future research on gut microbiota in neurodevelopmental disorders, it is necessary to consider the impact of ASD and ADHD co-occurrence, and strictly control for background information such as diet, to elucidate the gut-microbiota interaction in ASD and ADHD for exploring the potential of therapeutic interventions.
Collapse
Affiliation(s)
- Shunya Kurokawa
- Department of Neuropsychiatry, School of Medicine, Keio University, Tokyo, Japan
| | - Kensuke Nomura
- Department of Child Psychiatry, Shimada Ryoiku Medical Center for Challenged Children, Tokyo, Japan
| | - Kenji Sanada
- Department of Psychiatry, Showa University School of Medicine, Tokyo, Japan
| | - Katsuma Miyaho
- Department of Psychiatry, Showa University School of Medicine, Tokyo, Japan
| | - Chiharu Ishii
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
| | - Shinji Fukuda
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
- Laboratory for Regenerative Microbiology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Gut Environmental Design Group, Kanagawa Institute of Industrial Science and Technology, Kawasaki, Japan
- Transborder Medical Research Center, University of Tsukuba, Tsukuba, Japan
| | - Chiaki Iwamoto
- Division of Pharmacotherapeutics, Faculty of Pharmacy, Keio University, Tokyo, Japan
| | - Minori Naraoka
- Division of Pharmacotherapeutics, Faculty of Pharmacy, Keio University, Tokyo, Japan
| | - Shintaro Yoneda
- Division of Pharmacotherapeutics, Faculty of Pharmacy, Keio University, Tokyo, Japan
| | | | - Juntaro Matsuzaki
- Division of Pharmacotherapeutics, Faculty of Pharmacy, Keio University, Tokyo, Japan
| | - Yoshimasa Saito
- Division of Pharmacotherapeutics, Faculty of Pharmacy, Keio University, Tokyo, Japan
| | - Masaru Mimura
- Department of Neuropsychiatry, School of Medicine, Keio University, Tokyo, Japan
| | - Taishiro Kishimoto
- Department of Neuropsychiatry, School of Medicine, Keio University, Tokyo, Japan
| |
Collapse
|
3
|
Climent-Pérez P, Martínez-González AE, Andreo-Martínez P. Contributions of Artificial Intelligence to Analysis of Gut Microbiota in Autism Spectrum Disorder: A Systematic Review. CHILDREN (BASEL, SWITZERLAND) 2024; 11:931. [PMID: 39201866 PMCID: PMC11352523 DOI: 10.3390/children11080931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 07/26/2024] [Accepted: 07/27/2024] [Indexed: 09/03/2024]
Abstract
BACKGROUND Autism spectrum disorder (ASD) is a highly heterogeneous neurodevelopmental disorder whose etiology is not known today, but everything indicates that it is multifactorial. For example, genetic and epigenetic factors seem to be involved in the etiology of ASD. In recent years, there has been an increase in studies on the implications of gut microbiota (GM) on the behavior of children with ASD given that dysbiosis in GM may trigger the onset, development and progression of ASD through the microbiota-gut-brain axis. At the same time, significant progress has occurred in the development of artificial intelligence (AI). METHODS The aim of the present study was to perform a systematic review of articles using AI to analyze GM in individuals with ASD. In line with the PRISMA model, 12 articles using AI to analyze GM in ASD were selected. RESULTS Outcomes reveal that the majority of relevant studies on this topic have been conducted in China (33.3%) and Italy (25%), followed by the Netherlands (16.6%), Mexico (16.6%) and South Korea (8.3%). CONCLUSIONS The bacteria Bifidobacterium is the most relevant biomarker with regard to ASD. Although AI provides a very promising approach to data analysis, caution is needed to avoid the over-interpretation of preliminary findings. A first step must be taken to analyze GM in a representative general population and ASD samples in order to obtain a GM standard according to age, sex and country. Thus, more work is required to bridge the gap between AI in mental health research and clinical care in ASD.
Collapse
Affiliation(s)
- Pau Climent-Pérez
- Department of Computing Technology, University of Alicante, 03690 San Vicente del Raspeig, Alicante, Spain;
| | | | - Pedro Andreo-Martínez
- Department of Agricultural Chemistry, Faculty of Chemistry, Regional Campus of International Excellence “Campus Mare Nostrum”, University of Murcia, Campus of Espinardo, 30100 Murcia, Spain;
| |
Collapse
|
4
|
Phan J, Calvo DC, Nair D, Jain S, Montagne T, Dietsche S, Blanchard K, Treadwell S, Adams J, Krajmalnik-Brown R. Precision synbiotics increase gut microbiome diversity and improve gastrointestinal symptoms in a pilot open-label study for autism spectrum disorder. mSystems 2024; 9:e0050324. [PMID: 38661344 PMCID: PMC11097633 DOI: 10.1128/msystems.00503-24] [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: 04/08/2024] [Accepted: 04/08/2024] [Indexed: 04/26/2024] Open
Abstract
The efficacy of prebiotics and probiotics (synbiotics when combined) to improve symptoms associated with autism spectrum disorder (ASD) has shown considerable inter-study variation, likely due to the complex, heterogeneous nature of the disorder and its associated behavioral, developmental, and gastrointestinal symptoms. Here, we present a precision synbiotic supplementation study in 296 children and adults diagnosed with ASD versus 123 age-matched neurotypical controls. One hundred seventy ASD participants completed the study. Baseline and post-synbiotic assessment of ASD and gastrointestinal (GI) symptoms and deep metagenomic sequencing were performed. Within the ASD cohort, there were significant differences in microbes between subpopulations based on the social responsiveness scale (SRS2) survey (Prevotella spp., Bacteroides, Fusicatenibacter, and others) and gluten and dairy-free diets (Bifidobacterium spp., Lactococcus, Streptococcus spp., and others). At the baseline, the ASD cohort maintained a lower taxonomic alpha diversity and significant differences in taxonomic composition, metabolic pathways, and gene families, with a greater proportion of potential pathogens, including Shigella, Klebsiella, and Clostridium, and lower proportions of beneficial microbes, including Faecalibacterium compared to controls. Following the 3-month synbiotic supplementation, the ASD cohort showed increased taxonomic alpha diversity, shifts in taxonomy and metabolic pathway potential, and improvements in some ASD-related symptoms, including a significant reduction in GI discomfort and overall improved language, comprehension, cognition, thinking, and speech. However, the open-label study design may include some placebo effects. In summary, we found that precision synbiotics modulated the gut microbiome and could be used as supplementation to improve gastrointestinal and ASD-related symptoms. IMPORTANCE Autism spectrum disorder (ASD) is prevalent in 1 out of 36 children in the United States and contributes to health, financial, and psychological burdens. Attempts to identify a gut microbiome signature of ASD have produced varied results. The limited pre-clinical and clinical population sizes have hampered the success of these trials. To understand the microbiome associated with ASD, we employed whole metagenomic shotgun sequencing to classify microbial composition and genetic functional potential. Despite being one of the most extensive ASD post-synbiotic assessment studies, the results highlight the complexity of performing such a case-control supplementation study in this population and the potential for a future therapeutic approach in ASD.
Collapse
Affiliation(s)
- Joann Phan
- Sun Genomics, Inc., San Diego, California, USA
| | - Diana C. Calvo
- Department of Civil Engineering, Construction Management, and Environmental Engineering, Northern Arizona University, Flagstaff, Arizona, USA
| | - Divya Nair
- Sun Genomics, Inc., San Diego, California, USA
| | - Suneer Jain
- Sun Genomics, Inc., San Diego, California, USA
| | | | | | | | | | - James Adams
- Biodesign Center for Health Through Microbiomes, Arizona State University, Tempe, Arizona, USA
| | - Rosa Krajmalnik-Brown
- Biodesign Center for Health Through Microbiomes, Arizona State University, Tempe, Arizona, USA
| |
Collapse
|
5
|
Chen Y, Xue Y, Jia L, Yang M, Huang G, Xie J. Causal effects of gut microbiota on autism spectrum disorder: A two-sample mendelian randomization study. Medicine (Baltimore) 2024; 103:e37284. [PMID: 38428908 PMCID: PMC10906619 DOI: 10.1097/md.0000000000037284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 01/25/2024] [Indexed: 03/03/2024] Open
Abstract
There is increasing evidence that alterations in gut microbiota (GM) composition are associated with autism spectrum disorder (ASD), but no reliable causal relationship has been established. Therefore, a 2-sample Mendelian randomization (MR) study was conducted to reveal a potential causal relationship between GM and ASD. Instrumental variables for 211 GM taxa were obtained from genome-wide association studies (GWAS) and Mendelian randomization studies to estimate their impact on ASD risk in the iPSYCH-PGC GWAS dataset (18,382 ASD cases and 27,969 controls). Inverse variance weighted (IVW) is the primary method for causality analysis, and several sensitivity analyses validate MR results. Among 211 GM taxa, IVW results confirmed that Tenericutes (P value = .0369), Mollicutes (P value = .0369), Negativicutes (P value = .0374), Bifidobacteriales (P value = .0389), Selenomonadales (P value = .0374), Bifidobacteriaceae (P value = .0389), Family XIII (P value = .0149), Prevotella7 (P value = .0215), Ruminococcaceae NK4A214 group (P value = .0205) were potential protective factors for ASD. Eisenbergiella (P value = .0159) was a possible risk factor for ASD. No evidence of heterogeneous, pleiotropic, or outlier single-nucleotide polymorphism was detected. Additionally, further sensitivity analysis verified the robustness of the above results. We confirm a potential causal relationship between certain gut microbes and ASD, providing new insights into how gut microbes mediate ASD. The association between them needs to be further explored and will provide new ideas for the prevention and treatment of ASD.
