1
|
Alba C, Herranz C, Monroy MA, Aragón A, Jurado R, Díaz-Regañón D, Sánchez C, Tolín M, Miranda C, Gómez-Taylor B, Sempere F, Álvarez-Calatayud G, Rodríguez JM. Metataxonomic and Immunological Analysis of Feces from Children with or without Phelan-McDermid Syndrome. Microorganisms 2024; 12:2006. [PMID: 39458315 PMCID: PMC11509408 DOI: 10.3390/microorganisms12102006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 10/28/2024] Open
Abstract
Phelan-McDermid syndrome (PMS) is a neurodevelopmental disorder characterized by a developmental delay and autism spectrum disorder (ASD)-like behaviors. Emerging research suggests a link between gut microbiota and neuropsychiatric conditions, including PMS. This study aimed to investigate the fecal microbiota and immune profiles of children with PMS compared to healthy controls. Fecal samples were collected from children diagnosed with PMS and age-matched healthy controls. The bacterial composition was analyzed using 16S rRNA gene sequencing, while short-chain fatty acids (SCFAs) were quantified through gas chromatography. Immunological profiling was conducted using a multiplex cytokine assay. Significant differences were observed in the gut microbiota composition between PMS patients and controls, including a lower abundance of key bacterial genera such as Faecalibacterium and Agathobacter in PMS patients. SCFA levels were also reduced in PMS patients. Immunological analysis revealed higher levels of several pro-inflammatory cytokines in the PMS group, although these differences were not statistically significant. The findings indicate that children with PMS have distinct gut microbiota and SCFA profiles, which may contribute to the gastrointestinal and neurodevelopmental symptoms observed in this syndrome. These results suggest potential avenues for microbiota-targeted therapies in PMS.
Collapse
Affiliation(s)
- Claudio Alba
- Department Nutrition and Food Science, Complutense University of Madrid, 28040 Madrid, Spain; (D.D.-R.); (J.M.R.)
- Instituto Pluridisciplinar, Complutense University of Madrid, 28040 Madrid, Spain; (A.A.); (R.J.)
| | - Carmen Herranz
- Department Nutrition and Food Science, Complutense University of Madrid, 28040 Madrid, Spain; (D.D.-R.); (J.M.R.)
- Instituto Pluridisciplinar, Complutense University of Madrid, 28040 Madrid, Spain; (A.A.); (R.J.)
| | | | - Alberto Aragón
- Instituto Pluridisciplinar, Complutense University of Madrid, 28040 Madrid, Spain; (A.A.); (R.J.)
- Department Galenic Pharmacy and Food Technology, Complutense University of Madrid, 28040 Madrid, Spain
| | - Rubén Jurado
- Instituto Pluridisciplinar, Complutense University of Madrid, 28040 Madrid, Spain; (A.A.); (R.J.)
- Department Galenic Pharmacy and Food Technology, Complutense University of Madrid, 28040 Madrid, Spain
| | - David Díaz-Regañón
- Department Nutrition and Food Science, Complutense University of Madrid, 28040 Madrid, Spain; (D.D.-R.); (J.M.R.)
| | - César Sánchez
- Departamento de Nutrición Humana, Universidad Católica de Valencia, 46001 Valencia, Spain; (C.S.); (M.T.); (C.M.); (B.G.-T.); (F.S.)
| | - Mar Tolín
- Departamento de Nutrición Humana, Universidad Católica de Valencia, 46001 Valencia, Spain; (C.S.); (M.T.); (C.M.); (B.G.-T.); (F.S.)
| | - Carmen Miranda
- Departamento de Nutrición Humana, Universidad Católica de Valencia, 46001 Valencia, Spain; (C.S.); (M.T.); (C.M.); (B.G.-T.); (F.S.)
| | - Bárbara Gómez-Taylor
- Departamento de Nutrición Humana, Universidad Católica de Valencia, 46001 Valencia, Spain; (C.S.); (M.T.); (C.M.); (B.G.-T.); (F.S.)
| | - Francisca Sempere
- Departamento de Nutrición Humana, Universidad Católica de Valencia, 46001 Valencia, Spain; (C.S.); (M.T.); (C.M.); (B.G.-T.); (F.S.)
| | | | - Juan M. Rodríguez
- Department Nutrition and Food Science, Complutense University of Madrid, 28040 Madrid, Spain; (D.D.-R.); (J.M.R.)
- Instituto Pluridisciplinar, Complutense University of Madrid, 28040 Madrid, Spain; (A.A.); (R.J.)
| |
Collapse
|
2
|
Yang X, Li H, Yang C, Ge J. Supplementation with stigma maydis polysaccharide attenuates autism-like behaviors and improves gut function in valproic acid-induced autism model male rats. Int J Dev Neurosci 2024; 84:567-580. [PMID: 38923604 DOI: 10.1002/jdn.10354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/29/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Stigma maydis polysaccharide (SMPS) has regulatory effect on the intestinal microflora and promotes gastrointestinal peristalsis. Children with autism spectrum disorder (ASD) often experience gastrointestinal problems and dysbiosis in their gut microbiota. Our previous study revealed that SMPS interventions had an impact on the gut microbiota of valproic acid (VPA)-induced autism model rats. However, the effects of SMPS on the behavior and gut function of autism model rats remain poorly understood. Therefore, we gave different doses of SMPS intervention in the early stage of autism model rats to observe their developmental conditions and behavior performances. Through histological evaluation and real-time polymerase chain reaction (PCR), integrity of the intestinal structure and the expression of tight junction-related gene Zo-1 and Occludin were detected. The results indicated that SMPS intervention improved the physical development, learning and memory impairment, and social performance of autism model rats. Meanwhile, SMPS promoted intestinal peristalsis and restored the integrity of the intestinal structure, reduced the number of inflammatory cells, and increased the expression of the Zo-1 and Occludin genes. Furthermore, the expression levels of neurotransmitters (substance P, enkephalin, vasoactive intestinal peptide, and 5-hydroxytryptamine) in the hippocampal tissues were altered after SMPS treatment. In conclusion, SMPS could ameliorate ASD-like phenotypes and gut problems in autism model rats. Collectively, these results provide new evidence for the relationship between the gut-brain axis and ASD and suggest a novel therapeutic target for ASD treatment.
Collapse
Affiliation(s)
- Xiaolei Yang
- Department of Preventive Medicine, School of Public Health, Qiqihar Medical University, Qiqihar, China
| | - Hongjie Li
- Department of Preventive Medicine, School of Public Health, Qiqihar Medical University, Qiqihar, China
| | - Chao Yang
- Department of Preventive Treatment, Qiqihar Hospital of Traditional Chinese Medicine, Qiqihar, China
| | - Jie Ge
- Department of Preventive Medicine, School of Public Health, Qiqihar Medical University, Qiqihar, China
| |
Collapse
|
3
|
Akhlaghi E, Salari E, Mansouri M, Shafiei M, Kalantar-Neyestanaki D, Aghassi H, Fasihi Harandi M. Identification and comparison of intestinal microbial diversity in patients at different stages of hepatic cystic echinococcosis. Sci Rep 2024; 14:18912. [PMID: 39143364 PMCID: PMC11324937 DOI: 10.1038/s41598-024-70005-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 08/12/2024] [Indexed: 08/16/2024] Open
Abstract
There is a significant focus on the role of the host microbiome in different outcomes of human parasitic diseases, including cystic echinococcosis (CE). This study was conducted to identify the intestinal microbiome of patients with CE at different stages of hydatid cyst compared to healthy individuals. Stool samples from CE patients as well as healthy individuals were collected. The samples were divided into three groups representing various stages of hepatic hydatid cyst: active (CE1 and CE2), transitional (CE3), and inactive (CE4 and CE5). One family member from each group was selected to serve as a control. The gut microbiome of patients with different stages of hydatid cysts was investigated using metagenomic next-generation amplicon sequencing of the V3-V4 region of the 16S rRNA gene. In this study, we identified 4862 Operational Taxonomic Units from three stages of hydatid cysts in CE patients and healthy individuals with a combined frequency of 2,955,291. The most abundant genera observed in all the subjects were Blautia, Agathobacter, Faecalibacterium, Bacteroides, Bifidobacterium, and Prevotella. The highest microbial frequency was related to inactive forms of CE, and the lowest frequency was observed in the group with active forms. However, the lowest OTU diversity was found in patients with inactive cysts compared with those with active and transitional cyst stages. The genus Agatobacter had the highest OTU frequency. Pseudomonas, Gemella, and Ligilactobacillus showed significant differences among the patients with different stages of hydatid cysts. Additionally, Anaerostipes and Candidatus showed significantly different reads in CE patients compared to healthy individuals. Our findings indicate that several bacterial genera can play a role in the fate of hydatid cysts in patients at different stages of the disease.
Collapse
Affiliation(s)
- Elham Akhlaghi
- Research Center for Hydatid Disease in Iran, Department of Medical Parasitology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Elham Salari
- Department of Plant Protection, Faculty of Plant Production, Gorgan University of Agricultural Sciences and Natural Resources, Gorgan, Iran
| | - Mehdi Mansouri
- Department of Agricultural Biotechnology, Faculty of Agriculture, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Mohammad Shafiei
- Research Center for Hydatid Disease in Iran, Department of Surgery, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Davood Kalantar-Neyestanaki
- Medical Mycology and Bacteriology Research Center, Kerman University of Medical Sciences, Kerman, Iran
- Department of Medical Microbiology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Hossein Aghassi
- Research Center for Hydatid Disease in Iran, Department of Medical Parasitology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Majid Fasihi Harandi
- Research Center for Hydatid Disease in Iran, Department of Medical Parasitology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
4
|
Xu H, Luo Y, Li Q, Zhu H. Acupuncture influences multiple diseases by regulating gut microbiota. Front Cell Infect Microbiol 2024; 14:1371543. [PMID: 39040602 PMCID: PMC11260648 DOI: 10.3389/fcimb.2024.1371543] [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: 01/16/2024] [Accepted: 06/13/2024] [Indexed: 07/24/2024] Open
Abstract
Acupuncture, an important green and side effect-free therapy in traditional Chinese medicine, is widely use both domestically and internationally. Acupuncture can interact with the gut microbiota and influence various diseases, including metabolic diseases, gastrointestinal diseases, mental disorders, nervous system diseases, and other diseases. This review presents a thorough analysis of these interactions and their impacts and examines the alterations in the gut microbiota and the potential clinical outcomes following acupuncture intervention to establish a basis for the future utilization of acupuncture in clinical treatments.
