1
|
Teglas T, Marcos AC, Torices S, Toborek M. Circadian control of polycyclic aromatic hydrocarbon-induced dysregulation of endothelial tight junctions and mitochondrial bioenergetics. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 952:175886. [PMID: 39218115 PMCID: PMC11444715 DOI: 10.1016/j.scitotenv.2024.175886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 08/05/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
The study evaluates the impact of environmental toxicants, such as polycyclic aromatic hydrocarbons (PAHs), on circadian regulations and functions of brain endothelial cells, which form the main structural element of the blood-brain barrier (BBB). PAH are lipophilic and highly toxic environmental pollutants that accumulate in human and animal tissues. Environmental factors related to climate change, such as an increase in frequency and intensity of wildfires or enhanced strength of hurricanes or tropical cyclones, may lead to redistribution of these toxicants and enhanced human exposure. These natural disasters are also associated with disruption of circadian rhythms in affected populations, linking increased exposure to environmental toxicants to alterations of circadian rhythm pathways. Several vital physiological processes are coordinated by circadian rhythms, and disruption of the circadian clock can contribute to the development of several diseases. The blood-brain barrier (BBB) is crucial for protecting the brain from blood-borne harmful substances, and its integrity is influenced by circadian rhythms. Exposure of brain endothelial cells to a human and environmentally-relevant PAH mixture resulted in dose-dependent alterations of expression of critical circadian modulators, such as Clock, Bmal1, Cry1/2, and Per1/2. Moreover, silencing of the circadian Clock gene potentiated the impact of PAHs on the expression of the main tight junction genes and proteins (namely, claudin-5, occludin, JAM-2, and ZO-2), as well as mitochondrial bioenergetics. Findings from this study contribute to a better understanding of pathological influence of PAH-induced health effects, especially those related to circadian rhythm disruption.
Collapse
Affiliation(s)
- Timea Teglas
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street, Miami, FL 33136, USA
| | - Anne Caroline Marcos
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street, Miami, FL 33136, USA
| | - Silvia Torices
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street, Miami, FL 33136, USA
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street, Miami, FL 33136, USA; Institute of Physiotherapy and Health Sciences, The Jerzy Kukuczka Academy of Physical Education, Katowice, Poland.
| |
Collapse
|
2
|
Abuljadayel D, Alotibi A, Algothmi K, Basingab F, Alhazmi S, Almuhammadi A, Alharthi A, Alyoubi R, Bahieldin A. Gut microbiota of children with autism spectrum disorder and healthy siblings: A comparative study. Exp Ther Med 2024; 28:430. [PMID: 39328398 PMCID: PMC11425773 DOI: 10.3892/etm.2024.12719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 05/01/2024] [Indexed: 09/28/2024] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental abnormality that impairs social communication. The human gut microbiome (GM) influences a variety of local processes, including dysbiosis and the defense against pathogenic microorganisms. The aim of the present study was to categorize and identify molecular biomarkers for ASD. In the present study, metagenomics whole genome shotgun sequencing was used to identify the gut microbiota in autistic individuals. Fecal samples from four children with ASD and four healthy control siblings, aged 3-10 years old, were examined using bioinformatics analysis. A total of 673,091 genes were cataloged, encompassing 25 phyla and 2 kingdoms based on the taxonomy analysis. The results revealed 257 families, 34 classes, 84 orders, and 1,314 genera among 4,339 species. The top 10 most abundant genes and corresponding functional genes for each group were determined after the abundance profile was screened. The results showed that children with ASD had a higher abundance of certain gut microbiomes than their normal siblings and vice versa. The phyla Firmicutes and Proteobacteria were the most abundant in ASD. The Thermoanaerobacteria class was also restricted to younger healthy individuals. Moreover, the Lactobacillaceae family was more abundant in children with ASD. Additionally, it was discovered that children with ASD had a higher abundance of the Bacteroides genus and a lower abundance of the Bifidobacterium and Prevotella genera. In conclusion, there were more pathogenic genera and species and higher levels of biomass, diversity and richness in the GM of children with ASD.
Collapse
Affiliation(s)
- Dalia Abuljadayel
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Asalah Alotibi
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Khloud Algothmi
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Immunology Unit, King Fahad Medical Research Centre, King Abdulaziz University, Jeddah 22252, Saudi Arabia
| | - Fatemah Basingab
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Immunology Unit, King Fahad Medical Research Centre, King Abdulaziz University, Jeddah 22252, Saudi Arabia
| | - Safiah Alhazmi
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Immunology Unit, King Fahad Medical Research Centre, King Abdulaziz University, Jeddah 22252, Saudi Arabia
- Neuroscience and Geroscience Research Unit, King Fahad Medical Research Centre, King Abdulaziz University, Jeddah 22252, Saudi Arabia
- Central Lab of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Asma Almuhammadi
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Amani Alharthi
- Department of Biology, College of Science in Zulfi, Majmaaha University, Zulfi 11932, Saudi Arabia
| | - Reem Alyoubi
- College of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia
| | - Ahmad Bahieldin
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
3
|
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
|
4
|
Horecka-Lewitowicz A, Lewitowicz W, Wawszczak-Kasza M, Lim H, Lewitowicz P. Autism Spectrum Disorder Pathogenesis-A Cross-Sectional Literature Review Emphasizing Molecular Aspects. Int J Mol Sci 2024; 25:11283. [PMID: 39457068 PMCID: PMC11508848 DOI: 10.3390/ijms252011283] [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: 09/16/2024] [Revised: 10/14/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
The etiology of autism spectrum disorder (ASD) has not yet been completely elucidated. Through time, multiple attempts have been made to uncover the causes of ASD. Different theories have been proposed, such as being caused by alterations in the gut-brain axis with an emphasis on gut dysbiosis, post-vaccine complications, and genetic or even autoimmune causes. In this review, we present data covering the main streams that focus on ASD etiology. Data collection occurred in many countries covering ethnically diverse subjects. Moreover, we aimed to show how the progress in genetic techniques influences the explanation of medical White Papers in the ASD area. There is no single evidence-based pathway that results in symptoms of ASD. Patient management has constantly only been symptomatic, and there is no ASD screening apart from symptom-based diagnosis and parent-mediated interventions. Multigene sequencing or epigenetic alterations hold promise in solving the disjointed molecular puzzle. Further research is needed, especially in the field of biogenetics and metabolomic aspects, because young children constitute the patient group most affected by ASD. In summary, to date, molecular research has confirmed multigene dysfunction as the causative factor of ASD, the multigene model with metabolomic influence would explain the heterogeneity in ASD, and it is proposed that ion channel dysfunction could play a core role in ASD pathogenesis.
Collapse
Affiliation(s)
- Agata Horecka-Lewitowicz
- Institute of Medical Sciences, Jan Kochanowski University, Al. IX Wiekow Kielc 19A, 25-516 Kielce, Poland
| | - Wojciech Lewitowicz
- Student Scientific Society at Collegium Medicum, Jan Kochanowski University, Al. IX Wiekow Kielc 19A, 25-516 Kielce, Poland; (W.L.); (H.L.)
| | - Monika Wawszczak-Kasza
- Institute of Health Sciences, Jan Kochanowski University, Al. IX Wiekow Kielc 19A, 25-516 Kielce, Poland
| | - Hyebin Lim
- Student Scientific Society at Collegium Medicum, Jan Kochanowski University, Al. IX Wiekow Kielc 19A, 25-516 Kielce, Poland; (W.L.); (H.L.)
| | - Piotr Lewitowicz
- Institute of Medical Sciences, Jan Kochanowski University, Al. IX Wiekow Kielc 19A, 25-516 Kielce, Poland
| |
Collapse
|
5
|
Lin CH, Zeng T, Lu CW, Li DY, Liu YY, Li BM, Chen SQ, Deng YH. Efficacy and safety of Bacteroides fragilis BF839 for pediatric autism spectrum disorder: a randomized clinical trial. Front Nutr 2024; 11:1447059. [PMID: 39290561 PMCID: PMC11407114 DOI: 10.3389/fnut.2024.1447059] [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: 06/11/2024] [Accepted: 08/16/2024] [Indexed: 09/19/2024] Open
Abstract
Background The clinical utility of Bacteroides fragilis in treating autism spectrum disorder (ASD) remains unclear. Therefore, this randomized, double-blind, placebo-controlled study aimed to explore the therapeutic effects and safety of B. fragilis BF839 in the treatment of pediatric ASD. Methods We examined 60 children aged 2-10 years diagnosed with ASD, and participants received either BF839 powder (10 g/bar with ≥106 CFU/bar of viable bacteria, two bars/day) or placebo for 16 weeks. The primary outcomes was Autism Behavior Checklist (ABC) score. The secondary outcomes were Childhood Autism Rating Scale (CARS), Social Responsiveness Scale (SRS), Normal Development of Social Skills from Infants to Junior High School Children (S-M), Gastrointestinal Symptom Rating Scale (GSRS) scores, and fecal microbiome composition. Assessments were performed on day 0 and at weeks 8 and 16. Results Compared with the placebo group, the BF839 group showed significant improvement in the ABC body and object use scores at week 16, which was more pronounced in children with ASD aged <4 years. Among children with a baseline CARS score ≥30, the BF839 group showed significant improvements at week 16 in the ABC total score, ABC body and object use score, CARS score, and GSRS score compared to the placebo group. Only two patients (6.67%) in the BF839 group experienced mild diarrhea. Compared with baseline and placebo group levels, the BF839 group showed a significant post-intervention increase in abundance of bifidobacteria and change in the metabolic function of neuroactive compounds encoded by intestinal microorganisms. Conclusion BF839 significantly and safely improved abnormal behavior and gastrointestinal symptoms in children with ASD.
Collapse
Affiliation(s)
- Chu-Hui Lin
- Department of Clinical Nutrition, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ting Zeng
- Department of Clinical Nutrition, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Cui-Wei Lu
- Medical Administration College, Guangzhou Medical University, Guangzhou, China
| | - De-Yang Li
- Medical Administration College, Guangzhou Medical University, Guangzhou, China
- Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| | - Yi-Ying Liu
- Weierkang Specialist Outpatient Department, Guangzhou, China
| | - Bing-Mei Li
- Department of Neurology, Institute of Neuroscience, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - Sheng-Qiang Chen
- Medical Administration College, Guangzhou Medical University, Guangzhou, China
| | - Yu-Hong Deng
- Department of Clinical Nutrition, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Neurology, Institute of Neuroscience, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| |
Collapse
|
6
|
Devason AS, Thaiss CA, de la Fuente-Nunez C. Neuromicrobiology Comes of Age: The Multifaceted Interactions between the Microbiome and the Nervous System. ACS Chem Neurosci 2024; 15:2957-2965. [PMID: 39102500 DOI: 10.1021/acschemneuro.4c00307] [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] [Indexed: 08/07/2024] Open
Abstract
The past decade has seen an explosion in our knowledge about the interactions between gut microbiota, the central nervous system, and the immune system. The gut-brain axis has recently gained much attention due to its role in regulating host physiology. This review explores recent findings concerning potential pathways linking the gut-brain axis to the initiation, pathophysiology, and development of neurological disorders. Our objective of this work is to uncover causative factors and pinpoint particular pathways and therapeutic targets that may facilitate the translation of experimental animal research into practical applications for human patients. We highlight three distinct yet interrelated mechanisms: (1) disruptions of both the intestinal and blood-brain barriers, (2) persistent neuroinflammation, and (3) the role of the vagus nerve.
Collapse
Affiliation(s)
- Ashwarya S Devason
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Institute for Immunology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, United States
- Institute for Obesity, Diabetes and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia 19104, Pennsylvania United States
| | - Christoph A Thaiss
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Institute for Immunology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, United States
- Institute for Obesity, Diabetes and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia 19104, Pennsylvania United States
| | - Cesar de la Fuente-Nunez
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
7
|
Aldegheri L, Kharrat F, Conti A, Monica F, Busa F, Campisciano G, Zanotta N, Cason C, Comar M. Impact of Human Milk Oligosaccharides and Probiotics on Gut Microbiome and Mood in Autism: A Case Report. Microorganisms 2024; 12:1625. [PMID: 39203467 PMCID: PMC11356532 DOI: 10.3390/microorganisms12081625] [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: 07/22/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 09/03/2024] Open
Abstract
Recent evidence has highlighted the role of the gut-brain axis in the progression of autism spectrum disorder (ASD), with significant changes in the gut microbiome of individuals with this condition. This report investigates the effects of probiotics and human milk oligosaccharide (HMO) supplements on the gut microbiome, inflammatory cytokine profile, and clinical outcomes in an ASD adolescent with chronic gastrointestinal dysfunction and cognitive impairment. Following treatment, we observed a decrease in proinflammatory cytokines' concentration alongside Sutterella relative abundance, a bacterium reported to be linked with gastrointestinal diseases. Also, we reported a notable increase in mood stability. The study aims to evaluate the use of gut microbiome-based therapy in selected ASD patients, highlighting its potential to improve related clinical symptoms.
Collapse
Affiliation(s)
- Luana Aldegheri
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy; (L.A.); (F.K.); (A.C.); (G.C.); (N.Z.); (C.C.)
| | - Feras Kharrat
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy; (L.A.); (F.K.); (A.C.); (G.C.); (N.Z.); (C.C.)
| | - Andrea Conti
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy; (L.A.); (F.K.); (A.C.); (G.C.); (N.Z.); (C.C.)
| | - Fabio Monica
- Department of Gastroenterology and Endoscopy, Trieste University Hospital, Strada di Fiume 447, 34149 Trieste, Italy;
| | | | - Giuseppina Campisciano
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy; (L.A.); (F.K.); (A.C.); (G.C.); (N.Z.); (C.C.)
| | - Nunzia Zanotta
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy; (L.A.); (F.K.); (A.C.); (G.C.); (N.Z.); (C.C.)
| | - Carolina Cason
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy; (L.A.); (F.K.); (A.C.); (G.C.); (N.Z.); (C.C.)
| | - Manola Comar
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy; (L.A.); (F.K.); (A.C.); (G.C.); (N.Z.); (C.C.)
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy
| |
Collapse
|
8
|
Zhu G, Yan L, Fang L, Fan C, Sun H, Zhou X, Zhang Y, Shi Z. Possible immune mechanisms of gut microbiota and its metabolites in the occurrence and development of immune thrombocytopenia. Front Microbiol 2024; 15:1426911. [PMID: 39171254 PMCID: PMC11335631 DOI: 10.3389/fmicb.2024.1426911] [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: 05/02/2024] [Accepted: 07/26/2024] [Indexed: 08/23/2024] Open
Abstract
Immune thrombocytopenia (ITP) is an autoimmune disease characterized by increased platelet destruction and impaired production, leading to an elevated bleeding tendency. Recent studies have demonstrated an important link between the gut microbiota and the onset and progression of several immune diseases in humans, emphasizing that gut microbiota-derived metabolites play a non-negligible role in autoimmune diseases. The gut microbiota and its metabolites, such as short-chain fatty acids, oxidized trimethylamine, tryptophan metabolites, secondary bile acids and lipopolysaccharides, can alter intestinal barrier permeability by modulating immune cell differentiation and cytokine secretion, which in turn affects the systemic immune function of the host. It is therefore reasonable to hypothesize that ecological dysregulation of the gut microbiota may be an entirely new factor in the triggering of ITP. This article reviews the potential immune-related mechanisms of the gut microbiota and representative metabolites in ITP, as well as the important influence of leaky gut on the development of ITP, with a view to enriching the theoretical system of ITP-related gut microecology and providing new ideas for the study of ITP.