Collapse
Affiliation(s)
- Yajun Chen
- North Sichuan Medical College, Nanchong, Sichuan, China
- Department of Pediatrics, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Yan Xue
- Department of Pediatrics, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Lang Jia
- Department of Pediatrics, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Menghan Yang
- Department of Pediatrics, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Gelin Huang
- Sichuan University-The Chinese University of Hong Kong (SCU-CUHK) Joint Laboratory for Reproductive Medicine, Key Laboratory of Obstetric, Gynaecologic and Paediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jiang Xie
- Department of Pediatrics, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| |
Collapse
|
6
|
Lin P, Zhang Q, Sun J, Li Q, Li D, Zhu M, Fu X, Zhao L, Wang M, Lou X, Chen Q, Liang K, Zhu Y, Qu C, Li Z, Ma P, Wang R, Liu H, Dong K, Guo X, Cheng X, Sun Y, Sun J. A comparison between children and adolescents with autism spectrum disorders and healthy controls in biomedical factors, trace elements, and microbiota biomarkers: a meta-analysis. Front Psychiatry 2024; 14:1318637. [PMID: 38283894 PMCID: PMC10813399 DOI: 10.3389/fpsyt.2023.1318637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 12/13/2023] [Indexed: 01/30/2024] Open
Abstract
Introduction Autism spectrum disorder (ASD) is a multifaceted developmental condition that commonly appears during early childhood. The etiology of ASD remains multifactorial and not yet fully understood. The identification of biomarkers may provide insights into the underlying mechanisms and pathophysiology of the disorder. The present study aimed to explore the causes of ASD by investigating the key biomedical markers, trace elements, and microbiota factors between children with autism spectrum disorder (ASD) and control subjects. Methods Medline, PubMed, ProQuest, EMBASE, Cochrane Library, PsycINFO, Web of Science, and EMBSCO databases have been searched for publications from 2012 to 2023 with no language restrictions using the population, intervention, control, and outcome (PICO) approach. Keywords including "autism spectrum disorder," "oxytocin," "GABA," "Serotonin," "CRP," "IL-6," "Fe," "Zn," "Cu," and "gut microbiota" were used for the search. The Joanna Briggs Institute (JBI) critical appraisal checklist was used to assess the article quality, and a random model was used to assess the mean difference and standardized difference between ASD and the control group in all biomedical markers, trace elements, and microbiota factors. Results From 76,217 records, 43 studies met the inclusion and exclusion criteria and were included in this meta-analysis. The pooled analyses showed that children with ASD had significantly lower levels of oxytocin (mean differences, MD = -45.691, 95% confidence interval, CI: -61.667, -29.717), iron (MD = -3.203, 95% CI: -4.891, -1.514), and zinc (MD = -6.707, 95% CI: -12.691, -0.722), lower relative abundance of Bifidobacterium (MD = -1.321, 95% CI: -2.403, -0.238) and Parabacteroides (MD = -0.081, 95% CI: -0.148, -0.013), higher levels of c-reactive protein, CRP (MD = 0.401, 95% CI: 0.036, 0.772), and GABA (MD = 0.115, 95% CI: 0.045, 0.186), and higher relative abundance of Bacteroides (MD = 1.386, 95% CI: 0.717, 2.055) and Clostridium (MD = 0.281, 95% CI: 0.035, 0.526) when compared with controls. The results of the overall analyses were stable after performing the sensitivity analyses. Additionally, no substantial publication bias was observed among the studies. Interpretation Children with ASD have significantly higher levels of CRP and GABA, lower levels of oxytocin, iron, and zinc, lower relative abundance of Bifidobacterium and Parabacteroides, and higher relative abundance of Faecalibacterium, Bacteroides, and Clostridium when compared with controls. These results suggest that these indicators may be a potential biomarker panel for the diagnosis or determining therapeutic targets of ASD. Furthermore, large, sample-based, and randomized controlled trials are needed to confirm these results.
Collapse
Affiliation(s)
- Ping Lin
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qianwen Zhang
- Key Laboratory of Digital Technology in Medical Diagnostics of Zhejiang Province, Hangzhou, China
- Hangzhou Calibra Diagnostics, Hangzhou, China
| | - Junyu Sun
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Qingtian Li
- College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dan Li
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengyuan Zhu
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaomei Fu
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ling Zhao
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengxia Wang
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoyan Lou
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qing Chen
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kangyi Liang
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuxin Zhu
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Caiwei Qu
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenhua Li
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peijun Ma
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Renyu Wang
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huafen Liu
- Key Laboratory of Digital Technology in Medical Diagnostics of Zhejiang Province, Hangzhou, China
- Hangzhou Calibra Diagnostics, Hangzhou, China
| | - Ke Dong
- Institute for Global Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaokui Guo
- Institute for Global Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xunjia Cheng
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yang Sun
- Institute of Arthritis Research, Shanghai Academy of Chinese Medical Sciences, Shanghai, China
| | - Jing Sun
- School of Medicine and Dentistry, Institute for Integrated Intelligence and Systems, Griffith University, Gold Coast Campus, Gold Coast, QLD, Australia
- Charles Sturt University, Orange, NSW, Australia
| |
Collapse
|
7
|
Panzetta ME, Valdivia RH. Akkermansia in the gastrointestinal tract as a modifier of human health. Gut Microbes 2024; 16:2406379. [PMID: 39305271 PMCID: PMC11418289 DOI: 10.1080/19490976.2024.2406379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 09/25/2024] Open
Abstract
Akkermansia sp are common members of the human gut microbiota. Multiple reports have emerged linking the abundance of A. muciniphila to health benefits and disease risk in humans and animals. This review highlights findings linking Akkermansia species in the gastrointestinal (GI) tract to health outcomes across a spectrum of disorders, encompassing those that affect the digestive, respiratory, urinary, and central nervous systems. The mechanism through which Akkermansia exerts a beneficial versus a detrimental effect on health is likely dependent on the genetic makeup of the host metabolic capacity and immunomodulatory properties of the strain, the competition or cooperation with other members of the host microbiota, as well as synergy with co-administered therapies.
Collapse
Affiliation(s)
- Maria E. Panzetta
- Department of Integrative Immunobiology, Duke University, Durham, NC, USA
| | | |
Collapse
|
8
|
Ranieri A, Mennitti C, Falcone N, La Monica I, Di Iorio MR, Tripodi L, Gentile A, Vitale M, Pero R, Pastore L, D’Argenio V, Scudiero O, Lombardo B. Positive effects of physical activity in autism spectrum disorder: how influences behavior, metabolic disorder and gut microbiota. Front Psychiatry 2023; 14:1238797. [PMID: 38025444 PMCID: PMC10681626 DOI: 10.3389/fpsyt.2023.1238797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/25/2023] [Indexed: 12/01/2023] Open
Abstract
Autism spectrum disorder is a neurodevelopmental disorder characterized by social interactions and communication skills impairments that include intellectual disabilities, communication delays and self-injurious behaviors; often are present systemic comorbidities such as gastrointestinal disorders, obesity and cardiovascular disease. Moreover, in recent years has emerged a link between alterations in the intestinal microbiota and neurobehavioral symptoms in children with autism spectrum disorder. Recently, physical activity and exercise interventions are known to be beneficial for improving communication and social interaction and the composition of microbiota. In our review we intend to highlight how different types of sports can help to improve communication and social behaviors in children with autism and also show positive effects on gut microbiota composition.
Collapse
Affiliation(s)
| | - Cristina Mennitti
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Naples, Italy
| | - Noemi Falcone
- CEINGE-Biotecnologie Avanzate Franco Salvatore, Naples, Italy
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Naples, Italy
| | - Ilaria La Monica
- CEINGE-Biotecnologie Avanzate Franco Salvatore, Naples, Italy
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Naples, Italy
| | - Maria Rosaria Di Iorio
- CEINGE-Biotecnologie Avanzate Franco Salvatore, Naples, Italy
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Naples, Italy
| | - Lorella Tripodi
- CEINGE-Biotecnologie Avanzate Franco Salvatore, Naples, Italy
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Naples, Italy
| | - Alessandro Gentile
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Naples, Italy
| | - Maria Vitale
- CEINGE-Biotecnologie Avanzate Franco Salvatore, Naples, Italy
| | - Raffaella Pero
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Naples, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
| | - Lucio Pastore
- CEINGE-Biotecnologie Avanzate Franco Salvatore, Naples, Italy
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Naples, Italy
| | - Valeria D’Argenio
- CEINGE-Biotecnologie Avanzate Franco Salvatore, Naples, Italy
- Department of Human Sciences and Quality of Life Promotion, San Raffaele Open University, Rome, Italy
| | - Olga Scudiero
- CEINGE-Biotecnologie Avanzate Franco Salvatore, Naples, Italy
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Naples, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
| | - Barbara Lombardo
- CEINGE-Biotecnologie Avanzate Franco Salvatore, Naples, Italy
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Naples, Italy
| |
Collapse
|
9
|
Notting F, Pirovano W, Sybesma W, Kort R. The butyrate-producing and spore-forming bacterial genus Coprococcus as a potential biomarker for neurological disorders. GUT MICROBIOME (CAMBRIDGE, ENGLAND) 2023; 4:e16. [PMID: 39295905 PMCID: PMC11406416 DOI: 10.1017/gmb.2023.14] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 07/28/2023] [Accepted: 08/10/2023] [Indexed: 09/21/2024]
Abstract
The host-intestinal microbiome interaction has gained much scientific attention in the past two decades, boosted by advances in DNA sequencing and cultivation techniques. An accumulating amount of evidence shows that gut microbes play crucial roles in gut homeostasis, immune system education, and are associated with quality-of-life indicators. Beneficial health factors are associated with the digestion of dietary fibres in the colon and the subsequent production of short-chain fatty acids, including acetate, propionate, and butyrate. Coprococcus is a butyrate-producing genus in the phylum Firmicutes, and its abundance is inversely correlated with several neuropsychological and neurodegenerative disorders. Case-control studies provide strong evidence of decreased abundance of Coprococcus spp. in depressed individuals. The species Coprococcus eutactus has the unique capacity to use two separate pathways for butyrate synthesis and has been found to be depleted in children with delayed language development and adults with Parkinson's disease. The combined literature on Coprococcus and the gut microbiota-brain axis points towards enhanced butyrate production and reduced colonisation of pathogenic clades as factors explaining its association with health effects. The genus Coprococcus is a promising candidate for a mental health biomarker and an interesting lead for novel dietary-based preventive therapies for specific neurological disorders.
Collapse
Affiliation(s)
- Fleur Notting
- Amsterdam Institute for Life and Environment, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Walter Pirovano
- Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | | | - Remco Kort
- Amsterdam Institute for Life and Environment, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- ARTIS-Micropia, Amsterdam, The Netherlands
| |
Collapse
|
10
|
Morton JT, Jin DM, Mills RH, Shao Y, Rahman G, McDonald D, Zhu Q, Balaban M, Jiang Y, Cantrell K, Gonzalez A, Carmel J, Frankiensztajn LM, Martin-Brevet S, Berding K, Needham BD, Zurita MF, David M, Averina OV, Kovtun AS, Noto A, Mussap M, Wang M, Frank DN, Li E, Zhou W, Fanos V, Danilenko VN, Wall DP, Cárdenas P, Baldeón ME, Jacquemont S, Koren O, Elliott E, Xavier RJ, Mazmanian SK, Knight R, Gilbert JA, Donovan SM, Lawley TD, Carpenter B, Bonneau R, Taroncher-Oldenburg G. Multi-level analysis of the gut-brain axis shows autism spectrum disorder-associated molecular and microbial profiles. Nat Neurosci 2023; 26:1208-1217. [PMID: 37365313 PMCID: PMC10322709 DOI: 10.1038/s41593-023-01361-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 05/13/2023] [Indexed: 06/28/2023]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by heterogeneous cognitive, behavioral and communication impairments. Disruption of the gut-brain axis (GBA) has been implicated in ASD although with limited reproducibility across studies. In this study, we developed a Bayesian differential ranking algorithm to identify ASD-associated molecular and taxa profiles across 10 cross-sectional microbiome datasets and 15 other datasets, including dietary patterns, metabolomics, cytokine profiles and human brain gene expression profiles. We found a functional architecture along the GBA that correlates with heterogeneity of ASD phenotypes, and it is characterized by ASD-associated amino acid, carbohydrate and lipid profiles predominantly encoded by microbial species in the genera Prevotella, Bifidobacterium, Desulfovibrio and Bacteroides and correlates with brain gene expression changes, restrictive dietary patterns and pro-inflammatory cytokine profiles. The functional architecture revealed in age-matched and sex-matched cohorts is not present in sibling-matched cohorts. We also show a strong association between temporal changes in microbiome composition and ASD phenotypes. In summary, we propose a framework to leverage multi-omic datasets from well-defined cohorts and investigate how the GBA influences ASD.
