1
|
Qing L, Qian X, Zhu H, Wang J, Sun J, Jin Z, Tang X, Zhao Y, Wang G, Zhao J, Chen W, Tian P. Maternal-infant probiotic transmission mitigates early-life stress-induced autism in mice. Gut Microbes 2025; 17:2456584. [PMID: 39931863 DOI: 10.1080/19490976.2025.2456584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 12/14/2024] [Accepted: 01/13/2025] [Indexed: 02/14/2025] Open
Abstract
Autism, a disorder influenced by both genetic and environmental factors, presents significant challenges for prevention and treatment. While maternal-infant gut microbiota has been a focus in autism research, preventive strategies targeting maternal gut microbiota remain underexplored. This study demonstrates that prenatal probiotic intake can effectively prevent maternal separation-induced autistic-like behaviors in offspring without altering the embryonic neurodevelopment in mice. Using specific PCR primers and cross-fostering experiments, we traced the vertical transmission of probiotics, primarily via fecal/vaginal contamination. Early probiotic colonization conferred resilience against stress-induced gut pathogenic microbes and Th17-mediated peripheral inflammation while significantly inhibiting hypermyelination and neuroinflammation linked to systemic inflammation. Microbial metabolites like tyrosol and xanthurenic acid alleviated neuroinflammation and hypermyelination in vitro, though the causal relationship among neuroinflammation, hypermyelination, and autism in vivo requires further validation. These findings underscore the importance of the maternal-infant microbiota transmission window in autism prevention and highlight the clinical potential of prenatal probiotic interventions.
Collapse
Affiliation(s)
- Li Qing
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Xin Qian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Huiyue Zhu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Jingyu Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Jingge Sun
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Zhiying Jin
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Xinyu Tang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Yingqi Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Gang Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, P. R. China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou, P. R. China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Peijun Tian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| |
Collapse
|
2
|
Prince N, Peralta Marzal LN, Roussin L, Monnoye M, Philippe C, Maximin E, Ahmed S, Salenius K, Lin J, Autio R, Adolfs Y, Pasterkamp RJ, Garssen J, Naudon L, Rabot S, Kraneveld AD, Perez-Pardo P. Mouse strain-specific responses along the gut-brain axis upon fecal microbiota transplantation from children with autism. Gut Microbes 2025; 17:2447822. [PMID: 39773319 PMCID: PMC11730631 DOI: 10.1080/19490976.2024.2447822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 12/03/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025] Open
Abstract
Several factors are linked to the pathophysiology of autism spectrum disorders (ASD); however, the molecular mechanisms of the condition remain unknown. As intestinal problems and gut microbiota dysbiosis are associated with ASD development and severity, recent studies have focused on elucidating the microbiota-gut-brain axis' involvement. This study aims to explore mechanisms through which gut microbiota might influence ASD. Briefly, we depleted the microbiota of conventional male BALB/cAnNCrl (Balb/c) and C57BL/6J (BL/6) mice prior to human fecal microbiota transplantation (hFMT) with samples from children with ASD or their neurotypical siblings. We found mouse strain-specific responses to ASD hFMT. Notably, Balb/c mice exhibit decreased exploratory and social behavior, and show evidence of intestinal, systemic, and central inflammation accompanied with metabolic shifts. BL/6 mice show less changes after hFMT. Our results reveal that gut microbiota alone induce changes in ASD-like behavior, and highlight the importance of mouse strain selection when investigating multifactorial conditions like ASD.
Collapse
Affiliation(s)
- Naika Prince
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Lucia N. Peralta Marzal
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Léa Roussin
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Magali Monnoye
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Catherine Philippe
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Elise Maximin
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Sabbir Ahmed
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Karoliina Salenius
- Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Centre, Tampere, Finland
| | - Jake Lin
- Health Sciences, Faculty of Social Sciences, Tampere University, Tampere, Finland
| | - Reija Autio
- Health Sciences, Faculty of Social Sciences, Tampere University, Tampere, Finland
| | - Youri Adolfs
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, Netherlands
| | - R. Jeroen Pasterkamp
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, Netherlands
| | - Johan Garssen
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
- Danone Nutricia Research, Utrecht, Netherlands
| | - Laurent Naudon
- Université Paris-Saclay, INRAE, AgroParisTech, CNRS, Micalis Institute, Jouy-en-Josas, France
| | - Sylvie Rabot
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Aletta D. Kraneveld
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
- Department of Neuroscience, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Paula Perez-Pardo
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
3
|
Logan AC, Mishra P, Prescott SL. The Legalome: Microbiology, Omics and Criminal Justice. Microb Biotechnol 2025; 18:e70129. [PMID: 40072296 PMCID: PMC11898878 DOI: 10.1111/1751-7915.70129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 02/25/2025] [Accepted: 02/28/2025] [Indexed: 03/15/2025] Open
Abstract
Advances in neuromicrobiology and related omics technologies have reinforced the idea that unseen microbes play critical roles in human cognition and behaviour. Included in this research is evidence indicating that gut microbes, through direct and indirect pathways, can influence aggression, anger, irritability and antisocial behaviour. Moreover, gut microbes can manufacture chemicals that are known to compromise cognition. For example, recent court decisions in the United States and Europe acknowledge that gut microbes can produce high levels of ethanol, without consumption of alcohol by the defendants. The dismissal of driving while intoxicated charges in these cases-so-called auto-brewery syndrome-highlights the way in which microbiome knowledge will enhance the precision, objectivity and fairness of our legal systems. Here in this opinion essay, we introduce the concept of the 'legalome'-the application of microbiome and omics science to forensic psychiatry and criminal law. We argue that the rapid pace of microbial discoveries, including those that challenge ideas of free will and moral responsibility, will necessitate a reconsideration of traditional legal doctrines and justifications of retributive punishment. The implications extend beyond the courtroom, challenging us to reconsider how environmental factors-from diet to socioeconomic conditions-might shape preventative and rehabilitative efforts through their effects on the microbiome.
Collapse
Affiliation(s)
| | - Pragya Mishra
- University of Allahabad (A Central University)PrayagrajIndia
| | - Susan L. Prescott
- Nova Institute for HealthBaltimoreMarylandUSA
- University of Western AustraliaPerthWestern AustraliaAustralia
- University of MarylandBaltimoreMarylandUSA
| |
Collapse
|
4
|
Hu DX, Lu CM, Si XY, Wu QT, Wu LH, Zhong HJ, He XX. Effects of gastrointestinal symptoms on the efficacy of washed microbiota transplantation in patients with autism. Front Pediatr 2025; 13:1528167. [PMID: 40017709 PMCID: PMC11865235 DOI: 10.3389/fped.2025.1528167] [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: 11/14/2024] [Accepted: 01/24/2025] [Indexed: 03/01/2025] Open
Abstract
Objective Washed microbiota transplantation (WMT) has emerged as a promising therapeutic strategy for autism spectrum disorder (ASD), though the factors that influence its efficacy remain poorly understood. This study explores the impact of gastrointestinal (GI) symptoms on the effectiveness of WMT in ASD. Methods Clinical data encompassing ASD symptoms, GI disturbances, and sleep disorders were collected from patients with ASD undergoing WMT. The therapeutic impact of WMT and the contributing factors to its efficacy were assessed. Results WMT significantly reduced scores on the Aberrant Behavior Checklist (ABC), Childhood Autism Rating Scale (CARS), and Sleep Disturbance Scale for Children (SDSC), alongside a significant reduction in the incidence of constipation, abnormal stool forms, and diarrhea (all p < 0.05). After six courses of WMT, substantial reductions were observed in ABC, CARS, and SDSC scores, with increased treatment courses correlating with greater improvement (p < 0.05). Multiple linear regression analysis revealed that WMT efficacy was enhanced in patients with pre-existing GI symptoms (diarrhea: β = 0.119, p < 0.001; abnormal stool form: β = 0.201, p < 0.001) and those receiving a higher number of treatment courses (β = 0.116, p < 0.001). Additionally, the analysis indicated that treatment outcomes were more favorable in patients who had not undergone adjunct interventions (β = -0.041, p = 0.002), had a longer disease duration (β = 0.168, p = 0.007), and exhibited more severe disease symptoms (β = 0.125, p < 0.001). Conclusion WMT significantly alleviates both ASD and GI symptoms, along with sleep disturbances, in affected individuals. Six treatment courses resulted in notable improvement, with increased course numbers further improving therapeutic outcomes. Furthermore, pre-treatment GI symptoms, such as diarrhea and abnormal stool forms, may influence the effectiveness of WMT. Notably, patients who did not receive additional interventions, had a prolonged disease duration, and presented with more severe symptoms experienced markedly improved treatment responses.
Collapse
Affiliation(s)
- Dong-Xia Hu
- Department of Gastroenterology, Research Center for Engineering Techniques of Microbiota-Targeted Therapies of Guangdong Province, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Cai-Mei Lu
- Department of Gastroenterology, Research Center for Engineering Techniques of Microbiota-Targeted Therapies of Guangdong Province, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Xin-Yu Si
- Department of Gastroenterology, Research Center for Engineering Techniques of Microbiota-Targeted Therapies of Guangdong Province, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Qing-Ting Wu
- Department of Gastroenterology, Research Center for Engineering Techniques of Microbiota-Targeted Therapies of Guangdong Province, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Li-Hao Wu
- Department of Gastroenterology, Research Center for Engineering Techniques of Microbiota-Targeted Therapies of Guangdong Province, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Hao-Jie Zhong
- Department of Gastroenterology, Research Center for Engineering Techniques of Microbiota-Targeted Therapies of Guangdong Province, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Xing-Xiang He
- Department of Gastroenterology, Research Center for Engineering Techniques of Microbiota-Targeted Therapies of Guangdong Province, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
5
|
Hassib L, Kanashiro A, Pedrazzi JFC, Vercesi BF, Higa S, Arruda Í, Soares Y, de Jesus de Souza A, Jordão AA, Guimarães FS, Ferreira FR. Should we consider microbiota-based interventions as a novel therapeutic strategy for schizophrenia? A systematic review and meta-analysis. Brain Behav Immun Health 2025; 43:100923. [PMID: 39839986 PMCID: PMC11745983 DOI: 10.1016/j.bbih.2024.100923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 12/04/2024] [Accepted: 12/10/2024] [Indexed: 01/06/2025] Open
Abstract
Schizophrenia is a chronic psychiatric disorder characterized by a variety of symptoms broadly categorized into positive, negative, and cognitive domains. Its etiology is multifactorial, involving a complex interplay of genetic, neurobiological, and environmental factors, and its neurobiology is associated with abnormalities in different neurotransmitter systems. Due to this multifactorial etiology and neurobiology, leading to a wide heterogeneity of symptoms and clinical presentations, current antipsychotic treatments face challenges, underscoring the need for novel therapeutic approaches. Recent studies have revealed differences in the gut microbiome of individuals with schizophrenia compared to healthy controls, establishing an intricate link between this disorder and gastrointestinal health, and suggesting that microbiota-targeted interventions could help alleviate clinical symptoms. Therefore, this meta-analysis investigates whether gut microbiota manipulation can ameliorate psychotic outcomes in patients with schizophrenia receiving pharmacological treatment. Nine studies (n = 417 participants) were selected from 81 records, comprising seven randomized controlled trials and two open-label studies, all with a low risk of bias, included in this systematic review and meta-analysis. The overall combined effect size indicated significant symptom improvement following microbiota treatment (Hedges' g = 0.48, 95% CI = 0.09 to 0.88, p = 0.004, I2 = 62.35%). However, according to Hedges' g criteria, the effect size was small (approaching moderate), and study heterogeneity was moderate based on I2 criteria. This review also discusses clinical and preclinical studies to elucidate the neural, immune, and metabolic pathways by which microbiota manipulation, particularly with Lactobacillus and Bifidobacterium genera, may exert beneficial effects on schizophrenia symptoms via the gut-brain axis. Finally, we address the main confounding factors identified in our systematic review, highlight key limitations, and offer recommendations to guide future high-quality trials with larger participant cohorts to explore microbiome-based therapies as a primary or adjunctive treatment for schizophrenia.
Collapse
Affiliation(s)
- Lucas Hassib
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
- Oswaldo Cruz Foundation, Institute Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - Alexandre Kanashiro
- Faillace Department of Psychiatry and Behavioral Sciences, Translational Psychiatry Program, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | | | - Bárbara Ferreira Vercesi
- Department of Health Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Sayuri Higa
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of Sao Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Íris Arruda
- Oswaldo Cruz Foundation, Institute Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - Yago Soares
- Oswaldo Cruz Foundation, Institute Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - Adriana de Jesus de Souza
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of Sao Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Alceu Afonso Jordão
- Department of Health Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Francisco Silveira Guimarães
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of Sao Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | | |
Collapse
|
6
|
Tomaszek N, Urbaniak AD, Bałdyga D, Chwesiuk K, Modzelewski S, Waszkiewicz N. Unraveling the Connections: Eating Issues, Microbiome, and Gastrointestinal Symptoms in Autism Spectrum Disorder. Nutrients 2025; 17:486. [PMID: 39940343 PMCID: PMC11819948 DOI: 10.3390/nu17030486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/27/2025] [Accepted: 01/28/2025] [Indexed: 02/16/2025] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental condition characterized by challenges in social communication, restricted interests, and repetitive behaviors. It is also associated with a high prevalence of eating disorders, gastrointestinal (GI) symptoms, and alterations in gut microbiota composition. One of the most pressing concerns is food selectivity. Various eating disorders, such as food neophobia, avoidant/restrictive food intake disorder (ARFID), specific dietary patterns, and poor-quality diets, are commonly observed in this population, often leading to nutrient deficiencies. Additionally, gastrointestinal problems in children with ASD are linked to imbalances in gut microbiota and immune system dysregulation. The aim of this narrative review is to identify previous associations between the gut-brain axis and gastrointestinal problems in ASD. We discuss the impact of the "microbiome-gut-brain axis", a bidirectional connection between gut microbiota and brain function, on the development and symptoms of ASD. In gastrointestinal problems associated with ASD, a 'vicious cycle' may play a significant role: ASD symptoms contribute to the prevalence of ARFID, which in turn leads to microbiota degradation, ultimately worsening ASD symptoms. Current data suggest a link between gastrointestinal problems in ASD and the microbiota, but the amount of evidence is limited. Further research is needed, targeting the correlation of a patient's microbiota status, dietary habits, and disease course.