Collapse
Affiliation(s)
- Huimin Xu
- Department of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yingzhe Luo
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Qiaoqi Li
- Department of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hong Zhu
- Department of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
5
|
Deyang T, Baig MAI, Dolkar P, Hediyal TA, Rathipriya AG, Bhaskaran M, PandiPerumal SR, Monaghan TM, Mahalakshmi AM, Chidambaram SB. Sleep apnoea, gut dysbiosis and cognitive dysfunction. FEBS J 2024; 291:2519-2544. [PMID: 37712936 DOI: 10.1111/febs.16960] [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: 05/26/2023] [Revised: 08/14/2023] [Accepted: 09/13/2023] [Indexed: 09/16/2023]
Abstract
Sleep disorders are becoming increasingly common, and their distinct effects on physical and mental health require elaborate investigation. Gut dysbiosis (GD) has been reported in sleep-related disorders, but sleep apnoea is of particular significance because of its higher prevalence and chronicity. Cumulative evidence has suggested a link between sleep apnoea and GD. This review highlights the gut-brain communication axis that is mediated via commensal microbes and various microbiota-derived metabolites (e.g. short-chain fatty acids, lipopolysaccharide and trimethyl amine N-oxide), neurotransmitters (e.g. γ-aminobutyric acid, serotonin, glutamate and dopamine), immune cells and inflammatory mediators, as well as the vagus nerve and hypothalamic-pituitary-adrenal axis. This review also discusses the pathological role underpinning GD and altered gut bacterial populations in sleep apnoea and its related comorbid conditions, particularly cognitive dysfunction. In addition, the review examines the preclinical and clinical evidence, which suggests that prebiotics and probiotics may potentially be beneficial in sleep apnoea and its comorbidities through restoration of eubiosis or gut microbial homeostasis that regulates neural, metabolic and immune responses, as well as physiological barrier integrity via the gut-brain axis.
Collapse
Affiliation(s)
- Tenzin Deyang
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
| | - Md Awaise Iqbal Baig
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
| | - Phurbu Dolkar
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
| | - Tousif Ahmed Hediyal
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru, India
| | | | - Mahendran Bhaskaran
- College of Pharmacy and Pharmaceutical Sciences, Frederic and Mary Wolf Center, University of Toledo Health Science Campus, OH, USA
| | - Seithikuruppu R PandiPerumal
- Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Division of Research and Development, Lovely Professional University, Phagwara, India
| | - Tanya M Monaghan
- National Institute for Health Research Nottingham Biomedical Research Centre, University of Nottingham, UK
- Nottingham Digestive Diseases Centre, School of Medicine, University of Nottingham, UK
| | - Arehally M Mahalakshmi
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru, India
- SIG-Brain, Behaviour and Cognitive Neurosciences Research (BBRC), JSS Academy of Higher Education & Research, Mysuru, India
| | - Saravana Babu Chidambaram
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru, India
- SIG-Brain, Behaviour and Cognitive Neurosciences Research (BBRC), JSS Academy of Higher Education & Research, Mysuru, India
| |
Collapse
|
6
|
Esposito D, Cruciani G, Zaccaro L, Di Carlo E, Spitoni GF, Manti F, Carducci C, Fiori E, Leuzzi V, Pascucci T. A Systematic Review on Autism and Hyperserotonemia: State-of-the-Art, Limitations, and Future Directions. Brain Sci 2024; 14:481. [PMID: 38790459 PMCID: PMC11119126 DOI: 10.3390/brainsci14050481] [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: 04/10/2024] [Revised: 04/29/2024] [Accepted: 05/02/2024] [Indexed: 05/26/2024] Open
Abstract
Hyperserotonemia is one of the most studied endophenotypes in autism spectrum disorder (ASD), but there are still no unequivocal results about its causes or biological and behavioral outcomes. This systematic review summarizes the studies investigating the relationship between blood serotonin (5-HT) levels and ASD, comparing diagnostic tools, analytical methods, and clinical outcomes. A literature search on peripheral 5-HT levels and ASD was conducted. In total, 1104 publications were screened, of which 113 entered the present systematic review. Of these, 59 articles reported hyperserotonemia in subjects with ASD, and 26 presented correlations between 5-HT levels and ASD-core clinical outcomes. The 5-HT levels are increased in about half, and correlations between hyperserotonemia and clinical outcomes are detected in a quarter of the studies. The present research highlights a large amount of heterogeneity in this field, ranging from the characterization of ASD and control groups to diagnostic and clinical assessments, from blood sampling procedures to analytical methods, allowing us to delineate critical topics for future studies.
Collapse
Affiliation(s)
- Dario Esposito
- Department of Human Neuroscience, Unit of Child Neurology and Psychiatry, Sapienza University of Rome, Via dei Sabelli 108, 00185 Rome, Italy; (D.E.); (F.M.)
| | - Gianluca Cruciani
- Department of Dynamic and Clinical Psychology, and Health Studies, Sapienza University of Rome, Via degli Apuli 1, 00185 Rome, Italy; (G.C.); (G.F.S.)
| | - Laura Zaccaro
- Department of Psychology, Sapienza University, Via dei Marsi 78, 00185 Rome, Italy; (L.Z.); (T.P.)
| | - Emanuele Di Carlo
- Department of Experimental Medicine, Sapienza University, Viale del Policlinico 155, 00161 Rome, Italy; (E.D.C.); (C.C.)
| | - Grazia Fernanda Spitoni
- Department of Dynamic and Clinical Psychology, and Health Studies, Sapienza University of Rome, Via degli Apuli 1, 00185 Rome, Italy; (G.C.); (G.F.S.)
- Cognitive and Motor Rehabilitation and Neuroimaging Unit, IRCCS Fondazione Santa Lucia, Via Ardeatina 306-354, 00179 Rome, Italy
| | - Filippo Manti
- Department of Human Neuroscience, Unit of Child Neurology and Psychiatry, Sapienza University of Rome, Via dei Sabelli 108, 00185 Rome, Italy; (D.E.); (F.M.)
| | - Claudia Carducci
- Department of Experimental Medicine, Sapienza University, Viale del Policlinico 155, 00161 Rome, Italy; (E.D.C.); (C.C.)
| | - Elena Fiori
- Rome Technopole Foundation, P.le Aldo Moro, 5, 00185 Rome, Italy;
| | - Vincenzo Leuzzi
- Department of Human Neuroscience, Unit of Child Neurology and Psychiatry, Sapienza University of Rome, Via dei Sabelli 108, 00185 Rome, Italy; (D.E.); (F.M.)
| | - Tiziana Pascucci
- Department of Psychology, Sapienza University, Via dei Marsi 78, 00185 Rome, Italy; (L.Z.); (T.P.)
- Centro “Daniel Bovet”, Sapienza University, P.le Aldo Moro 5, 00185 Rome, Italy
- Fondazione Santa Lucia Istituto di Ricovero e Cura a Carattere Scientifico, Via Ardeatina 306, 00179 Rome, Italy
| |
Collapse
|
7
|
Korteniemi J, Karlsson L, Aatsinki A. Systematic Review: Autism Spectrum Disorder and the Gut Microbiota. FOCUS (AMERICAN PSYCHIATRIC PUBLISHING) 2024; 22:242-251. [PMID: 38680985 PMCID: PMC11046714 DOI: 10.1176/appi.focus.24022008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/01/2024]
Abstract
Objective Autism spectrum disorders (ASD) are a varying group of disorders characterized by deficiency in social interaction and restrictive patterns of behavior and interests. While there are several studies focusing on the neuro-psychiatric pathogenesis of ASD, its etiology remains unclear. The role of gut-brain-axis in ASD has been studied increasingly and a correlation between symptoms and the composition of gut microbiota has been documented in various works. Despite this, the significance of individual microbes and their function is still widely unknown. This work aims to elucidate the current knowledge of the interrelations between ASD and the gut microbiota in children based on scientific evidence. Methods This is a systematic review done by a literature search focusing on the main findings concerning the gut microbiota composition, interventions targeting the gut microbiota, and possible mechanisms explaining the results in children aged between 2 and 18 years of age. Results Most studies in this review found significant differences between microbial communities, while there was notable variation in results regarding diversity indices or taxonomic level abundance. The most consistent results regarding taxa differences in ASD children's gut microbiota were higher levels of Proteobacteria, Actinobacteria and Sutterella compared to controls. Conclusion These results show that the gut microbiota of children with ASD is altered compared to one of neurotypically developed children. More research is needed to discover whether some of these features could be used as potential biomarkers for ASD and how the gut microbiota could be targeted in therapeutical interventions.Appeared originally in Acta Psychiatr Scand 2023;148:242-254.