Collapse
Affiliation(s)
- James T Morton
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, NY, USA
- Biostatistics & Bioinformatics Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Dong-Min Jin
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, NY, USA
| | | | - Yan Shao
- Host-Microbiota Interactions Laboratory, Wellcome Sanger Institute, Hinxton, UK
| | - Gibraan Rahman
- Bioinformatics and Systems Biology Program, University of California, San Diego, La Jolla, CA, USA
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Daniel McDonald
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Qiyun Zhu
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, AZ, USA
| | - Metin Balaban
- Bioinformatics and Systems Biology Program, University of California, San Diego, La Jolla, CA, USA
| | - Yueyu Jiang
- Department of Electrical and Computer Engineering, University of California, San Diego, La Jolla, CA, USA
| | - Kalen Cantrell
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA, USA
- Department of Computer Science and Engineering, Jacobs School of Engineering, University of California, San Diego, La Jolla, CA, USA
| | - Antonio Gonzalez
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Julie Carmel
- Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
| | | | - Sandra Martin-Brevet
- Laboratory for Research in Neuroimaging, Centre for Research in Neurosciences, Department of Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Kirsten Berding
- Division of Nutritional Sciences, University of Illinois, Urbana, IL, USA
| | - Brittany D Needham
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - María Fernanda Zurita
- Microbiology Institute and Health Science College, Universidad San Francisco de Quito, Quito, Ecuador
| | - Maude David
- Departments of Microbiology & Pharmaceutical Sciences, Oregon State University, Corvallis, OR, USA
| | - Olga V Averina
- Vavilov Institute of General Genetics Russian Academy of Sciences, Moscow, Russia
| | - Alexey S Kovtun
- Vavilov Institute of General Genetics Russian Academy of Sciences, Moscow, Russia
- Skolkovo Institute of Science and Technology, Skolkovo, Russia
| | - Antonio Noto
- Department of Biomedical Sciences, School of Medicine, University of Cagliari, Cagliari, Italy
| | - Michele Mussap
- Laboratory Medicine, Department of Surgical Sciences, School of Medicine, University of Cagliari, Cagliari, Italy
| | - Mingbang Wang
- Shanghai Key Laboratory of Birth Defects, Division of Neonatology, Children's Hospital of Fudan University, National Center for Children's Health, Shanghai, China
- Microbiome Therapy Center, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, China
| | - Daniel N Frank
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Ellen Li
- Department of Medicine, Division of Gastroenterology and Hepatology, Stony Brook University, Stony Brook, NY, USA
| | - Wenhao Zhou
- Shanghai Key Laboratory of Birth Defects, Division of Neonatology, Children's Hospital of Fudan University, National Center for Children's Health, Shanghai, China
| | - Vassilios Fanos
- Neonatal Intensive Care Unit and Neonatal Pathology, Department of Surgical Sciences, School of Medicine, University of Cagliari, Cagliari, Italy
| | - Valery N Danilenko
- Vavilov Institute of General Genetics Russian Academy of Sciences, Moscow, Russia
| | - Dennis P Wall
- Pediatrics (Systems Medicine), Biomedical Data Science, and Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Paúl Cárdenas
- Institute of Microbiology, COCIBA, Universidad San Francisco de Quito, Quito, Ecuador
| | - Manuel E Baldeón
- Facultad de Ciencias Médicas, de la Salud y la Vida, Universidad Internacional del Ecuador, Quito, Ecuador
| | - Sébastien Jacquemont
- Sainte Justine Hospital Research Center, Montréal, QC, Canada
- Department of Pediatrics, Université de Montréal, Montréal, QC, Canada
| | - Omry Koren
- Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
| | - Evan Elliott
- Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar Ilan University, Ramat Gan, Israel
| | - Ramnik J Xavier
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston, MA, USA
| | - Sarkis K Mazmanian
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Rob Knight
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA, USA
- Department of Computer Science and Engineering, Jacobs School of Engineering, University of California, San Diego, La Jolla, CA, USA
- Department of Bioengineering, University of California, San Diego, La Jolla, California, USA
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, California, USA
| | - Jack A Gilbert
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA, USA
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, California, USA
- Scripps Institution of Oceanography, University of California, San Diego, La Jolla, CA, USA
| | - Sharon M Donovan
- Division of Nutritional Sciences, University of Illinois, Urbana, IL, USA
| | - Trevor D Lawley
- Host-Microbiota Interactions Laboratory, Wellcome Sanger Institute, Hinxton, UK
| | - Bob Carpenter
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, NY, USA
| | - Richard Bonneau
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, NY, USA
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, NY, USA
- Prescient Design, a Genentech Accelerator, New York, NY, USA
| | - Gaspar Taroncher-Oldenburg
- Gaspar Taroncher Consulting, Philadelphia, PA, USA.
- Simons Foundation Autism Research Initiative, Simons Foundation, New York, NY, USA.
| |
Collapse
|
11
|
Zhang F, Wang D. Potential of Akkermansia muciniphila and its outer membrane proteins as therapeutic targets for neuropsychological diseases. Front Microbiol 2023; 14:1191445. [PMID: 37440890 PMCID: PMC10333588 DOI: 10.3389/fmicb.2023.1191445] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 05/25/2023] [Indexed: 07/15/2023] Open
Abstract
The gut microbiota varies dramatically among individuals, and changes over time within the same individual, due to diversities in genetic backgrounds, diet, nutrient supplementations and use of antibiotics. Up until now, studies on dysbiosis of microbiota have expanded to a wider range of diseases, with Akkermansia muciniphila at the cross spot of many of these diseases. A. muciniphila is a Gram-negative bacterium that produces short-chain fatty acids (SCFAs), and Amuc_1100 is one of its most highly expressed outer membrane proteins. This review aims to summarize current knowledge on correlations between A. muciniphila and involved neuropsychological diseases published in the last decade, with a focus on the potential of this bacterium and its outer membrane proteins as therapeutic targets for these diseases, on the basis of evidence accumulated from animal and clinical studies, as well as mechanisms of action from peripheral to central nervous system (CNS).
Collapse
Affiliation(s)
- Fenghua Zhang
- Department of Laboratory Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Dali Wang
- Center for Clinical and Translational Medicine, Shanghai University of Medicine and Health Sciences, Shanghai, China
| |
Collapse
|
12
|
Levkova M, Chervenkov T, Pancheva R. Genus-Level Analysis of Gut Microbiota in Children with Autism Spectrum Disorder: A Mini Review. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1103. [PMID: 37508600 PMCID: PMC10377934 DOI: 10.3390/children10071103] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/20/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023]
Abstract
Autism is a global health problem, probably due to a combination of genetic and environmental factors. There is emerging data that the gut microbiome of autistic children differs from the one of typically developing children and it is important to know which bacterial genera may be related to autism. We searched different databases using specific keywords and inclusion criteria and identified the top ten bacterial genera from the selected articles that were significantly different between the studied patients and control subjects studied. A total of 34 studies that met the inclusion criteria were identified. The genera Bacteroides, Bifidobacterium, Clostridium, Coprococcus, Faecalibacterium, Lachnospira, Prevotella, Ruminococcus, Streptococcus, and Blautia exhibited the most substantial data indicating that their fluctuations in the gastrointestinal tract could be linked to the etiology of autism. It is probable that autism symptoms are influenced by both increased levels of harmful bacteria and decreased levels of beneficial bacteria. Interestingly, these genera demonstrated varying patterns of increased or decreased levels across different articles. To validate and eliminate the sources of this fluctuation, further research is needed. Consequently, future investigations on the causes of autism should prioritize the examination of the bacterial genera discussed in this publication.
Collapse
Affiliation(s)
- Mariya Levkova
- Department of Medical Genetics, Medical University Varna, Marin Drinov Str 55, 9000 Varna, Bulgaria
- Laboratory of Medical Genetics, St. Marina Hospital, Hristo Smirnenski Blv 1, 9000 Varna, Bulgaria
| | - Trifon Chervenkov
- Laboratory of Medical Genetics, St. Marina Hospital, Hristo Smirnenski Blv 1, 9000 Varna, Bulgaria
- Laboratory of Clinical Immunology, St. Marina Hospital, Hristo Smirnenski Blv 1, 9000 Varna, Bulgaria
| | - Rouzha Pancheva
- Department of Hygiene and Epidemiology, Medical University Varna, Marin Drinov Str 55, 9000 Varna, Bulgaria
| |
Collapse
|
13
|
Kamalmaz N, Ben Bacha A, Alonazi M, Albasher G, Khayyat AIA, El-Ansary A. Unveiling sex-based differences in developing propionic acid-induced features in mice as a rodent model of ASD. PeerJ 2023; 11:e15488. [PMID: 37334116 PMCID: PMC10274690 DOI: 10.7717/peerj.15488] [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: 01/25/2023] [Accepted: 05/10/2023] [Indexed: 06/20/2023] Open
Abstract
Background Males are more likely to develop autism as a neurodevelopmental disorder than females are, although the mechanisms underlying male vulnerability are not fully understood. Therefore, studying the role of autism etiologies considering sex differences in the propionic acid (PPA) rodent model of autism would build greater understanding of how females are protected from autism spectrum disorder, which may be used as a treatment strategy for males with autism. Objectives This study aimed to investigate the sex differences in oxidative stress, glutamate excitotoxicity, neuroinflammation, and gut microbiota impairment as etiological mechanisms for many neurological diseases, with specific reference to autism. Method Forty albino mice were divided into four groups of 10 animals each with two control and two treated groups of both sexes received only phosphate-buffered saline or a neurotoxic dose of PPA (250 mg/kg body weight) for 3 days, respectively. Biochemical markers of energy metabolism, oxidative stress, neuroinflammation, and excitotoxicity were measured in mouse brain homogenates, whereas the presence of pathogenic bacteria was assessed in mouse stool samples. Furthermore, the repetitive behavior, cognitive ability, and physical-neural coordination of the animals were examined. Results Collectively, selected variables related to oxidative stress, glutamate excitotoxicity, neuroinflammation, and gut bacteria were impaired concomitantly with altered behavior in PPA-induced rodent model, with males being more susceptible than females. Conclusion This study explains the role of sex in the higher vulnerability of males to develop autistic biochemical and behavioral features compared with females. Female sex hormones and the higher detoxification capacity and higher glycolytic flux in females serve as neuroprotective contributors in a rodent model of autism.