Collapse
Affiliation(s)
| | | | | | | | - Stefan Modzelewski
- Department of Psychiatry, Medical University of Bialystok, pl. Wołodyjowskiego 2, 15-272 Białystok, Poland; (N.T.); (A.D.U.); (D.B.); (K.C.); (N.W.)
| | | |
Collapse
|
7
|
Salia S, Burke FF, Hinks ME, Randell AM, Matheson MA, Walling SG, Swift-Gallant A. Gut microbiota transfer from the preclinical maternal immune activation model of autism is sufficient to induce sex-specific alterations in immune response and behavioural outcomes. Brain Behav Immun 2025; 123:813-823. [PMID: 39471905 DOI: 10.1016/j.bbi.2024.10.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/22/2024] [Accepted: 10/26/2024] [Indexed: 11/01/2024] Open
Abstract
The gut microbiome plays a vital role in health and disease, including neurodevelopmental disorders like autism spectrum disorder (ASD). ASD affects 4:1 males-to-females, and sex differences are apparent in gut microbiota composition among ASD individuals and in animal models of this condition, such as the maternal immune activation (MIA) mouse model. However, few studies have included sex as a biological variable when assessing the role of gut microbiota in mediating ASD symptoms. Using the MIA model of ASD, we assessed whether gut microbiota contributes to the sex differences in the presentation of ASD-like behaviors. Gut microbiota transplantation from MIA or vehicle/control male and female mice into healthy, otherwise unmanipulated, 4-week-old C57Bl/6 mice was performed for 6 treatments over 12 days. Colonization with male, but not female, MIA microbiota was sufficient to reduce sociability, decrease microbiota diversity and increase neuroinflammation with more pronounced deficits in male recipients. Colonization with both male and female donor microbiota altered juvenile ultrasonic vocalizations and anxiety-like behavior in recipients of both sexes, and there was an accompanied change in the gut microbiota and serum cytokine IL-4 and IL-7 levels of all recipients of MIA gut microbiota. In addition to the increases in gut microbes associated with pathological states, the female donor microbiota profile also had increases in gut microbes with known neural protective effects (e.g., Lactobacillus and Rikenella). These results suggest that gut reactivity to environmental insults, such as in the MIA model, may play a role in shaping the sex disparity in ASD development.
Collapse
Affiliation(s)
- Stephanie Salia
- Department of Psychology, Memorial University of Newfoundland, 232 Elizabeth Avenue, St. John's, NL A1B 3X9, Canada.
| | - Francine F Burke
- Department of Psychology, Memorial University of Newfoundland, 232 Elizabeth Avenue, St. John's, NL A1B 3X9, Canada
| | - Meagan E Hinks
- Department of Psychology, Memorial University of Newfoundland, 232 Elizabeth Avenue, St. John's, NL A1B 3X9, Canada
| | - Alison M Randell
- Department of Psychology, Memorial University of Newfoundland, 232 Elizabeth Avenue, St. John's, NL A1B 3X9, Canada
| | - Mairead Anna Matheson
- Department of Psychology, Memorial University of Newfoundland, 232 Elizabeth Avenue, St. John's, NL A1B 3X9, Canada
| | - Susan G Walling
- Department of Psychology, Memorial University of Newfoundland, 232 Elizabeth Avenue, St. John's, NL A1B 3X9, Canada
| | - Ashlyn Swift-Gallant
- Department of Psychology, Memorial University of Newfoundland, 232 Elizabeth Avenue, St. John's, NL A1B 3X9, Canada.
| |
Collapse
|
8
|
Mamun AA, Geng P, Wang S, Shao C, Xiao J. IUPHAR review: Targeted therapies of signaling pathways based on the gut microbiome in autism spectrum disorders: Mechanistic and therapeutic applications. Pharmacol Res 2025; 211:107559. [PMID: 39733842 DOI: 10.1016/j.phrs.2024.107559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 12/22/2024] [Accepted: 12/23/2024] [Indexed: 12/31/2024]
Abstract
Autism spectrum disorders (ASD) are complex neurodevelopmental disorders characterized by impairments in social interaction, communication and repetitive activities. Gut microbiota significantly influences behavior and neurodevelopment by regulating the gut-brain axis. This review explores gut microbiota-influenced treatments for ASD, focusing on their therapeutic applications and mechanistic insights. In addition, this review discusses the interactions between gut microbiota and the immune, metabolic and neuroendocrine systems, focusing on crucial microbial metabolites including short-chain fatty acids (SCFAs) and several neurotransmitters. Furthermore, the review explores various therapy methods including fecal microbiota transplantation, dietary modifications, probiotics and prebiotics and evaluates their safety and efficacy in reducing ASD symptoms. The discussion shows the potential of customized microbiome-based therapeutics and the integration of multi-omics methods to understand the underlying mechanisms. Moreover, the review explores the intricate relationship between gut microbiota and ASD, aiming to develop innovative therapies that utilize the gut microbiome to improve the clinical outcomes of ASD patients. Microbial metabolites such as neurotransmitter precursors, tryptophan metabolites and SCFAs affect brain development and behavior. Symptoms of ASD are linked to changes in these metabolites. Dysbiosis in the gut microbiome may impact neuroinflammatory processes linked to autism, negatively affecting immune signaling pathways. Research indicates that probiotics and prebiotics can improve gut microbiota and alleviate symptoms in ASD patients. Fecal microbiota transplantation may also improve behavioral symptoms and restore gut microbiota balance. The review emphasizes the need for further research on gut microbiota modification as a potential therapeutic approach for ASD, highlighting its potential in clinical settings.
Collapse
Affiliation(s)
- Abdullah Al Mamun
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People's Hospital, Lishui, Zhejiang 323000, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Peiwu Geng
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People's Hospital, Lishui, Zhejiang 323000, China
| | - Shuanghu Wang
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People's Hospital, Lishui, Zhejiang 323000, China
| | - Chuxiao Shao
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People's Hospital, Lishui, Zhejiang 323000, China.
| | - Jian Xiao
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People's Hospital, Lishui, Zhejiang 323000, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Department of Wound Healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| |
Collapse
|
9
|
Wang J, Yang R, Zhong H, Liu YJ. Fecal microbiota transplants in pediatric autism: opportunities and challenges. World J Pediatr 2024; 20:1201-1204. [PMID: 39548040 DOI: 10.1007/s12519-024-00855-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 10/23/2024] [Indexed: 11/17/2024]
Affiliation(s)
- Jian Wang
- Department of Pediatrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Rong Yang
- Department of Pediatrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hui Zhong
- Department of Pediatrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yan-Jun Liu
- Department of Pediatrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
10
|
Poupard L, Page G, Thoreau V, Kaouah Z. Relationships between Gut Microbiota and Autism Spectrum Disorders: Development and Treatment. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE : THE OFFICIAL SCIENTIFIC JOURNAL OF THE KOREAN COLLEGE OF NEUROPSYCHOPHARMACOLOGY 2024; 22:554-564. [PMID: 39420603 PMCID: PMC11494427 DOI: 10.9758/cpn.24.1179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 05/31/2024] [Accepted: 06/03/2024] [Indexed: 10/19/2024]
Abstract
Many studies have demonstrated the impact of intestinal microbiota on normal brain development. Moreover, the gut microbiota (GM) is impacted by multiple endogenous and environmental factors that may promote gut dysbiosis (GD). An increasing number of studies are investigating the possible role of the GD in the development of neurological and behavioral disorders. For autism spectrum disorders (ASD), specific intestinal bacterial signatures have been identified, knowing that gastrointestinal symptoms are frequently found in ASD. In this review, the peri and post-natal factors modulating the GM are described and the specific gut bacterial signature of ASD children is detailed. Through bidirectional communication between the GM and the brain, several mechanisms are involved in the development of ASD, such as cytokine-mediated neuroinflammation and decreased production of neuroprotective factors such as short-chain fatty acids by the GM. Imbalance of certain neurotransmitters such as serotonin or gamma-aminobutyric acid could also play a role in these gut-brain interactions. Some studies show that this GD in ASD is partly reversible by treatment with pre- and probiotics, or fecal microbiota transplantation with promising results. However, certain limitations have been raised, in particular concerning the short duration of treatment, the small sample sizes and the diversity of protocols. The development of standardized therapeutics acting on GD in large cohort could rescue the gastrointestinal symptoms and behavioral impairments, as well as patient management.
Collapse
Affiliation(s)
- Lisa Poupard
- Medicine and Pharmacy Faculty, University of Poitiers, Poitiers, France
| | - Guylène Page
- Medicine and Pharmacy Faculty, University of Poitiers, Poitiers, France
- Neurovascular Unit and Cognitive Disorders (NEUVACOD), Pôle Biologie Santé, University of Poitiers, Poitiers, France
| | - Vincent Thoreau
- Medicine and Pharmacy Faculty, University of Poitiers, Poitiers, France
- Neurovascular Unit and Cognitive Disorders (NEUVACOD), Pôle Biologie Santé, University of Poitiers, Poitiers, France
| | - Zahyra Kaouah
- Medicine and Pharmacy Faculty, University of Poitiers, Poitiers, France
- Neurovascular Unit and Cognitive Disorders (NEUVACOD), Pôle Biologie Santé, University of Poitiers, Poitiers, France
| |
Collapse
|
11
|
Finnegan YE, Neill HR, Prpa EJ, Pot B. "Gut" to grips with the science of the microbiome - a symposium report. GUT MICROBIOME (CAMBRIDGE, ENGLAND) 2024; 5:e11. [PMID: 39703540 PMCID: PMC11658944 DOI: 10.1017/gmb.2024.8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 06/18/2024] [Indexed: 12/21/2024]
Abstract
The latest Yakult Science Study Day was held virtually on 2 November 2023. Aimed at healthcare professionals, researchers, and students, a variety of experts explored the latest gut microbiome research and what it means in practice. The morning sessions discussed the role of the microbiome in health and disease, the rapid advancements in DNA sequencing and implications for personalised nutrition, the current state of evidence on health benefits associated with fermented foods, prebiotics and probiotics and the challenges involved in interpreting research in this area. The afternoon session considered the emerging research on the microbiota-gut-brain axis in mediating effects of food on mood, the bidirectional impact of menopause on the gut microbiota, and the interplay between the gut and skin with implications for the treatment of rare and common skin disorders. The session ended with an update on the use of faecal microbiota transplant in both research and clinical practice. Undoubtedly, the gut microbiome is emerging as a key conductor of human health, both in relation to gastrointestinal and non-gastrointestinal outcomes. As research continues to elucidate mechanisms of action and confirm their effects in human trials, the gut microbiome should be a key consideration within a holistic approach to health moving forward.
Collapse
Affiliation(s)
- Yvonne E. Finnegan
- Yvonne Finnegan FINNE Nutrition & Regulatory Consultancy, Kilkenny, Ireland
| | | | | | - Bruno Pot
- Yakult Europe BV, Science Department, Almere, The Netherlands
| |
Collapse
|
12
|
Wang K, Zhang S, Wang Y, Wu X, Wen L, Meng T, Jin X, Li S, Hong Y, Ke J, Xu Y, Yuan H, Hu F. Taprenepag restores maternal-fetal interface homeostasis for the treatment of neurodevelopmental disorders. J Neuroinflammation 2024; 21:307. [PMID: 39609821 PMCID: PMC11603931 DOI: 10.1186/s12974-024-03300-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 11/16/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND AND PURPOSE Neurodevelopmental disorders (NDDs) are characterized by abnormalities in brain development and neurobehaviors, including autism. The maternal-fetal interface (MFI) is a highly specialized tissue through which maternal factors affect fetal brain development. However, limited research exists on restoring and maintaining MFI homeostasis and its potential impact on NDDs. This study explores the role of placental indoleamine 2,3-dioxygenase (IDO-1) in MFI homeostasis and fetal brain development. EXPERIMENTAL APPROACH The maternal-fetal barrier was disrupted by sodium valproate (VPA) in pregnant mice, whose offspring show typical autism-like behaviors. Ultrastructural analysis and flow cytometric analysis were conducted to observe the morphological and immune system changes. Behavioral tests and immunofluorescence staining was used to investigate the ability and mechanism of taprenepag to alleviate the abnormal behaviors of VPA-exposed offspring and normalize the development of serotonergic neurons. KEY RESULTS In VPA-exposed pregnant mice, the downregulation of IDO-1 led to maternal immune overactivation and disruption of maternal-fetal barrier, resulting in excessive 5-HT synthesis in the placenta. This process disrupted the development of the serotonergic neuronal system in the offspring, resulting in impaired development of serotonergic neurons, thalamocortical axons, and NDDs in the progeny. However, a single injection of taprenepag at E13.5 ultimately upregulated placental IDO-1 through amplifying the positive feedback loop COX-2/PGE2/PTGER-2/IDO-1 and abolished these alterations. CONCLUSION Taprenepag improved autism-like behaviors in the offspring of VPA-exposed mice by addressing placental IDO-1 downregulation. This study highlights the potential of targeting IDO-1 to mitigate MFI disruption and NDD development.
Collapse
Affiliation(s)
- Kai Wang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, PR China
| | - Shufen Zhang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China
| | - Yunxia Wang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China
| | - Xiaomei Wu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China
| | - Lijuan Wen
- Department of Pharmacy, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, PR China
| | - Tingting Meng
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, PR China
| | - Xiangyu Jin
- Department of Pharmacy, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, PR China
| | - Sufen Li
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China
| | - Yiling Hong
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China
| | - Jia Ke
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China
| | - Yichong Xu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China
| | - Hong Yuan
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, PR China
| | - Fuqiang Hu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China.
- Jinhua Institute of Zhejiang University, Jinhua, 321299, PR China.
- National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, College of Pharmacy, Gannan Medical University, Ganzhou, 341000, PR China.
| |
Collapse
|
13
|
Mihailovich M, Tolinački M, Soković Bajić S, Lestarevic S, Pejovic-Milovancevic M, Golić N. The Microbiome-Genetics Axis in Autism Spectrum Disorders: A Probiotic Perspective. Int J Mol Sci 2024; 25:12407. [PMID: 39596472 PMCID: PMC11594817 DOI: 10.3390/ijms252212407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/15/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
Autism spectrum disorder (commonly known as autism) is a complex and prevalent neurodevelopmental condition characterized by challenges in social behavior, restricted interests, and repetitive behaviors. It is projected that the annual cost of autism spectrum disorder in the US will reach USD 461 billion by 2025. However, despite being a major public health problem, effective treatment for the underlying symptoms remains elusive. As numerous literature data indicate the role of gut microbiota in autism prognosis, particularly in terms of alleviating gastrointestinal (GI) symptoms, high hopes have been placed on probiotics for autism treatment. Approximately twenty clinical studies have been conducted using single or mixed probiotic cultures. However, unequivocal results on the effect of probiotics on people with autism have not been obtained. The small sample sizes, differences in age of participants, choice of probiotics, dose and duration of treatment, outcome measures, and analytical methods used are largely inconsistent, making it challenging to draw distinctive conclusions. Here, we discuss the experimental evidence for specific gut bacteria and their metabolites and how they affect autism in light of the phenotypic and etiological complexity and heterogeneity. We propose a personalized medicine approach for using probiotics to increase the quality of life of individuals with autism by selecting specific probiotics to improve particular features of the condition.
Collapse
Affiliation(s)
- Marija Mihailovich
- Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, 11042 Belgrade, Serbia; (M.T.); (S.S.B.); (N.G.)
- Human Technopole, 20157 Milan, Italy
| | - Maja Tolinački
- Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, 11042 Belgrade, Serbia; (M.T.); (S.S.B.); (N.G.)
| | - Svetlana Soković Bajić
- Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, 11042 Belgrade, Serbia; (M.T.); (S.S.B.); (N.G.)
| | - Sanja Lestarevic
- Institute of Mental Health, 11000 Belgrade, Serbia; (S.L.); (M.P.-M.)
| | - Milica Pejovic-Milovancevic
- Institute of Mental Health, 11000 Belgrade, Serbia; (S.L.); (M.P.-M.)