Collapse
Affiliation(s)
- Jenni Korteniemi
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, Psychiatry, University of Turku, Turku, Finland (Korteniemi, Karlsson, Aatsinki); Centre for Population Health Research, Turku University Hospital, University of Turku, Turku, Finland (Karlsson, Aatsinki); Department of Clinical Medicine, Paediatrics and Adolescent Medicine, Turku University Hospital, University of Turku, Turku, Finland (Karlsson)
| | - Linnea Karlsson
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, Psychiatry, University of Turku, Turku, Finland (Korteniemi, Karlsson, Aatsinki); Centre for Population Health Research, Turku University Hospital, University of Turku, Turku, Finland (Karlsson, Aatsinki); Department of Clinical Medicine, Paediatrics and Adolescent Medicine, Turku University Hospital, University of Turku, Turku, Finland (Karlsson)
| | - Anna Aatsinki
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, Psychiatry, University of Turku, Turku, Finland (Korteniemi, Karlsson, Aatsinki); Centre for Population Health Research, Turku University Hospital, University of Turku, Turku, Finland (Karlsson, Aatsinki); Department of Clinical Medicine, Paediatrics and Adolescent Medicine, Turku University Hospital, University of Turku, Turku, Finland (Karlsson)
| |
Collapse
|
8
|
Borghi E, Xynomilakis O, Ottaviano E, Ceccarani C, Viganò I, Tognini P, Vignoli A. Gut microbiota profile in CDKL5 deficiency disorder patients. Sci Rep 2024; 14:7376. [PMID: 38548767 PMCID: PMC10978852 DOI: 10.1038/s41598-024-56989-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 03/13/2024] [Indexed: 04/01/2024] Open
Abstract
CDKL5 deficiency disorder (CDD) is a neurodevelopmental condition characterized by global developmental delay, early-onset seizures, intellectual disability, visual and motor impairments. Unlike Rett Syndrome (RTT), CDD lacks a clear regression period. Patients with CDD frequently encounter gastrointestinal (GI) disturbances and exhibit signs of subclinical immune dysregulation. However, the underlying causes of these conditions remain elusive. Emerging studies indicate a potential connection between neurological disorders and gut microbiota, an area completely unexplored in CDD. We conducted a pioneering study, analyzing fecal microbiota composition in individuals with CDD (n = 17) and their healthy relatives (n = 17). Notably, differences in intestinal bacterial diversity and composition were identified in CDD patients. In particular, at genus level, CDD microbial communities were characterized by an increase in the relative abundance of Clostridium_AQ, Eggerthella, Streptococcus, and Erysipelatoclostridium, and by a decrease in Eubacterium, Dorea, Odoribacter, Intestinomonas, and Gemmiger, pointing toward a dysbiotic profile. We further investigated microbiota changes based on the severity of GI issues, seizure frequency, sleep disorders, food intake type, impairment in neuro-behavioral features and ambulation capacity. Enrichment in Lachnoclostridium and Enterobacteriaceae was observed in the microbiota of patients with more severe GI symptoms, while Clostridiaceae, Peptostreptococcaceae, Coriobacteriaceae, Erysipelotrichaceae, Christensenellaceae, and Ruminococcaceae were enriched in patients experiencing daily epileptic seizures. Our findings suggest a potential connection between CDD, microbiota and symptom severity. This study marks the first exploration of the gut-microbiota-brain axis in subjects with CDD. It adds to the growing body of research emphasizing the role of the gut microbiota in neurodevelopmental disorders and opens doors to potential interventions that target intestinal microbes with the aim of improving the lives of patients with CDD.
Collapse
Affiliation(s)
- Elisa Borghi
- Department of Health Sciences, Università Degli Studi di Milano, Milan, Italy
| | - Ornella Xynomilakis
- Department of Health Sciences, Università Degli Studi di Milano, Milan, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
- Dipartimento di Scienze Biomediche e Cliniche, Università Degli Studi di Milano, 20157, Milan, Italy
| | | | - Camilla Ceccarani
- Institute of Biomedical Technologies, National Research Council, Segrate, Milan, Italy
| | - Ilaria Viganò
- Epilepsy Center-Child Neuropsychiatric Unit, ASST Santi Paolo e Carlo, Milan, Italy
| | - Paola Tognini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy.
- Health Science Interdisciplinary Center, Sant'Anna School of Advanced Studies, Pisa, Italy.
| | - Aglaia Vignoli
- Department of Health Sciences, Università Degli Studi di Milano, Milan, Italy
- Childhood and Adolescence Neurology and Psychiatry Unit, ASST GOM Niguarda, Milan, Italy
| |
Collapse
|
9
|
Wang Q, Gao T, Zhang W, Liu D, Li X, Chen F, Mei J. Causal relationship between the gut microbiota and insomnia: a two-sample Mendelian randomization study. Front Cell Infect Microbiol 2024; 14:1279218. [PMID: 38500501 PMCID: PMC10945026 DOI: 10.3389/fcimb.2024.1279218] [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: 09/22/2023] [Accepted: 02/01/2024] [Indexed: 03/20/2024] Open
Abstract
Background Changes in the gut microbiota are closely related to insomnia, but the causal relationship between them is not yet clear. Objective To clarify the relationship between the gut microbiota and insomnia and provide genetic evidence for them, we conducted a two-sample Mendelian randomization study. Methods We used a Mendelian randomized two-way validation method to discuss the causal relationship. First, we downloaded the data of 462,341 participants relating to insomnia, and the data of 18,340 participants relating to the gut microbiota from a genome-wide association study (GWAS). Then, we used two regression models, inverse-variance weighted (IVW) and MR-Egger regression, to evaluate the relationship between exposure factors and outcomes. Finally, we took a reverse MR analysis to assess the possibility of reverse causality. Results The combined results show 19 gut microbiotas to have a causal relationship with insomnia (odds ratio (OR): 1.03; 95% confidence interval (CI): 1.01, 1.05; p=0.000 for class. Negativicutes; OR: 1.03; 95% CI: 1.01, 1.05; p=0.000 for order.Selenomonadales; OR: 1.01; 95% CI: 1.00, 1.02; p=0.003 for genus.RikenellaceaeRC9gutgroup). The results were consistent with sensitivity analyses for these bacterial traits. In reverse MR analysis, we found no statistical difference between insomnia and these gut microbiotas. Conclusion This study can provide a new direction for the causal relationship between the gut microbiota (class.Negativicutes, order.Selenomonadales, genus.Lactococcus) and insomnia and the treatment or prevention strategies of insomnia.
Collapse
Affiliation(s)
- Qianfei Wang
- The Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, China
- The First Affiliated Hospital, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Tianci Gao
- The Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, China
- The First Affiliated Hospital, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Weichao Zhang
- The Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, China
- The First Affiliated Hospital, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Dong Liu
- The Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, China
- The First Affiliated Hospital, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Xin Li
- The Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, China
- The First Affiliated Hospital, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Fenqiao Chen
- The First Affiliated Hospital, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Jianqiang Mei
- The First Affiliated Hospital, Hebei University of Chinese Medicine, Shijiazhuang, China
| |
Collapse
|
10
|
Hung LY, Margolis KG. Autism spectrum disorders and the gastrointestinal tract: insights into mechanisms and clinical relevance. Nat Rev Gastroenterol Hepatol 2024; 21:142-163. [PMID: 38114585 DOI: 10.1038/s41575-023-00857-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/11/2023] [Indexed: 12/21/2023]
Abstract
Autism spectrum disorders (ASDs) are recognized as central neurodevelopmental disorders diagnosed by impairments in social interactions, communication and repetitive behaviours. The recognition of ASD as a central nervous system (CNS)-mediated neurobehavioural disorder has led most of the research in ASD to be focused on the CNS. However, gastrointestinal function is also likely to be affected owing to the neural mechanistic nature of ASD and the nervous system in the gastrointestinal tract (enteric nervous system). Thus, it is unsurprising that gastrointestinal disorders, particularly constipation, diarrhoea and abdominal pain, are highly comorbid in individuals with ASD. Gastrointestinal problems have also been repeatedly associated with increased severity of the core symptoms diagnostic of ASD and other centrally mediated comorbid conditions, including psychiatric issues, irritability, rigid-compulsive behaviours and aggression. Despite the high prevalence of gastrointestinal dysfunction in ASD and its associated behavioural comorbidities, the specific links between these two conditions have not been clearly delineated, and current data linking ASD to gastrointestinal dysfunction have not been extensively reviewed. This Review outlines the established and emerging clinical and preclinical evidence that emphasizes the gut as a novel mechanistic and potential therapeutic target for individuals with ASD.
Collapse
Affiliation(s)
- Lin Y Hung
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, USA
| | - Kara Gross Margolis
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, USA.
- Department of Cell Biology, NYU Grossman School of Medicine and Langone Medical Center, New York, NY, USA.
- Department of Pediatrics, NYU Grossman School of Medicine and Langone Medical Center, New York, NY, USA.
| |
Collapse
|
11
|
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
|
12
|
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
|
13
|
Wiefels MD, Furar E, Eshraghi RS, Mittal J, Memis I, Moosa M, Mittal R, Eshraghi AA. Targeting Gut Dysbiosis and Microbiome Metabolites for the Development of Therapeutic Modalities for Neurological Disorders. Curr Neuropharmacol 2024; 22:123-139. [PMID: 36200211 PMCID: PMC10716879 DOI: 10.2174/1570159x20666221003085508] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/10/2022] [Accepted: 08/15/2022] [Indexed: 11/22/2022] Open
Abstract
The gut microbiota, composed of numerous species of microbes, works in synergy with the various organ systems in the body to bolster our overall health and well-being. The most well-known function of the gut microbiome is to facilitate the metabolism and absorption of crucial nutrients, such as complex carbohydrates, while also generating vitamins. In addition, the gut microbiome plays a crucial role in regulating the functioning of the central nervous system (CNS). Host genetics, including specific genes and single nucleotide polymorphisms (SNPs), have been implicated in the pathophysiology of neurological disorders, including Parkinson's disease (PD), Alzheimer's disease (AD), and autism spectrum disorder (ASD). The gut microbiome dysbiosis also plays a role in the pathogenesis of these neurodegenerative disorders, thus perturbing the gut-brain axis. Overproduction of certain metabolites synthesized by the gut microbiome, such as short-chain fatty acids (SCFAs) and p-cresyl sulfate, are known to interfere with microglial function and trigger misfolding of alpha-synuclein protein, which can build up inside neurons and cause damage. By determining the association of the gut microbiome and its metabolites with various diseases, such as neurological disorders, future research will pave the way for the development of effective preventive and treatment modalities.