Collapse
Affiliation(s)
- Nasreen Kamalmaz
- Biochemistry Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Abir Ben Bacha
- Biochemistry Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Mona Alonazi
- Biochemistry Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Gadah Albasher
- Zoology Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Arwa Ishaq A. Khayyat
- Biochemistry Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Afaf El-Ansary
- Central Research Laboratory, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
14
|
Zhu X, Shen J, Feng S, Huang C, Wang H, Huo F, Liu H. Akkermansia muciniphila, which is enriched in the gut microbiota by metformin, improves cognitive function in aged mice by reducing the proinflammatory cytokine interleukin-6. MICROBIOME 2023; 11:120. [PMID: 37254162 DOI: 10.1186/s40168-023-01567-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 05/05/2023] [Indexed: 06/01/2023]
Abstract
BACKGROUND Metformin, a type 2 diabetes treatment, improves the cognitive function of aged mice; however, whether the protective effects of metformin on cognitive function in aged mice are associated with the gut microbiome is poorly understood. Although some studies suggest that the gut microbe composition influences cognitive function and that manipulating the gut microbiota might protect against age-related cognitive dysfunction, there is no direct evidence to validate that the gut microbiota mediates the effect of metformin on cognitive improvement. RESULTS In this study, we show that the gut microbiota is altered by metformin, which is necessary for protection against ageing-associated cognitive function declines in aged mice. Mice treated with antibiotics did not exhibit metformin-mediated cognitive function protection. Moreover, treatment with Akkermansia muciniphila, which is enriched by metformin, improved cognitive function in aged mice. Mechanistically, A. muciniphila decreased pro-inflammatory-associated pathways, particularly that of the pro-inflammatory cytokine interleukin (IL)-6, in both the peripheral blood and hippocampal profiles, which was correlated with cognitive function improvement. An IL-6 antibody protected cognitive function, and an IL-6 recombinant protein abolished the protective effect of A. muciniphila on cognitive function in aged mice. CONCLUSION This study reveals that A. muciniphila, which is mediated in the gut microbiota by metformin, modulates inflammation-related pathways in the host and improves cognitive function in aged mice by reducing the pro-inflammatory cytokine IL-6. Video Abstract.
Collapse
Affiliation(s)
- Xiaoqi Zhu
- Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200123, China
| | - Junyan Shen
- Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200123, China
| | - Shengyu Feng
- Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200123, China
| | - Ce Huang
- Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200123, China
| | - Hao Wang
- Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200123, China
| | - Fengjiao Huo
- Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200123, China
| | - Hailiang Liu
- Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200123, China.
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization of Ministry of Education, College of Life Sciences, Shihezi University, Shihezi, 832003, China.
- Institute of Advanced Biotechnology, Southern University of Science and Technology, Shenzhen, 518055, China.
| |
Collapse
|
15
|
Rodrigues EL, Figueiredo PS, Marcelino G, de Cássia Avellaneda Guimarães R, Pott A, Santana LF, Hiane PA, do Nascimento VA, Bogo D, de Cássia Freitas K. Maternal Intake of Polyunsaturated Fatty Acids in Autism Spectrum Etiology and Its Relation to the Gut Microbiota: What Do We Know? Nutrients 2023; 15:nu15071551. [PMID: 37049390 PMCID: PMC10097097 DOI: 10.3390/nu15071551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/17/2023] [Accepted: 02/20/2023] [Indexed: 04/14/2023] Open
Abstract
Maternal food habits and gut microbiota composition have potential effects on fetal neurodevelopment, impacting Autism Spectrum Disorder (ASD). Our research aims to outline the relationship that ingestion of polyunsaturated fatty acids (PUFAs) and the composition of maternal gut microbiota have with the possible development of ASD in offspring. We suggest that genetic factors could be related to the different conversions between unsaturated fatty acids according to sex and, mainly, the impact of the pregnancy diet on the higher or lower risk of neurological impairments. The proportion of the phyla Firmicutes/Bacteroidetes is high with an increased consumption of linoleic acid (LA, n-6 PUFA), which is associated with maternal intestinal dysbiosis and consequently starts the inflammatory process, harming myelinization. In contrast, the consumption of α-linolenic acid (ALA, n-3 PUFA) tends to re-establish the balance of the maternal microbiota with anti-inflammatory action. Moreover, human observational studies showed a strong correlation between the consumption of n-3 PUFA, mainly above 340 g of fish per week, with beneficial effects on infant neurodevelopment. Therefore, we suggest that the proper intake of foods rich in n-3 PUFAs and their supplementation during pregnancy until lactation has an impact on reducing the development of ASD. Controlled studies with n-3 PUFA supplementation are still necessary to verify the ideal dose and the best form of administration.
Collapse
Affiliation(s)
- Elisana Lima Rodrigues
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul-UFMS, Campo Grande 79079-900, MS, Brazil
| | - Priscila Silva Figueiredo
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul-UFMS, Campo Grande 79079-900, MS, Brazil
| | - Gabriela Marcelino
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul-UFMS, Campo Grande 79079-900, MS, Brazil
| | - Rita de Cássia Avellaneda Guimarães
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul-UFMS, Campo Grande 79079-900, MS, Brazil
| | - Arnildo Pott
- Institute of Biosciences, Federal University of Mato Grosso do Sul-UFMS, Campo Grande 79079-900, MS, Brazil
| | - Lidiani Figueiredo Santana
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul-UFMS, Campo Grande 79079-900, MS, Brazil
| | - Priscila Aiko Hiane
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul-UFMS, Campo Grande 79079-900, MS, Brazil
| | - Valter Aragão do Nascimento
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul-UFMS, Campo Grande 79079-900, MS, Brazil
| | - Danielle Bogo
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul-UFMS, Campo Grande 79079-900, MS, Brazil
| | - Karine de Cássia Freitas
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul-UFMS, Campo Grande 79079-900, MS, Brazil
| |
Collapse
|
16
|
Dhanasekara CS, Ancona D, Cortes L, Hu A, Rimu AH, Robohm-Leavitt C, Payne D, Wakefield SM, Mastergeorge AM, Kahathuduwa CN. Association Between Autism Spectrum Disorders and Cardiometabolic Diseases: A Systematic Review and Meta-analysis. JAMA Pediatr 2023; 177:248-257. [PMID: 36716018 PMCID: PMC9887535 DOI: 10.1001/jamapediatrics.2022.5629] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 11/11/2022] [Indexed: 01/31/2023]
Abstract
Importance Although the increased risk of obesity among individuals with autism has been well established, evidence on the association between autism, cardiometabolic disorders, and obesity remains inconclusive. Objective To examine the association between autism spectrum disorders and cardiometabolic diseases in a systematic review and meta-analysis. Data Sources PubMed, Scopus, Web of Science, ProQuest, Embase, and Ovid databases were searched from inception through July 31, 2022, without restrictions on date of publication or language. Study Selection Observational or baseline data of interventional studies reporting the prevalence of cardiometabolic risk factors (ie, diabetes, hypertension, dyslipidemia, atherosclerotic macrovascular disease) among children and/or adults with autism and matched with participants without autism were included. Data Extraction and Synthesis Screening, data extraction, and quality assessment were performed independently by at least 2 researchers. DerSimonian-Laird random-effects meta-analyses were performed using the meta package in R. Main Outcomes and Measures Relative risks (RRs) of diabetes, hypertension, dyslipidemia, and atherosclerotic macrovascular disease among individuals with autism were the primary outcomes. Secondary outcomes included the RR of type 1 and type 2 diabetes, heart disease, stroke, and peripheral vascular disease. Results A total of 34 studies were evaluated and included 276 173 participants with autism and 7 733 306 participants without autism (mean [range] age, 31.2 [3.8-72.8] years; pooled proportion [range] of female individuals, 47% [0-66%]). Autism was associated with greater risks of developing diabetes overall (RR, 1.57; 95% CI, 1.23-2.01; 20 studies), type 1 diabetes (RR, 1.64; 95% CI, 1.06-2.54; 6 studies), and type 2 diabetes (RR, 2.47; 95% CI, 1.30-4.70; 3 studies). Autism was also associated with increased risks of dyslipidemia (RR, 1.69; 95% CI, 1.20-2.40; 7 studies) and heart disease (RR, 1.46; 95% CI, 1.42-1.50; 3 studies). Yet, there was no significantly associated increased risk of hypertension and stroke with autism (RR, 1.22; 95% CI, 0.98-1.52; 12 studies; and RR, 1.19; 95% CI, 0.63-2.24; 4 studies, respectively). Meta-regression analyses revealed that children with autism were at a greater associated risk of developing diabetes and hypertension compared with adults. High between-study heterogeneity was a concern for several meta-analyses. Conclusions and Relevance Results suggest that the associated increased risk of cardiometabolic diseases should prompt clinicians to vigilantly monitor individuals with autism for potential contributors, signs of cardiometabolic disease, and their complications.
Collapse
Affiliation(s)
- Chathurika S. Dhanasekara
- Department of Laboratory Science and Primary Care, School of Health Professions, Texas Tech University Health Sciences Center, Lubbock
- Department of Surgery, School of Medicine, Texas Tech University Health Sciences Center, Lubbock
| | - Dominic Ancona
- Department of Laboratory Science and Primary Care, School of Health Professions, Texas Tech University Health Sciences Center, Lubbock
| | - Leticia Cortes
- Department of Laboratory Science and Primary Care, School of Health Professions, Texas Tech University Health Sciences Center, Lubbock
| | - Amy Hu
- Department of Laboratory Science and Primary Care, School of Health Professions, Texas Tech University Health Sciences Center, Lubbock
| | - Afrina H. Rimu
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock
| | - Christina Robohm-Leavitt
- Department of Laboratory Science and Primary Care, School of Health Professions, Texas Tech University Health Sciences Center, Lubbock
| | - Drew Payne
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock
| | - Sarah M. Wakefield
- Department of Psychiatry, School of Medicine, Texas Tech University Health Sciences Center, Lubbock
| | - Ann M. Mastergeorge
- Department of Human Development and Family Sciences, College of Human Sciences, Texas Tech University, Lubbock
| | - Chanaka N. Kahathuduwa
- Department of Laboratory Science and Primary Care, School of Health Professions, Texas Tech University Health Sciences Center, Lubbock
- Department of Psychiatry, School of Medicine, Texas Tech University Health Sciences Center, Lubbock
- Department of Neurology, School of Medicine, Texas Tech University Health Sciences Center, Lubbock
| |
Collapse
|
17
|
Patel M, Atluri LM, Gonzalez NA, Sakhamuri N, Athiyaman S, Randhi B, Gutlapalli SD, Pu J, Zaidi MF, Khan S. A Systematic Review of Mixed Studies Exploring the Effects of Probiotics on Gut-Microbiome to Modulate Therapy in Children With Autism Spectrum Disorder. Cureus 2022; 14:e32313. [PMID: 36632246 PMCID: PMC9828007 DOI: 10.7759/cureus.32313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 12/08/2022] [Indexed: 12/13/2022] Open
Abstract
Autism spectrum disorder(ASD) is a complex neurodevelopmental disorder characterized by social deficits, repetitive typical behaviors, insistence on the same routines, and communication impairments. The prevalence of ASD has increased in the past decade. While we are aware that there is no cure for ASD, attempts are being made to reduce its symptoms and improve the learning, overall growth, and well-being of ASD patients. Gastrointestinal (GI) symptoms are frequent occurrences in patients with ASD, but the underlying mechanisms are unknown. Recent studies show that the microbiota-gut-brain axis is the key modulator of neuropsychiatric health. Although fecal transplants have shown positive outcomes in treating dysbiosis and symptoms of autism, lifestyle modifications such as dietary intervention will prevent and treat this disorder without causing major adverse effects. Probiotics enhance the microbiome to provide necessary metabolites, which help in gut permeability, cognitive function, and immunity. In some studies, children with increased GI symptoms have also shown increased behavioral disturbances. In this study, a systematic review of mixed studies is conducted to obtain more robust and conclusive results. We included randomized controlled studies with larger sample sizes and specifications on probiotics.