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Nataša Golić
- Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, 11042 Belgrade, Serbia; (M.T.); (S.S.B.); (N.G.)
| |
Collapse
|
14
|
Manghi P, Filosi M, Zolfo M, Casten LG, Garcia-Valiente A, Mattevi S, Heidrich V, Golzato D, Perini S, Thomas AM, Montalbano S, Cancellieri S, Waldron L, Hall JB, Xu S, Volfovsky N, Green Snyder L, Feliciano P, Asnicar F, Valles-Colomer M, Michaelson JJ, Segata N, Domenici E. Large-scale metagenomic analysis of oral microbiomes reveals markers for autism spectrum disorders. Nat Commun 2024; 15:9743. [PMID: 39528484 PMCID: PMC11555315 DOI: 10.1038/s41467-024-53934-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
The link between the oral microbiome and neurodevelopmental disorders remains a compelling hypothesis, still requiring confirmation in large-scale datasets. Leveraging over 7000 whole-genome sequenced salivary samples from 2025 US families with children diagnosed with autism spectrum disorders (ASD), our cross-sectional study shows that the oral microbiome composition can discriminate ASD subjects from neurotypical siblings (NTs, AUC = 0.66), with 108 differentiating species (q < 0.005). The relative abundance of these species is highly correlated with cognitive impairment as measured by Full-Scale Intelligence Quotient (IQ). ASD children with IQ < 70 also exhibit lower microbiome strain sharing with parents (p < 10-6) with respect to NTs. A two-pronged functional enrichment analysis suggests the contribution of enzymes from the serotonin, GABA, and dopamine degradation pathways to the distinct microbial community compositions observed between ASD and NT samples. Although measures of restrictive eating diet and proxies of oral hygiene show relatively minor effects on the microbiome composition, the observed associations with ASD and IQ may still represent unaccounted-for underlying differences in lifestyle among groups. While causal relationships could not be established, our study provides substantial support to the investigation of oral microbiome biomarkers in ASD.
Collapse
Affiliation(s)
- Paolo Manghi
- Department CIBIO, University of Trento, Trento, Italy.
- Computational Biology Unit, Research and Innovation Centre, Fondazione Edmund Mach, via Mach 1, 38098, San Michele all'Adige, Italy.
| | - Michele Filosi
- Department CIBIO, University of Trento, Trento, Italy
- EURAC Research Institute for Biomedicine BIO, Bolzano, Italy
| | - Moreno Zolfo
- Department CIBIO, University of Trento, Trento, Italy
- Okinawa Institute of Science and Technology (OIST), Okinawa, Japan
| | - Lucas G Casten
- Department of Psychiatry, University of Iowa, Iowa city, IA, USA
| | | | - Stefania Mattevi
- Department CIBIO, University of Trento, Trento, Italy
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | | | | | - Samuel Perini
- Department CIBIO, University of Trento, Trento, Italy
| | | | - Simone Montalbano
- Department CIBIO, University of Trento, Trento, Italy
- Institute of Biological Psychiatry, Copenhagen University Hospital, Copenhagen, Denmark
| | - Samuele Cancellieri
- Department CIBIO, University of Trento, Trento, Italy
- Norwegian Center of Molecular Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Levi Waldron
- CUNY Graduate School of Public Health and Health Policy, Institute for Implementation Science in Public Health, New York, NY, USA
| | | | - Simon Xu
- Simons Foundation, New York, NY, USA
| | | | - LeeAnne Green Snyder
- Simons Foundation, New York, NY, USA
- Department of Pediatrics, Division of Genetics & Genomics, Boston Children's Hospital, Boston, MA, USA
| | - Pamela Feliciano
- Simons Foundation, New York, NY, USA
- Department of Pediatrics, Division of Genetics & Genomics, Boston Children's Hospital, Boston, MA, USA
| | | | | | | | - Nicola Segata
- Department CIBIO, University of Trento, Trento, Italy.
- IEO, European Institute of Oncology IRCCS, Milan, Italy.
| | | |
Collapse
|
15
|
Yang T, Zhang Q, Chen L, Dai Y, Jia FY, Hao Y, Li L, Zhang J, Wu LJ, Ke XY, Yi MJ, Hong Q, Chen JJ, Fang SF, Wang YC, Wang Q, Jin CH, Chen J, Li TY. Intestinal Symptoms Among Children aged 2-7 Years with Autism Spectrum Disorder in 13 Cities of China. J Autism Dev Disord 2024; 54:4302-4310. [PMID: 38060105 DOI: 10.1007/s10803-023-06122-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2023] [Indexed: 12/08/2023]
Abstract
BACKGROUND Autism spectrum disorder (ASD) is a multifactorial, pervasive, neurodevelopmental disorder, of which intestinal symptoms collectively represent one of the most common comorbidities. METHODS In this study, 1,222 children with ASD and 1,206 typically developing (TD) children aged 2-7 years were enrolled from 13 cities in China. Physical measurement and basic information questionnaires were conducted in ASD and TD children. The Childhood Autism Rating Scale (CARS), Social Responsiveness Scale (SRS), and Autism Behavior Checklist (ABC) were used to evaluate the clinical symptoms of children with ASD. The six-item Gastrointestinal Severity Index (6-GSI) was used to evaluate the prevalence of intestinal symptoms in two groups. RESULTS The detection rates of constipation, stool odor, and total intestinal symptoms in ASD children were significantly higher than those in TD children (40.098% vs. 25.622%, 17.021% vs. 9.287%, and 53.601% vs. 41.294%, respectively). Autistic children presenting with intestinal comorbidity had significantly higher scores on the ABC, SRS, CARS, and multiple subscales than autistic children without intestinal symptoms, suggesting that intestinal comorbidity may exacerbates the core symptoms of ASD children. CONCLUSION Intestinal dysfunction was significantly more common in autistic than in TD children. This dysfunction may aggravate the core symptoms of children with ASD.
Collapse
Affiliation(s)
- Ting Yang
- Chongqing Key Laboratory of Childhood Nutrition and Health, Ministry of Education Key Laboratory of Child Development and Disorders, Department of Child Health Care, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Chongqing, China
| | - Qian Zhang
- Chongqing Key Laboratory of Childhood Nutrition and Health, Ministry of Education Key Laboratory of Child Development and Disorders, Department of Child Health Care, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Chongqing, China
| | - Li Chen
- Chongqing Key Laboratory of Childhood Nutrition and Health, Ministry of Education Key Laboratory of Child Development and Disorders, Department of Child Health Care, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Chongqing, China
| | - Ying Dai
- Chongqing Key Laboratory of Childhood Nutrition and Health, Ministry of Education Key Laboratory of Child Development and Disorders, Department of Child Health Care, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Chongqing, China
| | - Fei-Yong Jia
- Department of developmental and behavioral pediatrics, the First Hospital of Jilin University, Changchun, China
| | - Yan Hao
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ling Li
- Department of Children Rehabilitation, Hainan Women and Children's Medical Center, Haikou, China
| | - Jie Zhang
- Children Health Care Center, Xi'an Children's Hospital, Xi'an, China
| | - Li-Jie Wu
- Department of Children's and Adolescent Health, Public Health College of Harbin Medical University, Harbin, China
| | - Xiao-Yan Ke
- Child mental health research center of Nanjing Brain Hospital, Nanjing, China
| | - Ming-Ji Yi
- Department of Child Health Care, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qi Hong
- Maternal and Child Health Hospital of Baoan, Shenzhen, China
| | - Jin-Jin Chen
- Department of Child Healthcare, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Shuan-Feng Fang
- Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Yi-Chao Wang
- NHC Key Laboratory of Birth Defect for Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, China
| | - Qi Wang
- Deyang Maternity & Child Healthcare Hospital, Deyang, Sichuan, China
| | - Chun-Hua Jin
- Department of Children Health Care, Capital Institute of Pediatrics, Beijing, China
| | - Jie Chen
- Chongqing Key Laboratory of Childhood Nutrition and Health, Ministry of Education Key Laboratory of Child Development and Disorders, Department of Child Health Care, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Chongqing, China.
| | - Ting-Yu Li
- Chongqing Key Laboratory of Childhood Nutrition and Health, Ministry of Education Key Laboratory of Child Development and Disorders, Department of Child Health Care, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Chongqing, China.
| |
Collapse
|
16
|
Qu S, Yu Z, Zhou Y, Wang S, Jia M, Chen T, Zhang X. Gut microbiota modulates neurotransmitter and gut-brain signaling. Microbiol Res 2024; 287:127858. [PMID: 39106786 DOI: 10.1016/j.micres.2024.127858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 06/16/2024] [Accepted: 07/22/2024] [Indexed: 08/09/2024]
Abstract
Neurotransmitters, including 5-hydroxytryptamine (5-HT), dopamine (DA), gamma-aminobutyric acid (GABA), and glutamate, are essential transductors in the Gut-Brain Axis (GBA), playing critical roles both peripherally and centrally. Accumulating evidence suggests that the gut microbiota modulates intestinal neurotransmitter metabolism and gut-to-brain signaling, shedding light on the crucial role of the gut microbiota in brain function and the pathogenesis of various neuropsychiatric diseases, such as major depression disorder (MDD), anxiety, addiction and Parkinson's disease (PD). Despite the exciting findings, the mechanisms underlying the modulation of neurotransmitter metabolism and function by the gut microbiota are still being elucidated. In this review, we aim to provide a comprehensive overview of the existing knowledge about the role of the gut microbiota in neurotransmitter metabolism and function in animal and clinical experiments. Moreover, we will discuss the potential mechanisms through which gut microbiota-derived neurotransmitters contribute to the pathogenesis of neuropsychiatric diseases, thus highlighting a novel therapeutic target for these conditions.
Collapse
Affiliation(s)
- Shiyan Qu
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, China; National Clinic Research Center for Mental Disorders, Changsha, Hunan 410000, China; National Technology Institute on Mental Disorders, Changsha, Hunan 410000, China; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan 410000, China; Mental Health Institute, Second Xiangya Hospital, Central South University, Changsha 410000, China
| | - Zijin Yu
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, China; National Clinic Research Center for Mental Disorders, Changsha, Hunan 410000, China; National Technology Institute on Mental Disorders, Changsha, Hunan 410000, China; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan 410000, China; Mental Health Institute, Second Xiangya Hospital, Central South University, Changsha 410000, China
| | - Yaxuan Zhou
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, China; National Clinic Research Center for Mental Disorders, Changsha, Hunan 410000, China; National Technology Institute on Mental Disorders, Changsha, Hunan 410000, China; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan 410000, China; Mental Health Institute, Second Xiangya Hospital, Central South University, Changsha 410000, China
| | - Shiyi Wang
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, China; National Clinic Research Center for Mental Disorders, Changsha, Hunan 410000, China; National Technology Institute on Mental Disorders, Changsha, Hunan 410000, China; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan 410000, China; Mental Health Institute, Second Xiangya Hospital, Central South University, Changsha 410000, China
| | - Minqi Jia
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, China; National Clinic Research Center for Mental Disorders, Changsha, Hunan 410000, China; National Technology Institute on Mental Disorders, Changsha, Hunan 410000, China; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan 410000, China; Mental Health Institute, Second Xiangya Hospital, Central South University, Changsha 410000, China
| | - Ti Chen
- Clinical Laboratory, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410000, China
| | - Xiaojie Zhang
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, China; National Clinic Research Center for Mental Disorders, Changsha, Hunan 410000, China; National Technology Institute on Mental Disorders, Changsha, Hunan 410000, China; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan 410000, China; Mental Health Institute, Second Xiangya Hospital, Central South University, Changsha 410000, China.
| |
Collapse
|
17
|
Prescott SL, Holton KF, Lowry CA, Nicholson JJ, Logan AC. The Intersection of Ultra-Processed Foods, Neuropsychiatric Disorders, and Neurolaw: Implications for Criminal Justice. NEUROSCI 2024; 5:354-377. [PMID: 39483285 PMCID: PMC11477939 DOI: 10.3390/neurosci5030028] [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: 08/17/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 11/03/2024] Open
Abstract
Over the last decade there has been increasing interest in the links between the consumption of ultra-processed foods and various neuropsychiatric disorders, aggression, and antisocial behavior. Neurolaw is an interdisciplinary field that seeks to translate the rapid and voluminous advances in brain science into legal decisions and policy. An enhanced understanding of biophysiological mechanisms by which ultra-processed foods influence brain and behavior allows for a historical reexamination of one of forensic neuropsychiatry's most famous cases-The People v. White and its associated 'Twinkie Defense'. Here in this Viewpoint article, we pair original court transcripts with emergent research in neurolaw, including nutritional neuroscience, microbiome sciences (legalome), pre-clinical mechanistic research, and clinical intervention trials. Advances in neuroscience, and related fields such as the microbiome, are challenging basic assumptions in the criminal justice system, including notions of universal free will. Recent dismissals of criminal charges related to auto-brewery syndrome demonstrate that courts are open to advances at the intersection of neuromicrobiology and nutritional neuroscience, including those that relate to criminal intent and diminished capacity. As such, it is our contention that experts in the neurosciences will play an increasing role in shaping research that underpins 21st-century courtroom discourse, policy, and decision-making.
Collapse
Affiliation(s)
- Susan L Prescott
- Nova Institute for Health, Baltimore, MD 21231, USA;
- School of Medicine, University of Western Australia, Perth, WA 6009, Australia
- Department of Family and Community Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Kathleen F Holton
- Departments of Health Studies and Neuroscience, American University, Washington, DC 20016, USA;
| | - Christopher A Lowry
- Department of Integrative Physiology, Department of Psychology and Neuroscience, Center for Neuroscience, and Center for Microbial Exploration, University of Colorado Boulder, Boulder, CO 80309, USA;
| | - Jeffrey J Nicholson
- Law and Government, Humber College Institute of Technology & Advanced Learning, Toronto, ON M9W 5L7, Canada;
| | - Alan C Logan
- Nova Institute for Health, Baltimore, MD 21231, USA;
| |
Collapse
|
18
|
Cannon M, Toma R, Ganeshan S, de Jesus Alvarez Varela E, Vuyisich M, Banavar G. Salivary Transcriptome and Mitochondrial Analysis of Autism Spectrum Disorder Children Compared to Healthy Controls. NEUROSCI 2024; 5:276-290. [PMID: 39483288 PMCID: PMC11467968 DOI: 10.3390/neurosci5030022] [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: 05/13/2024] [Revised: 07/22/2024] [Accepted: 07/28/2024] [Indexed: 11/03/2024] Open
Abstract
Autism rates have been reported to be increasing rapidly in industrialized societies. The pathology most often combines neurological symptoms associated with language and social impairments with gastrointestinal symptoms. This study aimed to measure differences in oral metatranscriptome and mitochondrial health between ASD children and neurotypical USA and Colombia ("Blue Zone") children. In addition, this study aimed to determine whether using prebiotics and probiotics would change the oral microbiome and mitochondrial health of ASD children. Buccal swabs and saliva samples were obtained from 30 autistic individuals (USA) at three intervals: prior to intervention, post-prebiotic, and post-probiotic. In addition, a subject component who were neurotypical, which included individuals from the USA (30) and Colombia (30), had buccal swabbing and salivary sampling performed for metatranscriptomic and mitochondrial comparison. Significant differences were observed in the temporal data, demonstrating shifts that interventions with probiotics and polyols may have precipitated. Particular bacterial strains were significantly more prevalent in the autism group, including a strain that reduced neurotransmitter levels via enzymatic degradation. This supports the hypothesis that the microbiome may influence the occurrence and degree of autism. Verbal skills increased in six of the 30 ASD subjects following xylitol and three more after probiotic supplementation, according to both parental reports and the subjects' healthcare providers.