Collapse
Affiliation(s)
- Gengda Zhu
- National Medical Research Center of Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lixiang Yan
- National Medical Research Center of Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lijun Fang
- National Medical Research Center of Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Hematology and Blood Diseases Hospital, National Clinical Medical Research Center for Blood Diseases, Tianjin, China
| | - Chenyang Fan
- National Medical Research Center of Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Hui Sun
- National Medical Research Center of Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xinli Zhou
- National Medical Research Center of Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yucheng Zhang
- National Medical Research Center of Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhexin Shi
- National Medical Research Center of Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
9
|
Tao K, Yuan Y, Xie Q, Dong Z. Relationship between human oral microbiome dysbiosis and neuropsychiatric diseases: An updated overview. Behav Brain Res 2024; 471:115111. [PMID: 38871130 DOI: 10.1016/j.bbr.2024.115111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/24/2024] [Accepted: 06/10/2024] [Indexed: 06/15/2024]
Abstract
The role of the gut-brain axis in mental health disorders has been extensively studied. As the oral cavity is the starting point of the digestive tract, the role that the oral microbiota plays in mental health disorders has gained recent attention. Oral microbiota can enter the bloodstream and trigger inflammatory responses or translocate to the brain through the trigeminal nerve or olfactory system. Hence, the concept of the oral microbiota-brain axis has emerged. Several hypotheses have been suggested that the oral microbiota can enter the gastrointestinal tract and affect the gut-brain axis; however, literature describing oral-brain communication remains limited. This review summarizes the characteristics of oral microbiota and its mechanisms associated with mental health disorders. Through a comprehensive examination of the relationship between oral microbiota and various neuropsychiatric diseases, such as anxiety, depression, schizophrenia, autism spectrum disorder, epilepsy, Parkinson's disease, and dementia, this review seeks to identify promising avenues of future research.
Collapse
Affiliation(s)
- Kai Tao
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Yanling Yuan
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Qinglian Xie
- Department of Outpatient, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China; Department of Outpatient, West China Xiamen Hospital, Sichuan University, Fujian 361022, People's Republic of China.
| | - Zaiquan Dong
- Mental Health Center, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China.
| |
Collapse
|
10
|
Tu G, Jiang N, Chen W, Liu L, Hu M, Liao B. The neurobiological mechanisms underlying the effects of exercise interventions in autistic individuals. Rev Neurosci 2024; 0:revneuro-2024-0058. [PMID: 39083671 DOI: 10.1515/revneuro-2024-0058] [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: 04/22/2024] [Accepted: 07/15/2024] [Indexed: 08/02/2024]
Abstract
Autism spectrum disorder is a pervasive and heterogeneous neurodevelopmental condition characterized by social communication difficulties and rigid, repetitive behaviors. Owing to the complex pathogenesis of autism, effective drugs for treating its core features are lacking. Nonpharmacological approaches, including education, social-communication, behavioral and psychological methods, and exercise interventions, play important roles in supporting the needs of autistic individuals. The advantages of exercise intervention, such as its low cost, easy implementation, and high acceptance, have garnered increasing attention. Exercise interventions can effectively improve the core features and co-occurring conditions of autism, but the underlying neurobiological mechanisms are unclear. Abnormal changes in the gut microbiome, neuroinflammation, neurogenesis, and synaptic plasticity may individually or interactively be responsible for atypical brain structure and connectivity, leading to specific autistic experiences and characteristics. Interestingly, exercise can affect these biological processes and reshape brain network connections, which may explain how exercise alleviates core features and co-occurring conditions in autistic individuals. In this review, we describe the definition, diagnostic approach, epidemiology, and current support strategies for autism; highlight the benefits of exercise interventions; and call for individualized programs for different subtypes of autistic individuals. Finally, the possible neurobiological mechanisms by which exercise improves autistic features are comprehensively summarized to inform the development of optimal exercise interventions and specific targets to meet the needs of autistic individuals.
Collapse
Affiliation(s)
- Genghong Tu
- Department of Sports Medicine, 47878 Guangzhou Sport University , Guangzhou, Guangdong, 510500, P.R. China
- Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, 47878 Scientific Research Center, Guangzhou Sport University , Guangzhou, Guangdong, 510500, P.R. China
| | - Nan Jiang
- Graduate School, 47878 Guangzhou Sport University , Guangzhou, Guangdong, 510500, P.R. China
| | - Weizhong Chen
- Graduate School, 47878 Guangzhou Sport University , Guangzhou, Guangdong, 510500, P.R. China
| | - Lining Liu
- Graduate School, 47878 Guangzhou Sport University , Guangzhou, Guangdong, 510500, P.R. China
| | - Min Hu
- Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, 47878 Scientific Research Center, Guangzhou Sport University , Guangzhou, Guangdong, 510500, P.R. China
| | - Bagen Liao
- Department of Sports Medicine, 47878 Guangzhou Sport University , Guangzhou, Guangdong, 510500, P.R. China
- Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, 47878 Scientific Research Center, Guangzhou Sport University , Guangzhou, Guangdong, 510500, P.R. China
| |
Collapse
|
11
|
Aziz-Zadeh L, Mayer E, Labus J, Ringold S, Jayashankar A, Kilroy E, Butera C, Jacobs J, Tanartkit S, Joshi S, Dapretto M. Relationships between tryptophan-related gut metabolites, brain activity, and autism symptomatology. RESEARCH SQUARE 2024:rs.3.rs-4559624. [PMID: 39108481 PMCID: PMC11302680 DOI: 10.21203/rs.3.rs-4559624/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/11/2024]
Abstract
Gut microbial metabolites have been theorized to play a causative role in the pathophysiology of autism spectrum disorder (ASD). This hypothesis is based on results from mechanistic preclinical studies and several correlational studies showing differences in gut microbial composition between ASD subjects and neurotypical (NT) controls. However, alterations in how the human brain interacts with the gut microbiome in ASD have not been examined. In this cross-sectional, case-control observational study, fecal metabolomics, task-based functional magnetic resonance imaging (fMRI), and behavioral assessments were obtained from 43 ASD and 41 NT children aged 8-17. The fMRI tasks were based on socio-emotional and sensory paradigms that commonly show strong evoked brain differences in ASD participants. General linear models and mediational modeling were applied to examine the links between tryptophan metabolism and evoked brain activity and behavior. Results indicated that fecal levels of specific tryptophan-related metabolites were associated with: 1) brain activity atypicalities in regions previously implicated in ASD (i.e., insula and cingulate); and 2) ASD severity and symptomatology (i.e., ADOS scores, disgust propensity, and sensory sensitivities). Importantly, activity in the mid-insula and mid-cingulate significantly mediated relationships between the microbial tryptophan metabolites, indolelactate and tryptophan betaine, and ASD severity and disgust sensitivity. To our knowledge, this is the first study to elucidate how interactions between gut metabolites and brain activity may impact autism symptomatology, particularly in functional brain pathways associated with vagal and interoceptive/emotion processing.
Collapse
Affiliation(s)
| | - Emeran Mayer
- G. Oppenheimer Center for Neurobiology of Stress and Resilience at UCLA; Institute for Genomics and Bioinformatics, University of California, Irvine
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Vashisht A, Vashisht V, Singh H, Ahluwalia P, Mondal AK, Williams C, Farmaha J, Woodall J, Kolhe R. Neurological Complications of COVID-19: Unraveling the Pathophysiological Underpinnings and Therapeutic Implications. Viruses 2024; 16:1183. [PMID: 39205157 PMCID: PMC11359204 DOI: 10.3390/v16081183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/11/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease (COVID-19), induced a global pandemic with a diverse array of clinical manifestations. While the acute phase of the pandemic may be waning, the intricacies of COVID-19's impact on neurological health remain a crucial area of investigation. Early recognition of the spectrum of COVID-19 symptoms, ranging from mild fever and cough to life-threatening respiratory distress and multi-organ failure, underscored the significance of neurological complications, including anosmia, seizures, stroke, disorientation, encephalopathy, and paralysis. Notably, patients requiring intensive care unit (ICU) admission due to neurological challenges or due to them exhibiting neurological abnormalities in the ICU have shown increased mortality rates. COVID-19 can lead to a range of neurological complications such as anosmia, stroke, paralysis, cranial nerve deficits, encephalopathy, delirium, meningitis, seizures, etc., in affected patients. This review elucidates the burgeoning landscape of neurological sequelae associated with SARS-CoV-2 infection and explores the underlying neurobiological mechanisms driving these diverse manifestations. A meticulous examination of potential neuroinvasion routes by SARS-CoV-2 underscores the intricate interplay between the virus and the nervous system. Moreover, we dissect the diverse neurological manifestations emphasizing the necessity of a multifaceted approach to understanding the disease's neurological footprint. In addition to elucidating the pathophysiological underpinnings, this review surveys current therapeutic modalities and delineates prospective avenues for neuro-COVID research. By integrating epidemiological, clinical, and diagnostic parameters, we endeavor to foster a comprehensive analysis of the nexus between COVID-19 and neurological health, thereby laying the groundwork for targeted therapeutic interventions and long-term management strategies.
Collapse
Affiliation(s)
- Ashutosh Vashisht
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (A.V.); (V.V.); (H.S.); (P.A.); (A.K.M.); (J.F.); (J.W.)
| | - Vishakha Vashisht
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (A.V.); (V.V.); (H.S.); (P.A.); (A.K.M.); (J.F.); (J.W.)
| | - Harmanpreet Singh
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (A.V.); (V.V.); (H.S.); (P.A.); (A.K.M.); (J.F.); (J.W.)
| | - Pankaj Ahluwalia
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (A.V.); (V.V.); (H.S.); (P.A.); (A.K.M.); (J.F.); (J.W.)
| | - Ashis K. Mondal
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (A.V.); (V.V.); (H.S.); (P.A.); (A.K.M.); (J.F.); (J.W.)
| | - Colin Williams
- Lincoln Memorial DeBusk College of Osteopathic Medicine, Lincoln Memorial University, Knoxville, TN 37902, USA;
| | - Jaspreet Farmaha
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (A.V.); (V.V.); (H.S.); (P.A.); (A.K.M.); (J.F.); (J.W.)
| | - Jana Woodall
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (A.V.); (V.V.); (H.S.); (P.A.); (A.K.M.); (J.F.); (J.W.)
| | - Ravindra Kolhe
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (A.V.); (V.V.); (H.S.); (P.A.); (A.K.M.); (J.F.); (J.W.)
| |
Collapse
|
13
|
Wu Y, Su Q. Harnessing the Gut Microbiome: To What Extent Can Pre-/Probiotics Alleviate Immune Activation in Autism Spectrum Disorder? Nutrients 2024; 16:2382. [PMID: 39125263 PMCID: PMC11314583 DOI: 10.3390/nu16152382] [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: 06/26/2024] [Revised: 07/22/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Children diagnosed with autism spectrum disorder (ASD) are at an increased risk of experiencing gastrointestinal (GI) discomfort, which has been linked to dysfunctions in the microbiome-gut-brain axis. The bidirectional communication between gut and brain plays a crucial role in the overall health of individuals, and alterations in the gut microbiome can contribute to immune activation and gut-brain dysfunction in ASD. Despite the limited and controversial results of pre-/probiotic applications in ASD, this review comprehensively maps the association between ASD clinical symptoms and specific bacterial taxa and evaluates the efficacy of pre-/probiotics in modulating microbiota composition, reducing inflammatory biomarkers, alleviating difficulties in GI distress, sleep problems, core and other ASD-associated symptoms, as well as relieving parental concerns, separately, in individuals with ASD. Beyond simply targeting core ASD symptoms, this review highlights the potential of pre-/probiotic supplementations as a strategy to modulate gut homeostasis and immune response, and to delineate the potential mechanisms by which its direct or mediating effects can alleviate gut-brain dysfunction and poor nutritional status in ASD management. Further well-designed randomized controlled trials are needed to strengthen the existing evidence and establish optimal protocols for the use of pre-/probiotics in the context of ASD.
Collapse
Affiliation(s)
- Yuqi Wu
- Microbiota I-Center (MagIC), Hong Kong SAR, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qi Su
- Microbiota I-Center (MagIC), Hong Kong SAR, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
14
|
Yang C, Xiao H, Zhu H, Du Y, Wang L. Revealing the gut microbiome mystery: A meta-analysis revealing differences between individuals with autism spectrum disorder and neurotypical children. Biosci Trends 2024; 18:233-249. [PMID: 38897955 DOI: 10.5582/bst.2024.01123] [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] [Indexed: 06/21/2024]
Abstract
The brain-gut axis intricately links gut microbiota (GM) dysbiosis to the development or worsening of autism spectrum disorder (ASD). However, the precise GM composition in ASD and the effectiveness of probiotics are unclear. To address this, we performed a thorough meta-analysis of 28 studies spanning PubMed, PsycINFO, Web of Science, Scopus, and MEDLINE, involving 1,256 children with ASD and 1042 neurotypical children, up to February 2024. Using Revman 5.3, we analyzed the relative abundance of 8 phyla and 64 genera. While individuals with ASD did not exhibit significant differences in included phyla, they exhibited elevated levels of Parabacteroides, Anaerostipes, Faecalibacterium, Clostridium, Dorea, Phascolarctobacterium, Lachnoclostridium, Catenibacterium, and Collinsella along with reduced percentages of Barnesiella, Odoribacter, Paraprevotella, Blautia, Turicibacter, Lachnospira, Pseudomonas, Parasutterella, Haemophilus, and Bifidobacterium. Notably, discrepancies in Faecalibacterium, Clostridium, Dorea, Phascolarctobacterium, Catenibacterium, Odoribacter, and Bifidobacterium persisted even upon systematic exclusion of individual studies. Consequently, the GM of individuals with ASD demonstrates an imbalance, with potential increases or decreases in both beneficial and harmful bacteria. Therefore, personalized probiotic interventions tailored to ASD specifics are imperative, rather than a one-size-fits-all approach.