Collapse
Affiliation(s)
- Maithily Patel
- Family Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Lakshmi M Atluri
- General Surgery, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Natalie A Gonzalez
- Pediatrics, Medical University of Graz, Graz, AUT
- Pediatrics, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Navya Sakhamuri
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Sreekartthik Athiyaman
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, CA, USA
| | - Bhawna Randhi
- Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Sai Dheeraj Gutlapalli
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Jingxiong Pu
- Psychiatry and Behavioral Sciences, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Maheen F Zaidi
- Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Safeera Khan
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
18
|
Plaza-Diaz J, Radar AM, Baig AT, Leyba MF, Costabel MM, Zavala-Crichton JP, Sanchez-Martinez J, MacKenzie AE, Solis-Urra P. Physical Activity, Gut Microbiota, and Genetic Background for Children and Adolescents with Autism Spectrum Disorder. CHILDREN (BASEL, SWITZERLAND) 2022; 9:1834. [PMID: 36553278 PMCID: PMC9777368 DOI: 10.3390/children9121834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/19/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022]
Abstract
It is estimated that one in 100 children worldwide has been diagnosed with autism spectrum disorder (ASD). Children with ASD frequently suffer from gut dysbiosis and gastrointestinal issues, findings which possibly play a role in the pathogenesis and/or severity of their condition. Physical activity may have a positive effect on the composition of the intestinal microbiota of healthy adults. However, the effect of exercise both on the gastrointestinal problems and intestinal microbiota (and thus possibly on ASD) itself in affected children is unknown. In terms of understanding the physiopathology and manifestations of ASD, analysis of the gut-brain axis holds some promise. Here, we discuss the physiopathology of ASD in terms of genetics and microbiota composition, and how physical activity may be a promising non-pharmaceutical approach to improve ASD-related symptoms.
Collapse
Affiliation(s)
- Julio Plaza-Diaz
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain
| | - Ana Mei Radar
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Aiman Tariq Baig
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Marcos Federico Leyba
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Maria Macarena Costabel
- Children’s Hospital of Eastern Ontario, Division of Urology, Department of Surgery, University of Ottawa, Ottawa, ON K1H 8L6, Canada
| | | | - Javier Sanchez-Martinez
- Escuela de Kinesiología, Facultad de Salud, Universidad Santo Tomás, Viña del Mar 2520298, Chile
| | - Alex E. MacKenzie
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Patricio Solis-Urra
- Faculty of Education and Social Sciences, Universidad Andres Bello, Viña del Mar 2531015, Chile
- PROFITH “PROmoting FITness and Health through Physical Activity” Research Group, Sport and Health University Research Institute (iMUDS), Department of Physical Education and Sports, Faculty of Sport Sciences, University of Granada, 18071 Granada, Spain
- Servicio de Medicina Nuclear, Hospital Universitario Virgen de las Nieves, 18014 Granada, Spain
| |
Collapse
|
19
|
Soltysova M, Tomova A, Ostatnikova D. Gut Microbiota Profiles in Children and Adolescents with Psychiatric Disorders. Microorganisms 2022; 10:2009. [PMID: 36296284 PMCID: PMC9608804 DOI: 10.3390/microorganisms10102009] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/28/2022] [Accepted: 09/30/2022] [Indexed: 11/16/2022] Open
Abstract
The aim of our work is to summarize the current state of knowledge on gut microbiota differences in children and adolescents with psychiatric disorders. To find the relevant articles, the PubMed, Web of Science, and Google Scholar databases were searched. Articles in English presenting original data and comparing the composition of gut microbiota in child psychiatric patients with gut microbiota in healthy children and adolescents were selected. Finally, we identified 55 articles eligible for our purpose. The majority of patients with autism spectrum disorders (ASD) were investigated. A smaller number of studies evaluating the gut microbiota in children and adolescents with attention-deficit/hyperactivity disorder (ADHD), Rett syndrome, anorexia nervosa, depressive disorder (DD), and tic disorders were found. The main findings of this research are discussed in our review, focusing on the age-related gut microbiota specificity for psychiatric disorders and the differences between individual diagnosis. To conclude, the gut microbiota in children and adolescents with psychiatric disorders is evidently different from that in controls. The most pronounced differences are seen in children with ASD, less in ADHD. Moreover, the changes are not identical to those in adult psychiatric patients, as Ruminococcus, Turicibacter, and Bilophila were increased in adults, and decreased in children with ASD, and Parabacteroides and Alistipes were more frequently represented in adults, but less frequently represented in children with depression. The available data suggest some genera have a different abundance in individual psychiatric disorders (e.g., Bilophila, Bifidobacterium, Clostridium, Coprococcus, Faecalibacterium, and Ruminococcus), suggesting their importance for the gut-brain axis. Other bacterial genera might be more important for the pathophysiology of specific disorder in children and adolescents, as Akkermansia and Desulfovibrio for ASD, or Romboutsia for DD. Based on the research findings, we assume that gut microbiota corrections have the potential to improve clinical symptoms in psychiatric patients.
Collapse
Affiliation(s)
- Marcela Soltysova
- Academic Research Center for Autism, Institute of Physiology, Faculty of Medicine in Bratislava, Comenius University, 813 72 Bratislava, Slovakia
- Child Psychiatry Outpatient Care Unit, Zvolen Hospital, 960 01 Zvolen, Slovakia
| | - Aleksandra Tomova
- Child Psychiatry Outpatient Care Unit, Zvolen Hospital, 960 01 Zvolen, Slovakia
| | - Daniela Ostatnikova
- Child Psychiatry Outpatient Care Unit, Zvolen Hospital, 960 01 Zvolen, Slovakia
| |
Collapse
|
20
|
Pietrucci D, Teofani A, Milanesi M, Fosso B, Putignani L, Messina F, Pesole G, Desideri A, Chillemi G. Machine Learning Data Analysis Highlights the Role of Parasutterella and Alloprevotella in Autism Spectrum Disorders. Biomedicines 2022; 10:biomedicines10082028. [PMID: 36009575 PMCID: PMC9405825 DOI: 10.3390/biomedicines10082028] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/10/2022] [Accepted: 08/15/2022] [Indexed: 11/25/2022] Open
Abstract
In recent years, the involvement of the gut microbiota in disease and health has been investigated by sequencing the 16S gene from fecal samples. Dysbiotic gut microbiota was also observed in Autism Spectrum Disorder (ASD), a neurodevelopmental disorder characterized by gastrointestinal symptoms. However, despite the relevant number of studies, it is still difficult to identify a typical dysbiotic profile in ASD patients. The discrepancies among these studies are due to technical factors (i.e., experimental procedures) and external parameters (i.e., dietary habits). In this paper, we collected 959 samples from eight available projects (540 ASD and 419 Healthy Controls, HC) and reduced the observed bias among studies. Then, we applied a Machine Learning (ML) approach to create a predictor able to discriminate between ASD and HC. We tested and optimized three algorithms: Random Forest, Support Vector Machine and Gradient Boosting Machine. All three algorithms confirmed the importance of five different genera, including Parasutterella and Alloprevotella. Furthermore, our results show that ML algorithms could identify common taxonomic features by comparing datasets obtained from countries characterized by latent confounding variables.
Collapse
Affiliation(s)
- Daniele Pietrucci
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), University of Tuscia, 01100 Viterbo, Italy
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, IBIOM, CNR, 70126 Bari, Italy
| | - Adelaide Teofani
- Department of Biology, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Marco Milanesi
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), University of Tuscia, 01100 Viterbo, Italy
| | - Bruno Fosso
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari “A. Moro”, Piazza Umberto I, 1, 70121 Bari, Italy
| | - Lorenza Putignani
- Unit of Microbiology and Diagnostic Immunology, Units of Microbiomics, Department of Diagnostic and Laboratory Medicine, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy
| | - Francesco Messina
- Laboratory of Microbiology and Biological Bank National Institute for Infectious Diseases “Lazzaro Spallanzani” Istituto di Ricovero e Cura a Carattere Scientifico, 00149 Rome, Italy
| | - Graziano Pesole
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, IBIOM, CNR, 70126 Bari, Italy
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari “A. Moro”, Piazza Umberto I, 1, 70121 Bari, Italy
| | - Alessandro Desideri
- Department of Biology, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Giovanni Chillemi
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), University of Tuscia, 01100 Viterbo, Italy
- Correspondence: ; Tel.: +39-0761-357-429
| |
Collapse
|
21
|
Alamoudi MU, Hosie S, Shindler AE, Wood JL, Franks AE, Hill-Yardin EL. Comparing the Gut Microbiome in Autism and Preclinical Models: A Systematic Review. Front Cell Infect Microbiol 2022; 12:905841. [PMID: 35846755 PMCID: PMC9286068 DOI: 10.3389/fcimb.2022.905841] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 06/02/2022] [Indexed: 12/21/2022] Open
Abstract
Many individuals diagnosed with autism spectrum disorder (ASD) experience gastrointestinal (GI) dysfunction and show microbial dysbiosis. Variation in gut microbial populations is associated with increased risk for GI symptoms such as chronic constipation and diarrhoea, which decrease quality of life. Several preclinical models of autism also demonstrate microbial dysbiosis. Given that much pre-clinical research is conducted in mouse models, it is important to understand the similarities and differences between the gut microbiome in humans and these models in the context of autism. We conducted a systematic review of the literature using PubMed, ProQuest and Scopus databases to compare microbiome profiles of patients with autism and transgenic (NL3R451C, Shank3 KO, 15q dup), phenotype-first (BTBR) and environmental (Poly I:C, Maternal Inflammation Activation (MIA), valproate) mouse models of autism. Overall, we report changes in fecal microbial communities relevant to ASD based on both clinical and preclinical studies. Here, we identify an overlapping cluster of genera that are modified in both fecal samples from individuals with ASD and mouse models of autism. Specifically, we describe an increased abundance of Bilophila, Clostridium, Dorea and Lactobacillus and a decrease in Blautia genera in both humans and rodents relevant to this disorder. Studies in both humans and mice highlighted multidirectional changes in abundance (i.e. in some cases increased abundance whereas other reports showed decreases) for several genera including Akkermansia, Bacteroides, Bifidobacterium, Parabacteroides and Prevotella, suggesting that these genera may be susceptible to modification in autism. Identification of these microbial profiles may assist in characterising underlying biological mechanisms involving host-microbe interactions and provide future therapeutic targets for improving gut health in autism.