Collapse
Affiliation(s)
- Mark Cannon
- Ann and Robert Lurie Children’s Hospital, Northwestern University, Chicago, IL 60611, USA
| | - Ryan Toma
- Viome Research Institute, Los Alamos, NM 98011, USA (M.V.); (G.B.)
| | - Sri Ganeshan
- MITOSWAB Religen Labs, Plymouth Meeting, PA 19462, USA
| | | | | | - Guruduth Banavar
- Viome Research Institute, Los Alamos, NM 98011, USA (M.V.); (G.B.)
| |
Collapse
|
19
|
He Y, Wang K, Su N, Yuan C, Zhang N, Hu X, Fu Y, Zhao F. Microbiota-gut-brain axis in health and neurological disease: Interactions between gut microbiota and the nervous system. J Cell Mol Med 2024; 28:e70099. [PMID: 39300699 PMCID: PMC11412916 DOI: 10.1111/jcmm.70099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 09/03/2024] [Accepted: 09/09/2024] [Indexed: 09/22/2024] Open
Abstract
Along with mounting evidence that gut microbiota and their metabolites migrate endogenously to distal organs, the 'gut-lung axis,' 'gut-brain axis,' 'gut-liver axis' and 'gut-renal axis' have been established. Multiple animal recent studies have demonstrated gut microbiota may also be a key susceptibility factor for neurological disorders such as Alzheimer's disease, Parkinson's disease and autism. The gastrointestinal tract is innervated by the extrinsic sympathetic and vagal nerves and the intrinsic enteric nervous system, and the gut microbiota interacts with the nervous system to maintain homeostatic balance in the host gut. A total of 1507 publications on the interactions between the gut microbiota, the gut-brain axis and neurological disorders are retrieved from the Web of Science to investigate the interactions between the gut microbiota and the nervous system and the underlying mechanisms involved in normal and disease states. We provide a comprehensive overview of the effects of the gut microbiota and its metabolites on nervous system function and neurotransmitter secretion, as well as alterations in the gut microbiota in neurological disorders, to provide a basis for the possibility of targeting the gut microbiota as a therapeutic agent for neurological disorders.
Collapse
Affiliation(s)
- Yuhong He
- Department of Operating RoomChina‐Japan Union Hospital of Jilin UniversityChangchunJilinChina
- Department of Clinical Veterinary MedicineCollege of Veterinary Medicine, Jilin UniversityChangchunJilinChina
| | - Ke Wang
- Department of Operating RoomChina‐Japan Union Hospital of Jilin UniversityChangchunJilinChina
| | - Niri Su
- Department of Clinical Veterinary MedicineCollege of Veterinary Medicine, Jilin UniversityChangchunJilinChina
| | - Chongshan Yuan
- Department of Clinical Veterinary MedicineCollege of Veterinary Medicine, Jilin UniversityChangchunJilinChina
| | - Naisheng Zhang
- Department of Clinical Veterinary MedicineCollege of Veterinary Medicine, Jilin UniversityChangchunJilinChina
| | - Xiaoyu Hu
- Department of Clinical Veterinary MedicineCollege of Veterinary Medicine, Jilin UniversityChangchunJilinChina
| | - Yunhe Fu
- Department of Clinical Veterinary MedicineCollege of Veterinary Medicine, Jilin UniversityChangchunJilinChina
| | - Feng Zhao
- Department of Operating RoomChina‐Japan Union Hospital of Jilin UniversityChangchunJilinChina
| |
Collapse
|
20
|
Prince N, Peralta Marzal LN, Markidi A, Ahmed S, Adolfs Y, Pasterkamp RJ, Kumar H, Roeselers G, Garssen J, Kraneveld AD, Perez-Pardo P. Prebiotic diet normalizes aberrant immune and behavioral phenotypes in a mouse model of autism spectrum disorder. Acta Pharmacol Sin 2024; 45:1591-1603. [PMID: 38589690 PMCID: PMC11272935 DOI: 10.1038/s41401-024-01268-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 03/19/2024] [Indexed: 04/10/2024]
Abstract
Autism spectrum disorder (ASD) is a cluster of neurodevelopmental disorders characterized by deficits in communication and behavior. Increasing evidence suggests that the microbiota-gut-brain axis and the likely related immune imbalance may play a role in the development of this disorder. Gastrointestinal deficits and gut microbiota dysfunction have been linked to the development or severity of autistic behavior. Therefore, treatments that focus on specific diets may improve gastrointestinal function and aberrant behavior in individuals with ASD. In this study, we investigated whether a diet containing specific prebiotic fibers, namely, 3% galacto-oligosaccharide/fructo-oligosaccharide (GOS/FOS; 9:1), can mitigate the adverse effects of in utero exposure to valproic acid (VPA) in mice. Pregnant BALB/cByJ dams were injected with VPA (600 mg/kg, sc.) or phosphate-buffered saline (PBS) on gestational day 11 (G11). Male offspring were divided into four groups: (1) in utero PBS-exposed with a control diet, (2) in utero PBS-exposed with GOS/FOS diet, (3) in utero VPA-exposed with a control diet, and (4) in utero VPA-exposed with GOS/FOS diet. Dietary intervention started from birth and continued throughout the duration of the experiment. We showed that the prebiotic diet normalized VPA-induced alterations in male offspring, including restoration of key microbial taxa, intestinal permeability, peripheral immune homeostasis, reduction of neuroinflammation in the cerebellum, and impairments in social behavior and cognition in mice. Overall, our research provides valuable insights into the gut-brain axis involvement in ASD development. In addition, dietary interventions might correct the disbalance in gut microbiota and immune responses and, ultimately, might improve detrimental behavioral outcomes in ASD.
Collapse
Affiliation(s)
- Naika Prince
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Lucia N Peralta Marzal
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Anastasia Markidi
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, The Netherlands
- Division of Cell Biology, Metabolism & Cancer, Department of Biomolecular Health Sciences, Utrecht University, 3584 CL, Utrecht, The Netherlands
| | - Sabbir Ahmed
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Youri Adolfs
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Himanshu Kumar
- Danone Nutricia Research, 3584 CT, Utrecht, The Netherlands
| | - Guus Roeselers
- Danone Nutricia Research, 3584 CT, Utrecht, The Netherlands
| | - Johan Garssen
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, The Netherlands
- Danone Nutricia Research, 3584 CT, Utrecht, The Netherlands
| | - Aletta D Kraneveld
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, The Netherlands
- Department of Neuroscience, Faculty of Science, VU university, 1081 HV, Amsterdam, The Netherlands
| | - Paula Perez-Pardo
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, The Netherlands.
| |
Collapse
|
21
|
You M, Chen N, Yang Y, Cheng L, He H, Cai Y, Liu Y, Liu H, Hong G. The gut microbiota-brain axis in neurological disorders. MedComm (Beijing) 2024; 5:e656. [PMID: 39036341 PMCID: PMC11260174 DOI: 10.1002/mco2.656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 06/15/2024] [Accepted: 06/17/2024] [Indexed: 07/23/2024] Open
Abstract
Previous studies have shown a bidirectional communication between human gut microbiota and the brain, known as the microbiota-gut-brain axis (MGBA). The MGBA influences the host's nervous system development, emotional regulation, and cognitive function through neurotransmitters, immune modulation, and metabolic pathways. Factors like diet, lifestyle, genetics, and environment shape the gut microbiota composition together. Most research have explored how gut microbiota regulates host physiology and its potential in preventing and treating neurological disorders. However, the individual heterogeneity of gut microbiota, strains playing a dominant role in neurological diseases, and the interactions of these microbial metabolites with the central/peripheral nervous systems still need exploration. This review summarizes the potential role of gut microbiota in driving neurodevelopmental disorders (autism spectrum disorder and attention deficit/hyperactivity disorder), neurodegenerative diseases (Alzheimer's and Parkinson's disease), and mood disorders (anxiety and depression) in recent years and discusses the current clinical and preclinical gut microbe-based interventions, including dietary intervention, probiotics, prebiotics, and fecal microbiota transplantation. It also puts forward the current insufficient research on gut microbiota in neurological disorders and provides a framework for further research on neurological disorders.
Collapse
Affiliation(s)
- Mingming You
- Xiamen Key Laboratory of Genetic TestingThe Department of Laboratory MedicineThe First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen UniversityXiamenChina
| | - Nan Chen
- Master of Public HealthSchool of Public HealthXiamen UniversityXiamenChina
| | - Yuanyuan Yang
- Xiamen Key Laboratory of Genetic TestingThe Department of Laboratory MedicineThe First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen UniversityXiamenChina
| | - Lingjun Cheng
- Xiamen Key Laboratory of Genetic TestingThe Department of Laboratory MedicineThe First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen UniversityXiamenChina
| | - Hongzhang He
- Xiamen Key Laboratory of Genetic TestingThe Department of Laboratory MedicineThe First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen UniversityXiamenChina
| | - Yanhua Cai
- Master of Public HealthSchool of Public HealthXiamen UniversityXiamenChina
| | - Yating Liu
- Xiamen Key Laboratory of Genetic TestingThe Department of Laboratory MedicineThe First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen UniversityXiamenChina
| | - Haiyue Liu
- Xiamen Key Laboratory of Genetic TestingThe Department of Laboratory MedicineThe First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen UniversityXiamenChina
| | - Guolin Hong
- Xiamen Key Laboratory of Genetic TestingThe Department of Laboratory MedicineThe First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen UniversityXiamenChina
| |
Collapse
|
22
|
Logan AC, Prescott SL, LaFata EM, Nicholson JJ, Lowry CA. Beyond Auto-Brewery: Why Dysbiosis and the Legalome Matter to Forensic and Legal Psychology. LAWS 2024; 13:46. [DOI: 10.3390/laws13040046] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
Abstract
International studies have linked the consumption of ultra-processed foods with a variety of non-communicable diseases. Included in this growing body of research is evidence linking ultra-processed foods to mental disorders, aggression, and antisocial behavior. Although the idea that dietary patterns and various nutrients or additives can influence brain and behavior has a long history in criminology, in the absence of plausible mechanisms and convincing intervention trials, the topic was mostly excluded from mainstream discourse. The emergence of research across nutritional neuroscience and nutritional psychology/psychiatry, combined with mechanistic bench science, and human intervention trials, has provided support to epidemiological findings, and legitimacy to the concept of nutritional criminology. Among the emergent research, microbiome sciences have illuminated mechanistic pathways linking various socioeconomic and environmental factors, including the consumption of ultra-processed foods, with aggression and antisocial behavior. Here in this review, we examine this burgeoning research, including that related to ultra-processed food addiction, and explore its relevance across the criminal justice spectrum—from prevention to intervention—and in courtroom considerations of diminished capacity. We use auto-brewery syndrome as an example of intersecting diet and gut microbiome science that has been used to refute mens rea in criminal charges. The legalome—microbiome and omics science applied in forensic and legal psychology—appears set to emerge as an important consideration in matters of criminology, law, and justice.
Collapse
Affiliation(s)
| | - Susan L. Prescott
- Nova Institute for Health, Baltimore, MD 21231, USA
- School of Medicine, University of Western Australia, Perth, WA 6009, Australia
- Department of Family and Community Medicine, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
| | - Erica M. LaFata
- Center for Weight, Eating, and Lifestyle Science, Drexel University, 3141 Chestnut St, Philadelphia, PA 19104, USA
| | | | - Christopher A. Lowry
- Departments of Integrative Physiology and Psychology and Neuroscience, Center for Neuroscience and Center for Microbial Exploration, University of Colorado Boulder, Boulder, CO 80309, USA
| |
Collapse
|
23
|
Zheng L, Jiao Y, Zhong H, Tan Y, Yin Y, Liu Y, Liu D, Wu M, Wang G, Huang J, Wang P, Qin M, Wang M, Xiao Y, Lv T, Luo Y, Hu H, Hou ST, Kui L. Human-derived fecal microbiota transplantation alleviates social deficits of the BTBR mouse model of autism through a potential mechanism involving vitamin B 6 metabolism. mSystems 2024; 9:e0025724. [PMID: 38780265 PMCID: PMC11237617 DOI: 10.1128/msystems.00257-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 04/18/2024] [Indexed: 05/25/2024] Open
Abstract
Autism spectrum disorder (ASD) is a heterogeneous neurodevelopmental condition characterized by social communication deficiencies and stereotypic behaviors influenced by hereditary and/or environmental risk factors. There are currently no approved medications for treating the core symptoms of ASD. Human fecal microbiota transplantation (FMT) has emerged as a potential intervention to improve autistic symptoms, but the underlying mechanisms are not fully understood. In this study, we evaluated the effects of human-derived FMT on behavioral and multi-omics profiles of the BTBR mice, an established model for ASD. FMT effectively alleviated the social deficits in the BTBR mice and normalized their distinct plasma metabolic profile, notably reducing the elevated long-chain acylcarnitines. Integrative analysis linked these phenotypic changes to specific Bacteroides species and vitamin B6 metabolism. Indeed, vitamin B6 supplementation improved the social behaviors in BTBR mice. Collectively, these findings shed new light on the interplay between FMT and vitamin B6 metabolism and revealed a potential mechanism underlying the therapeutic role of FMT in ASD.IMPORTANCEAccumulating evidence supports the beneficial effects of human fecal microbiota transplantation (FMT) on symptoms associated with autism spectrum disorder (ASD). However, the precise mechanism by which FMT induces a shift in the microbiota and leads to symptom improvement remains incompletely understood. This study integrated data from colon-content metagenomics, colon-content metabolomics, and plasma metabolomics to investigate the effects of FMT treatment on the BTBR mouse model for ASD. The analysis linked the amelioration of social deficits following FMT treatment to the restoration of mitochondrial function and the modulation of vitamin B6 metabolism. Bacterial species and compounds with beneficial roles in vitamin B6 metabolism and mitochondrial function may further contribute to improving FMT products and designing novel therapies for ASD treatment.
Collapse
Affiliation(s)
- Lifeng Zheng
- Brain Research Centre and Department of Neuroscience, Southern University of Science and Technology, Shenzhen, China
- Xbiome Co. Ltd., Shenzhen, China
| | - Yinming Jiao
- Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, China
| | - Haolin Zhong
- Brain Research Centre and Department of Neuroscience, Southern University of Science and Technology, Shenzhen, China
| | - Yan Tan
- Xbiome Co. Ltd., Shenzhen, China
| | | | | | - Ding Liu
- Xbiome Co. Ltd., Shenzhen, China
| | - Manli Wu
- Xbiome Co. Ltd., Shenzhen, China
| | - Guoyun Wang
- Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, China
| | | | - Ping Wang
- Shenzhen Institute for Drug Control, Shenzhen, China
| | - Meirong Qin
- Shenzhen Institute for Drug Control, Shenzhen, China
| | - Mingbang Wang
- Microbiome Therapy Center, South China Hospital, Medical School, Shenzhen University, Shenzhen, China
- Shanghai Key Laboratory of Birth Defects, Division of Neonatology, Children’s Hospital of Fudan University, National Center for Children’s Health, Shanghai, China
| | - Yang Xiao
- Department of Hematology, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, China
| | - Tiying Lv
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yangzi Luo
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Han Hu
- Xbiome Co. Ltd., Shenzhen, China
| | - Sheng-Tao Hou
- Brain Research Centre and Department of Neuroscience, Southern University of Science and Technology, Shenzhen, China
| | - Ling Kui
- Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, China
| |
Collapse
|
24
|
Laue HE, Bonham KS, Coker MO, Moroishi Y, Pathmasiri W, McRitchie S, Sumner S, Hoen AG, Karagas MR, Klepac-Ceraj V, Madan JC. Prospective association of the infant gut microbiome with social behaviors in the ECHO consortium. Mol Autism 2024; 15:21. [PMID: 38760865 PMCID: PMC11101342 DOI: 10.1186/s13229-024-00597-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 04/11/2024] [Indexed: 05/19/2024] Open
Abstract
BACKGROUND Identifying modifiable risk factors of autism spectrum disorders (ASDs) may inform interventions to reduce financial burden. The infant/toddler gut microbiome is one such feature that has been associated with social behaviors, but results vary between cohorts. We aimed to identify consistent overall and sex-specific associations between the early-life gut microbiome and autism-related behaviors. METHODS Utilizing the Environmental influences on Children Health Outcomes (ECHO) consortium of United States (U.S.) pediatric cohorts, we gathered data on 304 participants with fecal metagenomic sequencing between 6-weeks to 2-years postpartum (481 samples). ASD-related social development was assessed with the Social Responsiveness Scale (SRS-2). Linear regression, PERMANOVA, and Microbiome Multivariable Association with Linear Models (MaAsLin2) were adjusted for sociodemographic factors. Stratified models estimated sex-specific effects. RESULTS Genes encoding pathways for synthesis of short-chain fatty acids were associated with higher SRS-2 scores, indicative of ASDs. Fecal concentrations of butyrate were also positively associated with ASD-related SRS-2 scores, some of which may be explained by formula use. LIMITATIONS The distribution of age at outcome assessment differed in the cohorts included, potentially limiting comparability between cohorts. Stool sample collection methods also differed between cohorts. Our study population reflects the general U.S. population, and thus includes few participants who met the criteria for being at high risk of developing ASD. CONCLUSIONS Our study is among the first multicenter studies in the U.S. to describe prospective microbiome development from infancy in relation to neurodevelopment associated with ASDs. Our work contributes to clarifying which microbial features associate with subsequent diagnosis of neuropsychiatric outcomes. This will allow for future interventional research targeting the microbiome to change neurodevelopmental trajectories.