Collapse
Affiliation(s)
- Changjiang Yang
- Faculty of Education, East China Normal University, Shanghai, China
| | - Hongli Xiao
- Faculty of Education, East China Normal University, Shanghai, China
| | - Han Zhu
- Faculty of Education, East China Normal University, Shanghai, China
| | - Yijie Du
- Qingpu Traditional Chinese Medicine Hospital, Shanghai, China
| | - Ling Wang
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- The Academy of Integrative Medicine, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
| |
Collapse
|
15
|
Zhao T, Huang C, Zhang Y, Zhu Y, Chen X, Wang T, Shao J, Meng X, Huang Y, Wang H, Wang H, Wang B, Xu D. Prenatal 1-Nitropyrene Exposure Causes Autism-Like Behavior Partially by Altering DNA Hydroxymethylation in Developing Brain. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306294. [PMID: 38757379 PMCID: PMC11267330 DOI: 10.1002/advs.202306294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 03/13/2024] [Indexed: 05/18/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder, characterized by social communication disability and stereotypic behavior. This study aims to investigate the impact of prenatal exposure to 1-nitropyrene (1-NP), a key component of motor vehicle exhaust, on autism-like behaviors in a mouse model. Three-chamber test finds that prenatal 1-NP exposure causes autism-like behaviors during the weaning period. Patch clamp shows that inhibitory synaptic transmission is reduced in medial prefrontal cortex of 1-NP-exposed weaning pups. Immunofluorescence finds that prenatal 1-NP exposure reduces the number of prefrontal glutamate decarboxylase 67 (GAD67) positive interneurons in fetuses and weaning pups. Moreover, prenatal 1-NP exposure retards tangential migration of GAD67-positive interneurons and downregulates interneuron migration-related genes, such as Nrg1, Erbb4, and Sema3F, in fetal forebrain. Mechanistically, prenatal 1-NP exposure reduces hydroxymethylation of interneuron migration-related genes through inhibiting ten-eleven translocation (TET) activity in fetal forebrain. Supplement with alpha-ketoglutarate (α-KG), a cofactor of TET enzyme, reverses 1-NP-induced hypohydroxymethylation at specific sites of interneuron migration-related genes. Moreover, α-KG supplement alleviates 1-NP-induced migration retardation of interneurons in fetal forebrain. Finally, maternal α-KG supplement improves 1-NP-induced autism-like behaviors in weaning offspring. In conclusion, prenatal 1-NP exposure causes autism-like behavior partially by altering DNA hydroxymethylation of interneuron migration-related genes in developing brain.
Collapse
Affiliation(s)
- Ting Zhao
- Department of ToxicologySchool of Public HealthAnhui Medical UniversityHefei230022China
- Key Laboratory of Environmental Toxicology of Anhui Higher Education InstitutesAnhui Medical UniversityHefei230032China
| | - Cheng‐Qing Huang
- School of Food and BioengineeringHefei University of TechnologyHefei230009China
| | - Yi‐Hao Zhang
- Department of ToxicologySchool of Public HealthAnhui Medical UniversityHefei230022China
- Key Laboratory of Environmental Toxicology of Anhui Higher Education InstitutesAnhui Medical UniversityHefei230032China
| | - Yan‐Yan Zhu
- Department of ToxicologySchool of Public HealthAnhui Medical UniversityHefei230022China
- Key Laboratory of Environmental Toxicology of Anhui Higher Education InstitutesAnhui Medical UniversityHefei230032China
| | - Xiao‐Xi Chen
- Department of ToxicologySchool of Public HealthAnhui Medical UniversityHefei230022China
- Key Laboratory of Environmental Toxicology of Anhui Higher Education InstitutesAnhui Medical UniversityHefei230032China
| | - Tao Wang
- Key Laboratory of Environmental Toxicology of Anhui Higher Education InstitutesAnhui Medical UniversityHefei230032China
| | - Jing Shao
- Key Laboratory of Environmental Toxicology of Anhui Higher Education InstitutesAnhui Medical UniversityHefei230032China
| | - Xiu‐Hong Meng
- Key Laboratory of Environmental Toxicology of Anhui Higher Education InstitutesAnhui Medical UniversityHefei230032China
| | - Yichao Huang
- Department of ToxicologySchool of Public HealthAnhui Medical UniversityHefei230022China
- Key Laboratory of Environmental Toxicology of Anhui Higher Education InstitutesAnhui Medical UniversityHefei230032China
| | - Hua Wang
- Department of ToxicologySchool of Public HealthAnhui Medical UniversityHefei230022China
- Key Laboratory of Environmental Toxicology of Anhui Higher Education InstitutesAnhui Medical UniversityHefei230032China
| | - Hui‐Li Wang
- School of Food and BioengineeringHefei University of TechnologyHefei230009China
| | - Bo Wang
- Department of ToxicologySchool of Public HealthAnhui Medical UniversityHefei230022China
- Key Laboratory of Environmental Toxicology of Anhui Higher Education InstitutesAnhui Medical UniversityHefei230032China
| | - De‐Xiang Xu
- Department of ToxicologySchool of Public HealthAnhui Medical UniversityHefei230022China
- Key Laboratory of Environmental Toxicology of Anhui Higher Education InstitutesAnhui Medical UniversityHefei230032China
| |
Collapse
|
16
|
Indika NLR, Senarathne UD, Malvaso A, Darshana D, Owens SC, Mansouri B, Semenova Y, Bjørklund G. Abnormal Porphyrin Metabolism in Autism Spectrum Disorder and Therapeutic Implications. Mol Neurobiol 2024; 61:3851-3866. [PMID: 38032468 DOI: 10.1007/s12035-023-03722-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 10/17/2023] [Indexed: 12/01/2023]
Abstract
Autism spectrum disorder (ASD) is a mosaic of neurodevelopmental conditions composed of early-onset social interaction and communication deficits, along with repetitive and/or restricted patterns of activities, behavior, and interests. ASD affects around 1% of children worldwide, with a male predominance. Energy, porphyrin, and neurotransmitter homeostasis are the key metabolic pathways affected by heavy metal exposure, potentially implicated in the pathogenesis of ASD. Exposure to heavy metals can lead to an altered porphyrin metabolism due to enzyme inhibition by heavy metals. Heavy metal exposure, inborn genetic susceptibility, and abnormal thiol and selenol metabolism may play a significant role in the urinary porphyrin profile anomalies observed in ASD. Altered porphyrin metabolism in ASD may also be associated with, vitamin B6 deficiency, hyperoxalemia, hyperhomocysteinemia, and hypomagnesemia. The present review considers the abnormal porphyrin metabolism in ASD in relation to the potential pathogenic mechanism and discusses the possible metabolic therapies such as vitamins, minerals, cofactors, and antioxidants that need to be explored in future research. Such targeted therapeutic therapies would bring about favorable outcomes such as improvements in core and co-occurring symptoms.
Collapse
Affiliation(s)
- Neluwa-Liyanage R Indika
- Department of Biochemistry, Faculty of Medical Sciences, University of Sri Jayewardenepura, Nugegoda, 10250, Sri Lanka.
| | - Udara D Senarathne
- Department of Biochemistry, Faculty of Medical Sciences, University of Sri Jayewardenepura, Nugegoda, 10250, Sri Lanka
- Department of Chemical Pathology, Monash Health Pathology, Monash Health, Clayton, Victoria, Australia
| | - Antonio Malvaso
- IRCCS "C. Mondino" Foundation, National Neurological Institute, Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Dhanushka Darshana
- Department of Pharmacy, Faculty of Allied Health Sciences, University of Ruhuna, Galle, Sri Lanka
| | - Susan C Owens
- Autism Oxalate Project, Autism Research Institute, San Diego, CA, USA
| | - Borhan Mansouri
- Substance Abuse Prevention Research Center, Research Institute for Health, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Yuliya Semenova
- Nazarbayev University School of Medicine, Astana, Kazakhstan
| | - Geir Bjørklund
- Council for Nutritional and Environmental Medicine, Toften 24, 8610, Mo i Rana, Norway.
| |
Collapse
|
17
|
Singh J, Vanlallawmzuali, Singh A, Biswal S, Zomuansangi R, Lalbiaktluangi C, Singh BP, Singh PK, Vellingiri B, Iyer M, Ram H, Udey B, Yadav MK. Microbiota-brain axis: Exploring the role of gut microbiota in psychiatric disorders - A comprehensive review. Asian J Psychiatr 2024; 97:104068. [PMID: 38776563 DOI: 10.1016/j.ajp.2024.104068] [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: 11/22/2023] [Revised: 02/28/2024] [Accepted: 05/01/2024] [Indexed: 05/25/2024]
Abstract
Mental illness is a hidden epidemic in modern science that has gradually spread worldwide. According to estimates from the World Health Organization (WHO), approximately 10% of the world's population suffers from various mental diseases each year. Worldwide, financial and health burdens on society are increasing annually. Therefore, understanding the different factors that can influence mental illness is required to formulate novel and effective treatments and interventions to combat mental illness. Gut microbiota, consisting of diverse microbial communities residing in the gastrointestinal tract, exert profound effects on the central nervous system through the gut-brain axis. The gut-brain axis serves as a conduit for bidirectional communication between the two systems, enabling the gut microbiota to affect emotional and cognitive functions. Dysbiosis, or an imbalance in the gut microbiota, is associated with an increased susceptibility to mental health disorders and psychiatric illnesses. Gut microbiota is one of the most diverse and abundant groups of microbes that have been found to interact with the central nervous system and play important physiological functions in the human gut, thus greatly affecting the development of mental illnesses. The interaction between gut microbiota and mental health-related illnesses is a multifaceted and promising field of study. This review explores the mechanisms by which gut microbiota influences mental health, encompassing the modulation of neurotransmitter production, neuroinflammation, and integrity of the gut barrier. In addition, it emphasizes a thorough understanding of how the gut microbiome affects various psychiatric conditions.
Collapse
Affiliation(s)
- Jawahar Singh
- Department of Psychiatry, All India Institute of Medical Sciences (AIIMS), Bathinda, Punjab, India
| | - Vanlallawmzuali
- Department of Biotechnology, Mizoram Central University, Pachhunga University College Campus, Aizawl, Mizoram, India
| | - Amit Singh
- Department of Microbiology Central University of Punjab, Bathinda 151401, India
| | - Suryanarayan Biswal
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda 151401, India
| | - Ruth Zomuansangi
- Department of Microbiology Central University of Punjab, Bathinda 151401, India
| | - C Lalbiaktluangi
- Department of Microbiology Central University of Punjab, Bathinda 151401, India
| | - Bhim Pratap Singh
- Department of Agriculture and Environmental Sciences (AES), National Institute of Food Technology Entrepreneurship and Management (NIFTEM), Sonepat, Haryana, India
| | - Prashant Kumar Singh
- Department of Biotechnology, Pachhunga University College Campus, Mizoram University (A Central University), Aizawl 796001, Mizoram, India
| | - Balachandar Vellingiri
- Stem cell and Regenerative Medicine/Translational Research, Department of Zoology, School of Basic Sciences, Central University of Punjab (CUPB), Bathinda, Punjab 151401, India
| | - Mahalaxmi Iyer
- Department of Microbiology Central University of Punjab, Bathinda 151401, India
| | - Heera Ram
- Department of Zoology, Jai Narain Vyas University, Jodhpur, Rajasthan 342001, India
| | - Bharat Udey
- Department of Psychiatry, All India Institute of Medical Sciences (AIIMS), Bathinda, Punjab, India
| | - Mukesh Kumar Yadav
- Department of Microbiology Central University of Punjab, Bathinda 151401, India.
| |
Collapse
|
18
|
Młynarska E, Wasiak J, Gajewska A, Steć G, Jasińska J, Rysz J, Franczyk B. Exploring the Significance of Gut Microbiota in Diabetes Pathogenesis and Management-A Narrative Review. Nutrients 2024; 16:1938. [PMID: 38931292 PMCID: PMC11206785 DOI: 10.3390/nu16121938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/13/2024] [Accepted: 06/16/2024] [Indexed: 06/28/2024] Open
Abstract
Type 2 diabetes is a disease with significant health consequences for the individual. Currently, new mechanisms and therapeutic approaches that may affect this disease are being sought. One of them is the association of type 2 diabetes with microbiota. Through the enteric nervous system and the gut-microbiota axis, the microbiota affects the functioning of the body. It has been proven to have a real impact on influencing glucose and lipid metabolism and insulin sensitivity. With dysbiosis, there is increased bacterial translocation through the disrupted intestinal barrier and increased inflammation in the body. In diabetes, the microbiota's composition is altered with, for example, a more abundant class of Betaproteobacteria. The consequences of these disorders are linked to mechanisms involving short-chain fatty acids, branched-chain amino acids, and bacterial lipopolysaccharide, among others. Interventions focusing on the gut microbiota are gaining traction as a promising approach to diabetes management. Studies are currently being conducted on the effects of the supply of probiotics and prebiotics, as well as fecal microbiota transplantation, on the course of diabetes. Further research will allow us to fully develop our knowledge on the subject and possibly best treat and prevent type 2 diabetes.
Collapse
Affiliation(s)
- Ewelina Młynarska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Jakub Wasiak
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Agata Gajewska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Greta Steć
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Joanna Jasińska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Beata Franczyk
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| |
Collapse
|
19
|
Dai H, Jiang Y, Liu S, Li D, Zhang X. Dietary flavonoids modulate the gut microbiota: A new perspective on improving autism spectrum disorder through the gut-brain axis. Food Res Int 2024; 186:114404. [PMID: 38729686 DOI: 10.1016/j.foodres.2024.114404] [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: 12/19/2023] [Revised: 04/18/2024] [Accepted: 04/20/2024] [Indexed: 05/12/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder with an unknown etiology. It is associated with various factors and causes great inconvenience to the patient's life. The gut-brain axis (GBA), which serves as a bidirectional information channel for exchanging information between the gut microbiota and the brain, is vital in studying many neurodegenerative diseases. Dietary flavonoids provide anti-inflammatory and antioxidant benefits, as well as regulating the structure and function of the gut microbiota. The occurrence and development of ASD are associated with dysbiosis of the gut microbiota. Modulation of gut microbiota can effectively improve the severity of ASD. This paper reviews the links between gut microbiota, flavonoids, and ASD, focusing on the mechanism of dietary flavonoids in regulating ASD through the GBA.
Collapse
Affiliation(s)
- Haochen Dai
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, PR China
| | - Yuhan Jiang
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, PR China
| | - Shuxun Liu
- College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, PR China.
| | - Dandan Li
- Sinograin Chengdu Storage Research Institute Co., Ltd, Chengdu 610091, PR China
| | - Xin Zhang
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, PR China.
| |
Collapse
|
20
|
Xie X, Zhou R, Fang Z, Zhang Y, Wang Q, Liu X. Seeing beyond words: Visualizing autism spectrum disorder biomarker insights. Heliyon 2024; 10:e30420. [PMID: 38694128 PMCID: PMC11061761 DOI: 10.1016/j.heliyon.2024.e30420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/04/2024] Open
Abstract
Objective This study employs bibliometric and visual analysis to elucidate global research trends in Autism Spectrum Disorder (ASD) biomarkers, identify critical research focal points, and discuss the potential integration of diverse biomarker modalities for precise ASD assessment. Methods A comprehensive bibliometric analysis was conducted using data from the Web of Science Core Collection database until December 31, 2022. Visualization tools, including R, VOSviewer, CiteSpace, and gCLUTO, were utilized to examine collaborative networks, co-citation patterns, and keyword associations among countries, institutions, authors, journals, documents, and keywords. Results ASD biomarker research emerged in 2004, accumulating a corpus of 4348 documents by December 31, 2022. The United States, with 1574 publications and an H-index of 213, emerged as the most prolific and influential country. The University of California, Davis, contributed significantly with 346 publications and an H-index of 69, making it the leading institution. Concerning journals, the Journal of Autism and Developmental Disorders, Autism Research, and PLOS ONE were the top three publishers of ASD biomarker-related articles among a total of 1140 academic journals. Co-citation and keyword analyses revealed research hotspots in genetics, imaging, oxidative stress, neuroinflammation, gut microbiota, and eye tracking. Emerging topics included "DNA methylation," "eye tracking," "metabolomics," and "resting-state fMRI." Conclusion The field of ASD biomarker research is dynamically evolving. Future endeavors should prioritize individual stratification, methodological standardization, the harmonious integration of biomarker modalities, and longitudinal studies to advance the precision of ASD diagnosis and treatment.