Collapse
Affiliation(s)
- Mohammed U. Alamoudi
- School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology (RMIT), Bundoora, VIC, Australia
- Medical Laboratory Technology Department, Faculty of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Suzanne Hosie
- School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology (RMIT), Bundoora, VIC, Australia
| | - Anya E. Shindler
- Department of Microbiology, Anatomy, Physiology and Pharmacology, School of Life Sciences, La Trobe University, Bundoora, VIC, Australia
| | - Jennifer L. Wood
- Department of Microbiology, Anatomy, Physiology and Pharmacology, School of Life Sciences, La Trobe University, Bundoora, VIC, Australia
| | - Ashley E. Franks
- Department of Microbiology, Anatomy, Physiology and Pharmacology, School of Life Sciences, La Trobe University, Bundoora, VIC, Australia
| | - Elisa L. Hill-Yardin
- School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology (RMIT), Bundoora, VIC, Australia
- *Correspondence: Elisa L. Hill-Yardin,
| |
Collapse
|
22
|
Leveraging Existing 16SrRNA Microbial Data to Define a Composite Biomarker for Autism Spectrum Disorder. Microbiol Spectr 2022; 10:e0033122. [PMID: 35762814 PMCID: PMC9431227 DOI: 10.1128/spectrum.00331-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Cumulative studies have utilized high-throughput sequencing of the 16SrRNA gene to characterize the composition and structure of the microbiota in autism spectrum disorder (ASD). However, they do not always obtain consistent results; thus, conducting cross-study comparisons is necessary. This study sought to analyze the alteration of fecal microbiota and the diagnostic capabilities of gut microbiota biomarkers in individuals with ASD using the existing 16SrRNA microbial data and explore heterogeneity among studies. The raw sequence and metadata from 10 studies, including 1,019 samples, were reanalyzed. Results showed no significant difference in alpha diversity of fecal microbiota between ASD and the control group. However, a significant difference in the composition structure of fecal microbiota was observed. Given the large differences in sample selection and technical differences, the separation of fecal microbiota between ASD and controls was not observed. Subgroup analysis was performed on the basis of different country of origin, hypervariable regions, and sequencing platforms, and the dominant genera in ASD and healthy control groups were determined by linear discriminant analysis (LDA) of the effect size (LEfSe) algorithm and Wilcoxon rank-sum test. Machine learning analyses were carried out to determine the diagnostic capabilities of potential microbial biomarkers. A total of 12 genera were identified to distinguish ASD from control, and the AUC of the training set and verification set was 0.757 and 0.761, respectively. Despite cohort heterogeneity, gut microbial dysbiosis of ASD has been proven to be a widespread phenomenon. Therefore, fecal microbial markers are of great significance in diagnosing ASD diseases and possible candidates for further mechanistic study of the role of intestinal microbiota in ASD. IMPORTANCE This study provides an updated analysis to characterize the gut microbiota in ASD using 16SrRNA gene high-throughput sequencing data from 10 publicly available studies. Our analysis suggests an association between the fecal microbiota and ASD. Sample selection and technical differences between studies may interfere with the species composition analysis of the ASD group and control group. By summarizing the results of 16SrRNA gene sequencing from multiple fecal samples, we can provide evidence to support the use of microbial biomarkers to diagnose the occurrence of ASD. Our study provides a new perspective for further revealing the correlation between gut microbiota and ASD from the perspective of 16SrRNA sequencing in larger samples.
Collapse
|
23
|
Liu Z, Wang J, Xu Q, Wu Z, You L, Hong Q, Zhu J, Chi X. Vitamin A supplementation ameliorates prenatal valproic acid-induced autism-like behaviors in rats. Neurotoxicology 2022; 91:155-165. [PMID: 35594946 DOI: 10.1016/j.neuro.2022.05.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 03/28/2022] [Accepted: 05/09/2022] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by social deficits and repetitive stereotyped behaviors. Prenatal exposure to the anticonvulsant drug valproic acid (VPA) is reported to induce ASD in human and ASD-like phenotypes in rodents. Unfortunately, the etiology and pathogenesis of ASD remains unclear. METHODS Pregnant rats received an intraperitoneal injection of 600 mg/kg VPA on E12.5 to construct the ASD rat model in offspring. The different expression of long non-coding RNA (lncRNA) and mRNA profiles in the hippocampus were determined by RNA sequencing to investigate potential mechanisms of VPA-induced ASD. Gene Ontology (GO) and pathway enrichment analysis were performed to predict the function of dysregulated lncRNAs. Co-expression network and real-time polymerase chain reaction (RT-PCR) analysis were conducted to validate the potential regulatory lncRNA-mRNA network. RESULTS VPA increased the total distance, time spent in the central zone and self-grooming (open field test) in rats. Meanwhile, VPA induced social impairment (three-chamber sociability test) and repetitive behaviors (marble burying test). A total of 238 lncRNAs and 354 mRNAs were differentially expressed in the VPA group. In addition, the dysregulated lncRNAs were involved in neural function and developmental processes of ASD. 5 lncRNAs and 7 mRNAs were differently expressed and included in the lncRNA-mRNA co-expression network. RT-PCR confirmed the upregulation of 4 lncRNAs and 6 mRNAs, and identified a potential regulatory network of NONRATT021475.2 (lncRNA) and Desert hedgehog (Dhh). Moreover, VPA decreased the serum vitamin A (VA) levels in offspring rats on postnatal day (PND) 21 and 49. Importantly, VA supplementation significantly restored VPA-induced autism-related behaviors and upregulation of NONRATT021475.2 and Dhh in the hippocampus of ASD rats. CONCLUSION This study not only contributed to understand the importance of lncRNAs and mRNAs in the progression of ASD, but also identified VA as a potential therapy for the condition. DATA AVAILABILITY The data that support the findings of this study are available from the corresponding author with reasonable request.
Collapse
Affiliation(s)
- Zhonghui Liu
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, 123 Tianfei Alley, Mochou Road, Nanjing 210004, PR China; Institute of Pediatrics, Nanjing Medical University, 72 Guangzhou Road, Nanjing 210008, PR China
| | - Jingyu Wang
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, 123 Tianfei Alley, Mochou Road, Nanjing 210004, PR China; Institute of Pediatrics, Nanjing Medical University, 72 Guangzhou Road, Nanjing 210008, PR China
| | - Qu Xu
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, 123 Tianfei Alley, Mochou Road, Nanjing 210004, PR China
| | - Zhenggang Wu
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, 123 Tianfei Alley, Mochou Road, Nanjing 210004, PR China
| | - Lianghui You
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, 123 Tianfei Alley, Mochou Road, Nanjing 210004, PR China
| | - Qin Hong
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, 123 Tianfei Alley, Mochou Road, Nanjing 210004, PR China
| | - Jiansheng Zhu
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, 123 Tianfei Alley, Mochou Road, Nanjing 210004, PR China.
| | - Xia Chi
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, 123 Tianfei Alley, Mochou Road, Nanjing 210004, PR China; Institute of Pediatrics, Nanjing Medical University, 72 Guangzhou Road, Nanjing 210008, PR China.
| |
Collapse
|
24
|
Zhang L, Xu Y, Li H, Li B, Duan G, Zhu C. The role of probiotics in children with autism spectrum disorders: A study protocol for a randomised controlled trial. PLoS One 2022; 17:e0263109. [PMID: 35202432 PMCID: PMC8870536 DOI: 10.1371/journal.pone.0263109] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 01/11/2022] [Indexed: 12/20/2022] Open
Abstract
Background Autism spectrum disorder (ASD) is a neurological and developmental condition that begins in infancy or earlier and lasts through the individual’s lifetime. The aetiology and mechanisms of ASD are not yet fully understood, and current treatment comprises mainly education and rehabilitation, without significant improvement in the core symptoms. Recent studies suggest that microbiota change in children with ASD after the ingestion of probiotics may improve the balance of microbiota and thus ASD symptoms. Objective The objectives of this study are to evaluate the efficacy of probiotics on the symptoms of children with ASD and the possible mechanisms involved. Methods This is a prospective controlled trial. A total of 160 children with ASD will be stratified and allocated to placebo and probiotics groups randomised according to the severity of their ASD symptoms. The probiotics group will be given probiotics supplements orally twice a day for 3 months and the control group will be given a placebo at the same amount, in addition to the baseline therapy of education and rehabilitation. All the children will be evaluated systematically by using different scales, questionnaires before, during, and after 3 months’ treatment, as well as 3 months after discontinuation. The potential impact of probiotics on immunity and inflammation, metabolism, and metagenome will also be investigated. Discussion Our previous study showed that the abundance of intestinal flora was greatly different in children with ASD, and that Bifidobacterium was associated with the severity of ASD. In the present study, we will investigate the impact of probiotics supplementation on the symptoms of Children with ASD, with the purpose of evaluating the possible therapeutic effects of additives on ASD and of providing a reference for clinical treatment. The results will help to disclose as yet unknown relationship between probiotics and ASD. Trial registration This study has been registered with Chinese Clinical Trial Registry (ChiCTR-2000037941).
Collapse
Affiliation(s)
- Lingling Zhang
- Henan Key Laboratory of Child Brain Injury and Henan Clinical Research Center for Child Neurological Disorders, Institute of Neuroscience and The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yiran Xu
- Henan Key Laboratory of Child Brain Injury and Henan Clinical Research Center for Child Neurological Disorders, Institute of Neuroscience and The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongwei Li
- Henan Key Laboratory of Child Brain Injury and Henan Clinical Research Center for Child Neurological Disorders, Institute of Neuroscience and The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Bingbing Li
- Henan Key Laboratory of Child Brain Injury and Henan Clinical Research Center for Child Neurological Disorders, Institute of Neuroscience and The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guiqin Duan
- Center for Child Behavioral Development, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Changlian Zhu
- Henan Key Laboratory of Child Brain Injury and Henan Clinical Research Center for Child Neurological Disorders, Institute of Neuroscience and The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Göteborg, Sweden
- Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
25
|
Abbas-Egbariya H, Haberman Y, Braun T, Hadar R, Denson L, Gal-Mor O, Amir A. Meta-analysis defines predominant shared microbial responses in various diseases and a specific inflammatory bowel disease signal. Genome Biol 2022; 23:61. [PMID: 35197084 PMCID: PMC8867743 DOI: 10.1186/s13059-022-02637-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 02/15/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Gut microbial alteration is implicated in inflammatory bowel disease but is noted in other diseases. Systematic comparison to define similarities and specificities is hampered since most studies focus on a single disease. RESULTS We develop a pipeline to compare between disease cohorts starting from the raw V4 16S amplicon sequence variants. Including 12,838 subjects, from 59 disease cohorts, we demonstrate a predominant shared signature across diseases, indicating a common bacterial response to different diseases. We show that classifiers trained on one disease cohort predict relatively well other diseases due to this shared signal, and hence, caution should be taken when using such classifiers in real-world scenarios, where diseases are intermixed. Based on this common signature across a large array of diseases, we develop a universal dysbiosis index that successfully differentiates between cases and controls across various diseases and can be used for prioritizing fecal donors and samples with lower disease probability. Finally, we identify a set of IBD-specific bacteria, which can direct mechanistic studies and design of IBD-specific microbial interventions. CONCLUSIONS A robust non-specific general response of the gut microbiome is detected in a large array of diseases. Disease classifiers may confuse between different diseases due to this shared microbial response. Our universal dysbiosis index can be used as a tool to prioritize fecal samples and donors. Finally, the IBD-specific taxa may indicate a more direct association to gut inflammation and disease pathogenesis, and those can be further used as biomarkers and as future targets for interventions.