Collapse
Affiliation(s)
- Matthew D Wiefels
- Hearing Research and Communication Disorders Laboratory, Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Emily Furar
- Hearing Research and Communication Disorders Laboratory, Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Rebecca S Eshraghi
- Hearing Research and Communication Disorders Laboratory, Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Jeenu Mittal
- Hearing Research and Communication Disorders Laboratory, Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Idil Memis
- Hearing Research and Communication Disorders Laboratory, Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Moeed Moosa
- Hearing Research and Communication Disorders Laboratory, Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Rahul Mittal
- Hearing Research and Communication Disorders Laboratory, Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Adrien A Eshraghi
- Hearing Research and Communication Disorders Laboratory, Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami, Florida, USA
- Department of Pediatrics, Miller School of Medicine, University of Miami, Miami, Florida, 33136, USA
- Department of Neurological Surgery, Miller School of Medicine, University of Miami, Miami, Florida, USA
| |
Collapse
|
14
|
Bonmatí-Carrión MÁ, Rol MA. Melatonin as a Mediator of the Gut Microbiota-Host Interaction: Implications for Health and Disease. Antioxidants (Basel) 2023; 13:34. [PMID: 38247459 PMCID: PMC10812647 DOI: 10.3390/antiox13010034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/23/2024] Open
Abstract
In recent years, the role played by melatonin on the gut microbiota has gained increasingly greater attention. Additionally, the gut microbiota has been proposed as an alternative source of melatonin, suggesting that this antioxidant indoleamine could act as a sort of messenger between the gut microbiota and the host. This review analyses the available scientific literature about possible mechanisms involved in this mediating role, highlighting its antioxidant effects and influence on this interaction. In addition, we also review the available knowledge on the effects of melatonin on gut microbiota composition, as well as its ability to alleviate dysbiosis related to sleep deprivation or chronodisruptive conditions. The melatonin-gut microbiota relationship has also been discussed in terms of its role in the development of different disorders, from inflammatory or metabolic disorders to psychiatric and neurological conditions, also considering oxidative stress and the reactive oxygen species-scavenging properties of melatonin as the main factors mediating this relationship.
Collapse
Affiliation(s)
- María-Ángeles Bonmatí-Carrión
- Chronobiology Laboratory, Department of Physiology, College of Biology, Mare Nostrum Campus, University of Murcia, Instituto Universitario de Investigación en Envejecimiento, Instituto Murciano de Investigación Biosanitaria-Arrixaca, 30100 Murcia, Spain;
- Ciber Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Maria-Angeles Rol
- Chronobiology Laboratory, Department of Physiology, College of Biology, Mare Nostrum Campus, University of Murcia, Instituto Universitario de Investigación en Envejecimiento, Instituto Murciano de Investigación Biosanitaria-Arrixaca, 30100 Murcia, Spain;
- Ciber Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
15
|
Jing Y, Yang D, Bai F, Wang Q, Zhang C, Yan Y, Li Z, Li Y, Chen Z, Li J, Yu Y. Spinal cord injury-induced gut dysbiosis influences neurological recovery partly through short-chain fatty acids. NPJ Biofilms Microbiomes 2023; 9:99. [PMID: 38092763 PMCID: PMC10719379 DOI: 10.1038/s41522-023-00466-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 11/23/2023] [Indexed: 12/17/2023] Open
Abstract
Spinal cord injury (SCI) can reshape gut microbial composition, significantly affecting clinical outcomes in SCI patients. However, mechanisms regarding gut-brain interactions and their clinical implications have not been elucidated. We hypothesized that short-chain fatty acids (SCFAs), intestinal microbial bioactive metabolites, may significantly affect the gut-brain axis and enhance functional recovery in a mouse model of SCI. We enrolled 59 SCI patients and 27 healthy control subjects and collected samples. Thereafter, gut microbiota and SCFAs were analyzed using 16 S rDNA sequencing and gas chromatography-mass spectrometry, respectively. We observed an increase in Actinobacteriota abundance and a decrease in Firmicutes abundance. Particularly, the SCFA-producing genera, such as Faecalibacterium, Megamonas, and Agathobacter were significantly downregulated among SCI patients compared to healthy controls. Moreover, SCI induced downregulation of acetic acid (AA), propionic acid (PA), and butyric acid (BA) in the SCI group. Fecal SCFA contents were altered in SCI patients with different injury course and injury segments. Main SCFAs (AA, BA, and PA) were administered in combination to treat SCI mice. SCFA supplementation significantly improved locomotor recovery in SCI mice, enhanced neuronal survival, promoted axonal formation, reduced astrogliosis, and suppressed microglial activation. Furthermore, SCFA supplementation downregulated NF-κB signaling while upregulating neurotrophin-3 expression following SCI. Microbial sequencing and metabolomics analysis showed that SCI patients exhibited a lower level of certain SCFAs and related bacterial strains than healthy controls. SCFA supplementation can reduce inflammation and enhance nourishing elements, facilitating the restoration of neurological tissues and the improvement of functional recuperation. Trial registration: This study was registered in the China Clinical Trial Registry ( www.chictr.org.cn ) on February 13, 2017 (ChiCTR-RPC-17010621).
Collapse
Affiliation(s)
- Yingli Jing
- China Rehabilitation Science Institute, China Rehabilitation Research Center, Beijing Key Laboratory of Neural Injury and Rehabilitation, and School of Rehabilitation Medicine, Capital Medical University, Beijing, 100068, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, China
| | - Degang Yang
- China Rehabilitation Science Institute, China Rehabilitation Research Center, Beijing Key Laboratory of Neural Injury and Rehabilitation, and School of Rehabilitation Medicine, Capital Medical University, Beijing, 100068, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, China
- Department of Spinal and Neural Function Reconstruction, Beijing Bo'ai Hospital, Beijing, 100068, China
| | - Fan Bai
- China Rehabilitation Science Institute, China Rehabilitation Research Center, Beijing Key Laboratory of Neural Injury and Rehabilitation, and School of Rehabilitation Medicine, Capital Medical University, Beijing, 100068, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, China
| | - Qiuying Wang
- China Rehabilitation Science Institute, China Rehabilitation Research Center, Beijing Key Laboratory of Neural Injury and Rehabilitation, and School of Rehabilitation Medicine, Capital Medical University, Beijing, 100068, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, China
| | - Chao Zhang
- Department of Neurosurgery, Linyi People's Hospital, Shangdong, 276034, China
| | - Yitong Yan
- China Rehabilitation Science Institute, China Rehabilitation Research Center, Beijing Key Laboratory of Neural Injury and Rehabilitation, and School of Rehabilitation Medicine, Capital Medical University, Beijing, 100068, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, China
| | - Zihan Li
- China Rehabilitation Science Institute, China Rehabilitation Research Center, Beijing Key Laboratory of Neural Injury and Rehabilitation, and School of Rehabilitation Medicine, Capital Medical University, Beijing, 100068, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, China
| | - Yan Li
- China Rehabilitation Science Institute, China Rehabilitation Research Center, Beijing Key Laboratory of Neural Injury and Rehabilitation, and School of Rehabilitation Medicine, Capital Medical University, Beijing, 100068, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, China
| | - Zhiguo Chen
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, China.
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China.
| | - Jianjun Li
- China Rehabilitation Science Institute, China Rehabilitation Research Center, Beijing Key Laboratory of Neural Injury and Rehabilitation, and School of Rehabilitation Medicine, Capital Medical University, Beijing, 100068, China.
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, China.
| | - Yan Yu
- China Rehabilitation Science Institute, China Rehabilitation Research Center, Beijing Key Laboratory of Neural Injury and Rehabilitation, and School of Rehabilitation Medicine, Capital Medical University, Beijing, 100068, China.
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, China.
| |
Collapse
|
16
|
Santi D, Debbi V, Costantino F, Spaggiari G, Simoni M, Greco C, Casarini L. Microbiota Composition and Probiotics Supplementations on Sleep Quality-A Systematic Review and Meta-Analysis. Clocks Sleep 2023; 5:770-792. [PMID: 38131749 PMCID: PMC10742335 DOI: 10.3390/clockssleep5040050] [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: 11/13/2023] [Revised: 11/29/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023] Open
Abstract
The gut microbiota (GM) plays a crucial role in human health. The bidirectional interaction between GM and the central nervous system may occur via the microbiota-gut-brain axis, possibly regulating the sleep/wake cycle. Recent reports highlight associations between intestinal dysbiosis and sleep disorders, suggesting that probiotics could ameliorate this condition. However, data are poor and inconsistent. The aim of this quantitative metanalytic study is to assess the GM composition in sleep disturbances and evaluate probiotics' effectiveness for managing sleep disorders. A systematic review was carried out until July 2022 in online databases, limiting the literature research to human studies and English language articles. No significant GM diversity between patients with sleep disturbances versus healthy controls was found, revealed by α-diversity, while β-diversity is missing due to lack of proper reporting. However, probiotics supplementation significantly reduced the self-assessed parameter of sleep quality and disturbances Pittsburgh Sleep Quality Index (PSQI) score compared with the placebo. No difference in the Epworth Sleepiness Scale (ESS) score was found. While available data suggest that GM diversity is not related to sleep disturbances, probiotics administration strongly improves sleep quality as a subjective perception. However, heterogeneity of data reporting in the scientific literature should be considered as a limitation.