Collapse
Affiliation(s)
- Xinyue Xie
- The First Affiliated Hospital of Henan University of Chinese Medicine, Pediatrics Hospital, Zhengzhou, Henan, 450000, China
- Henan University of Chinese Medicine, School of Pediatrics, Zhengzhou, Henan, 450046, China
| | - Rongyi Zhou
- The First Affiliated Hospital of Henan University of Chinese Medicine, Pediatrics Hospital, Zhengzhou, Henan, 450000, China
- Henan University of Chinese Medicine, School of Pediatrics, Zhengzhou, Henan, 450046, China
| | - Zihan Fang
- Henan University of Chinese Medicine, School of Pediatrics, Zhengzhou, Henan, 450046, China
| | - Yongting Zhang
- The First Affiliated Hospital of Henan University of Chinese Medicine, Pediatrics Hospital, Zhengzhou, Henan, 450000, China
- Henan University of Chinese Medicine, School of Pediatrics, Zhengzhou, Henan, 450046, China
| | - Qirong Wang
- Henan University of Chinese Medicine, School of Pediatrics, Zhengzhou, Henan, 450046, China
| | - Xiaomian Liu
- Henan University of Chinese Medicine, School of Medicine, Zhengzhou, Henan, 450046, China
| |
Collapse
|
21
|
Shi Z, Wang S, Chen M, Hu A, Long Q, Lee Y. The effect of music therapy on language communication and social skills in children with autism spectrum disorder: a systematic review and meta-analysis. Front Psychol 2024; 15:1336421. [PMID: 38774719 PMCID: PMC11106491 DOI: 10.3389/fpsyg.2024.1336421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 04/12/2024] [Indexed: 05/24/2024] Open
Abstract
Background Studies have shown that music therapy can be used as a therapeutic aid for clinical disorders. To evaluate the effects of music therapy (MT) on language communication and social skills in children with autism spectrum disorder (ASD), a meta-analysis was performed on eligible studies in this field. Methods A systematic search was conducted in eight databases: PubMed, Embase, Web of Science, Cochrane Library databases, the China National Knowledge Infrastructure (CNKI), Wanfang Data, the Chinese Biomedical Literature (CBM) Database, and the VIP Chinese Science and Technology Periodicals Database. The standard mean difference (SMD) values were used to evaluate outcomes, and the pooled proportions and SMD with their 95% confidence intervals (CIs) were also calculated. Results Eighteen randomized controlled trial (RCT) studies were included, with a total of 1,457 children with ASD. This meta-analysis revealed that music therapy improved their language communication [SMD = -1.20; 95%CI -1.45, -0.94; χ2 (17) = 84.17, I2 = 80%, p < 0.001] and social skills [SMD = -1. 13; 95%CI -1.49, -0.78; χ2 (17) = 162.53, I2 = 90%, p < 0.001]. In addition, behavior [SMD = -1.92; 94%CI -2.56, -1.28; χ2 (13) = 235.08, I2 = 95%, p < 0.001], sensory perception [SMD = -1.62; 95%CI -2.17, -1.08; χ2 (16) = 303.80, I2 = 95%, p < 0.001], self-help [SMD = -2. 14; 95%CI -3.17, -1.10; χ2 (6) = 173.07, I2 = 97%, p < 0.001] were all improved. Conclusion Music therapy has a positive effect on the improvement of symptoms in children with ASD. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/.
Collapse
Affiliation(s)
- Zijuan Shi
- North Sichuan Medical College, Nanchong, China
| | - Si Wang
- North Sichuan Medical College, Nanchong, China
| | | | - Aimin Hu
- North Sichuan Medical College, Nanchong, China
| | | | - Yujun Lee
- North Sichuan Medical College, Nanchong, China
- The Graduate School of Xi’an International Studies University, Shaanxi, China
| |
Collapse
|
22
|
Hazan S, Haroon J, Jordan S, Walker SJ. Improvements in Gut Microbiome Composition and Clinical Symptoms Following Familial Fecal Microbiota Transplantation in a Nineteen-Year-Old Adolescent With Severe Autism. J Med Cases 2024; 15:82-91. [PMID: 38715916 PMCID: PMC11073461 DOI: 10.14740/jmc4209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 04/11/2024] [Indexed: 06/11/2024] Open
Abstract
This case report describes a novel therapy for patients with severe autism spectrum disorder (ASD) that is worth further investigation. A 19-year-old male adolescent with ASD, who was not responding to standard treatment received fecal microbiota transplant (FMT) using donor material from his typically developing female sibling. The patient's ASD symptoms were assessed by assessors who were blind to the patient's past ASD symptomatology. Assessors used the Childhood Autism Rating Scale (CARS), an observation-based rating scale to assess developmental delay in children with autism (range of CARS scores is 15 - 60; a score > 28 is indicative of autism; higher score is positively correlated with degree of severity), at baseline and again at six timepoints post-FMT. The patient experienced marked improvements in microbiome diversity and composition over the year and a half period that followed the FMT procedure. Additionally, the patient who was previously nonverbal said his first two words and experienced a reduction in aggression 1-month post-FMT. To the authors' knowledge, this is the first report to demonstrate the use of familial FMT in an adolescent patient with ASD. Given that ASD symptom improvements post-FMT tend to occur in younger patients, the authors hypothesize that the use of a familial donor may be an important factor that contributed to the improved outcomes experienced by this older child.
Collapse
Affiliation(s)
- Sabine Hazan
- ProgenaBiome, LLC, Ventura, CA, USA
- Microbiome Research Foundation, Ventura, CA, USA
| | | | | | - Stephen J. Walker
- Wake Forest Institute for Regenerative Medicine, Winston Salem, NC, USA
| |
Collapse
|
23
|
Lam EWF, Ip BPI. The prevalence of constipation in adult psychiatric out-patients on clozapine treatment at a regional public hospital in Hong Kong. Hum Psychopharmacol 2024:e2897. [PMID: 38605548 DOI: 10.1002/hup.2897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/20/2024] [Accepted: 03/28/2024] [Indexed: 04/13/2024]
Abstract
OBJECTIVE To determine the occurrence of constipation in local patients on clozapine treatment, and to compare the demographical and clinical characteristics of patients on clozapine treatment with or without constipation. METHODS This is a cross-sectional, observational study. All adult psychiatric out-patients on clozapine treatment attending follow-up at a regional hospital were recruited for clinical interview and medical record review. The Enhanced Asian Rome III Questionnaire (EAR3Q) was used to define patients with constipation. The Bristol Stool Form Scale (BSFS) was used to assess stool form. The Brief Psychiatric Rating Scale-Anchored (BPRS-A) was used to measure psychiatric symptoms. The Brief Medication Adherence Scale (BMAS) was used to assess treatment adherence. Logistic regression was conducted to identify independent associating factors of constipation in patients on clozapine treatment. RESULTS The prevalence of constipation in patients on clozapine treatment was 26.3%, (95% CI [21.5%, 31.6%]). Independent associating factors included disorder of psychological development (aOR = 6.98, 95% CI [1.24, 39.18]), anxiety (very mild: aOR = 9.23, 95% CI [2.59, 32.87]; mild: aOR = 2.66, 95% CI [1.26, 5.62]), prescription with combination of laxatives (aOR = 0.40, 95% CI [0.17, 0.95]), and concomitant use of amisulpride (aOR = 2.52, 95% CI [1.09, 5.82]), quetiapine (aOR = 5.92, 95% CI [1.11, 31.56]) and metamucil (aOR = 9.30, 95% CI [1.53, 56.58]). CONCLUSION This study examined the prevalence of clozapine-associated constipation in Hong Kong using a validated questionnaire. The identification of independent factors associated with constipation could facilitate better risk stratification and risk modification in clinical practice.
Collapse
Affiliation(s)
- Eric Wai-Fung Lam
- Department of Psychiatry, Pamela Youde Nethersole Eastern Hospital, Hong Kong, China
| | - Brian Pak-In Ip
- Department of Psychiatry, Pamela Youde Nethersole Eastern Hospital, Hong Kong, China
| |
Collapse
|
24
|
Salari Z, Moslemizadeh A, Tezerji SS, Sabet N, Parizi AS, Khaksari M, Sheibani V, Jafari E, Shafieipour S, Bashiri H. Sex-dependent alterations of inflammatory factors, oxidative stress, and histopathology of the brain-gut axis in a VPA-induced autistic-like model of rats. Birth Defects Res 2024; 116:e2310. [PMID: 38563145 DOI: 10.1002/bdr2.2310] [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: 08/03/2023] [Revised: 12/23/2023] [Accepted: 01/21/2024] [Indexed: 04/04/2024]
Abstract
INTRODUCTION In this study, we aimed to investigate the inflammatory factors, oxidative stress, and histopathological consequences of the brain-gut axis in male and female rats prenatally exposed to VPA. METHODS Pregnant Wistar rats were randomly divided into two groups. The animals received saline, and valproic acid (VPA) (600 mg/kg, i.p.) on embryonic day 12.5 (E12.5). All offspring were weaned on postnatal day 21, and the experiments were done in male and female rats on day 60. The brain and intestine tissues were extracted to assess histopathology, inflammation, and oxidative stress. RESULTS An increase of interleukin-1β (IL-1β) and interleukin-6 (IL-6) and a decrease of interleukin-10 (IL-10) were observed in the two sexes and two tissues of the autistic rats. In the VPA-exposed animals, malondialdehyde (MDA) and protein carbonyl (PC) increased in the brain of both sexes and the intestines of only the males. The total antioxidant capacity (TAC), superoxide dismutase (SOD), and catalase (CAT) significantly decreased in both tissues of male and female autistic groups. Histopathological evaluation showed that the %apoptosis of the cortex in the autistic male and female groups was more than in controls whereas this parameter in the CA1 and CA3 was significant only in the male rats. In the intestine, histopathologic changes were seen only in the male autistic animals. CONCLUSION The inflammatory and antioxidant factors were in line in the brain-gut axis in male and female rats prenatally exposed to VPA. Histopathological consequences were more significant in the VPA-exposed male animals.
Collapse
Affiliation(s)
- Zahra Salari
- Gastroenterology and Hepatology Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Sara Sheibani Tezerji
- Department of Behavioural and Molecular Neurobiology, Regensburg Center for Neuroscience, University of Regensburg, Regensburg, Germany
| | - Nazanin Sabet
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Ali Saeidpour Parizi
- Gastroenterology and Hepatology Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Khaksari
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Vahid Sheibani
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
- Department of Physiology and Pharmacology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Elham Jafari
- Pathology and Stem Cells Research Center, Department of Pathology, Afzalipour School of Medicine, Kerman University of Medical Science, Kerman, Iran
| | - Sara Shafieipour
- Gastroenterology and Hepatology Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Hamideh Bashiri
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
- Department of Physiology and Pharmacology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
25
|
Fu Z, Yang X, Jiang Y, Mao X, Liu H, Yang Y, Chen J, Chen Z, Li H, Zhang XS, Mao X, Li N, Wang D, Jiang J. Microbiota profiling reveals alteration of gut microbial neurotransmitters in a mouse model of autism-associated 16p11.2 microduplication. Front Microbiol 2024; 15:1331130. [PMID: 38596370 PMCID: PMC11002229 DOI: 10.3389/fmicb.2024.1331130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/27/2024] [Indexed: 04/11/2024] Open
Abstract
The gut-brain axis is evident in modulating neuropsychiatric diseases including autism spectrum disorder (ASD). Chromosomal 16p11.2 microduplication 16p11.2dp/+ is among the most prevalent genetic copy number variations (CNV) linked with ASD. However, the implications of gut microbiota status underlying the development of ASD-like impairments induced by 16p11.2dp/+ remains unclear. To address this, we initially investigated a mouse model of 16p11.2dp/+, which exhibits social novelty deficit and repetitive behavior characteristic of ASD. Subsequently, we conducted a comparative analysis of the gut microbial community and metabolomic profiles between 16p11.2dp/+ and their wild-type counterparts using 16S rRNA sequencing and liquid chromatography-mass spectrometry (LC/MS). Our microbiota analysis revealed structural dysbiosis in 16p11.2dp/+ mice, characterized by reduced biodiversity and alterations in species abundance, as indicated by α/β-diversity analysis. Specifically, we observed reduced relative abundances of Faecalibaculum and Romboutsia, accompanied by an increase in Turicibacter and Prevotellaceae UCG_001 in 16p11.2dp/+ group. Metabolomic analysis identified 19 significantly altered metabolites and unveiled enriched amino acid metabolism pathways. Notably, a disruption in the predominantly histamine-centered neurotransmitter network was observed in 16p11.2dp/+ mice. Collectively, our findings delineate potential alterations and correlations among the gut microbiota and microbial neurotransmitters in 16p11.2dp/+ mice, providing new insights into the pathogenesis of and treatment for 16p11.2 CNV-associated ASD.