Collapse
Affiliation(s)
- Hannah E Laue
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - Kevin S Bonham
- Department of Biological Sciences, Wellesley College, 106 Central Street, Wellesley, MA, 02481, USA
| | - Modupe O Coker
- School of Dental Medicine, Rutgers University, Newark, NJ, USA
| | - Yuka Moroishi
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - Wimal Pathmasiri
- Department of Nutrition, Nutrition Research Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Susan McRitchie
- Department of Nutrition, Nutrition Research Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Susan Sumner
- Department of Nutrition, Nutrition Research Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Anne G Hoen
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - Margaret R Karagas
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - Vanja Klepac-Ceraj
- Department of Biological Sciences, Wellesley College, 106 Central Street, Wellesley, MA, 02481, USA.
| | - Juliette C Madan
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Hanover, NH, USA.
- Departments of Pediatrics and Psychiatry, One Medical Center Drive, Dartmouth Hitchcock Medical Center, Lebanon, NH, 03756, USA.
| |
Collapse
|
25
|
Zhuang H, Liang Z, Ma G, Qureshi A, Ran X, Feng C, Liu X, Yan X, Shen L. Autism spectrum disorder: pathogenesis, biomarker, and intervention therapy. MedComm (Beijing) 2024; 5:e497. [PMID: 38434761 PMCID: PMC10908366 DOI: 10.1002/mco2.497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 01/27/2024] [Accepted: 01/30/2024] [Indexed: 03/05/2024] Open
Abstract
Autism spectrum disorder (ASD) has become a common neurodevelopmental disorder. The heterogeneity of ASD poses great challenges for its research and clinical translation. On the basis of reviewing the heterogeneity of ASD, this review systematically summarized the current status and progress of pathogenesis, diagnostic markers, and interventions for ASD. We provided an overview of the ASD molecular mechanisms identified by multi-omics studies and convergent mechanism in different genetic backgrounds. The comorbidities, mechanisms associated with important physiological and metabolic abnormalities (i.e., inflammation, immunity, oxidative stress, and mitochondrial dysfunction), and gut microbial disorder in ASD were reviewed. The non-targeted omics and targeting studies of diagnostic markers for ASD were also reviewed. Moreover, we summarized the progress and methods of behavioral and educational interventions, intervention methods related to technological devices, and research on medical interventions and potential drug targets. This review highlighted the application of high-throughput omics methods in ASD research and emphasized the importance of seeking homogeneity from heterogeneity and exploring the convergence of disease mechanisms, biomarkers, and intervention approaches, and proposes that taking into account individuality and commonality may be the key to achieve accurate diagnosis and treatment of ASD.
Collapse
Affiliation(s)
- Hongbin Zhuang
- College of Life Science and OceanographyShenzhen UniversityShenzhenP. R. China
| | - Zhiyuan Liang
- College of Life Science and OceanographyShenzhen UniversityShenzhenP. R. China
| | - Guanwei Ma
- College of Life Science and OceanographyShenzhen UniversityShenzhenP. R. China
| | - Ayesha Qureshi
- College of Life Science and OceanographyShenzhen UniversityShenzhenP. R. China
| | - Xiaoqian Ran
- College of Life Science and OceanographyShenzhen UniversityShenzhenP. R. China
| | - Chengyun Feng
- Maternal and Child Health Hospital of BaoanShenzhenP. R. China
| | - Xukun Liu
- College of Life Science and OceanographyShenzhen UniversityShenzhenP. R. China
| | - Xi Yan
- College of Life Science and OceanographyShenzhen UniversityShenzhenP. R. China
| | - Liming Shen
- College of Life Science and OceanographyShenzhen UniversityShenzhenP. R. China
- Shenzhen‐Hong Kong Institute of Brain Science‐Shenzhen Fundamental Research InstitutionsShenzhenP. R. China
| |
Collapse
|
26
|
Hung LY, Margolis KG. Autism spectrum disorders and the gastrointestinal tract: insights into mechanisms and clinical relevance. Nat Rev Gastroenterol Hepatol 2024; 21:142-163. [PMID: 38114585 DOI: 10.1038/s41575-023-00857-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/11/2023] [Indexed: 12/21/2023]
Abstract
Autism spectrum disorders (ASDs) are recognized as central neurodevelopmental disorders diagnosed by impairments in social interactions, communication and repetitive behaviours. The recognition of ASD as a central nervous system (CNS)-mediated neurobehavioural disorder has led most of the research in ASD to be focused on the CNS. However, gastrointestinal function is also likely to be affected owing to the neural mechanistic nature of ASD and the nervous system in the gastrointestinal tract (enteric nervous system). Thus, it is unsurprising that gastrointestinal disorders, particularly constipation, diarrhoea and abdominal pain, are highly comorbid in individuals with ASD. Gastrointestinal problems have also been repeatedly associated with increased severity of the core symptoms diagnostic of ASD and other centrally mediated comorbid conditions, including psychiatric issues, irritability, rigid-compulsive behaviours and aggression. Despite the high prevalence of gastrointestinal dysfunction in ASD and its associated behavioural comorbidities, the specific links between these two conditions have not been clearly delineated, and current data linking ASD to gastrointestinal dysfunction have not been extensively reviewed. This Review outlines the established and emerging clinical and preclinical evidence that emphasizes the gut as a novel mechanistic and potential therapeutic target for individuals with ASD.
Collapse
Affiliation(s)
- Lin Y Hung
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, USA
| | - Kara Gross Margolis
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, USA.
- Department of Cell Biology, NYU Grossman School of Medicine and Langone Medical Center, New York, NY, USA.
- Department of Pediatrics, NYU Grossman School of Medicine and Langone Medical Center, New York, NY, USA.
| |
Collapse
|
27
|
Miao Z, Chen L, Zhang Y, Zhang J, Zhang H. Bifidobacterium animalis subsp. lactis Probio-M8 alleviates abnormal behavior and regulates gut microbiota in a mouse model suffering from autism. mSystems 2024; 9:e0101323. [PMID: 38108654 PMCID: PMC10804959 DOI: 10.1128/msystems.01013-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 11/08/2023] [Indexed: 12/19/2023] Open
Abstract
Probiotics can effectively improve a variety of neurological diseases, but there is little research on autism, and the specific mechanism is unclear. In this study, shotgun metagenomics analysis was used to investigate the preventive and therapeutic effects of Bifidobacterium animalis subsp. lactis Probio-M8 on autism. The results showed that Probio-M8 treatment significantly alleviated valproate (VPA)-induced autism in mice, with autistic symptoms characterized by increased stereotyped behaviors such as grooming, reduced learning ability, and decreased desire to socialize. Further studies have found that Probio-M8 can alleviate autism by optimizing gut microbiota diversity and regulating metabolic levels. Probio-M8 regulates gut microbiota structure by increasing the abundance of beneficial bacteria such as Bifidobacterium globosum and Akkermansia muciniphila. In addition, Probio-M8 regulates metabolic activity by increasing levels of choline, which corrects CAZy disorders. In conclusion, Probio-M8 is therapeutic in the VPA-induced autism mouse model by regulating the gut microbiome and metabolic levels.IMPORTANCEIndividuals with autism often exhibit symptoms of social invariance, obsessive-compulsive tendencies, and repetitive behaviors. However, early intervention and treatment can be effective in improving social skills and mitigating autism symptoms, including behaviors related to irritability. Although taking medication for autism may lead to side effects such as weight gain, probiotics can be an ideal intervention for alleviating these symptoms. In this study, we investigated the effects of Probio-M8 intervention on the behavior of autistic mice using an open-field test, a three-chamber sociability test, and a novel object recognition test. Metagenomic analysis revealed differences in gut microbiota diversity among groups, predicted changes in metabolite levels, and functionally annotated CAZy. Additionally, we analyzed serum neurotransmitter levels and found that probiotics were beneficial in mitigating neurotransmitter imbalances in mice with autism.
Collapse
Affiliation(s)
- Zhuangzhuang Miao
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Lin Chen
- School of Food Science and Engineering, Hainan University, Haikou, China
| | - Yong Zhang
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing (USTB), Beijing, China
| | - Jiachao Zhang
- School of Food Science and Engineering, Hainan University, Haikou, China
| | - Heping Zhang
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| |
Collapse
|
28
|
Gonçalves CL, Doifode T, Rezende VL, Costa MA, Rhoads JM, Soutullo CA. The many faces of microbiota-gut-brain axis in autism spectrum disorder. Life Sci 2024; 337:122357. [PMID: 38123016 DOI: 10.1016/j.lfs.2023.122357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/02/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023]
Abstract
The gut-brain axis is gaining more attention in neurodevelopmental disorders, especially autism spectrum disorder (ASD). Many factors can influence microbiota in early life, including host genetics and perinatal events (infections, mode of birth/delivery, medications, nutritional supply, and environmental stressors). The gut microbiome can influence blood-brain barrier (BBB) permeability, drug bioavailability, and social behaviors. Developing microbiota-based interventions such as probiotics, gastrointestinal (GI) microbiota transplantation, or metabolite supplementation may offer an exciting approach to treating ASD. This review highlights that RNA sequencing, metabolomics, and transcriptomics data are needed to understand how microbial modulators can influence ASD pathophysiology. Due to the substantial clinical heterogeneity of ASD, medical caretakers may be unlikely to develop a broad and effective general gut microbiota modulator. However, dietary modulation followed by administration of microbiota modulators is a promising option for treating ASD-related behavioral and gastrointestinal symptoms. Future work should focus on the accuracy of biomarker tests and developing specific psychobiotic agents tailored towards the gut microbiota seen in ASD patients, which may include developing individualized treatment options.
Collapse
Affiliation(s)
- Cinara L Gonçalves
- Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil.
| | - Tejaswini Doifode
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health (UTHealth), Houston, TX, USA
| | - Victoria L Rezende
- Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Maiara A Costa
- Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - J Marc Rhoads
- Department of Pediatrics, Division of Pediatric Gastroenterology, McGovern Medical School, The University of Texas Health (UTHealth), Houston, TX, USA
| | - Cesar A Soutullo
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health (UTHealth), Houston, TX, USA
| |
Collapse
|
29
|
Peralta-Marzal LN, Rojas-Velazquez D, Rigters D, Prince N, Garssen J, Kraneveld AD, Perez-Pardo P, Lopez-Rincon A. A robust microbiome signature for autism spectrum disorder across different studies using machine learning. Sci Rep 2024; 14:814. [PMID: 38191575 PMCID: PMC10774349 DOI: 10.1038/s41598-023-50601-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 12/21/2023] [Indexed: 01/10/2024] Open
Abstract
Autism spectrum disorder (ASD) is a highly complex neurodevelopmental disorder characterized by deficits in sociability and repetitive behaviour, however there is a great heterogeneity within other comorbidities that accompany ASD. Recently, gut microbiome has been pointed out as a plausible contributing factor for ASD development as individuals diagnosed with ASD often suffer from intestinal problems and show a differentiated intestinal microbial composition. Nevertheless, gut microbiome studies in ASD rarely agree on the specific bacterial taxa involved in this disorder. Regarding the potential role of gut microbiome in ASD pathophysiology, our aim is to investigate whether there is a set of bacterial taxa relevant for ASD classification by using a sibling-controlled dataset. Additionally, we aim to validate these results across two independent cohorts as several confounding factors, such as lifestyle, influence both ASD and gut microbiome studies. A machine learning approach, recursive ensemble feature selection (REFS), was applied to 16S rRNA gene sequencing data from 117 subjects (60 ASD cases and 57 siblings) identifying 26 bacterial taxa that discriminate ASD cases from controls. The average area under the curve (AUC) of this specific set of bacteria in the sibling-controlled dataset was 81.6%. Moreover, we applied the selected bacterial taxa in a tenfold cross-validation scheme using two independent cohorts (a total of 223 samples-125 ASD cases and 98 controls). We obtained average AUCs of 74.8% and 74%, respectively. Analysis of the gut microbiome using REFS identified a set of bacterial taxa that can be used to predict the ASD status of children in three distinct cohorts with AUC over 80% for the best-performing classifiers. Our results indicate that the gut microbiome has a strong association with ASD and should not be disregarded as a potential target for therapeutic interventions. Furthermore, our work can contribute to use the proposed approach for identifying microbiome signatures across other 16S rRNA gene sequencing datasets.