Collapse
Affiliation(s)
- Daniele Santi
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41126 Modena, Italy; (D.S.); (V.D.); (M.S.); (L.C.)
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, 41126 Modena, Italy
- Unit of Andrology and Sexual Medicine of the Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, 41126 Modena, Italy
| | - Valentina Debbi
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41126 Modena, Italy; (D.S.); (V.D.); (M.S.); (L.C.)
| | - Francesco Costantino
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41126 Modena, Italy; (D.S.); (V.D.); (M.S.); (L.C.)
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, 41126 Modena, Italy
| | - Giorgia Spaggiari
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, 41126 Modena, Italy
- Unit of Andrology and Sexual Medicine of the Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, 41126 Modena, Italy
| | - Manuela Simoni
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41126 Modena, Italy; (D.S.); (V.D.); (M.S.); (L.C.)
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, 41126 Modena, Italy
- Unit of Andrology and Sexual Medicine of the Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, 41126 Modena, Italy
- Center for Genomic Research, University of Modena and Reggio Emilia, 41126 Modena, Italy
| | - Carla Greco
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41126 Modena, Italy; (D.S.); (V.D.); (M.S.); (L.C.)
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, 41126 Modena, Italy
| | - Livio Casarini
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41126 Modena, Italy; (D.S.); (V.D.); (M.S.); (L.C.)
- Center for Genomic Research, University of Modena and Reggio Emilia, 41126 Modena, Italy
| |
Collapse
|
17
|
Nguyen TQ, Martínez-Álvaro M, Lima J, Auffret MD, Rutherford KMD, Simm G, Dewhurst RJ, Baima ET, Roehe R. Identification of intestinal and fecal microbial biomarkers using a porcine social stress model. Front Microbiol 2023; 14:1197371. [PMID: 38029169 PMCID: PMC10670831 DOI: 10.3389/fmicb.2023.1197371] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Understanding the relationships between social stress and the gastrointestinal microbiota, and how they influence host health and performance is expected to have many scientific and commercial implementations in different species, including identification and improvement of challenges to animal welfare and health. In particular, the study of the stress impact on the gastrointestinal microbiota of pigs may be of interest as a model for human health. A porcine stress model based on repeated regrouping and reduced space allowance during the last 4 weeks of the finishing period was developed to identify stress-induced changes in the gut microbiome composition. The application of the porcine stress model resulted in a significant increase in salivary cortisol concentration over the course of the trial and decreased growth performance and appetite. The applied social stress resulted in 32 bacteria being either enriched (13) or depleted (19) in the intestine and feces. Fecal samples showed a greater number of microbial genera influenced by stress than caecum or colon samples. Our trial revealed that the opportunistic pathogens Treponema and Clostridium were enriched in colonic and fecal samples from stressed pigs. Additionally, genera such as Streptococcus, Parabacteroides, Desulfovibrio, Terrisporobacter, Marvinbryantia, and Romboutsia were found to be enriched in response to social stress. In contrast, the genera Prevotella, Faecalibacterium, Butyricicoccus, Dialister, Alloprevotella, Megasphaera, and Mitsuokella were depleted. These depleted bacteria are of great interest because they synthesize metabolites [e.g., short-chain fatty acids (SCFA), in particular, butyrate] showing beneficial health benefits due to inhibitory effects on pathogenic bacteria in different animal species. Of particular interest are Dialister and Faecalibacterium, as their depletion was identified in a human study to be associated with inferior quality of life and depression. We also revealed that some pigs were more susceptible to pathogens as indicated by large enrichments of opportunistic pathogens of Clostridium, Treponema, Streptococcus and Campylobacter. Generally, our results provide further evidence for the microbiota-gut-brain axis as indicated by an increase in cortisol concentration due to social stress regulated by the hypothalamic-pituitary-adrenal axis, and a change in microbiota composition, particularly of bacteria known to be associated with pathogenicity and mental health diseases.
Collapse
Affiliation(s)
- Tuan Q. Nguyen
- Scotland’s Rural College, Edinburgh, United Kingdom
- Department of Animal Breeding, Faculty of Animal Science and Veterinary Medicine, Nong Lam University – Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | | | - Joana Lima
- Scotland’s Rural College, Edinburgh, United Kingdom
| | | | | | - Geoff Simm
- Global Academy of Agriculture and Food Security, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Eric T. Baima
- Zoetis Inc., Parsippany-Troy Hills, NJ, United States
| | - Rainer Roehe
- Scotland’s Rural College, Edinburgh, United Kingdom
| |
Collapse
|
18
|
Widjaja F, Rietjens IMCM. From-Toilet-to-Freezer: A Review on Requirements for an Automatic Protocol to Collect and Store Human Fecal Samples for Research Purposes. Biomedicines 2023; 11:2658. [PMID: 37893032 PMCID: PMC10603957 DOI: 10.3390/biomedicines11102658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 09/22/2023] [Accepted: 09/24/2023] [Indexed: 10/29/2023] Open
Abstract
The composition, viability and metabolic functionality of intestinal microbiota play an important role in human health and disease. Studies on intestinal microbiota are often based on fecal samples, because these can be sampled in a non-invasive way, although procedures for sampling, processing and storage vary. This review presents factors to consider when developing an automated protocol for sampling, processing and storing fecal samples: donor inclusion criteria, urine-feces separation in smart toilets, homogenization, aliquoting, usage or type of buffer to dissolve and store fecal material, temperature and time for processing and storage and quality control. The lack of standardization and low-throughput of state-of-the-art fecal collection procedures promote a more automated protocol. Based on this review, an automated protocol is proposed. Fecal samples should be collected and immediately processed under anaerobic conditions at either room temperature (RT) for a maximum of 4 h or at 4 °C for no more than 24 h. Upon homogenization, preferably in the absence of added solvent to allow addition of a buffer of choice at a later stage, aliquots obtained should be stored at either -20 °C for up to a few months or -80 °C for a longer period-up to 2 years. Protocols for quality control should characterize microbial composition and viability as well as metabolic functionality.
Collapse
Affiliation(s)
- Frances Widjaja
- Division of Toxicology, Wageningen University & Research, 6708 WE Wageningen, The Netherlands;
| | | |
Collapse
|
19
|
Olaguez-Gonzalez JM, Chairez I, Breton-Deval L, Alfaro-Ponce M. Machine Learning Algorithms Applied to Predict Autism Spectrum Disorder Based on Gut Microbiome Composition. Biomedicines 2023; 11:2633. [PMID: 37893007 PMCID: PMC10604849 DOI: 10.3390/biomedicines11102633] [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: 08/10/2023] [Revised: 09/01/2023] [Accepted: 09/18/2023] [Indexed: 10/29/2023] Open
Abstract
The application of machine learning (ML) techniques stands as a reliable method for aiding in the diagnosis of complex diseases. Recent studies have related the composition of the gut microbiota to the presence of autism spectrum disorder (ASD), but until now, the results have been mostly contradictory. This work proposes using machine learning to study the gut microbiome composition and its role in the early diagnosis of ASD. We applied support vector machines (SVMs), artificial neural networks (ANNs), and random forest (RF) algorithms to classify subjects as neurotypical (NT) or having ASD, using published data on gut microbiome composition. Naive Bayes, k-nearest neighbors, ensemble learning, logistic regression, linear regression, and decision trees were also trained and validated; however, the ones presented showed the best performance and interpretability. All the ML methods were developed using the SAS Viya software platform. The microbiome's composition was determined using 16S rRNA sequencing technology. The application of ML yielded a classification accuracy as high as 90%, with a sensitivity of 96.97% and specificity reaching 85.29%. In the case of the ANN model, no errors occurred when classifying NT subjects from the first dataset, indicating a significant classification outcome compared to traditional tests and data-based approaches. This approach was repeated with two datasets, one from the USA and the other from China, resulting in similar findings. The main predictors in the obtained models differ between the analyzed datasets. The most important predictors identified from the analyzed datasets are Bacteroides, Lachnospira, Anaerobutyricum, and Ruminococcus torques. Notably, among the predictors in each model, there is the presence of bacteria that are usually considered insignificant in the microbiome's composition due to their low relative abundance. This outcome reinforces the conventional understanding of the microbiome's influence on ASD development, where an imbalance in the composition of the microbiota can lead to disrupted host-microbiota homeostasis. Considering that several previous studies focused on the most abundant genera and neglected smaller (and frequently not statistically significant) microbial communities, the impact of such communities has been poorly analyzed. The ML-based models suggest that more research should focus on these less abundant microbes. A novel hypothesis explains the contradictory results in this field and advocates for more in-depth research to be conducted on variables that may not exhibit statistical significance. The obtained results seem to contribute to an explanation of the contradictory findings regarding ASD and its relation with gut microbiota composition. While some research correlates higher ratios of Bacillota/Bacteroidota, others find the opposite. These discrepancies are closely linked to the minority organisms in the microbiome's composition, which may differ between populations but share similar metabolic functions. Therefore, the ratios of Bacillota/Bacteroidota regarding ASD may not be determinants in the manifestation of ASD.
Collapse
Affiliation(s)
- Juan M. Olaguez-Gonzalez
- School of Engineering and Science, Tecnologico de Monterrey, Monterrey 64849, Mexico; (J.M.O.-G.); (I.C.)
| | - Isaac Chairez
- School of Engineering and Science, Tecnologico de Monterrey, Monterrey 64849, Mexico; (J.M.O.-G.); (I.C.)