Collapse
Affiliation(s)
- Zhang Fu
- Tomas Lindhal Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Xiuyan Yang
- Tomas Lindhal Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Youheng Jiang
- Tomas Lindhal Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Xinliang Mao
- Guangdong Perfect Life Health Science and Technology Research Institute Co., Ltd., Zhongshan, Guangdong, China
| | - Hualin Liu
- Guangdong Perfect Life Health Science and Technology Research Institute Co., Ltd., Zhongshan, Guangdong, China
| | - Yanming Yang
- Tomas Lindhal Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Jia Chen
- Tomas Lindhal Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Zhumei Chen
- Tomas Lindhal Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
- Department of Anesthesiology, The Seventh Affiliated Hospital of Sun Yat-Sen University (SYSU), Shenzhen, Guangdong, China
| | - Huiliang Li
- Division of Medicine, Wolfson Institute for Biomedical Research, Faculty of Medical Sciences, University College London, London, United Kingdom
- China-UK Institute for Frontier Science, Shenzhen, Guangdong, China
| | - Xue-Song Zhang
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, United States
| | - Xinjun Mao
- Department of Anesthesiology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Ningning Li
- Tomas Lindhal Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
- China-UK Institute for Frontier Science, Shenzhen, Guangdong, China
| | - Dilong Wang
- Department of Pediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jian Jiang
- Tomas Lindhal Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| |
Collapse
|
26
|
Barba-Vila O, García-Mieres H, Ramos B. Probiotics in autism spectrum disorders: a systematic review of clinical studies and future directions. Nutr Rev 2024:nuae010. [PMID: 38497979 DOI: 10.1093/nutrit/nuae010] [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] [Indexed: 03/19/2024] Open
Abstract
CONTEXT It is hypothesized that gut dysbiosis, a typical feature of patients with autism spectrum disorder (ASD), could be involved in the origin of this neurodevelopmental disorder. Therefore, the use of probiotics to restore gastrointestinal (GI) equilibrium might be a promising therapeutic strategy due to its capacity to balance the gut-brain axis and behavioral responses. OBJECTIVE To summarize current knowledge on the use of probiotics to treat core clinical ASD symptoms and concomitant GI signs, compare the design of published studies with those of ongoing trials, assess the near future of this field, and provide recommendations for improving novel studies. DATA SOURCES The literature search was conducted in February 2020 and updated in March 2021, using a broad range of bibliographic and clinical trial-specific databases. DATA EXTRACTION Data were extracted using a standardized form, and articles reporting on 28 clinical studies (already published or still ongoing) were included. The risk of bias in clinical studies was evaluated using the Cochrane Collaboration Risk of Bias Assessment tool for randomized trials and the Risk of Bias in Nonrandomized Studies-Interventions tool for nonrandomized trials. RESULTS The results suggest that probiotics improve ASD-like social deficits, GI symptoms, and gut microbiota profile. However, inconsistencies among studies and their methodological limitations make it difficult to draw any conclusions regarding the efficacy of probiotics in ASD. This review provides specific suggestions for future research to improve the quality of the studies. CONCLUSIONS Although ongoing studies have improved designs, the available knowledge does not permit solid conclusions to be made regarding the efficacy of probiotics in ameliorating the symptoms (psychiatric and/or GI) associated with ASD. Thus, more high-quality research and new approaches are needed to design effective probiotic strategies for ASD.
Collapse
Affiliation(s)
- Olga Barba-Vila
- Department de Bioquímica i Biología Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona Bellaterra, Barcelona, Spain
| | - Helena García-Mieres
- Etiopathogenesis and Treatment of Severe Mental Disorders, Teaching, Research, and Innovation Unit, Institut de Recerca Sant Joan de Déu, Parc Sanitari Sant Joan de Déu Sant Boi de Llobregat, Barcelona, Spain
- Centro Investigación Biomédica en Red Salud Mental, Madrid, Spain
- Health Services Research Unit, Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona, Spain
- CIBER Epidemiología y Salud Pública, Madrid, Spain
- Department of Medicine and Health Sciences, Pompeu Fabra University, Barcelona, Spain
- Faculty of Medicine, University of Vic-Central University of Catalonia, Vic, Spain
| | - Belén Ramos
- Department de Bioquímica i Biología Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona Bellaterra, Barcelona, Spain
- Etiopathogenesis and Treatment of Severe Mental Disorders, Teaching, Research, and Innovation Unit, Institut de Recerca Sant Joan de Déu, Parc Sanitari Sant Joan de Déu Sant Boi de Llobregat, Barcelona, Spain
- Centro Investigación Biomédica en Red Salud Mental, Madrid, Spain
| |
Collapse
|
27
|
Smolko NA, Valiev RI, Kabdesh IM, Fayzullina RA, Mukhamedshina YO. Eating disorder in children: Impact on quality of life, with a spotlight on autism spectrum disorder. Nutr Res 2024; 123:38-52. [PMID: 38241984 DOI: 10.1016/j.nutres.2023.12.006] [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: 07/17/2023] [Revised: 12/18/2023] [Accepted: 12/18/2023] [Indexed: 01/21/2024]
Abstract
Eating behavior, which includes eating habits and preferences, frequency of eating, and other features related to diet, is a major characteristic not only of a person's nutritional status, but also of health in general. In recent years, the prevalence of eating disorders in children has tended to increase; they also require cross-system approaches in diagnosis by a variety of specialists and correction requires appropriate selection of optimal methods. Maladaptive eating attitudes formed at an early age can contribute to the formation of eating disorders, which can lead to or worsen various neuropsychiatric diseases, digestive diseases, and other related conditions. In children with autism spectrum disorder (ASD), eating disorders often appear earlier than other major symptoms of the condition. However, the clinical manifestations of eating disorders in children with ASD are varied and differ in severity and duration, whereas these disorders in neurotypical children might present as short-lived and may not lead to serious consequences. Nevertheless, cases of progressive eating disorders accompanied by a child presenting as under- or overweight and/or with macronutrient and micronutrient deficiencies cannot be excluded. Given the high prevalence of eating disorders in children, many researchers have highlighted the lack of a valid and universally accepted instruments to assess atypical eating behaviors in this population. Therefore, in this review, we wanted to highlight the problems and causes of eating disorders in children, and also to analyze the existing approaches to the validation of these problems, taking into account the existing behavioral features in children with ASD.
Collapse
Affiliation(s)
- Natalia A Smolko
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia; Department of Propaedeutics of Pediatric Diseases and Faculty Pediatrics, Kazan State Medical University, Kazan, Russia
| | - Rushan I Valiev
- Department of General Hygiene, Kazan State Medical University, Kazan, Russia
| | - Ilyas M Kabdesh
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia.
| | - Rezeda A Fayzullina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia; Department of Propaedeutics of Pediatric Diseases and Faculty Pediatrics, Kazan State Medical University, Kazan, Russia
| | - Yana O Mukhamedshina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia; Department of Histology, Cytology and Embryology, Kazan State Medical University, Kazan, Russia
| |
Collapse
|
28
|
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
|
29
|
Breda C, Santero S, Conti MV, Cena H. Programmes to manage food selectivity in individuals with autism spectrum disorder. Nutr Res Rev 2024:1-14. [PMID: 38385245 DOI: 10.1017/s0954422424000052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
Autism spectrum disorder (ASD) is a complex group of neurodevelopmental disorders characterised by impaired social communication and restricted interests/repetitive behaviours. In this regard, sensory processing difficulties and delayed oral motor skills often predispose individuals with ASD to food selectivity (FS). It is usually associated with repetitive eating patterns that can lead to multiple malnutrition conditions. The objective of this narrative review is to present an overview about the existing nutritional interventions aiming at promoting a healthy eating pattern and addressing food selectivity among individuals with ASD. Regarding the interventions targeting nutrition education, the majority of the analysed studies failed to demonstrate their effectiveness. On the other hand, many educational interventions involving taste or cooking sessions, as well as behavioural interventions for FS, demonstrated effective results. Moreover, multidisciplinary in tailoring such programmes, including psychology speech therapy and nutritional skills, is acknowledged as a key approach.
Collapse
Affiliation(s)
- Chiara Breda
- Laboratory of Dietetics and Clinical Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, via Bassi 21, Pavia, Italy
| | - Sara Santero
- Laboratory of Dietetics and Clinical Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, via Bassi 21, Pavia, Italy
| | - Maria Vittoria Conti
- Laboratory of Dietetics and Clinical Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, via Bassi 21, Pavia, Italy
| | - Hellas Cena
- Laboratory of Dietetics and Clinical Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, via Bassi 21, Pavia, Italy
- Clinical Nutrition and Dietetics Service, Unit of Internal Medicine and Endocrinology, ICS Maugeri IRCCS, University of Pavia, Pavia, Italy
| |
Collapse
|
30
|
Feng P, Zhang Y, Zhao Y, Zhao P, Li E. Combined repetitive transcranial magnetic stimulation and gut microbiota modulation through the gut-brain axis for prevention and treatment of autism spectrum disorder. Front Immunol 2024; 15:1341404. [PMID: 38455067 PMCID: PMC10918007 DOI: 10.3389/fimmu.2024.1341404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 02/07/2024] [Indexed: 03/09/2024] Open
Abstract
Autism spectrum disorder (ASD) encompasses a range of neurodevelopmental conditions characterized by enduring impairments in social communication and interaction together with restricted repetitive behaviors, interests, and activities. No targeted pharmacological or physical interventions are currently available for ASD. However, emerging evidence has indicated a potential association between the development of ASD and dysregulation of the gut-brain axis. Repetitive transcranial magnetic stimulation (rTMS), a noninvasive diagnostic and therapeutic approach, has demonstrated positive outcomes in diverse psychiatric disorders; however, its efficacy in treating ASD and its accompanying gastrointestinal effects, particularly the effects on the gut-brain axis, remain unclear. Hence, this review aimed to thoroughly examine the existing research on the application of rTMS in the treatment of ASD. Additionally, the review explored the interplay between rTMS and the gut microbiota in children with ASD, focusing on the gut-brain axis. Furthermore, the review delved into the integration of rTMS and gut microbiota modulation as a targeted approach for ASD treatment based on recent literature. This review emphasizes the potential synergistic effects of rTMS and gut microbiota interventions, describes the underlying mechanisms, and proposes a potential therapeutic strategy for specific subsets of individuals with ASD.
Collapse
Affiliation(s)
- Pengya Feng
- Department of Children Rehabilitation, Key Laboratory of Rehabilitation Medicine in Henan, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- The American Psychiatric Association, Key Laboratory of Helicobacter pylori, Microbiota and Gastrointestinal Cancer of Henan Province, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yangyang Zhang
- Department of Children Rehabilitation, Key Laboratory of Rehabilitation Medicine in Henan, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yonghong Zhao
- Department of Children Rehabilitation, Key Laboratory of Rehabilitation Medicine in Henan, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Pengju Zhao
- Department of Children Rehabilitation, Key Laboratory of Rehabilitation Medicine in Henan, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Enyao Li
- Department of Children Rehabilitation, Key Laboratory of Rehabilitation Medicine in Henan, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
31
|
Suprunowicz M, Tomaszek N, Urbaniak A, Zackiewicz K, Modzelewski S, Waszkiewicz N. Between Dysbiosis, Maternal Immune Activation and Autism: Is There a Common Pathway? Nutrients 2024; 16:549. [PMID: 38398873 PMCID: PMC10891846 DOI: 10.3390/nu16040549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/05/2024] [Accepted: 02/15/2024] [Indexed: 02/25/2024] Open
Abstract
Autism spectrum disorder (ASD) is a neuropsychiatric condition characterized by impaired social interactions and repetitive stereotyped behaviors. Growing evidence highlights an important role of the gut-brain-microbiome axis in the pathogenesis of ASD. Research indicates an abnormal composition of the gut microbiome and the potential involvement of bacterial molecules in neuroinflammation and brain development disruptions. Concurrently, attention is directed towards the role of short-chain fatty acids (SCFAs) and impaired intestinal tightness. This comprehensive review emphasizes the potential impact of maternal gut microbiota changes on the development of autism in children, especially considering maternal immune activation (MIA). The following paper evaluates the impact of the birth route on the colonization of the child with bacteria in the first weeks of life. Furthermore, it explores the role of pro-inflammatory cytokines, such as IL-6 and IL-17a and mother's obesity as potentially environmental factors of ASD. The purpose of this review is to advance our understanding of ASD pathogenesis, while also searching for the positive implications of the latest therapies, such as probiotics, prebiotics or fecal microbiota transplantation, targeting the gut microbiota and reducing inflammation. This review aims to provide valuable insights that could instruct future studies and treatments for individuals affected by ASD.
Collapse
Affiliation(s)
| | | | | | | | - Stefan Modzelewski
- Department of Psychiatry, Medical University of Bialystok, pl. Wołodyjowskiego 2, 15-272 Białystok, Poland; (M.S.); (N.T.); (A.U.); (K.Z.); (N.W.)
| | | |
Collapse
|
32
|
Ozorio Dutra SV, Sarkar A, Yoo JY, Shaffer-Hudkins E, Groer M. Premature Infant Gut Microbiome relationships with childhood behavioral scales: preliminary insights. Front Nutr 2024; 10:1294549. [PMID: 38419643 PMCID: PMC10899318 DOI: 10.3389/fnut.2023.1294549] [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: 09/14/2023] [Accepted: 11/20/2023] [Indexed: 03/02/2024] Open
Abstract
Introduction Very Low Birth Weight (VLBW) infants, born weighing less than 1,500 grams, are at risk for both gut dysbiosis and later neuropsychological developmental deficits. Behavioral effects, while related to neurodevelopment, are often more subtle and difficult to measure. The extent of later neurobehavioral consequences associated with such microbial dysbiosis has yet to be determined. We explored associations between the infants' gut microbiome and early childhood behavior at 4 years of age and identified the bacterial taxa through a multivariate analysis by linear models. Methods Parents completed the Child Behavior Checklist (CBCL) focused on different DSM diagnostic categories: affective, anxiety, pervasive developmental, attention deficit/hyperactivity, and oppositional defiant. All the CBCL scores were corrected for gender, delivery method, gestational age, infant birth weight, occurrence of sepsis, and days on antibiotics prior statistical analyses. Canonical correlation analysis (CCA) was performed to determine the relationship between early life gut microbiome and the adjusted CBCL scores. The association of bacterial Amplicon sequence Variants (ASVs) to the CBCL scores were tested with multivariate analysis by linear models (MaAsLin). Results Nineteen children who were previously born with very low birth weight and studied while hospitalized in the Neonatal Intensive Care Unit (NICU) were included in this study. Statistically significant associations were observed between early life gut bacteria such as Veillonella dispar, Enterococcus, Escherichia coli, and Rumincococcus to later behavior at 4 years. No significant association could be observed with early-life gut microbiome alpha diversity and behavioral measures at 4 years. Discussion These preliminary observational data provide insight into the relationships between VLBW gut microbiome dysbiosis and childhood behavior. This study contributes to the literature on gut microbiome analysis by examining various behavioral domains using a standardized tool linked to the Diagnostic and Statistical Manual of Mental Disorders (DSM).