Collapse
Affiliation(s)
- Lucia N Peralta-Marzal
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, The Netherlands
| | - David Rojas-Velazquez
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, The Netherlands
- Department of Data Science, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Douwe Rigters
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, The Netherlands
| | - Naika Prince
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, The Netherlands
| | - Johan Garssen
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, The Netherlands
- Global Centre of Excellence Immunology, Danone Nutricia Research, Utrecht, The Netherlands
| | - Aletta D Kraneveld
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, The Netherlands
- Department of Neuroscience, Faculty of Science, VU University, Amsterdam, The Netherlands
| | - Paula Perez-Pardo
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, The Netherlands.
| | - Alejandro Lopez-Rincon
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, The Netherlands
- Department of Data Science, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
30
|
Cavanaugh G, Bai J, Tartar JL, Lin J, Nunn T, Sangwan N, Patel D, Stanis S, Patel RK, Rrukiqi D, Murphy H. Enteric Dysbiosis in Children With Autism Spectrum Disorder and Associated Response to Stress. Cureus 2024; 16:e53305. [PMID: 38435887 PMCID: PMC10905207 DOI: 10.7759/cureus.53305] [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: 01/02/2024] [Accepted: 01/31/2024] [Indexed: 03/05/2024] Open
Abstract
Background Microbiome studies in humans, though limited, have facilitated the evaluation of the potential connection between the microbiome and brain function. Children with autism spectrum disorder (ASD) have several behavioral challenges and avoidant/restrictive food intake disorder, which may contribute to gut microbiome dysbiosis. Aim The aim of this study is to examine the extent to which the gut microbiome of children with ASD differs in comparison to children with neurotypical development (CWND) and to assess whether a probiotic intervention has the potential to influence the gut microbiome in mediating positive behavior change and stress regulation. Methods This pilot study collected data from three children with ASD and four CWND before and after a four-week probiotic intervention. Data collection included microbiome diversity screening from stool samples as well as the following biophysiological measures: salivary alpha-amylase (sAA) levels, response to simulated stressor and calming stimulus (behavior), including pulse rate, galvanic skin response, and pupil diameter (PD). In addition, telomere length was assessed. All measures, except for telomere length, were repeated after the four-week intervention on the ASD and CWND groups for pre-/post-comparison. Data analysis consisted of multivariate analyses, including ANOVA. Results While greater heterogeneity in the ASD group was evident in all measures, the gut microbiome of participants who received probiotic intervention differed from pretreatment results within and across the groups investigated. Further, the biophysiological parameter sAA displayed a significant increase between baseline and exposure to stress in both groups, whereas PD increased in both groups from baseline, F(11, 26615) = 123.43, p = 0.00. Conclusion Though gut microbiome diversity is diminished in children with ASD compared to CWND, the gap is narrowed following a brief probiotic intervention. The results suggest that probiotic interventions have the potential to rescue microbiome diversity and abundance, potentially supporting stress regulation in pediatric populations.
Collapse
Affiliation(s)
- Gesulla Cavanaugh
- Department of Nursing Research, Ron and Kathy Assaf College of Nursing, Nova Southeastern University, Davie, USA
| | - Jinbing Bai
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, USA
| | - Jaime L Tartar
- Department of Psychology and Neuroscience, Nova Southeastern University, Davie, USA
| | - Jue Lin
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, USA
| | - Tina Nunn
- Lerner Research Institute, Case Western Reserve University, Cleveland, USA
| | - Naseer Sangwan
- Lerner Research Institute, Case Western Reserve University, Cleveland, USA
| | - Diti Patel
- Department of Allopathic Medicine, Nova Southeastern University Dr. Kiran C. Patel College of Allopathic Medicine, Fort Lauderdale, USA
| | - Stachyse Stanis
- Department of Allopathic Medicine, Nova Southeastern University Dr. Kiran C. Patel College of Allopathic Medicine, Fort Lauderdale, USA
| | - Raina K Patel
- Department of Allopathic Medicine, Nova Southeastern University Dr. Kiran C. Patel College of Allopathic Medicine, Fort Lauderdale, USA
| | - Djellza Rrukiqi
- Department of Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Davie, USA
| | - Hannah Murphy
- Department of Psychology and Neuroscience, Nova Southeastern University, Davie, USA
| |
Collapse
|
31
|
Hou Y, Li J, Ying S. Tryptophan Metabolism and Gut Microbiota: A Novel Regulatory Axis Integrating the Microbiome, Immunity, and Cancer. Metabolites 2023; 13:1166. [PMID: 37999261 PMCID: PMC10673612 DOI: 10.3390/metabo13111166] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 11/16/2023] [Accepted: 11/18/2023] [Indexed: 11/25/2023] Open
Abstract
Tryptophan metabolism and gut microbiota form an integrated regulatory axis that impacts immunity, metabolism, and cancer. This review consolidated current knowledge on the bidirectional interactions between microbial tryptophan processing and the host. We focused on how the gut microbiome controls tryptophan breakdown via the indole, kynurenine, and serotonin pathways. Dysbiosis of the gut microbiota induces disruptions in tryptophan catabolism which contribute to disorders like inflammatory conditions, neuropsychiatric diseases, metabolic syndromes, and cancer. These disruptions affect immune homeostasis, neurotransmission, and gut-brain communication. Elucidating the mechanisms of microbial tryptophan modulation could enable novel therapeutic approaches like psychobiotics and microbiome-targeted dietary interventions. Overall, further research on the microbiota-tryptophan axis has the potential to revolutionize personalized diagnostics and treatments for improving human health.
Collapse
Affiliation(s)
- Yingjian Hou
- Target Discovery Center, China Pharmaceutical University, Nanjing 211198, China;
| | - Jing Li
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha 410000, China
| | - Shuhuan Ying
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
- Shanghai Bocimed Pharmaceutical Research Co., Ltd., Shanghai 201203, China
| |
Collapse
|
32
|
Wang T, Chen B, Luo M, Xie L, Lu M, Lu X, Zhang S, Wei L, Zhou X, Yao B, Wang H, Xu D. Microbiota-indole 3-propionic acid-brain axis mediates abnormal synaptic pruning of hippocampal microglia and susceptibility to ASD in IUGR offspring. MICROBIOME 2023; 11:245. [PMID: 37932832 PMCID: PMC10629055 DOI: 10.1186/s40168-023-01656-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 08/23/2023] [Indexed: 11/08/2023]
Abstract
BACKGROUND Autism spectrum disorder (ASD) has been associated with intrauterine growth restriction (IUGR), but the underlying mechanisms are unclear. RESULTS We found that the IUGR rat model induced by prenatal caffeine exposure (PCE) showed ASD-like symptoms, accompanied by altered gut microbiota and reduced production of indole 3-propionic acid (IPA), a microbiota-specific metabolite and a ligand of aryl hydrocarbon receptor (AHR). IUGR children also had a reduced serum IPA level consistent with the animal model. We demonstrated that the dysregulated IPA/AHR/NF-κB signaling caused by disturbed gut microbiota mediated the hippocampal microglia hyperactivation and neuronal synapse over-pruning in the PCE-induced IUGR rats. Moreover, postnatal IPA supplementation restored the ASD-like symptoms and the underlying hippocampal lesions in the IUGR rats. CONCLUSIONS This study suggests that the microbiota-IPA-brain axis regulates ASD susceptibility in PCE-induced IUGR offspring, and supplementation of microbiota-derived IPA might be a promising interventional strategy for ASD with a fetal origin. Video Abstract.
Collapse
Affiliation(s)
- Tingting Wang
- Department of Obstetrics, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Beidi Chen
- Department of Rheumatology and Immunology, Peking University Third Hospital, Beijing, 100191, China
| | - Mingcui Luo
- Department of Obstetrics, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Lulu Xie
- Department of Pediatrics, Renmin Hospital of Wuhan University, Wuhan, 430071, China
| | - Mengxi Lu
- Department of Obstetrics, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Xiaoqian Lu
- Department of Pharmacology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, China
| | - Shuai Zhang
- Department of Obstetrics, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Liyi Wei
- Department of Obstetrics, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Xinli Zhou
- Department of Pharmacology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, China
| | - Baozhen Yao
- Department of Pediatrics, Renmin Hospital of Wuhan University, Wuhan, 430071, China
| | - Hui Wang
- Department of Pharmacology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Dan Xu
- Department of Obstetrics, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China.
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| |
Collapse
|
33
|
Dubey H, Roychoudhury R, Alex A, Best C, Liu S, White A, Carlson A, Azcarate-Peril MA, Mansfield LS, Knickmeyer R. Effect of Human Infant Gut Microbiota on Mouse Behavior, Dendritic Complexity, and Myelination. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.24.563309. [PMID: 37961091 PMCID: PMC10634763 DOI: 10.1101/2023.10.24.563309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
The mammalian gut microbiome influences numerous developmental processes. In human infants it has been linked with cognition, social skills, hormonal responses to stress, and brain connectivity. Yet, these associations are not necessarily causal. The present study tested whether two microbial stool communities, common in human infants, affected behavior, myelination, dendritic morphology, and spine density when used to colonize mouse models. Humanized animals were more like specific-pathogen free mice than germ-free mice for most phenotypes, although in males, both humanized groups were less social. Both humanized groups had thinner myelin sheaths in the hippocampus, than did germ-free animals. Humanized animals were similar to each other except for dendritic morphology and spine density where one group had greater dendritic length in the prefrontal cortex, greater dendritic volume in the nucleus accumbens, and greater spine density in both regions, compared to the other. Results add to a body of literature suggesting the gut microbiome impacts brain development. Teaser Fecal transplants from human infants with highly abundant Bifidobacterium , an important inhabitant of the intestinal tract of breastfed newborns, may promote brain connectivity in mice.
Collapse
|
34
|
Hai Q, Wang J, Kang W, Cheng S, Li J, Lyu N, Li Y, Luo Z, Liu Z. Metagenomic and metabolomic analysis of changes in intestinal contents of rainbow trout ( Oncorhynchus mykiss) infected with infectious hematopoietic necrosis virus at different culture water temperatures. Front Microbiol 2023; 14:1275649. [PMID: 37908544 PMCID: PMC10614001 DOI: 10.3389/fmicb.2023.1275649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/02/2023] [Indexed: 11/02/2023] Open
Abstract
Infectious hematopoietic necrosis (IHN) is a major disease that limits the culture of rainbow trout. In practical production, it has been found that the temperature of the culture water is a crucial factor affecting its mortality. Currently, little is known about how temperature affects the immune response of rainbow trout gut microbiota and metabolites to IHNV. In this study, our main objective is to analyze the changes in gut microorganisms of rainbow trout (juvenile fish with a consistent genetic background) after 14 days of infection with IHNV (5 × 105 pfu/fish) at 12-13°C (C: injected with saline, A: injected with IHNV) and 16-17°C (D: injected with saline, B: injected with IHNV) using metagenomic and metabolomic analyses, and to screen for probiotics that are effective against IHNV. The results showed that infection with IHNV at 12-13°C caused Eukaryote loss. Compared to Group C, Group A showed a significant increase in harmful pathogens, such as Yersiniaceae, and a significant alteration of 4,087 gut metabolites. Compared to group D, group B showed a significant increase in the abundance of Streptococcaceae and Lactococcus lactis, along with significant changes in 4,259 intestinal metabolites. Compared with their respective groups, the levels of two immune-related metabolites, 1-Octadecanoyl-glycero-3-phosphoethanolamine and L-Glutamate, were significantly upregulated in groups A and B. Compared to group B, Group A showed significantly higher pathogenic bacteria including Aeromonas, Pseudomonas, and Yersiniaceae, while group B showed a significant increase in Streptococcaceae and Lactococcus lactis. Additionally, there were 4,018 significantly different metabolites between the two groups. Interestingly, 1-Octadecanoyl-sn-glycero-3-phosphoethanolamine and L-Glutamate were significantly higher in group A than in group B. Some of the different metabolites in C vs. A are correlated with Fomitopsis pinicola, while in D vs. B they were correlated with Lactococcus raffinolactis, and in A vs. B they were correlated with Hypsizygus marmoreus. This study exposed how rainbow trout gut microbiota and metabolites respond to IHNV at different temperatures, and screens beneficial bacteria with potential resistance to IHN, providing new insights and scientific basis for the prevention and treatment of IHN.
Collapse
Affiliation(s)
| | - Jianfu Wang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Yuan C, He Y, Xie K, Feng L, Gao S, Cai L. Review of microbiota gut brain axis and innate immunity in inflammatory and infective diseases. Front Cell Infect Microbiol 2023; 13:1282431. [PMID: 37868345 PMCID: PMC10585369 DOI: 10.3389/fcimb.2023.1282431] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 09/20/2023] [Indexed: 10/24/2023] Open
Abstract
The microbiota gut brain (MGB) axis has been shown to play a significant role in the regulation of inflammatory and infective diseases. Exploring the structure and communication mode of MGB axis is crucial for understanding its role in diseases, and studying the signaling pathways and regulatory methods of MGB axis regulation in diseases is also of profound significance for future clinical research. This article reviews the composition, communication mechanism of MGB axis and its role in inflammatory and infective diseases, including Parkinson's disease (PD), Alzheimer's disease (AD), multiple sclerosis (MS), autism spectrum disorder (ASD), depression, psoriasis, irritable bowel syndrome (IBS), and inflammatory bowel diseases (IBD). In addition, our investigation delved into the regulatory functions of the inflammasome, IFN-I, NF-κB, and PARK7/DJ-1 innate immune signaling pathway in the context of inflammatory and infective diseases. Ultimately, we discussed the efficacy of various interventions, including fecal microbiota transplantation (FMT), antibiotics, probiotics, prebiotics, synbiotics, and postbiotics, in the management of inflammatory and infective diseases. Understanding the role and mechanism of the MGB axis might make positive effects in the treatment of inflammatory and infective diseases.
Collapse
Affiliation(s)
- Chongshan Yuan
- Department of Obstetrics, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Yuhong He
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Kunyu Xie
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Lianjun Feng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Shouyang Gao
- Department of Obstetrics, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Lifu Cai
- Department of Obstetrics, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
36
|
Gao J, Yang T, Song B, Ma X, Ma Y, Lin X, Wang H. Abnormal tryptophan catabolism in diabetes mellitus and its complications: Opportunities and challenges. Biomed Pharmacother 2023; 166:115395. [PMID: 37657259 DOI: 10.1016/j.biopha.2023.115395] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 08/20/2023] [Accepted: 08/26/2023] [Indexed: 09/03/2023] Open
Abstract
In recent years, the incidence rate of diabetes mellitus (DM), including type 1 diabetes mellitus(T1DM), type 2 diabetes mellitus(T2DM), and gestational diabetes mellitus (GDM), has increased year by year and has become a major global health problem. DM can lead to serious complications of macrovascular and microvascular. Tryptophan (Trp) is an essential amino acid for the human body. Trp is metabolized in the body through the indole pathway, kynurenine (Kyn) pathway and serotonin (5-HT) pathway, and is regulated by intestinal microorganisms to varying degrees. These three metabolic pathways have extensive regulatory effects on the immune, endocrine, neural, and energy metabolism systems of the body, and are related to the physiological and pathological processes of various diseases. The key enzymes and metabolites in the Trp metabolic pathway are also deeply involved in the pathogenesis of DM, playing an important role in pancreatic function, insulin resistance (IR), intestinal barrier, and angiogenesis. In DM and its complications, there is a disruption of Trp metabolic balance. Several therapy approaches for DM and complications have been proven to modify tryptophan metabolism. The metabolism of Trp is becoming a new area of focus for DM prevention and care. This paper reviews the impact of the three metabolic pathways of Trp on the pathogenesis of DM and the alterations in Trp metabolism in these diseases, expecting to provide entry points for the treatment of DM and its complications.
Collapse
Affiliation(s)
- Jialiang Gao
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ting Yang
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Bohan Song
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xiaojie Ma
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yichen Ma
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xiaowei Lin
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Hongwu Wang
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
37
|
Shaw C, Hess M, Weimer BC. Microbial-Derived Tryptophan Metabolites and Their Role in Neurological Disease: Anthranilic Acid and Anthranilic Acid Derivatives. Microorganisms 2023; 11:1825. [PMID: 37512997 PMCID: PMC10384668 DOI: 10.3390/microorganisms11071825] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/13/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
The gut microbiome provides the host access to otherwise indigestible nutrients, which are often further metabolized by the microbiome into bioactive components. The gut microbiome can also shift the balance of host-produced compounds, which may alter host health. One precursor to bioactive metabolites is the essential aromatic amino acid tryptophan. Tryptophan is mostly shunted into the kynurenine pathway but is also the primary metabolite for serotonin production and the bacterial indole pathway. Balance between tryptophan-derived bioactive metabolites is crucial for neurological homeostasis and metabolic imbalance can trigger or exacerbate neurological diseases. Alzheimer's, depression, and schizophrenia have been linked to diverging levels of tryptophan-derived anthranilic, kynurenic, and quinolinic acid. Anthranilic acid from collective microbiome metabolism plays a complex but important role in systemic host health. Although anthranilic acid and its metabolic products are of great importance for host-microbe interaction in neurological health, literature examining the mechanistic relationships between microbial production, host regulation, and neurological diseases is scarce and at times conflicting. This narrative review provides an overview of the current understanding of anthranilic acid's role in neurological health and disease, with particular focus on the contribution of the gut microbiome, the gut-brain axis, and the involvement of the three major tryptophan pathways.