- Institute of Advanced Materials for Sustainable Manufacturing, Tecnologico de Monterrey, Monterrey 64849, Mexico
| | - Luz Breton-Deval
- Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca 62210, Mexico;
- Consejo Nacional de Ciencia y Tecnologia, Mexico City 03940, Mexico
| | - Mariel Alfaro-Ponce
- School of Engineering and Science, Tecnologico de Monterrey, Monterrey 64849, Mexico; (J.M.O.-G.); (I.C.)
- Institute of Advanced Materials for Sustainable Manufacturing, Tecnologico de Monterrey, Monterrey 64849, Mexico
| |
Collapse
|
20
|
Korteniemi J, Karlsson L, Aatsinki A. Systematic review: Autism spectrum disorder and the gut microbiota. Acta Psychiatr Scand 2023; 148:242-254. [PMID: 37395517 DOI: 10.1111/acps.13587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 05/02/2023] [Accepted: 06/10/2023] [Indexed: 07/04/2023]
Abstract
OBJECTIVE Autism spectrum disorders (ASD) are a varying group of disorders characterized by deficiency in social interaction and restrictive patterns of behavior and interests. While there are several studies focusing on the neuropsychiatric pathogenesis of ASD, its etiology remains unclear. The role of gut-brain-axis in ASD has been studied increasingly and a correlation between symptoms and the composition of gut microbiota has been documented in various works. Despite this, the significance of individual microbes and their function is still widely unknown. This work aims to elucidate the current knowledge of the interrelations between ASD and the gut microbiota in children based on scientific evidence. METHODS This is a systematic review done by a literature search focusing on the main findings concerning the gut microbiota composition, interventions targeting the gut microbiota, and possible mechanisms explaining the results in children aged between 2 and 18 years of age. RESULTS Most studies in this review found significant differences between microbial communities, while there was notable variation in results regarding diversity indices or taxonomic level abundance. The most consistent results regarding taxa differences in ASD children's gut microbiota were higher levels of Proteobacteria, Actinobacteria and Sutterella compared to controls. CONCLUSION These results show that the gut microbiota of children with ASD is altered compared to one of neurotypically developed children. More research is needed to discover whether some of these features could be used as potential biomarkers for ASD and how the gut microbiota could be targeted in therapeutical interventions.
Collapse
Affiliation(s)
- Jenni Korteniemi
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, Psychiatry, University of Turku, Turku, Finland
| | - Linnea Karlsson
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, Psychiatry, University of Turku, Turku, Finland
- Centre for Population Health Research, Turku University Hospital, University of Turku, Turku, Finland
- Department of Clinical Medicine, Paediatrics and Adolescent Medicine, Turku University Hospital, University of Turku, Turku, Finland
| | - Anna Aatsinki
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, Psychiatry, University of Turku, Turku, Finland
- Centre for Population Health Research, Turku University Hospital, University of Turku, Turku, Finland
| |
Collapse
|
21
|
Siracusano M, Arturi L, Riccioni A, Noto A, Mussap M, Mazzone L. Metabolomics: Perspectives on Clinical Employment in Autism Spectrum Disorder. Int J Mol Sci 2023; 24:13404. [PMID: 37686207 PMCID: PMC10487559 DOI: 10.3390/ijms241713404] [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: 07/06/2023] [Revised: 08/09/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
Precision medicine is imminent, and metabolomics is one of the main actors on stage. We summarize and discuss the current literature on the clinical application of metabolomic techniques as a possible tool to improve early diagnosis of autism spectrum disorder (ASD), to define clinical phenotypes and to identify co-occurring medical conditions. A review of the current literature was carried out after PubMed, Medline and Google Scholar were consulted. A total of 37 articles published in the period 2010-2022 was included. Selected studies involve as a whole 2079 individuals diagnosed with ASD (1625 males, 394 females; mean age of 10, 9 years), 51 with other psychiatric comorbidities (developmental delays), 182 at-risk individuals (siblings, those with genetic conditions) and 1530 healthy controls (TD). Metabolomics, reflecting the interplay between genetics and environment, represents an innovative and promising technique to approach ASD. The metabotype may mirror the clinical heterogeneity of an autistic condition; several metabolites can be expressions of dysregulated metabolic pathways thus liable of leading to clinical profiles. However, the employment of metabolomic analyses in clinical practice is far from being introduced, which means there is a need for further studies for the full transition of metabolomics from clinical research to clinical diagnostic routine.
Collapse
Affiliation(s)
- Martina Siracusano
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
- Child Neurology and Psychiatry Unit, Department of Neurosciences, Policlinico Tor Vergata Hospital, Viale Oxford 81, 00133 Rome, Italy; (L.A.); (A.R.); (L.M.)
| | - Lucrezia Arturi
- Child Neurology and Psychiatry Unit, Department of Neurosciences, Policlinico Tor Vergata Hospital, Viale Oxford 81, 00133 Rome, Italy; (L.A.); (A.R.); (L.M.)
| | - Assia Riccioni
- Child Neurology and Psychiatry Unit, Department of Neurosciences, Policlinico Tor Vergata Hospital, Viale Oxford 81, 00133 Rome, Italy; (L.A.); (A.R.); (L.M.)
| | - Antonio Noto
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria, SS 554, Km 4.5, 09042 Monserrato, Italy
| | - Michele Mussap
- Department of Surgical Sciences, School of Medicine, University of Cagliari, Cittadella Universitaria, SS 554, Km 4.5, 09042 Monserrato, Italy
| | - Luigi Mazzone
- Child Neurology and Psychiatry Unit, Department of Neurosciences, Policlinico Tor Vergata Hospital, Viale Oxford 81, 00133 Rome, Italy; (L.A.); (A.R.); (L.M.)
- Systems Medicine Department, University of Rome Tor Vergata, Montpellier Street 1, 00133 Rome, Italy
| |
Collapse
|
22
|
Liu NH, Liu HQ, Zheng JY, Zhu ML, Wu LH, Pan HF, He XX. Fresh Washed Microbiota Transplantation Alters Gut Microbiota Metabolites to Ameliorate Sleeping Disorder Symptom of Autistic Children. J Microbiol 2023; 61:741-753. [PMID: 37665552 DOI: 10.1007/s12275-023-00069-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 07/25/2023] [Accepted: 07/30/2023] [Indexed: 09/05/2023]
Abstract
Accumulating studies have raised concerns about gut dysbiosis associating autism spectrum disorder (ASD) and its related symptoms. However, the effect of gut microbiota modification on the Chinese ASD population and its underlying mechanism were still elusive. Herein, we enrolled 24 ASD children to perform the first course of fresh washed microbiota transplantation (WMT), 18 patients decided to participate the second course, 13 of which stayed to participate the third course, and there were 8 patients at the fourth course. Then we evaluated the effects of fresh WMT on these patients and their related symptoms. Our results found that the sleeping disorder symptom was positively interrelated to ASD, fresh WMT significantly alleviated ASD and its sleeping disorder and constipation symptoms. In addition, WMT stably and continuously downregulated Bacteroides/Flavonifractor/Parasutterella while upregulated Prevotella_9 to decrease toxic metabolic production and improve detoxification by regulating glycolysis/myo-inositol/D-glucuronide/D-glucarate degradation, L-1,2-propanediol degradation, fatty acid β-oxidation. Thus, our results suggested that fresh WMT moderated gut microbiome to improve the behavioral and sleeping disorder symptoms of ASD via decrease toxic metabolic production and improve detoxification. Which thus provides a promising gut ecological strategy for ASD children and its related symptoms treatments.
Collapse
Affiliation(s)
- Nai-Hua Liu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, People's Republic of China
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Nonglin Down Street 19, Guangzhou, 510080, People's Republic of China
| | - Hong-Qian Liu
- Department of Gastroenterology, Research Center for Engineering Techniques of Microbiota-Targeted Therapies of Guangdong Province, The First Affiliated Hospital of Guangdong Pharmaceutical University, Nonglin Down Street 19, Guangzhou, 510080, People's Republic of China
| | - Jia-Yi Zheng
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, People's Republic of China
| | - Meng-Lu Zhu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, People's Republic of China
| | - Li-Hao Wu
- Department of Gastroenterology, Research Center for Engineering Techniques of Microbiota-Targeted Therapies of Guangdong Province, The First Affiliated Hospital of Guangdong Pharmaceutical University, Nonglin Down Street 19, Guangzhou, 510080, People's Republic of China
| | - Hua-Feng Pan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, People's Republic of China.
| | - Xing-Xiang He
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Nonglin Down Street 19, Guangzhou, 510080, People's Republic of China.
- Department of Gastroenterology, Research Center for Engineering Techniques of Microbiota-Targeted Therapies of Guangdong Province, The First Affiliated Hospital of Guangdong Pharmaceutical University, Nonglin Down Street 19, Guangzhou, 510080, People's Republic of China.
| |
Collapse
|
23
|
Nohesara S, Abdolmaleky HM, Thiagalingam S. Epigenetic Aberrations in Major Psychiatric Diseases Related to Diet and Gut Microbiome Alterations. Genes (Basel) 2023; 14:1506. [PMID: 37510410 PMCID: PMC10379841 DOI: 10.3390/genes14071506] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 07/30/2023] Open
Abstract
Nutrition and metabolism modify epigenetic signatures like histone acetylation and DNA methylation. Histone acetylation and DNA methylation in the central nervous system (CNS) can be altered by bioactive nutrients and gut microbiome via the gut-brain axis, which in turn modulate neuronal activity and behavior. Notably, the gut microbiome, with more than 1000 bacterial species, collectively contains almost three million functional genes whose products interact with millions of human epigenetic marks and 30,000 genes in a dynamic manner. However, genetic makeup shapes gut microbiome composition, food/nutrient metabolism, and epigenetic landscape, as well. Here, we first discuss the effect of changes in the microbial structure and composition in shaping specific epigenetic alterations in the brain and their role in the onset and progression of major mental disorders. Afterward, potential interactions among maternal diet/environmental factors, nutrition, and gastrointestinal microbiome, and their roles in accelerating or delaying the onset of severe mental illnesses via epigenetic changes will be discussed. We also provide an overview of the association between the gut microbiome, oxidative stress, and inflammation through epigenetic mechanisms. Finally, we present some underlying mechanisms involved in mediating the influence of the gut microbiome and probiotics on mental health via epigenetic modifications.