Collapse
Affiliation(s)
- Samia Valeria Ozorio Dutra
- Nancy Atmospera-Walch School of Nursing, University of Hawaii at Manoa, Honolulu, HI, United States
- College of Nursing, University of South Florida, Tampa, FL, United States
- College of Nursing, University of Tennessee-Knoxville, Knoxville, TN, United States
| | - Anujit Sarkar
- College of Nursing, University of South Florida, Tampa, FL, United States
- College of Nursing, University of Tennessee-Knoxville, Knoxville, TN, United States
| | - Ji Youn Yoo
- College of Nursing, University of South Florida, Tampa, FL, United States
- College of Nursing, University of Tennessee-Knoxville, Knoxville, TN, United States
| | - Emily Shaffer-Hudkins
- College of Medicine Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Maureen Groer
- College of Nursing, University of South Florida, Tampa, FL, United States
- College of Nursing, University of Tennessee-Knoxville, Knoxville, TN, United States
| |
Collapse
|
33
|
Li H, Guo W, Li S, Sun B, Li N, Xie D, Dong Z, Luo D, Chen W, Fu W, Zheng J, Zhu J. Alteration of the gut microbiota profile in children with autism spectrum disorder in China. Front Microbiol 2024; 14:1326870. [PMID: 38420215 PMCID: PMC10899803 DOI: 10.3389/fmicb.2023.1326870] [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: 10/24/2023] [Accepted: 12/18/2023] [Indexed: 03/02/2024] Open
Abstract
Background Autism spectrum disorder (ASD) is associated with alterations in the gut microbiome. However, there are few studies on gut microbiota of children with ASD in China, and there is a lack of consensus on the changes of bacterial species. Purpose Autism spectrum disorder (ASD) is associated with alterations in the gut microbiome. However, there are few studies on gut microbiota of children with ASD in China, and there is a lack of consensus on the changes of bacterial species. Methods We used 16S rRNA sequencing to analyze ASD children (2 to 12 years), HC (2 to 12 years). Results Our findings showed that the α-diversity, composition, and relative abundance of gut microbiota in the ASD group were significantly different from those in the HC groups. Compared with the HC group, the α-diversity in the ASD group was significantly decreased. At the genus level, the relative abundance of g_Faecalibacterium, g_Blautia, g_Eubacterium_eligens_group, g_Parasutterella, g_Lachnospiraceae_NK4A136_group and g_Veillonella in ASD group was significantly increased than that in HC groups, while the relative abundance of g_Prevotella 9 and g_Agathobacter was significantly decreased than that in HC groups. In addition, KEGG pathway analysis showed that the microbial functional abnormalities in ASD patients were mainly concentrated in metabolic pathways related to fatty acid, amino acid metabolism and aromatic compound metabolism, and were partially involved in neurotransmitter metabolism. Conclusion This study revealed the characteristics of gut microbiota of Chinese children with ASD and provided further evidence of gut microbial dysbiosis in ASD.
Collapse
Affiliation(s)
- Hui Li
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Wei Guo
- Stroke Center, Puyang People's Hospital, Puyang, China
| | - Sijie Li
- Department of Pediatrics, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Bishao Sun
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Ningshan Li
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Dongjing Xie
- Department of Neurology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Zongming Dong
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Dan Luo
- Department of Neurology, Yunyang People's Hospital, Yunyang, China
| | - Wei Chen
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Weihua Fu
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Ji Zheng
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Jingzhen Zhu
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
34
|
Mhanna A, Martini N, Hmaydoosh G, Hamwi G, Jarjanazi M, Zaifah G, Kazzazo R, Haji Mohamad A, Alshehabi Z. The correlation between gut microbiota and both neurotransmitters and mental disorders: A narrative review. Medicine (Baltimore) 2024; 103:e37114. [PMID: 38306525 PMCID: PMC10843545 DOI: 10.1097/md.0000000000037114] [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: 01/09/2024] [Indexed: 02/04/2024] Open
Abstract
The gastrointestinal tract is embedded with microorganisms of numerous genera, referred to as gut microbiota. Gut microbiota has multiple effects on many body organs, including the brain. There is a bidirectional connection between the gut and brain called the gut-brain-axis, and these connections are formed through immunological, neuronal, and neuroendocrine pathways. In addition, gut microbiota modulates the synthesis and functioning of neurotransmitters. Therefore, the disruption of the gut microbiota in the composition or function, which is known as dysbiosis, is associated with the pathogenesis of many mental disorders, such as schizophrenia, depression, and other psychiatric disorders. This review aims to summarize the modulation role of the gut microbiota in 4 prominent neurotransmitters (tryptophan and serotonergic system, dopamine, gamma-aminobutyric acid, and glutamate), as well as its association with 4 psychiatric disorders (schizophrenia, depression, anxiety disorders, and autism spectrum disorder). More future research is required to develop efficient gut-microbiota-based therapies for these illnesses.
Collapse
Affiliation(s)
- Amjad Mhanna
- Faculty of Medicine, Tishreen University, Latakia, Syrian Arab Republic
- Stemosis for Scientific Research, Damascus, Syrian Arab Republic
| | - Nafiza Martini
- Stemosis for Scientific Research, Damascus, Syrian Arab Republic
- Damascus University, Faculty of Medicine, Damascus, Syrian Arab Republic
| | - Ghefar Hmaydoosh
- Faculty of Medicine, Tishreen University, Latakia, Syrian Arab Republic
- Stemosis for Scientific Research, Damascus, Syrian Arab Republic
| | - George Hamwi
- Faculty of Medicine, Tishreen University, Latakia, Syrian Arab Republic
- Stemosis for Scientific Research, Damascus, Syrian Arab Republic
| | - Mulham Jarjanazi
- Pediatric Surgery Resident, Pediatric Surgery Department, Aleppo University Hospital, Aleppo, Syrian Arab Republic
| | - Ghaith Zaifah
- Faculty of Medicine, Tishreen University, Latakia, Syrian Arab Republic
- Stemosis for Scientific Research, Damascus, Syrian Arab Republic
| | - Reem Kazzazo
- Faculty of Medicine, Tishreen University, Latakia, Syrian Arab Republic
- Stemosis for Scientific Research, Damascus, Syrian Arab Republic
| | - Aya Haji Mohamad
- Stemosis for Scientific Research, Damascus, Syrian Arab Republic
- Faculty of Medicine, Aleppo University, Aleppo University Hospital, Aleppo, Syrian Arab Republic
| | - Zuheir Alshehabi
- Department of Pathology, Tishreen University Hospital, Latakia, Syrian Arab Republic
| |
Collapse
|
35
|
Lin P, Zhang Q, Sun J, Li Q, Li D, Zhu M, Fu X, Zhao L, Wang M, Lou X, Chen Q, Liang K, Zhu Y, Qu C, Li Z, Ma P, Wang R, Liu H, Dong K, Guo X, Cheng X, Sun Y, Sun J. A comparison between children and adolescents with autism spectrum disorders and healthy controls in biomedical factors, trace elements, and microbiota biomarkers: a meta-analysis. Front Psychiatry 2024; 14:1318637. [PMID: 38283894 PMCID: PMC10813399 DOI: 10.3389/fpsyt.2023.1318637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 12/13/2023] [Indexed: 01/30/2024] Open
Abstract
Introduction Autism spectrum disorder (ASD) is a multifaceted developmental condition that commonly appears during early childhood. The etiology of ASD remains multifactorial and not yet fully understood. The identification of biomarkers may provide insights into the underlying mechanisms and pathophysiology of the disorder. The present study aimed to explore the causes of ASD by investigating the key biomedical markers, trace elements, and microbiota factors between children with autism spectrum disorder (ASD) and control subjects. Methods Medline, PubMed, ProQuest, EMBASE, Cochrane Library, PsycINFO, Web of Science, and EMBSCO databases have been searched for publications from 2012 to 2023 with no language restrictions using the population, intervention, control, and outcome (PICO) approach. Keywords including "autism spectrum disorder," "oxytocin," "GABA," "Serotonin," "CRP," "IL-6," "Fe," "Zn," "Cu," and "gut microbiota" were used for the search. The Joanna Briggs Institute (JBI) critical appraisal checklist was used to assess the article quality, and a random model was used to assess the mean difference and standardized difference between ASD and the control group in all biomedical markers, trace elements, and microbiota factors. Results From 76,217 records, 43 studies met the inclusion and exclusion criteria and were included in this meta-analysis. The pooled analyses showed that children with ASD had significantly lower levels of oxytocin (mean differences, MD = -45.691, 95% confidence interval, CI: -61.667, -29.717), iron (MD = -3.203, 95% CI: -4.891, -1.514), and zinc (MD = -6.707, 95% CI: -12.691, -0.722), lower relative abundance of Bifidobacterium (MD = -1.321, 95% CI: -2.403, -0.238) and Parabacteroides (MD = -0.081, 95% CI: -0.148, -0.013), higher levels of c-reactive protein, CRP (MD = 0.401, 95% CI: 0.036, 0.772), and GABA (MD = 0.115, 95% CI: 0.045, 0.186), and higher relative abundance of Bacteroides (MD = 1.386, 95% CI: 0.717, 2.055) and Clostridium (MD = 0.281, 95% CI: 0.035, 0.526) when compared with controls. The results of the overall analyses were stable after performing the sensitivity analyses. Additionally, no substantial publication bias was observed among the studies. Interpretation Children with ASD have significantly higher levels of CRP and GABA, lower levels of oxytocin, iron, and zinc, lower relative abundance of Bifidobacterium and Parabacteroides, and higher relative abundance of Faecalibacterium, Bacteroides, and Clostridium when compared with controls. These results suggest that these indicators may be a potential biomarker panel for the diagnosis or determining therapeutic targets of ASD. Furthermore, large, sample-based, and randomized controlled trials are needed to confirm these results.
Collapse
Affiliation(s)
- Ping Lin
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qianwen Zhang
- Key Laboratory of Digital Technology in Medical Diagnostics of Zhejiang Province, Hangzhou, China
- Hangzhou Calibra Diagnostics, Hangzhou, China
| | - Junyu Sun
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Qingtian Li
- College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dan Li
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengyuan Zhu
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaomei Fu
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ling Zhao
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengxia Wang
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoyan Lou
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qing Chen
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kangyi Liang
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuxin Zhu
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Caiwei Qu
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenhua Li
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peijun Ma
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Renyu Wang
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huafen Liu
- Key Laboratory of Digital Technology in Medical Diagnostics of Zhejiang Province, Hangzhou, China
- Hangzhou Calibra Diagnostics, Hangzhou, China
| | - Ke Dong
- Institute for Global Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaokui Guo
- Institute for Global Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xunjia Cheng
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yang Sun
- Institute of Arthritis Research, Shanghai Academy of Chinese Medical Sciences, Shanghai, China
| | - Jing Sun
- School of Medicine and Dentistry, Institute for Integrated Intelligence and Systems, Griffith University, Gold Coast Campus, Gold Coast, QLD, Australia
- Charles Sturt University, Orange, NSW, Australia
| |
Collapse
|
36
|
Traetta ME, Chaves Filho AM, Akinluyi ET, Tremblay MÈ. Neurodevelopmental and Neuropsychiatric Disorders. ADVANCES IN NEUROBIOLOGY 2024; 37:457-495. [PMID: 39207708 DOI: 10.1007/978-3-031-55529-9_26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
This chapter will focus on microglial involvement in neurodevelopmental and neuropsychiatric disorders, particularly autism spectrum disorder (ASD), schizophrenia and major depressive disorder (MDD). We will describe the neuroimmune risk factors that contribute to the etiopathology of these disorders across the lifespan, including both in early life and adulthood. Microglia, being the resident immune cells of the central nervous system, could play a key role in triggering and determining the outcome of these disorders. This chapter will review preclinical and clinical findings where microglial morphology and function were examined in the contexts of ASD, schizophrenia and MDD. Clinical evidence points out to altered microglial morphology and reactivity, as well as increased expression of pro-inflammatory cytokines, supporting the idea that microglial abnormalities are involved in these disorders. Indeed, animal models for these disorders found altered microglial morphology and homeostatic functions which resulted in behaviours related to these disorders. Additionally, as microglia have emerged as promising therapeutic targets, we will also address in this chapter therapies involving microglial mechanisms for the treatment of neurodevelopmental and neuropsychiatric disorders.
Collapse
Affiliation(s)
| | | | - Elizabeth Toyin Akinluyi
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Pharmacology and Therapeutics, Afe Babalola University, Ado-Ekiti, Nigeria
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
- Département de Médecine Moléculaire, Université Laval, Quebec City, QC, Canada.
- Axe Neurosciences, Center de Recherche du CHU de Québec, Université Laval, Quebec City, QC, Canada.
- Neurology and Neurosurgery Department, McGill University, Montréal, QC, Canada.
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada.
- Center for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada.
- Institute on Aging and Lifelong Health (IALH), University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
37
|
Jingyi L, Lin W, Yuan C, Lingling Z, Qianqian J, Anlong X, Yansong G. Intravenous transplantation of bone marrow-derived mesenchymal stem cells improved behavioral deficits and altered fecal microbiota composition of BTBR mice. Life Sci 2024; 336:122330. [PMID: 38065352 DOI: 10.1016/j.lfs.2023.122330] [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: 10/01/2023] [Revised: 11/27/2023] [Accepted: 12/02/2023] [Indexed: 12/24/2023]
Abstract
AIMS It is recognized that autism spectrum disorder (ASD) is a highly complex neurodevelopmental disorder with communication deficits as well as multiple social barriers. The core symptoms of ASD are not treatable with current therapeutics. Therefore, finding new treatment strategies for ASD is urgently needed. Mesenchymal stem cells (MSC) have been shown to be a promising therapeutic approach in previous studies. However, the underlying mechanisms of MSC treatment for ASD through gut microbiota remain unclear and require further investigation. MAIN METHODS BTBR mice were used as ASD model and then randomly assigned to the human bone marrow-derived mesenchymal stem cell (hBMMSC) intravenous treatment group or vehicle treatment group. C57BL/6J (C57) mice served as control. Multiple social behavioral tests were performed during the 6-week period and fecal samples were collected at different time points for 16 s rRNA sequencing analysis. KEY FINDINGS The administration of hBMMSC improved social deficits of BTBR mice in the open field test (OFT), light-dark box test (LBT), novel object recognition (NOR), and free social test (FST), while also significantly reducing stereotypic behaviors. Additionally, hBMMSC administration notably reversed the alterations of microbiota abundance in BTBR mice, particularly the Firmicutes/Bacteroidetes ratio. Several specific differential taxa were further selected and showed a correlation with the prognosis and behavioral scores of ASD. SIGNIFICANCE Overall, intravenous treatment with hBMMSC had a beneficial impact on ASD by ameliorating social deficits and modifying microbiota compositions. This outcome indicates that hBMMSC intravenous transplantation could be a promising therapeutic strategy for enhancing ASD symptoms improvements.