Collapse
Affiliation(s)
- Claire Shaw
- Department of Population Health and Reproduction, 100K Pathogen Genome Project, University of California Davis, Davis, CA 95616, USA
- Department of Animal Science, College of Agricultural and Environmental Sciences, University of California Davis, Davis, CA 95616, USA
| | - Matthias Hess
- Department of Animal Science, College of Agricultural and Environmental Sciences, University of California Davis, Davis, CA 95616, USA
| | - Bart C Weimer
- Department of Population Health and Reproduction, 100K Pathogen Genome Project, University of California Davis, Davis, CA 95616, USA
| |
Collapse
|
38
|
Yang Y, Zhou S, Xing Y, Yang G, You M. Impact of pesticides exposure during neurodevelopmental period on autism spectrum disorders - A focus on gut microbiota. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 260:115079. [PMID: 37262968 DOI: 10.1016/j.ecoenv.2023.115079] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/03/2023]
Abstract
Accumulating evidence indicates exposure to pesticides during the crucial neurodevelopmental period increases susceptibility to many diseases, including the neurodevelopmental disorder known as autism spectrum disorder (ASD). In the last few years, it has been hypothesized that gut microbiota dysbiosis is strongly implicated in the aetiopathogenesis of ASD. Recently, new studies have suggested that the gut microbiota may be involved in the neurological and behavioural defects caused by pesticides, including ASD symptoms. This review highlights the available evidence from recent animal and human studies on the relationship between pesticides that have the potential to disturb intestinal microbiota homeostasis, and ASD symptoms. The mechanisms through which gut microbiota dysbiosis may trigger ASD-like behaviours induced by pesticides exposure during the neurodevelopmental period via the altered production of bacterial metabolites (short chain fatty acids, lipids, retinol, and amino acid) are also described. According to recent research, gut microbiota dysbiosis may be a major contributor to the symptoms of ASD associated with pesticides exposure. However, to determine the detailed mechanism of action of gut microbiota on pesticide-induced ASD behaviours, actual population exposure scenarios from epidemiological studies should be used as the basis for the appropriate exposure pattern and dosage to be used in animal studies.
Collapse
Affiliation(s)
- Yongyong Yang
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 550025, China
| | - Shun Zhou
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 550025, China
| | - Ying Xing
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 550025, China; Guizhou Provincial Center for Disease Control and Prevention, Guiyang, Guizhou 550004, China; School of Public Health, Guizhou Medical University, Guiyang, Guizhou 550025, China
| | - Guanghong Yang
- Guizhou Provincial Center for Disease Control and Prevention, Guiyang, Guizhou 550004, China; School of Public Health, Guizhou Medical University, Guiyang, Guizhou 550025, China.
| | - Mingdan You
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 550025, China.
| |
Collapse
|
39
|
Naufel MF, Truzzi GDM, Ferreira CM, Coelho FMS. The brain-gut-microbiota axis in the treatment of neurologic and psychiatric disorders. ARQUIVOS DE NEURO-PSIQUIATRIA 2023. [PMID: 37402401 PMCID: PMC10371417 DOI: 10.1055/s-0043-1767818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/06/2023]
Abstract
The human gut microbiota is a complex ecosystem made of trillions of microorganisms. The composition can be affected by diet, metabolism, age, geography, stress, seasons, temperature, sleep, and medications. The increasing evidence about the existence of a close and bi-directional correlation between the gut microbiota and the brain indicates that intestinal imbalance may play a vital role in the development, function, and disorders of the central nervous system. The mechanisms of interaction between the gut-microbiota on neuronal activity are widely discussed. Several potential pathways are involved with the brain-gut-microbiota axis, including the vagus nerve, endocrine, immune, and biochemical pathways. Gut dysbiosis has been linked to neurological disorders in different ways that involve activation of the hypothalamic-pituitary-adrenal axis, imbalance in neurotransmitter release, systemic inflammation, and increase in the permeability of the intestinal and the blood-brain barrier. Mental and neurological diseases have become more prevalent during the coronavirus disease 2019pandemic and are an essential issue in public health globally. Understanding the importance of diagnosing, preventing, and treating dysbiosis is critical because gut microbial imbalance is a significant risk factor for these disorders. This review summarizes evidence demonstrating the influence of gut dysbiosis on mental and neurological disorders.
Collapse
Affiliation(s)
| | | | | | - Fernando Morgadinho Santos Coelho
- Universidade Federal de São Paulo, Departamento de Psicobiologia, São Paulo SP, Brazil
- Universidade Federal de São Paulo, Departamento de Neurologia e Neurocirurgia, São Paulo SP, Brazil
| |
Collapse
|
40
|
Muhamadali H, Winder CL, Dunn WB, Goodacre R. Unlocking the secrets of the microbiome: exploring the dynamic microbial interplay with humans through metabolomics and their manipulation for synthetic biology applications. Biochem J 2023; 480:891-908. [PMID: 37378961 PMCID: PMC10317162 DOI: 10.1042/bcj20210534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/12/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023]
Abstract
Metabolomics is a powerful research discovery tool with the potential to measure hundreds to low thousands of metabolites. In this review, we discuss the application of GC-MS and LC-MS in discovery-based metabolomics research, we define metabolomics workflows and we highlight considerations that need to be addressed in order to generate robust and reproducible data. We stress that metabolomics is now routinely applied across the biological sciences to study microbiomes from relatively simple microbial systems to their complex interactions within consortia in the host and the environment and highlight this in a range of biological species and mammalian systems including humans. However, challenges do still exist that need to be overcome to maximise the potential for metabolomics to help us understanding biological systems. To demonstrate the potential of the approach we discuss the application of metabolomics in two broad research areas: (1) synthetic biology to increase the production of high-value fine chemicals and reduction in secondary by-products and (2) gut microbial interaction with the human host. While burgeoning in importance, the latter is still in its infancy and will benefit from the development of tools to detangle host-gut-microbial interactions and their impact on human health and diseases.
Collapse
Affiliation(s)
- Howbeer Muhamadali
- Centre for Metabolomics Research, Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, U.K
| | - Catherine L. Winder
- Centre for Metabolomics Research, Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, U.K
| | - Warwick B. Dunn
- Centre for Metabolomics Research, Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, U.K
| | - Royston Goodacre
- Centre for Metabolomics Research, Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, U.K
| |
Collapse
|
41
|
Lin F, Wang X, Luo R, Yuan B, Ye S, Yang T, Xiao L, Chen J. Maternal LPS Exposure Enhances the 5-HT Level in the Prefrontal Cortex of Autism-like Young Offspring. Brain Sci 2023; 13:958. [PMID: 37371436 DOI: 10.3390/brainsci13060958] [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: 04/24/2023] [Revised: 05/25/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by reduced social interactions, impaired communication, and stereotyped behavior. The aim of this research is to investigate the changes in serotonin (5-HT) in the medial prefrontal cortex (PFC) of autism-like offspring induced by maternal lipopolysaccharide (LPS) exposure. Pregnant Sprague-Dawley rats were intraperitoneally injected with LPS to establish an autism-like model in their offspring. Offspring prenatally exposed to LPS showed autism-like behavior. The serotonin level in the mPFC of 2-week-old offspring was noticeably increased after maternal LPS exposure. Differentially expressed genes (DEGs) were enriched in pathways related to tryptophan metabolism and the serotonin system, as shown in RNA-seq findings. Consistently, tryptophan and serotonin metabolisms were altered in 2-week-old LPS-exposed offspring. The mRNA expression levels of 5-HT catabolic enzymes were remarkably reduced or tended to decrease. Moreover, maternal LPS exposure resulted in a higher serotonin 1B receptor (5-HT1BR) expression level in the mPFC but no difference in tryptophan hydroxylase 2 (TPH2) or serotonin reuptake transporter (SERT). The concentrations of 5-HT in serum and colon were increased in LPS-exposed offspring. Meanwhile, the expression level of tryptophan hydroxylase 1 (TPH1) in the colon was increased after maternal LPS treatment, whereas SERT was reduced. Furthermore, Golgi-Cox staining showed that neuronal dendritic length and spine density were significantly reduced in the mPFC of LPS-exposed offspring. The current study reveals that maternal LPS treatment resulted in an exaltation of the 5-HT of mPFC in ASD-like young rats, which may partly be caused by the abnormal elevation of 5-HT metabolism in its colon.
Collapse
Affiliation(s)
- Fang Lin
- Chongqing Key Laboratory of Childhood Nutrition and Health, Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing 400015, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing 400015, China
| | - Xinyuan Wang
- Chongqing Key Laboratory of Childhood Nutrition and Health, Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing 400015, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing 400015, China
| | - Ruifang Luo
- Chongqing Key Laboratory of Childhood Nutrition and Health, Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing 400015, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing 400015, China
| | - Binlin Yuan
- Chongqing Key Laboratory of Childhood Nutrition and Health, Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing 400015, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing 400015, China
| | - Shasha Ye
- Chongqing Key Laboratory of Childhood Nutrition and Health, Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing 400015, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing 400015, China
| | - Ting Yang
- Chongqing Key Laboratory of Childhood Nutrition and Health, Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing 400015, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing 400015, China
| | - Lu Xiao
- Chongqing Key Laboratory of Childhood Nutrition and Health, Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing 400015, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing 400015, China
- Department of Gastroenterology, Children's Hospital of Chongqing Medical University, Chongqing 400015, China
| | - Jie Chen
- Chongqing Key Laboratory of Childhood Nutrition and Health, Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing 400015, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing 400015, China
| |
Collapse
|
42
|
Zhang M, Zhang W, Chen Y, Zhao J, Wu S, Su X. Flex Meta-Storms elucidates the microbiome local beta-diversity under specific phenotypes. Bioinformatics 2023; 39:btad148. [PMID: 36946295 PMCID: PMC10082668 DOI: 10.1093/bioinformatics/btad148] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 03/13/2023] [Accepted: 03/19/2023] [Indexed: 03/23/2023] Open
Abstract
MOTIVATION Beta-diversity quantitatively measures the difference among microbial communities thus enlightening the association between microbiome composition and environment properties or host phenotypes. The beta-diversity analysis mainly relies on distances among microbiomes that are calculated by all microbial features. However, in some cases, only a small fraction of members in a community plays crucial roles. Such a tiny proportion is insufficient to alter the overall distance, which is always missed by end-to-end comparison. On the other hand, beta-diversity pattern can also be interfered due to the data sparsity when only focusing on nonabundant microbes. RESULTS Here, we develop Flex Meta-Storms (FMS) distance algorithm that implements the "local alignment" of microbiomes for the first time. Using a flexible extraction that considers the weighted phylogenetic and functional relations of microbes, FMS produces a normalized phylogenetic distance among members of interest for microbiome pairs. We demonstrated the advantage of FMS in detecting the subtle variations of microbiomes among different states using artificial and real datasets, which were neglected by regular distance metrics. Therefore, FMS effectively discriminates microbiomes with higher sensitivity and flexibility, thus contributing to in-depth comprehension of microbe-host interactions, as well as promoting the utilization of microbiome data such as disease screening and prediction. AVAILABILITY AND IMPLEMENTATION FMS is implemented in C++, and the source code is released at https://github.com/qdu-bioinfo/flex-meta-storms.
Collapse
Affiliation(s)
- Mingqian Zhang
- College of Computer Science and Technology, Qingdao University, Qingdao, China
| | - Wenke Zhang
- College of Computer Science and Technology, Qingdao University, Qingdao, China
| | - Yuzhu Chen
- College of Computer Science and Technology, Qingdao University, Qingdao, China
| | - Jin Zhao
- College of Computer Science and Technology, Qingdao University, Qingdao, China
| | - Shunyao Wu
- College of Computer Science and Technology, Qingdao University, Qingdao, China
| | - Xiaoquan Su
- College of Computer Science and Technology, Qingdao University, Qingdao, China
| |
Collapse
|
43
|
Zhong JG, Lan WT, Feng YQ, Li YH, Shen YY, Gong JH, Zou Z, Hou X. Associations between dysbiosis gut microbiota and changes of neurotransmitters and short-chain fatty acids in valproic acid model rats. Front Physiol 2023; 14:1077821. [PMID: 37035670 PMCID: PMC10073564 DOI: 10.3389/fphys.2023.1077821] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 03/03/2023] [Indexed: 04/11/2023] Open
Abstract
Introduction: The microbiota-gut-brain axis plays an important role in the pathophysiology of autism spectrum disorder, but its specific mechanisms remain unclear. This study aimed to explore the associations of changes in neurotransmitters and short-chain fatty acids with alterations in gut microbiota in valproic acid model rats. Methods: The autism model rats were established by prenatal exposure to valproic acid (VPA). The Morris water maze test, open field test, and three-chamber test were conducted to assess the behaviors of rats. 16S rRNA gene sequences extracted from fecal samples were used to assess the gut microbial composition. Gas and liquid chromatography-mass spectroscopy was used to identify short-chain fatty acids in fecal samples and neurotransmitters in the prefrontal cortex (PFC). Results: The results showed that 28 bacterial taxa between valproic acid model rats and control rats were identified, and the most differential bacterial taxa in valproic acid model rats and control rats belonged to metagenomic species and Lactobacillus intestinalis. Acetic acid, butyric acid, valeric acid, isobutyric acid, and isovaleric acid were significantly decreased in the valproic acid model rats compared to those in control rats. Five neurotransmitters (threonine, kynurenine, tryptophan, 5-hydroxyindoleacetic acid, denoted as 5-HIAA, and betaine aldehyde chloride, denoted as BAC) were significantly decreased, whereas betaine was increased in the prefrontal cortex of valproic acid model rats compared to control rats. A variety of neurotransmitters (≥4) were correlated with Pseudomonas, Collisella, and Streptococcus at the genus level, and they were also related to the decrease of short-chain fatty acids. Discussion: According to this study, we can preliminarily infer that gut microbiota or their metabolic productions (such as SCFAs) may influence central neurotransmitter metabolism through related pathways of the gut-brain axis. These results provide microbial and short-chain fatty acid (SCFA) frameworks for understanding the role of the microbiota-gut-brain axis in autism spectrum disorder and shed new light on autism spectrum disorder treatment.