Collapse
Affiliation(s)
- Shabnam Nohesara
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA 02218, USA; (S.N.); (S.T.)
| | - Hamid Mostafavi Abdolmaleky
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA 02218, USA; (S.N.); (S.T.)
- Nutrition/Metabolism Laboratory, Beth Israel Deaconess Medical Center, Harvard Medical School, Boson, MA 02215, USA
| | - Sam Thiagalingam
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA 02218, USA; (S.N.); (S.T.)
- Department of Pathology & Laboratory Medicine, Boston University School of Medicine, Boston, MA 02218, USA
| |
Collapse
|
24
|
Xiong RG, Li J, Cheng J, Zhou DD, Wu SX, Huang SY, Saimaiti A, Yang ZJ, Gan RY, Li HB. The Role of Gut Microbiota in Anxiety, Depression, and Other Mental Disorders as Well as the Protective Effects of Dietary Components. Nutrients 2023; 15:3258. [PMID: 37513676 PMCID: PMC10384867 DOI: 10.3390/nu15143258] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 07/20/2023] [Accepted: 07/21/2023] [Indexed: 07/30/2023] Open
Abstract
The number of individuals experiencing mental disorders (e.g., anxiety and depression) has significantly risen in recent years. Therefore, it is essential to seek prevention and treatment strategies for mental disorders. Several gut microbiota, especially Firmicutes and Bacteroidetes, are demonstrated to affect mental health through microbiota-gut-brain axis, and the gut microbiota dysbiosis can be related to mental disorders, such as anxiety, depression, and other mental disorders. On the other hand, dietary components, including probiotics (e.g., Lactobacillus and Bifidobacterium), prebiotics (e.g., dietary fiber and alpha-lactalbumin), synbiotics, postbiotics (e.g., short-chain fatty acids), dairy products, spices (e.g., Zanthoxylum bungeanum, curcumin, and capsaicin), fruits, vegetables, medicinal herbs, and so on, could exert protective effects against mental disorders by enhancing beneficial gut microbiota while suppressing harmful ones. In this paper, the mental disorder-associated gut microbiota are summarized. In addition, the protective effects of dietary components on mental health through targeting the gut microbiota are discussed. This paper can be helpful to develop some dietary natural products into pharmaceuticals and functional foods to prevent and treat mental disorders.
Collapse
Affiliation(s)
- Ruo-Gu Xiong
- School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (R.-G.X.); (J.C.); (D.-D.Z.); (S.-X.W.); (S.-Y.H.); (A.S.); (Z.-J.Y.)
| | - Jiahui Li
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China;
| | - Jin Cheng
- School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (R.-G.X.); (J.C.); (D.-D.Z.); (S.-X.W.); (S.-Y.H.); (A.S.); (Z.-J.Y.)
| | - Dan-Dan Zhou
- School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (R.-G.X.); (J.C.); (D.-D.Z.); (S.-X.W.); (S.-Y.H.); (A.S.); (Z.-J.Y.)
| | - Si-Xia Wu
- School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (R.-G.X.); (J.C.); (D.-D.Z.); (S.-X.W.); (S.-Y.H.); (A.S.); (Z.-J.Y.)
| | - Si-Yu Huang
- School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (R.-G.X.); (J.C.); (D.-D.Z.); (S.-X.W.); (S.-Y.H.); (A.S.); (Z.-J.Y.)
| | - Adila Saimaiti
- School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (R.-G.X.); (J.C.); (D.-D.Z.); (S.-X.W.); (S.-Y.H.); (A.S.); (Z.-J.Y.)
| | - Zhi-Jun Yang
- School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (R.-G.X.); (J.C.); (D.-D.Z.); (S.-X.W.); (S.-Y.H.); (A.S.); (Z.-J.Y.)
| | - Ren-You Gan
- Singapore Institute of Food and Biotechnology Innovation (SIFBI), Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, Singapore 138669, Singapore
| | - Hua-Bin Li
- School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (R.-G.X.); (J.C.); (D.-D.Z.); (S.-X.W.); (S.-Y.H.); (A.S.); (Z.-J.Y.)
| |
Collapse
|
25
|
Naufel MF, Truzzi GDM, Ferreira CM, Coelho FMS. The brain-gut-microbiota axis in the treatment of neurologic and psychiatric disorders. ARQUIVOS DE NEURO-PSIQUIATRIA 2023. [PMID: 37402401 PMCID: PMC10371417 DOI: 10.1055/s-0043-1767818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/06/2023]
Abstract
The human gut microbiota is a complex ecosystem made of trillions of microorganisms. The composition can be affected by diet, metabolism, age, geography, stress, seasons, temperature, sleep, and medications. The increasing evidence about the existence of a close and bi-directional correlation between the gut microbiota and the brain indicates that intestinal imbalance may play a vital role in the development, function, and disorders of the central nervous system. The mechanisms of interaction between the gut-microbiota on neuronal activity are widely discussed. Several potential pathways are involved with the brain-gut-microbiota axis, including the vagus nerve, endocrine, immune, and biochemical pathways. Gut dysbiosis has been linked to neurological disorders in different ways that involve activation of the hypothalamic-pituitary-adrenal axis, imbalance in neurotransmitter release, systemic inflammation, and increase in the permeability of the intestinal and the blood-brain barrier. Mental and neurological diseases have become more prevalent during the coronavirus disease 2019pandemic and are an essential issue in public health globally. Understanding the importance of diagnosing, preventing, and treating dysbiosis is critical because gut microbial imbalance is a significant risk factor for these disorders. This review summarizes evidence demonstrating the influence of gut dysbiosis on mental and neurological disorders.
Collapse
Affiliation(s)
| | | | | | - Fernando Morgadinho Santos Coelho
- Universidade Federal de São Paulo, Departamento de Psicobiologia, São Paulo SP, Brazil
- Universidade Federal de São Paulo, Departamento de Neurologia e Neurocirurgia, São Paulo SP, Brazil
| |
Collapse
|
26
|
Yano N, Hosokawa K. The importance of comprehensive support based on the three pillars of exercise, nutrition, and sleep for improving core symptoms of autism spectrum disorders. Front Psychiatry 2023; 14:1119142. [PMID: 37260760 PMCID: PMC10228143 DOI: 10.3389/fpsyt.2023.1119142] [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: 12/08/2022] [Accepted: 03/15/2023] [Indexed: 06/02/2023] Open
Abstract
Autism spectrum disorder (ASD) is classified as a neurodevelopmental disorder. The Diagnostic and Statistical Manual of Mental Disorders (DSM)-V, which first described ASD, lists persistent deficits in social communication and interrelationships, as well as limited and recurrent modes of behavior, interests, and activities as diagnostic items. Until recently, understanding the pathophysiology of ASD has been mostly from a neurophysiological perspective, and interventions have been mostly behavioral and psychological. In recent years, however, it has become clear that ASD also affects many bodily systems, including the immune system, the sensorimotor system, and the gut-brain axis, and that these factors simultaneously influence it. In light of this background, a new "connectivome theory" has been proposed as a hypothesis for understanding ASD. "Exercise," "nutrition," and "sleep," which are discussed in this mini-review, have a particularly strong relationship with the immune, musculoskeletal, and gut systems among the pathologies mentioned in the "connectivome theory," furthermore, many reports suggest improvements in stereo-responsive behavior and social and communication skills, which are the core symptoms of ASD. In addition, these interventions are characterized by being less subject to location and cost limitations and excel in the continuity of therapeutic intervention, and the three interventions may have a reciprocal positive impact and may function as three pillars to support ASD.
Collapse
Affiliation(s)
- Nozomu Yano
- Graduate School of Health Sciences, Doctoral Course, Kagoshima University Graduate School, Kagoshima, Japan
| | - Kenji Hosokawa
- Department of Child Care and Education, Odawara Junior College, Nagoya, Japan
| |
Collapse
|
27
|
López-García E, Benítez-Cabello A, Arenas-de Larriva AP, Gutierrez-Mariscal FM, Pérez-Martínez P, Yubero-Serrano EM, Garrido-Fernández A, Arroyo-López FN. Oral intake of Lactiplantibacillus pentosus LPG1 Produces a Beneficial Regulation of Gut Microbiota in Healthy Persons: A Randomised, Placebo-Controlled, Single-Blind Trial. Nutrients 2023; 15:nu15081931. [PMID: 37111150 PMCID: PMC10144437 DOI: 10.3390/nu15081931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/03/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
The search for vegetable-origin probiotic microorganisms is a recent area of interest. This study conducted a phase I clinical trial to assess the effects of oral administration of Lactiplantibacillus pentosus LPG1, a natural strain with probiotic potential isolated from table olive fermentations, on the gut microbiota. The trial was a randomised, placebo-controlled, single-blind study involving 39 healthy volunteers. Group A (n = 20) ingested one capsule/day of L. pentosus LPG1 containing 1 × 1010 UFC/capsule, while Group B (n = 19) received one capsule/day containing only dextrose (placebo). The capsules were taken during breakfast for 30 consecutive days. Human stool samples were collected from all volunteers at the beginning (baseline) and at the end of the study (post-intervention) and were subjected to 16S rRNA metataxonomic analysis using Illumina MiSeq. Sequencing data at the genus level were statistically analysed using traditional methods and compositional data analysis (CoDA). After treatment, the alpha diversity in Group B (placebo) decreased according to an increase in the Berger and Parker dominance index (p-value < 0.05); moreover, dominance D increased and Simpson 1-D index decreased (p-value < 0.10). The Lactobacillus genus in the faeces was included in the CoDA signature balances (selbal and coda4microbiome) and played a notable role in distinguishing samples from baseline and post-intervention in Group A (LPG1). Additionally, ingesting L. pentosus LPG1 modified the gut microbiota post-intervention, increasing the presence of Parabacteroides and Agathobacter, but reducing Prevotella. These findings suggest that L. pentosus LPG1 is a potentially beneficial gut microbiota modulator in healthy persons.