Collapse
Affiliation(s)
- Haya Abbas-Egbariya
- Sheba Medical Center, Tel-HaShomer, affiliated with the Tel-Aviv University, Tel Aviv, Israel
| | - Yael Haberman
- Sheba Medical Center, Tel-HaShomer, affiliated with the Tel-Aviv University, Tel Aviv, Israel
- Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Tzipi Braun
- Sheba Medical Center, Tel-HaShomer, affiliated with the Tel-Aviv University, Tel Aviv, Israel
| | - Rotem Hadar
- Sheba Medical Center, Tel-HaShomer, affiliated with the Tel-Aviv University, Tel Aviv, Israel
| | - Lee Denson
- Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Ohad Gal-Mor
- The Infectious Diseases Research Laboratory, Sheba Medical Center, Tel-Hashomer, and the Department of Clinical Microbiology and Immunology, Tel Aviv University, Tel Aviv, Israel
| | - Amnon Amir
- Sheba Medical Center, Tel-HaShomer, affiliated with the Tel-Aviv University, Tel Aviv, Israel
| |
Collapse
|
26
|
Abuaish S, Al-Otaibi NM, Aabed K, Abujamel TS, Alzahrani SA, Alotaibi SM, Bhat RS, Arzoo S, Algahtani N, Moubayed NM, El-Ansary A. The Efficacy of Fecal Transplantation and Bifidobacterium Supplementation in Ameliorating Propionic Acid-Induced Behavioral and Biochemical Autistic Features in Juvenile Male Rats. J Mol Neurosci 2022; 72:372-381. [PMID: 35094316 DOI: 10.1007/s12031-021-01959-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 12/16/2021] [Indexed: 02/06/2023]
Abstract
Gut microbiota plays a major role in neurological disorders, including autism. Modulation of the gut microbiota through fecal microbiota transplantation (FMT) or probiotic administration, such as Bifidobacteria, is suggested to alleviate autistic symptoms; however, their effects on the brain are not fully examined. We tested both approaches in a propionic acid (PPA) rodent model of autism as treatment strategies. Autism was induced in Sprague-Dawley rats by administering PPA orally (250 mg/kg) for 3 days. Animals were later treated with either saline, FMT, or Bifidobacteria for 22 days. Control animals were treated with saline throughout the study. Social behavior and selected brain biochemical markers related to stress hormones, inflammation, and oxidative stress were assessed. PPA treatment induced social impairments, which was rescued by the treatments. In the brain, Bifidobacteria treatment increased oxytocin relative to control and PPA groups. Moreover, Bifidobacteria treatment rescued the PPA-induced increase in IFN-γ levels. Both treatments increased GST levels, which was diminished by the PPA treatment. These findings indicate the potential of gut microbiota-targeted therapeutics in ameliorating behavioral deficit and underlying neural biochemistry.
Collapse
Affiliation(s)
- Sameera Abuaish
- Department of Basic Sciences, College of Medicine, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Norah M Al-Otaibi
- Department of Biology, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Kawther Aabed
- Department of Biology, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Turki S Abujamel
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, 21589, Saudi Arabia.,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Saleha Ahmad Alzahrani
- Department of Biology, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Sohailah Masoud Alotaibi
- Department of Biology, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Ramesa Shafi Bhat
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Shaista Arzoo
- Department of Food Science and Nutrition, King Saud University, Riyadh, Saudi Arabia
| | - Norah Algahtani
- Central Research Laboratory, King Saud University Female Campus, P O Box 22452, Prince Turki Road, Riyadh, 22452, Saudi Arabia
| | - Nadine Ms Moubayed
- Botany and Microbiology Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Afaf El-Ansary
- Central Research Laboratory, King Saud University Female Campus, P O Box 22452, Prince Turki Road, Riyadh, 22452, Saudi Arabia.
| |
Collapse
|
27
|
Liu Z, Mao X, Dan Z, Pei Y, Xu R, Guo M, Liu K, Zhang F, Chen J, Su C, Zhuang Y, Tang J, Xia Y, Qin L, Hu Z, Liu X. Gene variations in autism spectrum disorder are associated with alteration of gut microbiota, metabolites and cytokines. Gut Microbes 2022; 13:1-16. [PMID: 33412999 PMCID: PMC7808426 DOI: 10.1080/19490976.2020.1854967] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The genetic variations and dysbiosis of gut microbiota are associated with ASD. However, the role of the microbiota in the etiology of ASD in terms of host genetic susceptibility remains unclear. This study aims to systematically explore the interplay between host genetic variation and gut microbiota in ASD children. Whole-exon sequencing was applied to 26 ASD children and 26 matched controls to identify the single nucleotide variations (SNVs) in ASD. Our previous study revealed alteration in gut microbiota and disorder of metabolism activity in ASD for this cohort. Systematic bioinformatic analyses were further performed to identify associations between SNVs and gut microbiota, as well as their metabolites. The ASD SNVs were significantly enriched in genes associated with innate immune response, protein glycosylation process, and retrograde axonal transport. These SNVs were also correlated with the microbiome composition and a broad aspect of microbial functions, especially metabolism. Additionally, the abundance of metabolites involved in the metabolic network of neurotransmitters was inferred to be causally related to specific SNVs and microbes. Furthermore, our data suggested that the interaction of host genetics and gut microbes may play a crucial role in the immune and metabolism homeostasis of ASD. This study may provide valuable clues to investigate the interaction of host genetic variations and gut microbiota in the pathogenesis of ASD.
Collapse
Affiliation(s)
- Zhi Liu
- State Key Laboratory of Reproductive Medicine, Center of Global Health, Nanjing Medical University, Nanjing, China,Key Laboratory of Pathogen Biology of Jiangsu Province, Department of Pathogen Biology, Nanjing Medical University, Nanjing, China,Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, China
| | - Xuhua Mao
- Department of Clinical Laboratory, Affiliated Yixing People’s Hospital, Jiangsu University, Wuxi, China
| | - Zhou Dan
- State Key Laboratory of Reproductive Medicine, Center of Global Health, Nanjing Medical University, Nanjing, China,Key Laboratory of Pathogen Biology of Jiangsu Province, Department of Pathogen Biology, Nanjing Medical University, Nanjing, China,Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, China,Key Laboratory of Holistic Integrative Enterology, Second Affiliated Hospital of Nanjing Medical University, Najing, China
| | - Yang Pei
- State Key Laboratory of Reproductive Medicine, Center of Global Health, Nanjing Medical University, Nanjing, China,Key Laboratory of Pathogen Biology of Jiangsu Province, Department of Pathogen Biology, Nanjing Medical University, Nanjing, China,Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, China
| | - Rui Xu
- State Key Laboratory of Reproductive Medicine, Center of Global Health, Nanjing Medical University, Nanjing, China,Key Laboratory of Pathogen Biology of Jiangsu Province, Department of Pathogen Biology, Nanjing Medical University, Nanjing, China,Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, China
| | - Mengchen Guo
- State Key Laboratory of Reproductive Medicine, Center of Global Health, Nanjing Medical University, Nanjing, China,Key Laboratory of Pathogen Biology of Jiangsu Province, Department of Pathogen Biology, Nanjing Medical University, Nanjing, China,Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, China
| | - Kangjian Liu
- Key Laboratory of Holistic Integrative Enterology, Second Affiliated Hospital of Nanjing Medical University, Najing, China
| | - Faming Zhang
- Key Laboratory of Holistic Integrative Enterology, Second Affiliated Hospital of Nanjing Medical University, Najing, China
| | - Junyu Chen
- State Key Laboratory of Reproductive Medicine, Center of Global Health, Nanjing Medical University, Nanjing, China,Key Laboratory of Pathogen Biology of Jiangsu Province, Department of Pathogen Biology, Nanjing Medical University, Nanjing, China,Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, China
| | - Chuan Su
- State Key Laboratory of Reproductive Medicine, Center of Global Health, Nanjing Medical University, Nanjing, China,Key Laboratory of Pathogen Biology of Jiangsu Province, Department of Pathogen Biology, Nanjing Medical University, Nanjing, China
| | - Yaoyao Zhuang
- State Key Laboratory of Reproductive Medicine, Center of Global Health, Nanjing Medical University, Nanjing, China,Key Laboratory of Pathogen Biology of Jiangsu Province, Department of Pathogen Biology, Nanjing Medical University, Nanjing, China,Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, China
| | - Junming Tang
- Department of Clinical Laboratory, Affiliated Yixing People’s Hospital, Jiangsu University, Wuxi, China
| | - Yankai Xia
- State Key Laboratory of Reproductive Medicine, Center of Global Health, Nanjing Medical University, Nanjing, China
| | - Lianhong Qin
- Children Growth Center of Bo’ai Homestead in Yixing, Yixing, China
| | - Zhibin Hu
- State Key Laboratory of Reproductive Medicine, Center of Global Health, Nanjing Medical University, Nanjing, China
| | - Xingyin Liu
- State Key Laboratory of Reproductive Medicine, Center of Global Health, Nanjing Medical University, Nanjing, China,Key Laboratory of Pathogen Biology of Jiangsu Province, Department of Pathogen Biology, Nanjing Medical University, Nanjing, China,Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, China,Key Laboratory of Holistic Integrative Enterology, Second Affiliated Hospital of Nanjing Medical University, Najing, China,CONTACT Xingyin Liu State Key Laboratory of Reproductive Medicine, Center of Gobal Health, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing 211166, P.R. China
| |
Collapse
|
28
|
Wang S, Ma X, Wu L, Yu H, Shan Y, Tian Y, Liu T, Gu X. Effect of gastrointestinal heat retention syndrome on gut microbiota in children with upper respiratory tract infection and lung-heat syndrome. JOURNAL OF TRADITIONAL CHINESE MEDICAL SCIENCES 2022. [DOI: 10.1016/j.jtcms.2022.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
29
|
Pan ZY, Zhong HJ, Huang DN, Wu LH, He XX. Beneficial Effects of Repeated Washed Microbiota Transplantation in Children With Autism. Front Pediatr 2022; 10:928785. [PMID: 35783298 PMCID: PMC9249087 DOI: 10.3389/fped.2022.928785] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 05/30/2022] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE While fecal microbiota transplantation is demonstrated to improve symptoms of autism spectrum disorder (ASD), it remains unclear whether additional treatment courses yield better results. This study sought to evaluate the efficacy of repeated washed microbiota transplantation (WMT) in children with ASD. METHODS Retrospective data from children who were serially treated with WMT, including ASD symptoms, sleep disorders, gastrointestinal (GI) symptoms, and white blood cell (WBC) and globulin levels were obtained. The effect of WMT on children with ASD and whether additional WMT courses led to a further improvement in symptoms were assessed. RESULTS Aberrant Behavior Checklist (ABC), Childhood Autism Rating Scale, and Sleep Disturbance Scale for Children (SDSC) scores, the proportion of children with constipation and abnormal fecal forms, and WBC and globulin levels were all significantly lower in ASD children after WMT. More WMT treatment courses led to significantly lower scores on the ABC and SDSC. CONCLUSION WMT significantly improved ASD and GI symptoms and sleep disorders in children with ASD, and reduced systemic inflammation. Additional WMT courses led to more obvious improvements in ASD symptoms within three treatment courses.