Collapse
Affiliation(s)
- Li Jingyi
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China; Jiuzhitang Maker (Beijing) Cell Technology Co., LTD., Beijing 102600, China
| | - Wang Lin
- Jiuzhitang Maker (Beijing) Cell Technology Co., LTD., Beijing 102600, China
| | - Chen Yuan
- Jiuzhitang Maker (Beijing) Cell Technology Co., LTD., Beijing 102600, China
| | - Zhang Lingling
- Jiuzhitang Maker (Beijing) Cell Technology Co., LTD., Beijing 102600, China
| | - Jiang Qianqian
- Jiuzhitang Maker (Beijing) Cell Technology Co., LTD., Beijing 102600, China
| | - Xu Anlong
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China.
| | - Gao Yansong
- Jiuzhitang Maker (Beijing) Cell Technology Co., LTD., Beijing 102600, China; Hunan Provincial Key Laboratory of Critical Quality Attribute of Cell Therapy Products, Changsha, Hunan Province, China.
| |
Collapse
|
38
|
Butera A, De Simone R, Potenza RL, Sanchez M, Armida M, Campanile D, Di Carlo N, Trenta F, Boirivant M, Ricceri L. Effects of a gut-selective integrin-targeted therapy in male mice exposed to early immune activation, a model for the study of autism spectrum disorder. Brain Behav Immun 2024; 115:89-100. [PMID: 37793488 DOI: 10.1016/j.bbi.2023.09.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 09/11/2023] [Accepted: 09/30/2023] [Indexed: 10/06/2023] Open
Abstract
To clarify the role of gut mucosal immunity in ASD, we evaluated, in the early-life immune activation (EIA) mouse model, the effects of administration of a monoclonal antibody directed against the integrin alpha4 beta7 (α4β7 mAb), blocking the leukocyte homing into the gut mucosa. EIA is a double-hit variant of the maternal immune-activation (MIA) model, including both prenatal (Poly I:C) and postnatal (LPS) immune challenges. In C57BL6/J EIA male adult offspring mice, IL-1β and IL-17A mRNA colonic tissue content increased when compared with controls. Cytofluorimetric analyses of lymphocytes isolated from mesenteric lymph-nodes (MLN) and spleens of EIA mice show increased percentage of total and CD4+α4β7+, unstimulated and stimulated IL-17A+ and stimulated IFN-γ+ lymphocytes in MLN and CD4+α4β7+ unstimulated and stimulated IL-17A+ and stimulated IFN-γ+ lymphocytes in the spleen. Treatment with anti-α4β7 mAb in EIA male mice was associated with colonic tissue IL-1β, and IL-17A mRNA content and percentage of CD4+ IL-17A+ and IFN-γ+ lymphocytes in MLN and spleens comparable to control mice. The anti-α4β7 mAb treatment rescue social novelty deficit showed in the three-chamber test by EIA male mice. Increased levels of IL-6 and IL-1β and decreased CD68 and TGF-β mRNAs were also observed in hippocampus and prefrontal cortex of EIA male mice together with a reduction of BDNF mRNA levels in all brain regions examined. Anti-α4β7 mAb treatment restored the expression of BDNF, TGF-β and CD68 in hippocampus and prefrontal cortex. Improvement of the gut inflammatory status, obtained by a pharmacological agent acting exclusively at gut level, ameliorates some ASD behavioral features and the neuroinflammatory status. Data provide the first preclinical indication for a therapeutic strategy against gut-immune activation in ASD subjects with peripheral increase of gut-derived (α4β7+) lymphocytes expressing IL-17A.
Collapse
Affiliation(s)
- Alessia Butera
- National Center for Drug Research and Evaluation Istituto Superiore di Sanità, Rome, Italy
| | - Roberta De Simone
- National Center for Drug Research and Evaluation Istituto Superiore di Sanità, Rome, Italy
| | - Rosa Luisa Potenza
- National Center for Drug Research and Evaluation Istituto Superiore di Sanità, Rome, Italy
| | - Massimo Sanchez
- Cytometry Unit-Core Facilities, Istituto Superiore di Sanità, Rome, Italy
| | - Monica Armida
- National Center for Drug Research and Evaluation Istituto Superiore di Sanità, Rome, Italy
| | - Doriana Campanile
- National Center for Drug Research and Evaluation Istituto Superiore di Sanità, Rome, Italy
| | - Nazzareno Di Carlo
- National Center for Drug Research and Evaluation Istituto Superiore di Sanità, Rome, Italy
| | - Francesco Trenta
- Center for Behavioral Science and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| | - Monica Boirivant
- National Center for Drug Research and Evaluation Istituto Superiore di Sanità, Rome, Italy.
| | - Laura Ricceri
- Center for Behavioral Science and Mental Health, Istituto Superiore di Sanità, Rome, Italy.
| |
Collapse
|
39
|
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
|
40
|
Davies MR, Greenberg Z, van Vuurden DG, Cross CB, Zannettino ACW, Bardy C, Wardill HR. More than a small adult brain: Lessons from chemotherapy-induced cognitive impairment for modelling paediatric brain disorders. Brain Behav Immun 2024; 115:229-247. [PMID: 37858741 DOI: 10.1016/j.bbi.2023.10.013] [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: 04/19/2023] [Revised: 10/10/2023] [Accepted: 10/14/2023] [Indexed: 10/21/2023] Open
Abstract
Childhood is recognised as a period of immense physical and emotional development, and this, in part, is driven by underlying neurophysiological transformations. These neurodevelopmental processes are unique to the paediatric brain and are facilitated by augmented rates of neuroplasticity and expanded neural stem cell populations within neurogenic niches. However, given the immaturity of the developing central nervous system, innate protective mechanisms such as neuroimmune and antioxidant responses are functionally naïve which results in periods of heightened sensitivity to neurotoxic insult. This is highly relevant in the context of paediatric cancer, and in particular, the neurocognitive symptoms associated with treatment, such as surgery, radio- and chemotherapy. The vulnerability of the developing brain may increase susceptibility to damage and persistent symptomology, aligning with reports of more severe neurocognitive dysfunction in children compared to adults. It is therefore surprising, given this intensified neurocognitive burden, that most of the pre-clinical, mechanistic research focuses exclusively on adult populations and extrapolates findings to paediatric cohorts. Given this dearth of age-specific research, throughout this review we will draw comparisons with neurodevelopmental disorders which share comparable pathways to cancer treatment related side-effects. Furthermore, we will examine the unique nuances of the paediatric brain along with the somatic systems which influence neurological function. In doing so, we will highlight the importance of developing in vitro and in vivo paediatric disease models to produce age-specific discovery and clinically translatable research.
Collapse
Affiliation(s)
- Maya R Davies
- School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia; Supportive Oncology Research Group, Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia.
| | - Zarina Greenberg
- South Australian Health and Medical Research Institute (SAHMRI), Laboratory of Human Neurophysiology and Genetics, Adelaide, SA, Australia
| | - Dannis G van Vuurden
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, the weNetherlands
| | - Courtney B Cross
- Supportive Oncology Research Group, Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
| | - Andrew C W Zannettino
- School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Cedric Bardy
- South Australian Health and Medical Research Institute (SAHMRI), Laboratory of Human Neurophysiology and Genetics, Adelaide, SA, Australia; Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Hannah R Wardill
- School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia; Supportive Oncology Research Group, Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
| |
Collapse
|
41
|
Gong XR, You XR, Guo MR, Ding XY, Ma BX. Children autism spectrum disorder and gut microbiota: A bibliometric and visual analysis from 2000 to 2023. Medicine (Baltimore) 2023; 102:e36794. [PMID: 38206702 PMCID: PMC10754604 DOI: 10.1097/md.0000000000036794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 12/05/2023] [Indexed: 01/13/2024] Open
Abstract
Autism spectrum disorder (ASD) has evolved from a narrow and rare childhood-onset disorder to a widely publicized and researched lifelong disease recognized as common and significantly heterogeneous. Researchers have suggested that gastrointestinal symptoms in ASD may be a manifestation of an underlying inflammatory process. However, there is a lack of bibliometric analysis of ASD and gut microbiota in children. Accordingly, this study conducts a bibliometric analysis of ASD and gut microbiota in children from 2000 to 2023, explores the current status and cutting-edge trends in the field of ASD and gut microbiota in children, and identifies new directions for future research. The literature on ASD and gut microbiota in children was screened using the Web of Science Core Collection from 2000 to 2023. Annual publications, countries, institutions, authors, journals, keywords, and references were visualized and analyzed using CiteSpace 5.8. R3 and VOSviewer1.6.18. This study included 1071 publications. Since the beginning of 2011, the overall number of articles shows an upward trend. The most productive country and institution are the United States and the University of California system, respectively. The most frequently cited author is Kang Dae-Wook, with 790 citations, who has contributed significantly to this field. Timothy Dinan is the most prolific author, with 34 articles. The journal with the most published articles on this topic is Nutrients, whereas PLOS One is the most cited journal. The most used keyword is "gut microbiota," and the reference for the highest outbreak intensity is Hsiao. The research hotspots and trends predicted in this study provide a reference for further in-depth research in this field.
Collapse
Affiliation(s)
- Xing-Ruo Gong
- Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Xiao-Rui You
- Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Mei-Ran Guo
- Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Xue-Ying Ding
- Henan University of Chinese Medicine, Zhengzhou, China
| | - Bing-Xiang Ma
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
42
|
Mishra V, Yadav D, Solanki KS, Koul B, Song M. A Review on the Protective Effects of Probiotics against Alzheimer's Disease. BIOLOGY 2023; 13:8. [PMID: 38248439 PMCID: PMC10813289 DOI: 10.3390/biology13010008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/01/2023] [Accepted: 12/13/2023] [Indexed: 01/23/2024]
Abstract
This review summarizes the protective effects of probiotics against Alzheimer's disease (AD), one of the most common neurodegenerative disorders affecting older adults. This disease is characterized by the deposition of tau and amyloid β peptide (Aβ) in different parts of the brain. Symptoms observed in patients with AD include struggles with writing, speech, memory, and knowledge. The gut microbiota reportedly plays an important role in brain functioning due to its bidirectional communication with the gut via the gut-brain axis. The emotional and cognitive centers in the brain are linked to the functions of the peripheral intestinal system via this gut-brain axis. Dysbiosis has been linked to neurodegenerative disorders, indicating the significance of gut homeostasis for proper brain function. Probiotics play an important role in protecting against the symptoms of AD as they restore gut-brain homeostasis to a great extent. This review summarizes the characteristics, status of gut-brain axis, and significance of gut microbiota in AD. Review and research articles related to the role of probiotics in the treatment of AD were searched in the PubMed database. Recent studies conducted using animal models were given preference. Recent clinical trials were searched for separately. Several studies conducted on animal and human models clearly explain the benefits of probiotics in improving cognition and memory in experimental subjects. Based on these studies, novel therapeutic approaches can be designed for the treatment of patients with AD.
Collapse
Affiliation(s)
- Vibhuti Mishra
- School of Studies in Biochemistry, Jiwaji University, Gwalior 474003, India;
| | - Dhananjay Yadav
- Department of Life Science, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| | - Kavita Singh Solanki
- Department of Neuroscience and Pharmacology, Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52242, USA;
| | - Bhupendra Koul
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, India;
| | - Minseok Song
- Department of Life Science, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| |
Collapse
|
43
|
Mussap M, Beretta P, Esposito E, Fanos V. Once upon a Time Oral Microbiota: A Cinderella or a Protagonist in Autism Spectrum Disorder? Metabolites 2023; 13:1183. [PMID: 38132865 PMCID: PMC10745349 DOI: 10.3390/metabo13121183] [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: 11/06/2023] [Revised: 11/28/2023] [Accepted: 11/30/2023] [Indexed: 12/23/2023] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder evolving over the lifetime of individuals. The oral and gut microbial ecosystems are closely connected to each other and the brain and are potentially involved in neurodevelopmental diseases. This narrative review aims to identify all the available evidence emerging from observational studies focused on the role of the oral microbiome in ASD. A literature search was conducted using PubMed and the Cochrane Library for relevant studies published over the last ten years. Overall, in autistic children, the oral microbiota is marked by the abundance of several microbial species belonging to the Proteobacteria phylum and by the depletion of species belonging to the Bacteroidetes phylum. In mouse models, the oral microbiota is marked by the abundance of the Bacteroidetes phylum. Oral dysbiosis in ASD induces changes in the human metabolome, with the overexpression of metabolites closely related to the pathogenesis of ASD, such as acetate, propionate, and indoles, together with the underexpression of butyrate, confirming the central role of tryptophan metabolism. The analysis of the literature evidences the close relationship between oral dysbiosis and autistic core symptoms; the rebuilding of the oral and gut ecosystems by probiotics may significantly contribute to mitigating the severity of ASD symptoms.
Collapse
Affiliation(s)
- Michele Mussap
- Laboratory Unit, Department of Surgical Sciences, AOU Cagliari, University of Cagliari, 09124 Cagliari, Italy;
| | - Paola Beretta
- Neonatal Intensive Care Unit, Department of Surgical Sciences, AOU Cagliari, University of Cagliari, 09124 Cagliari, Italy; (E.E.); (V.F.)
| | - Elena Esposito
- Neonatal Intensive Care Unit, Department of Surgical Sciences, AOU Cagliari, University of Cagliari, 09124 Cagliari, Italy; (E.E.); (V.F.)
| | - Vassilios Fanos
- Neonatal Intensive Care Unit, Department of Surgical Sciences, AOU Cagliari, University of Cagliari, 09124 Cagliari, Italy; (E.E.); (V.F.)
| |
Collapse
|
44
|
Lewandowska-Pietruszka Z, Figlerowicz M, Mazur-Melewska K. Microbiota in Autism Spectrum Disorder: A Systematic Review. Int J Mol Sci 2023; 24:16660. [PMID: 38068995 PMCID: PMC10706819 DOI: 10.3390/ijms242316660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/19/2023] [Accepted: 11/21/2023] [Indexed: 12/18/2023] Open
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental condition characterized by several core symptoms: restricted interests, communication difficulties, and impaired social interactions. Many ASD children experience gastrointestinal functional disorders, impacting their well-being. Emerging evidence suggests that a gut microbiota imbalance may exacerbate core and gastrointestinal symptoms. Our review assesses the gut microbiota in children with ASD and interventions targeting microbiota modulation. The analysis of forty-four studies (meta-analyses, reviews, original research) reveals insights into the gut microbiota-ASD relationship. While specific microbiota alterations are mixed, some trends emerge. ASD children exhibit increased Firmicutes (36-81%) and Pseudomonadota (78%) and decreased Bacteroidetes (56%). The Bacteroidetes to Firmicutes ratio tends to be lower (56%) compared to children without ASD, which correlates with behavioral and gastrointestinal abnormalities. Probiotics, particularly Lactobacillus, Bifidobacterium, and Streptococcus strains, show promise in alleviating behavioral and gastrointestinal symptoms (66%). Microbiota transfer therapy (MTT) seems to have lasting benefits for the microbiota and symptoms in one longitudinal study. Prebiotics can potentially help with gastrointestinal and behavioral issues, needing further research for conclusive efficacy due to different interventions being used. This review highlights the gut microbiota-ASD interplay, offering potential therapeutic avenues for the gut-brain axis. However, study heterogeneity, small sample sizes, and methodological variations emphasize the need for comprehensive, standardized research. Future investigations may unveil complex mechanisms linking the gut microbiota to ASD, ultimately enhancing the quality of life for affected individuals.