Collapse
Affiliation(s)
- Jiu-Gen Zhong
- School of Sport and Health, Guangzhou Sport University, Guangzhou, Guangdong, China
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Wan-Ting Lan
- School of Sport and Health, Guangzhou Sport University, Guangzhou, Guangdong, China
| | - Yan-Qing Feng
- School of Sport and Health, Guangzhou Sport University, Guangzhou, Guangdong, China
| | - Yin-Hua Li
- School of Sport and Health, Guangzhou Sport University, Guangzhou, Guangdong, China
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Ying-Ying Shen
- School of Sport and Health, Guangzhou Sport University, Guangzhou, Guangdong, China
| | - Jia-Heng Gong
- School of Sport and Health, Guangzhou Sport University, Guangzhou, Guangdong, China
| | - Zhi Zou
- School of Sport and Health, Guangzhou Sport University, Guangzhou, Guangdong, China
- *Correspondence: Zhi Zou, ; Xiaohui Hou,
| | - Xiaohui Hou
- School of Sport and Health, Guangzhou Sport University, Guangzhou, Guangdong, China
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
- *Correspondence: Zhi Zou, ; Xiaohui Hou,
| |
Collapse
|
44
|
Guseinova RM, Shestakova EA. Possibilities of Autologous Fecal Microbiota Transplantation in patients with obesity and diabetes mellitus. OBESITY AND METABOLISM 2022. [DOI: 10.14341/omet12901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Obesity and type 2 diabetes mellitus (T2DM) are major problems for public health all over the world. According to retrospective research, the prevalence of obesity has doubled in more than 70 countries since 1980, as well as the prevalence of obesity complications (atherosclerotic cardiovascular diseases, nonalcoholic fatty liver disease and their complications. There are many drug therapies for T2DM, but it is difficult to achieve a stable, clinically relevant effect on a long-term basis. The fact that a patient has both conditions makes it difficult to optimize carbohydrate metabolism and to achieve normal body weight. Many antidiabetic drugs cause weight again, which, in turn, contributes to the growth of insulin resistance (IR) and requires further intensification of therapy.In the last few years, there is a growing evidence of the relationship between the gut microbiota (GM), obesity and T2DM. There has been a steady growth of interest in such medical technology as fecal microbiota transplantation (FMT) in the world. Since there is data on the association of the gut microbiota (GM) with the development of IR and T2DM, the possibility of FMT can potentially be one of the new methods of treatment. This review presents the current state of the problem and discusses the possibility of modifying GM as a therapeutic strategy in obesity and T2DM with an accent on autologous fecal microbiota transplantation.
Collapse
|
45
|
Qu Z, Tian P, Yang B, Zhao J, Wang G, Chen W. Fecal microbiota transplantation for diseases: Therapeutic potential, methodology, risk management in clinical practice. Life Sci 2022; 304:120719. [PMID: 35716734 DOI: 10.1016/j.lfs.2022.120719] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/02/2022] [Accepted: 06/12/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND More than 95 % of human diseases may be related to the disturbance of gut microbes. As a treatment method that extensively regulates the gut microbes, fecal microbiota transplantation (FMT) has proven to be an effective therapy for some diseases, becoming a topic of interest among clinicians, patients and scientists. AIM To review the latest clinical research results of FMT in the treatment of various diseases and the methodology and risk management in clinical application. METHODS Search PubMed and Web of Science for reliable research results of clinical treatment of FMT within 5-10 years, as well as application guidelines and risk management policies in different regions. RESULTS As a measure of allogeneic/autologous microbiota transplantation, FMT has been used to treat a variety of diseases. By reviewing the clinical studies of FMT in gastrointestinal diseases, metabolic diseases, neurological diseases and malignant tumors, the various mechanisms in the treatment of diseases are summarized. Such as regulation of receptor microbiota composition, specific metabolites, phage function and immune response. In addition, potential risk factors, donor stool screening indicators, recipient self-specificity and possible prognostic marker molecules in the course of FMT treatment were generalized. CONCLUSIONS The potential regulatory mechanisms, risk factors and targets of FMT in gastrointestinal diseases, metabolic diseases, malignancies and neurological diseases were reviewed and proposed. It provides a theoretical basis for the establishment of a standardized treatment system for FMT and a breakthrough in treatment technology.
Collapse
Affiliation(s)
- Zhihao Qu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Peijun Tian
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Bo Yang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Gang Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China.
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
46
|
Zhao F, Zhang H, Wang P, Cui W, Xu K, Chen D, Hu M, Li Z, Geng X, Wei S. Oxytocin and serotonin in the modulation of neural function: Neurobiological underpinnings of autism-related behavior. Front Neurosci 2022; 16:919890. [PMID: 35937893 PMCID: PMC9354980 DOI: 10.3389/fnins.2022.919890] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 06/27/2022] [Indexed: 12/12/2022] Open
Abstract
Autism spectrum disorders (ASD) is a group of generalized neurodevelopmental disorders. Its main clinical features are social communication disorder and repetitive stereotyped behavioral interest. The abnormal structure and function of brain network is the basis of social dysfunction and stereotyped performance in patients with autism spectrum disorder. The number of patients diagnosed with ASD has increased year by year, but there is a lack of effective intervention and treatment. Oxytocin has been revealed to effectively improve social cognitive function and significantly improve the social information processing ability, empathy ability and social communication ability of ASD patients. The change of serotonin level also been reported affecting the development of brain and causes ASD-like behavioral abnormalities, such as anxiety, depression like behavior, stereotyped behavior. Present review will focus on the research progress of serotonin and oxytocin in the pathogenesis, brain circuit changes and treatment of autism. Revealing the regulatory effect and neural mechanism of serotonin and oxytocin on patients with ASD is not only conducive to a deeper comprehension of the pathogenesis of ASD, but also has vital clinical significance.
Collapse
Affiliation(s)
- Feng Zhao
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China
- TAIYUE Postdoctoral Innovation and Practice Base, Jinan, China
- Chinese Medicine and Brain Science Core Facility, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hao Zhang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China
- TAIYUE Postdoctoral Innovation and Practice Base, Jinan, China
- Chinese Medicine and Brain Science Core Facility, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Peng Wang
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Wenjie Cui
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Kaiyong Xu
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
- Chinese Medicine and Brain Science Core Facility, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Dan Chen
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
- Chinese Medicine and Brain Science Core Facility, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Minghui Hu
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
- TAIYUE Postdoctoral Innovation and Practice Base, Jinan, China
- Chinese Medicine and Brain Science Core Facility, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zifa Li
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China
- TAIYUE Postdoctoral Innovation and Practice Base, Jinan, China
- Chinese Medicine and Brain Science Core Facility, Shandong University of Traditional Chinese Medicine, Jinan, China
- Zifa Li,
| | - Xiwen Geng
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China
- TAIYUE Postdoctoral Innovation and Practice Base, Jinan, China
- Chinese Medicine and Brain Science Core Facility, Shandong University of Traditional Chinese Medicine, Jinan, China
- Xiwen Geng,
| | - Sheng Wei
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China
- TAIYUE Postdoctoral Innovation and Practice Base, Jinan, China
- Chinese Medicine and Brain Science Core Facility, Shandong University of Traditional Chinese Medicine, Jinan, China
- *Correspondence: Sheng Wei,
| |
Collapse
|
47
|
Avolio E, Olivito I, Rosina E, Romano L, Angelone T, Bartolo Anna D, Scimeca M, Bellizzi D, D'Aquila P, Passarino G, Alò R, Maria Facciolo R, Bagni C, De Lorenzo A, Canonaco M. Modifications of behavior and inflammation in mice following transplant with fecal microbiota from children with autism. Neuroscience 2022; 498:174-189. [DOI: 10.1016/j.neuroscience.2022.06.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 06/23/2022] [Accepted: 06/28/2022] [Indexed: 10/17/2022]
|
48
|
Liu J, Liu C, Gao Z, Zhou L, Gao J, Luo Y, Liu T, Fan X. GW4064 Alters Gut Microbiota Composition and Counteracts Autism-Associated Behaviors in BTBR T+tf/J Mice. Front Cell Infect Microbiol 2022; 12:911259. [PMID: 35811667 PMCID: PMC9257030 DOI: 10.3389/fcimb.2022.911259] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 05/13/2022] [Indexed: 11/13/2022] Open
Abstract
Autism spectrum disorder (ASD) is considered a heterogeneous neurodevelopmental disorder characterized by significant social, communication, and behavioral impairments. The gut microbiota is increasingly considered a promising therapeutic target in ASD. Farnesoid X receptor (FXR) has recently been shown to modulate the gut microbiota. We hypothesized that FXR agonist GW4064 could ameliorate behavioral deficits in an animal model for autism: BTBR T+Itpr3tf/J (BTBR) mouse. As expected, administration of GW4064 rescued the sociability of BTBR mice in the three-chamber sociability test and male-female social reciprocal interaction test, while no effects were observed in C57BL/6J mice. We also found that GW4064 administration increased fecal microbial abundance and counteracted the common ASD phenotype of a high Firmicutes to Bacteroidetes ratio in BTBR mice. In addition, GW4064 administration reversed elevated Lactobacillus and decreased Allobaculum content in the fecal matter of BTBR animals. Our findings show that GW4064 administration alleviates social deficits in BTBR mice and modulates selective aspects of the composition of the gut microbiota, suggesting that GW4064 supplementation might prove a potential strategy for improving ASD symptoms.
Collapse
Affiliation(s)
- Jiayin Liu
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
- Battalion 5 of Cadet Brigade, Third Military Medical University (Army Medical University), Chongqing, China
| | - Chuanqi Liu
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
- Battalion 5 of Cadet Brigade, Third Military Medical University (Army Medical University), Chongqing, China
| | - Zhanyuan Gao
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
- Battalion 5 of Cadet Brigade, Third Military Medical University (Army Medical University), Chongqing, China
| | - Lianyu Zhou
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
- Battalion 5 of Cadet Brigade, Third Military Medical University (Army Medical University), Chongqing, China
| | - Junwei Gao
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yi Luo
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Tianyao Liu
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xiaotang Fan
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
49
|
Bian J, Liebert A, Bicknell B, Chen XM, Huang C, Pollock CA. Faecal Microbiota Transplantation and Chronic Kidney Disease. Nutrients 2022; 14:nu14122528. [PMID: 35745257 PMCID: PMC9228952 DOI: 10.3390/nu14122528] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/10/2022] [Accepted: 06/14/2022] [Indexed: 02/05/2023] Open
Abstract
Faecal microbiota transplantation (FMT) has attracted increasing attention as an intervention in many clinical conditions, including autoimmune, enteroendocrine, gastroenterological, and neurological diseases. For years, FMT has been an effective second-line treatment for Clostridium difficile infection (CDI) with beneficial outcomes. FMT is also promising in improving bowel diseases, such as ulcerative colitis (UC). Pre-clinical and clinical studies suggest that this microbiota-based intervention may influence the development and progression of chronic kidney disease (CKD) via modifying a dysregulated gut–kidney axis. Despite the high morbidity and mortality due to CKD, there are limited options for treatment until end-stage kidney disease occurs, which results in death, dialysis, or kidney transplantation. This imposes a significant financial and health burden on the individual, their families and careers, and the health system. Recent studies have suggested that strategies to reverse gut dysbiosis using FMT are a promising therapy in CKD. This review summarises the preclinical and clinical evidence and postulates the potential therapeutic effect of FMT in the management of CKD.
Collapse
Affiliation(s)
- Ji Bian
- Kolling Institute, Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Royal North Shore Hospital, St Leonards, NSW 2065, Australia; (J.B.); (X.-M.C.)
| | - Ann Liebert
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2006, Australia;
| | - Brian Bicknell
- College of Health and Medicine, Australian National University, Deacon, ACT 2600, Australia;
| | - Xin-Ming Chen
- Kolling Institute, Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Royal North Shore Hospital, St Leonards, NSW 2065, Australia; (J.B.); (X.-M.C.)
| | - Chunling Huang
- Kolling Institute, Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Royal North Shore Hospital, St Leonards, NSW 2065, Australia; (J.B.); (X.-M.C.)
- Correspondence: (C.H.); (C.A.P.); Tel.: +61-2-9926-4784 (C.H.); +61-2-9926-4652 (C.A.P.)
| | - Carol A. Pollock
- Kolling Institute, Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Royal North Shore Hospital, St Leonards, NSW 2065, Australia; (J.B.); (X.-M.C.)
- Correspondence: (C.H.); (C.A.P.); Tel.: +61-2-9926-4784 (C.H.); +61-2-9926-4652 (C.A.P.)
| |
Collapse
|
50
|
Xiang M, Zheng L, Pu D, Lin F, Ma X, Ye H, Pu D, Zhang Y, Wang D, Wang X, Zou K, Chen L, Zhang Y, Sun Z, Zhang T, Wu G. Intestinal Microbes in Patients With Schizophrenia Undergoing Short-Term Treatment: Core Species Identification Based on Co-Occurrence Networks and Regression Analysis. Front Microbiol 2022; 13:909729. [PMID: 35783418 PMCID: PMC9247572 DOI: 10.3389/fmicb.2022.909729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/12/2022] [Indexed: 01/12/2023] Open
Abstract
Schizophrenia, a common mental disorder, has a tremendous impact on the health and economy of people worldwide. Evidence suggests that the microbial-gut-brain axis is an important pathway for the interaction between the gut microbiome and the development of schizophrenia. What is not clear is how changes in the gut microbiota composition and structure during antipsychotic treatment improve the symptoms of schizophrenia. In this study, 25 patients with schizophrenia were recruited. Their fecal samples were collected before and after hospital treatment for 14–19 days. The composition and structure of the intestinal microbiota were evaluated by 16S rRNA sequencing analysis, and the results showed significant differences in fecal microbiota before and after treatment. Firmicutes (relative abundances of 82.60 and 86.64%) and Gemminger (relative abundances of 14.17 and 13.57%) were the first dominant species at the phylum and genus levels, respectively. The random forest algorithm and co-occurrence network analysis demonstrated that intestinal flora (especially the core species ASV57) could be used as biomarkers to distinguish different clinical states and match treatment regimens accordingly. In addition, after fecal microbiota transplantation, antibiotic-treated recipient mice showed multiple behavioral improvements. These included decreased psychomotor hyperactivity, increased social interaction, and memory. In conclusion, this study suggests that differences in the composition and structure of gut microbiota after treatment are associated with the development and severity of schizophrenia. Results may provide a potential target for the treatment of this disorder.
Collapse
Affiliation(s)
- Min Xiang
- Medical Laboratory, The Fourth People's Hospital of Ya'an, Ya'an, China
| | - Liqin Zheng
- MOE Key Lab for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, China
| | - Daoshen Pu
- The Fourth People's Hospital of Ya'an, Ya'an, China
| | - Feng Lin
- Medical Laboratory, The Fourth People's Hospital of Ya'an, Ya'an, China
| | - Xiaodong Ma
- Medical Laboratory, The Fourth People's Hospital of Ya'an, Ya'an, China
| | - Huiqian Ye
- Medical Laboratory, The Fourth People's Hospital of Ya'an, Ya'an, China
| | - Daoqiong Pu
- Medical Laboratory, The Fourth People's Hospital of Ya'an, Ya'an, China
| | - Ying Zhang
- Medical Laboratory, The Fourth People's Hospital of Ya'an, Ya'an, China
| | - Dong Wang
- Psychiatry Department, The Fourth People's Hospital of Ya'an, Ya'an, China
| | - Xiaoli Wang
- Internal Medicine, The Fourth People's Hospital of Ya'an, Ya'an, China
| | - Kaiqing Zou
- The Outpatient Department, The Fourth People's Hospital of Ya'an, Ya'an, China
| | - Linqi Chen
- MOE Key Lab for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, China
| | - Yong Zhang
- MOE Key Lab for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, China
| | - Zhanjiang Sun
- MOE Key Lab for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, China
| | - Tao Zhang
- MOE Key Lab for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, China
- Tao Zhang
| | - Guolin Wu
- Medical Laboratory, The Fourth People's Hospital of Ya'an, Ya'an, China
- *Correspondence: Guolin Wu
| |
Collapse
|