Collapse
Affiliation(s)
- Zhao-Yu Pan
- Department of Gastroenterology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Research Center for Engineering Techniques of Microbiota-Targeted Therapies of Guangdong Province, Guangzhou, China
| | - Hao-Jie Zhong
- Department of Gastroenterology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Research Center for Engineering Techniques of Microbiota-Targeted Therapies of Guangdong Province, Guangzhou, China
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
- *Correspondence: Hao-Jie Zhong,
| | - Dong-Ni Huang
- Department of Gastroenterology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Research Center for Engineering Techniques of Microbiota-Targeted Therapies of Guangdong Province, Guangzhou, China
| | - Li-Hao Wu
- Department of Gastroenterology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Research Center for Engineering Techniques of Microbiota-Targeted Therapies of Guangdong Province, Guangzhou, China
| | - Xing-Xiang He
- Department of Gastroenterology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Research Center for Engineering Techniques of Microbiota-Targeted Therapies of Guangdong Province, Guangzhou, China
- Xing-Xiang He,
| |
Collapse
|
30
|
Chen LL, Abbaspour A, Mkoma GF, Bulik CM, Rück C, Djurfeldt D. Gut Microbiota in Psychiatric Disorders: A Systematic Review. Psychosom Med 2021; 83:679-692. [PMID: 34117156 PMCID: PMC8428865 DOI: 10.1097/psy.0000000000000959] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 02/04/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVE This systematic review sought to comprehensively summarize gut microbiota research in psychiatric disorders following PRISMA guidelines. METHODS Literature searches were performed on databases using keywords involving gut microbiota and psychiatric disorders. Articles in English with human participants up until February 13, 2020, were reviewed. Risk of bias was assessed using a modified Newcastle-Ottawa Scale for microbiota studies. RESULTS Sixty-nine of 4231 identified studies met the inclusion criteria for extraction. In most studies, gut microbiota composition differed between individuals with psychiatric disorders and healthy controls; however, limited consistency was observed in the taxonomic profiles. At the genus level, the most replicated findings were higher abundance of Bifidobacterium and lower abundance of Roseburia and Faecalibacterium among patients with psychiatric disorders. CONCLUSIONS Gut bacteria that produce short-chain fatty acids, such as Roseburia and Faecalibacterium, could be less abundant in patients with psychiatric disorders, whereas commensal genera, for example, Bifidobacterium, might be more abundant compared with healthy controls. However, most included studies were hampered by methodological shortcomings including small sample size, unclear diagnostics, failure to address confounding factors, and inadequate bioinformatic processing, which might contribute to inconsistent results. Based on our findings, we provide recommendations to improve quality and comparability of future microbiota studies in psychiatry.
Collapse
|
31
|
Abdulhaq A, Halboub E, Homeida HE, Kumar Basode V, Ghzwani AH, Zain KA, Baraniya D, Chen T, Al-Hebshi NN. Tongue microbiome in children with autism spectrum disorder. J Oral Microbiol 2021; 13:1936434. [PMID: 34211637 PMCID: PMC8221129 DOI: 10.1080/20002297.2021.1936434] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background: A few recent studies have characterized the salivary microbiome in association with Autism Spectrum Disorder (ASD). Here, we sought to assess if there is an association between the tongue microbiome and ASD. Methods: Tongue scrapping samples were obtained from 25 children with ASD and 38 neurotypical controls. The samples were sequenced for the 16S rRNA gene (V1-V3) and the resultant high-quality reads were assigned to the species-level using our previously described BLASTn-based algorithm. Downstream analyses of microbial profiles were conducted using QIIME, LEfSe, and R. Results: Independent of grouping, Prevotella, Streptococcus, Leptotrichia, Veillonella, Haemophilus and Rothia accounted for > 60% of the average microbiome. Haemophilus parainfluenzae, Rothia mucilaginosa, Prevotella melaninogenica and Neisseria flavescens/subflava were the most abundant species. Species richness and diversity did not significantly differ between the study groups. Thirteen species and three genera were differentially abundant between the two groups, e.g. enrichment of Actinomyces odontolyticus and Actinomyces lingnae and depletion of Campylobacter concisus and Streptococcus vestibularis in the ASD group. However, none of them withstood adjustment for multiple comparisons. Conclusion: The tongue microbiome of children with ASD was not significantly different from that of healthy control children, which is largely consistent with results from the literature.
Collapse
Affiliation(s)
- Ahmed Abdulhaq
- Department of Medical Laboratory Technology, College of Applied Medical Sciences, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Esam Halboub
- Department of Maxillofacial Surgery and Diagnostic Sciences, College of Dentistry, Jazan University, Jazan, Kingdom of Saudi Arabia.,Department of Oral Medicine, Oral Pathology and Oral Radiology, Faculty of Dentistry, Sana'a University, Yemen
| | - Husham E Homeida
- Department of Maxillofacial Surgery and Diagnostic Sciences, College of Dentistry, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Vinod Kumar Basode
- Department of Medical Laboratory Technology, College of Applied Medical Sciences, Jazan University, Jazan, Kingdom of Saudi Arabia
| | | | - Khalid Ammash Zain
- Medical Research Centre, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Divyashri Baraniya
- Department of Oral Health Sciences, Oral Microbiome Research Laboratory, Maurice H. Kornberg School of Dentistry, Temple University, Philadelphia, PA, USA
| | - Tsute Chen
- Department of Microbiology, Forsyth Institute, Cambridge, MA, USA
| | - Nezar Noor Al-Hebshi
- Department of Oral Health Sciences, Oral Microbiome Research Laboratory, Maurice H. Kornberg School of Dentistry, Temple University, Philadelphia, PA, USA
| |
Collapse
|
32
|
Chok KC, Ng KY, Koh RY, Chye SM. Role of the gut microbiome in Alzheimer's disease. Rev Neurosci 2021; 32:767-789. [PMID: 33725748 DOI: 10.1515/revneuro-2020-0122] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 02/19/2021] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia, affecting millions of individuals each year and this number is expected to significantly increase. The complicated microorganisms residing in human gut are closely associated with our health. Emerging evidence has suggested possible involvement of human gut microbiome in AD. Symbiotic gut microbiomes are known to maintain brain health by modulating host's barriers integrity, metabolic system, immune system, nervous system and endocrine system. However, in the event of gut dysbiosis and barriers disruption, gut pathobionts disrupt homeostasis of the metabolic system, immune system, nervous system, and endocrine system, resulting in deterioration of neurological functions and subsequently promoting development of AD. Multiple therapeutic approaches, such as fecal microbiome transplant, antibiotics, prebiotics, probiotics, symbiotic, and diet are discussed as potential treatment options for AD by manipulating the gut microbiome to reverse pathological alteration in the systems above.
Collapse
Affiliation(s)
- Kian Chung Chok
- School of Health Science, International Medical University, 57000Kuala Lumpur, Malaysia
| | - Khuen Yen Ng
- School of Pharmacy, Monash University Malaysia, 47500Selangor, Malaysia
| | - Rhun Yian Koh
- Division of Biomedical Science and Biotechnology, School of Health Science, International Medical University, No. 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000Kuala Lumpur, Malaysia
| | - Soi Moi Chye
- Division of Biomedical Science and Biotechnology, School of Health Science, International Medical University, No. 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000Kuala Lumpur, Malaysia
| |
Collapse
|
33
|
Hao X, Pan J, Gao X, Zhang S, Li Y. Gut microbiota on gender bias in autism spectrum disorder. Rev Neurosci 2020; 32:/j/revneuro.ahead-of-print/revneuro-2020-0042/revneuro-2020-0042.xml. [PMID: 32887209 DOI: 10.1515/revneuro-2020-0042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Accepted: 05/29/2020] [Indexed: 12/15/2022]
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder. Its three core symptoms are social communication disorder, communication disorder, narrow interest and stereotyped repetitive behavior. The proportion of male and female autistic patients is 4:1. Many researchers have studied this phenomenon, but the mechanism is still unclear. This review mainly discusses the related mechanism from the perspective of gut microbiota and introduces the influence of gut microbiota on the difference of ASD between men and women, as well as how gut microbiota may affect the gender dimorphism of ASD through metabolite of microbiota, immunity, and genetics, which provide some useful information for those who are interested in this research and find more gender-specific treatment for autistic men and women.
Collapse
Affiliation(s)
- Xia Hao
- State Key Laboratory of Component-Based Chinese Medicine, Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medicine Formulae, Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin301617,China
- College of Traditional Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin301617,China
| | - Jiao Pan
- Department of Microbiology, Ministry of Education Key Laboratory of Molecular Microbiology and Technology, Nankai University, Tianjin300071,China
| | - Xiumei Gao
- State Key Laboratory of Component-Based Chinese Medicine, Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medicine Formulae, Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin301617,China
| | - Shiyu Zhang
- College of Traditional Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin301617,China
| | - Yue Li
- State Key Laboratory of Component-Based Chinese Medicine, Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medicine Formulae, Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin301617,China
| |
Collapse
|
34
|
Abstract
While there are numerous medical comorbidities associated with ASD, gastrointestinal (GI) issues have a significant impact on quality of life for these individuals. Recent findings continue to support the relationship between the gut microbiome and both GI symptoms and behavior, but the heterogeneity within the autism spectrum requires in-depth clinical characterization of these clinical cohorts. Large, diverse, well-controlled studies in this area of research are still needed. Although there is still much to discover about the brain-gut-microbiome axis in ASD, microbially mediated therapies, specifically probiotics and fecal microbiota transplantation have shown promise in the treatment of GI symptoms in ASD, with potential benefit to the core behavioral symptoms of ASD as well. Future research and clinical trials must increasingly consider complex phenotypes in ASD in stratification of large datasets as well as in design of inclusion criteria for individual therapeutic interventions.
Collapse
Affiliation(s)
- Virginia Saurman
- Department of Pediatrics, Columbia University Medical Center, 620 West 168th Street, New York, NY 10032, USA
| | - Kara G. Margolis
- Department of Pediatrics, Columbia University Medical Center, 620 West 168th Street, New York, NY 10032, USA
| | - Ruth Ann Luna
- Department of Pathology and Immunology, Texas Children’s Microbiome Center, Baylor College of Medicine, Texas Children’s Hospital, Feigin Tower, 1102 Bates Avenue, Suite 955, Houston, TX 77030, USA
| |
Collapse
|