Collapse
Affiliation(s)
| | | | - Katarzyna Mazur-Melewska
- Department of Infectious Diseases and Child Neurology, Poznan University of Medical Sciences, 60-572 Poznan, Poland; (Z.L.-P.); (M.F.)
| |
Collapse
|
45
|
Önal S, Sachadyn-Król M, Kostecka M. A Review of the Nutritional Approach and the Role of Dietary Components in Children with Autism Spectrum Disorders in Light of the Latest Scientific Research. Nutrients 2023; 15:4852. [PMID: 38068711 PMCID: PMC10708497 DOI: 10.3390/nu15234852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/15/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder that affects several areas of mental development. The onset of ASD occurs in the first few years of life, usually before the age of 3 years. Proper nutrition is important to ensure that an individual's nutrient and energy requirements are met, and it can also have a moderating effect on the progression of the disorder. A systematic database search was conducted as a narrative review to determine whether nutrition and specific diets can potentially alter gastrointestinal symptoms and neurobehavioral disorders. Databases such as Science Direct, PubMed, Scopus, Web of Science (WoS), and Google Scholar were searched to find studies published between 2000 and September 2023 on the relationship between ASD, dietary approaches, and the role of dietary components. The review may indicate that despite extensive research into dietary interventions, there is a general lack of conclusive scientific data about the effect of therapeutic diets on ASD; therefore, no definitive recommendation can be made for any specific nutritional therapy as a standard treatment for ASD. An individualized dietary approach and the dietician's role in the therapeutic team are very important elements of every therapy. Parents and caregivers should work with nutrition specialists, such as registered dietitians or healthcare providers, to design meal plans for autistic individuals, especially those who would like to implement an elimination diet.
Collapse
Affiliation(s)
- Seda Önal
- Department of Nutrition and Dietetics, Health Sciences Institute, Ankara University, 06110 Ankara, Turkey;
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Fırat University, 23200 Elazığ, Turkey
| | - Monika Sachadyn-Król
- Faculty of Food Science and Biotechnology, University of Life Sciences in Lublin, 20-950 Lublin, Poland;
| | - Małgorzata Kostecka
- Faculty of Food Science and Biotechnology, University of Life Sciences in Lublin, 20-950 Lublin, Poland;
| |
Collapse
|
46
|
He J, Gong X, Hu B, Lin L, Lin X, Gong W, Zhang B, Cao M, Xu Y, Xia R, Zheng G, Wu S, Zhang Y. Altered Gut Microbiota and Short-chain Fatty Acids in Chinese Children with Constipated Autism Spectrum Disorder. Sci Rep 2023; 13:19103. [PMID: 37925571 PMCID: PMC10625580 DOI: 10.1038/s41598-023-46566-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 11/02/2023] [Indexed: 11/06/2023] Open
Abstract
Gastrointestinal symptoms are more prevalent in children with autism spectrum disorder (ASD) than in typically developing (TD) children. Constipation is a significant gastrointestinal comorbidity of ASD, but the associations among constipated autism spectrum disorder (C-ASD), microbiota and short-chain fatty acids (SCFAs) are still debated. We enrolled 80 children, divided into the C-ASD group (n = 40) and the TD group (n = 40). In this study, an integrated 16S rRNA gene sequencing and gas chromatography-mass spectrometry-based metabolomics approach was applied to explore the association of the gut microbiota and SCFAs in C-ASD children in China. The community diversity estimated by the Observe, Chao1, and ACE indices was significantly lower in the C-ASD group than in the TD group. We observed that Ruminococcaceae_UCG_002, Erysipelotrichaceae_UCG_003, Phascolarctobacterium, Megamonas, Ruminiclostridium_5, Parabacteroides, Prevotella_2, Fusobacterium, and Prevotella_9 were enriched in the C-ASD group, and Anaerostipes, Lactobacillus, Ruminococcus_gnavus_group, Lachnospiraceae_NK4A136_group, Ralstonia, Eubacterium_eligens_group, and Ruminococcus_1 were enriched in the TD group. The propionate levels, which were higher in the C-ASD group, were negatively correlated with the abundance of Lactobacillus taxa, but were positively correlated with the severity of ASD symptoms. The random forest model, based on the 16 representative discriminant genera, achieved a high accuracy (AUC = 0.924). In conclusion, we found that C-ASD is related to altered gut microbiota and SCFAs, especially decreased abundance of Lactobacillus and excessive propionate in faeces, which provide new clues to understand C-ASD and biomarkers for the diagnosis and potential strategies for treatment of the disorder. This study was registered in the Chinese Clinical Trial Registry ( www.chictr.org.cn ; trial registration number ChiCTR2100052106; date of registration: October 17, 2021).
Collapse
Affiliation(s)
- Jianquan He
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Department of Rehabilitation, School of Medicine, Zhongshan Hospital of Xiamen University, Xiamen University, Xiamen, China
- Xiamen Institute of Big Data of TCM Constitution and PreventiveTreatment for Disease, Xiamen, China
| | - Xiuhua Gong
- School of Nursing, Qingdao University, Qingdao, China
| | - Bing Hu
- Department of Pediatrics, Yichun People's Hospital, Yichun, China
| | - Lin Lin
- Xiamen Institute of Big Data of TCM Constitution and PreventiveTreatment for Disease, Xiamen, China
| | - Xiujuan Lin
- Xiamen Institute of Big Data of TCM Constitution and PreventiveTreatment for Disease, Xiamen, China
| | - Wenxiu Gong
- Xiamen Institute of Big Data of TCM Constitution and PreventiveTreatment for Disease, Xiamen, China
| | | | - Man Cao
- Xiamen Treatgut Biotechnology Co., Ltd, Xiamen, China
| | - Yanzhi Xu
- Xiamen Treatgut Biotechnology Co., Ltd, Xiamen, China
| | - Rongmu Xia
- Clinical Research Institute, The Second Affiliated Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Guohua Zheng
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China.
- College of Nursing and Health Management, Shanghai University of Medicine and Health Sciences, Shanghai, China.
| | - Shuijin Wu
- Xiamen Food and Drug Evaluation and Adverse Reaction Monitoring Center, Xiamen, China.
| | - Yuying Zhang
- Department of Gastroenterology, Weifang People's Hospital, Weifang, China.
| |
Collapse
|
47
|
Dovgan K, Gynegrowski K, Ferguson BJ. Bidirectional relationship between internalizing symptoms and gastrointestinal problems in youth with Autism Spectrum Disorder. J Autism Dev Disord 2023; 53:4488-4494. [PMID: 35441914 DOI: 10.1007/s10803-022-05539-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2022] [Indexed: 11/25/2022]
Abstract
Many youth with autism spectrum disorder (ASD) experience co-occurring conditions, such as gastrointestinal (GI) problems and internalizing symptoms. However, the relationship among these co-occurring problems is not well-understood. We analyzed parent reports of GI problems and internalizing symptoms of 621 youth with ASD using path models in a structural equation modeling framework. The best-fitting model was a bidirectional model wherein internalizing symptoms, including withdrawn and anxious behavior, were correlated with GI problems, including constipation, diarrhea, nausea, and stomach pain. This study provides a better understanding of the relationship among co-occurring conditions in youth with ASD and should encourage clinicians to consider treatment of underlying internalizing symptoms or GI problems when providing services for individuals with ASD.
Collapse
Affiliation(s)
- Kristen Dovgan
- Marist College, Department of Psychology, Poughkeepsie, NY, United States.
| | - Kyra Gynegrowski
- Marist College, Department of Psychology, Poughkeepsie, NY, United States
| | - Bradley J Ferguson
- Department of Health Psychology, University of Missouri, Columbia, MO, United States
- Thompson Center for Autism & Neurodevelopmental Disorders, University of Missouri, Columbia, MO, United States
- Department of Radiology, University of Missouri, Columbia, MO, United States
| |
Collapse
|
48
|
Parolisi S, Montanari C, Borghi E, Cazzorla C, Zuvadelli J, Tosi M, Barone R, Bensi G, Bonfanti C, Dionisi Vici C, Biasucci G, Burlina A, Carbone MT, Verduci E. Possible role of tryptophan metabolism along the microbiota-gut-brain axis on cognitive & behavioral aspects in Phenylketonuria. Pharmacol Res 2023; 197:106952. [PMID: 37804926 DOI: 10.1016/j.phrs.2023.106952] [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: 07/28/2023] [Revised: 10/01/2023] [Accepted: 10/04/2023] [Indexed: 10/09/2023]
Abstract
Cognitive and psychiatric disorders are well documented across the lifetime of patients with inborn errors of metabolism (IEMs). Gut microbiota impacts behavior and cognitive functions through the gut-brain axis (GBA). According to recent research, a broad spectrum of GBA disorders may be influenced by a perturbed Tryptophan (Trp) metabolism and are associated with alterations in composition or function of the gut microbiota. Furthermore, early-life diets may influence children's neurodevelopment and cognitive deficits in adulthood. In Phenylketonuria (PKU), since the main therapeutic intervention is based on a life-long restrictive diet, important alterations of gut microbiota have been observed. Studies on PKU highlight the impact of alterations of gut microbiota on the central nervous system (CNS), also investigating the involvement of metabolic pathways, such as Trp and kynurenine (KYN) metabolisms, involved in numerous neurodegenerative disorders. An alteration of Trp metabolism with an imbalance of the KYN pathway towards the production of neurotoxic metabolites implicated in numerous neurodegenerative and inflammatory diseases has been observed in PKU patients supplemented with Phe-free amino acid medical foods (AA-MF). The present review investigates the possible link between gut microbiota and the brain in IEMs, focusing on Trp metabolism in PKU. Considering the evidence collected, cognitive and behavioral well-being should always be monitored in routine IEMs clinical management. Further studies are required to evaluate the possible impact of Trp metabolism, through gut microbiota, on cognitive and behavioral functions in IEMs, to identify innovative dietetic strategies and improve quality of life and mental health of these patients.
Collapse
Affiliation(s)
- Sara Parolisi
- UOSD Metabolic Diseases, AORN Santobono-Pausilipon, Naples, Italy
| | - Chiara Montanari
- Department of Pediatrics, Vittore Buzzi Children's Hospital, University of Milan, Milan, Italy; Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Elisa Borghi
- Department of Health Science, University of Milan, Milan, Italy
| | - Chiara Cazzorla
- Division of Inherited Metabolic Diseases, DIDAS Department of Diagnostic Services, University Hospital, Padua, Italy
| | - Juri Zuvadelli
- Clinical Department of Pediatrics, San Paolo Hospital, ASST Santi Paolo e Carlo, Milan, Italy
| | - Martina Tosi
- Department of Pediatrics, Vittore Buzzi Children's Hospital, University of Milan, Milan, Italy; Department of Health Science, University of Milan, Milan, Italy
| | - Rita Barone
- Child Neuropsychiatry Unit, Department of Clinical and Experimental Medicine, AOU Policlinico "G.Rodolico-San Marco", University of Catania, Catania, Italy
| | - Giulia Bensi
- Paediatrics & Neonatology Unit, Guglielmo da Saliceto Hospital, Piacenza, Italy
| | - Cristina Bonfanti
- Rare metabolic disease unit, Pediatric Department, San Gerardo Hospital, Monza, Italy
| | | | - Giacomo Biasucci
- Paediatrics & Neonatology Unit, Guglielmo da Saliceto Hospital, Piacenza, Italy
| | - Alberto Burlina
- Division of Inherited Metabolic Diseases, DIDAS Department of Diagnostic Services, University Hospital, Padua, Italy
| | - Maria T Carbone
- UOSD Metabolic Diseases, AORN Santobono-Pausilipon, Naples, Italy
| | - Elvira Verduci
- Department of Pediatrics, Vittore Buzzi Children's Hospital, University of Milan, Milan, Italy; Department of Health Science, University of Milan, Milan, Italy.
| |
Collapse
|
49
|
Açıkel SB, Kara A, Bağcı Z, Can Ü. Serum trimethylamine N-oxide and lipopolysaccharide binding protein levels among children diagnosed with autism spectrum disorder. Int J Dev Neurosci 2023; 83:571-577. [PMID: 37525434 DOI: 10.1002/jdn.10287] [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: 05/25/2023] [Revised: 06/21/2023] [Accepted: 07/03/2023] [Indexed: 08/02/2023] Open
Abstract
In the literature, there have been several studies available investigating the relationship between autism spectrum disorder and intestinal permeability. In this study, it is aimed to examine the relationship between the levels of trimethylamine N-oxide (TMAO), which is a parameter associated with intestinal permeability, and lipopolysaccharide binding protein (LBP), which is a marker associated with bacterial translocation from the intestine, in patients with autism spectrum disorder (ASD) and healthy controls. Fifty-three children with ASD as the patient group and 30 healthy children as the control group have been included in the study. The diagnostic evaluation has been made according to DSM-5 criteria. According to the obtained results, there has been no significant difference between groups in terms of serum TMAO and LBP levels. Considering the existence of various studies that found different results on ASD and intestinal permeability, it is thought that the studies conducted in this field that did not find statistically different results will also make a contribution to the literature.
Collapse
Affiliation(s)
- Sadettin Burak Açıkel
- Faculty of Medicine, Department of Child and Adolescent Psychiatry, Ankara University, Ankara, Turkey
| | - Aziz Kara
- Faculty of Medicine, Department of Child and Adolescent Psychiatry, Afyon University of Health Sciences, Afyon, Turkey
| | - Zafer Bağcı
- Department of Pediatrics, Konya City Hospital, Konya, Turkey
| | - Ümmügülsüm Can
- Department of Biochemistry, Konya City Hospital, Konya, Turkey
| |
Collapse
|
50
|
Fujishiro S, Tsuji S, Akagawa S, Akagawa Y, Yamanouchi S, Ishizaki Y, Hashiyada M, Akane A, Kaneko K. Dysbiosis in Gut Microbiota in Children Born Preterm Who Developed Autism Spectrum Disorder: A Pilot Study. J Autism Dev Disord 2023; 53:4012-4020. [PMID: 35909184 DOI: 10.1007/s10803-022-05682-0] [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] [Accepted: 07/10/2022] [Indexed: 10/16/2022]
Abstract
The gut microbiota was reported to differ between children with autism spectrum disorder (ASD) and typically developing (TD) children, and dysbiosis of the gut microbiota in preterm infants is common. Here, we explored the characteristics of gut microbiota in children born preterm with ASD. We performed 16S rRNA gene sequencing using stool samples from ASD children born preterm and TD children born preterm. Alpha diversity was significantly greater in the ASD group. A comparison of beta diversity showed different clusters. Linear discriminant analysis effect size analysis revealed significantly more Firmicutes in the ASD group compared with the TD group. In conclusion, the gut microbiota in children born preterm differs between children with ASD and TD.
Collapse
Affiliation(s)
- Sadayuki Fujishiro
- Department of Pediatrics, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka, 573-1010, Japan
| | - Shoji Tsuji
- Department of Pediatrics, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka, 573-1010, Japan
| | - Shohei Akagawa
- Department of Pediatrics, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka, 573-1010, Japan
| | - Yuko Akagawa
- Department of Pediatrics, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka, 573-1010, Japan
| | - Sohsaku Yamanouchi
- Department of Pediatrics, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka, 573-1010, Japan
| | - Yuko Ishizaki
- Department of Pediatrics, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka, 573-1010, Japan
| | - Masaki Hashiyada
- Department of Legal Medicine, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka, 573-1010, Japan
| | - Atsushi Akane
- Department of Legal Medicine, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka, 573-1010, Japan
| | - Kazunari Kaneko
- Department of Pediatrics, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka, 573-1010, Japan.
| |
Collapse
|