Collapse
Affiliation(s)
- Elio López-García
- Food Biotechnology Department. Instituto de la Grasa (CSIC), Carretera Utrera km 1, Campus Universitario Pablo de Olavide, Building 46, 41013 Seville, Spain
| | - Antonio Benítez-Cabello
- Food Biotechnology Department. Instituto de la Grasa (CSIC), Carretera Utrera km 1, Campus Universitario Pablo de Olavide, Building 46, 41013 Seville, Spain
| | - Antonio Pablo Arenas-de Larriva
- Unidad de Gestión Clinica Medicina Interna, Lipids and Atherosclerosis Unit, Maimonides Institute for Biomedical Research in Córdoba, Reina Sofia University Hospital, University of Córdoba, 14004 Córdoba, Spain
| | - Francisco Miguel Gutierrez-Mariscal
- Unidad de Gestión Clinica Medicina Interna, Lipids and Atherosclerosis Unit, Maimonides Institute for Biomedical Research in Córdoba, Reina Sofia University Hospital, University of Córdoba, 14004 Córdoba, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
| | - Pablo Pérez-Martínez
- Unidad de Gestión Clinica Medicina Interna, Lipids and Atherosclerosis Unit, Maimonides Institute for Biomedical Research in Córdoba, Reina Sofia University Hospital, University of Córdoba, 14004 Córdoba, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
| | - Elena María Yubero-Serrano
- Unidad de Gestión Clinica Medicina Interna, Lipids and Atherosclerosis Unit, Maimonides Institute for Biomedical Research in Córdoba, Reina Sofia University Hospital, University of Córdoba, 14004 Córdoba, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
| | - Antonio Garrido-Fernández
- Food Biotechnology Department. Instituto de la Grasa (CSIC), Carretera Utrera km 1, Campus Universitario Pablo de Olavide, Building 46, 41013 Seville, Spain
| | - Francisco Noé Arroyo-López
- Food Biotechnology Department. Instituto de la Grasa (CSIC), Carretera Utrera km 1, Campus Universitario Pablo de Olavide, Building 46, 41013 Seville, Spain
| |
Collapse
|
28
|
Zheng L, Cao T, Xiong P, Ma Y, Wei L, Wang J. Characterization of the oral microbiome and gut microbiome of dental caries and extrinsic black stain in preschool children. Front Microbiol 2023; 14:1081629. [PMID: 37065128 PMCID: PMC10103904 DOI: 10.3389/fmicb.2023.1081629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 03/10/2023] [Indexed: 04/03/2023] Open
Abstract
IntroductionA lower prevalence of dental caries (hereafter termed “caries”) has been observed in children with dental extrinsic black stain (EBS).MethodsWe investigated the epidemiologic characterization of EBS and explored the possible role of the oral microbiome (OM) and gut microbiome (GM) in EBS formation and caries prevention. In an epidemiologic survey, 2,675 children aged 3–6 years were included. Thirty-eight of these children (7 children had both caries and EBS, 10 had EBS only, 11 had caries only, and 10 were healthy children) were recruited for 16S rRNA sequencing and collection of samples of supragingival plaque and feces. Collected plaque samples were divided into four groups: BCP (EBS+, caries+), BP (EBS+, caries−), CP (EBS−, caries+), and P (EBS−, caries−). Fecal samples were also divided into four groups: BCF (EBS+, caries+), BF (EBS+, caries−), CF (EBS−, caries+), and F (EBS−, caries−).ResultsEBS was observed in 12.10% of this population. Children with EBS had a significantly reduced prevalence of caries and a lower mean value of decayed–missing–filled teeth (dmft; p < 0.01). According to analyses of dental plaque, the P group had the most complex microbiome. The BCP group exhibited greater operational taxonomic unit (OTU) richness but a reduced evenness compared with the BP group, and the CP group showed greater OTU richness than the BP group. At the genus level, higher abundance of Actinomyces and Cardiobacterium species was observed in the BCP group. Higher abundance of Lautropia and Pesudopropionibacteriumin species was observed in the BP group compared with P and CP groups, respectively (p < 0.05). Veillonella species were significantly more common in P and CP groups than in BP groups, whereas Porphyromonas and Fusobacterium species were more common in the CP group (p < 0.05). With regard to the GM, the CF group exhibited greater OTU diversity than the BF group. The GM in the BCF group exhibited the most complex relationships across all fecal groups. GM groups could be distinguished by various unique biomarkers, such as Escherichia and Shigella species in the BCF group, Agathobacter and Ruminococcus species in the CF group, Lactobacillus species in the BF group, and Roseburia species in the F group. Our results suggest that EBS is a possible protective factor against early-childhood caries. Dental plaque and the GM may be relevant to EBS in primary dentition.
Collapse
Affiliation(s)
- Luoyuan Zheng
- School and Hospital of Stomatology, Wenzhou Medical University,, Wenzhou, China
| | - Tingting Cao
- School and Hospital of Stomatology, Wenzhou Medical University,, Wenzhou, China
| | - Puling Xiong
- School and Hospital of Stomatology, Wenzhou Medical University,, Wenzhou, China
| | - Yulian Ma
- School and Hospital of Stomatology, Wenzhou Medical University,, Wenzhou, China
| | - Limin Wei
- Department of Preventive Dentistry, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
- *Correspondence: Limin Wei, ; Jianfeng Wang,
| | - Jianfeng Wang
- Department of Orthodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
- *Correspondence: Limin Wei, ; Jianfeng Wang,
| |
Collapse
|
29
|
Kuo YJ, Chen CJ, Hussain B, Tsai HC, Hsu GJ, Chen JS, Asif A, Fan CW, Hsu BM. Inferring Bacterial Community Interactions and Functionalities Associated with Osteopenia and Osteoporosis in Taiwanese Postmenopausal Women. Microorganisms 2023; 11:234. [PMID: 36838199 PMCID: PMC9959971 DOI: 10.3390/microorganisms11020234] [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: 12/02/2022] [Revised: 12/30/2022] [Accepted: 01/13/2023] [Indexed: 01/19/2023] Open
Abstract
Growing evidence suggests that the gut microbiota and their metabolites are associated with bone homeostasis and fragility. However, this association is limited to microbial taxonomic differences. This study aimed to explore whether gut bacterial community associations, composition, and functions are associated with osteopenia and osteoporosis. We compared the gut bacterial community composition and interactions of healthy postmenopausal women with normal bone density (n = 8) with those of postmenopausal women with osteopenia (n = 18) and osteoporosis (n = 21) through 16S rRNA sequencing coupled with network biology and statistical analyses. The results of this study showed reduced alpha diversity in patients with osteoporosis, followed by that in patients with osteopenia, then in healthy controls. Taxonomic analysis revealed that significantly enriched bacterial genera with higher abundance was observed in patients with osteoporosis and osteopenia than in healthy subjects. Additionally, a co-occurrence network revealed that, compared to healthy controls, bacterial interactions were higher in patients with osteoporosis, followed by those with osteopenia. Further, NetShift analysis showed that a higher number of bacteria drove changes in the microbial community structure of patients with osteoporosis than osteopenia. Correlation analysis revealed that most of these driver bacteria had a significant positive relationship with several significant metabolic pathways. Further, ordination analysis revealed that height and T-score were the primary variables influencing the gut microbial community structure. Taken together, this study evaluated that microbial community interaction is more important than the taxonomic differences in knowing the critical role of gut microbiota in postmenopausal women associated with osteopenia and osteoporosis. Additionally, the significantly enriched bacteria and functional pathways might be potential biomarkers for the prognosis and treatment of postmenopausal women with osteopenia and osteoporosis.
Collapse
Affiliation(s)
- Yi-Jie Kuo
- Department of Orthopedic Surgery, Wan Fang Hospital, Taipei Medical University, Taipei 116, Taiwan
| | - Chia-Jung Chen
- Department of Chinese Medicine, Dalin Tzu Chi Hospital, The Buddhist Tzu Chi Medical Foundation, Chiayi 622, Taiwan
| | - Bashir Hussain
- Department of Earth and Environmental Sciences, National Chung Cheng University, Chiayi 621, Taiwan
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi 621, Taiwan
| | - Hsin-Chi Tsai
- Department of Psychiatry, School of Medicine, Tzu Chi University, Hualien 970, Taiwan
- Department of Psychiatry, Tzu-Chi General Hospital, Hualien 970, Taiwan
| | - Gwo-Jong Hsu
- Division of Infectious Disease, Department of Internal Medicine, Chia-Yi Christian Hospital, Chiayi 621, Taiwan
| | - Jung-Sheng Chen
- Department of Medical Research, E-Da Hospital, I-Shou University, Kaohsiung 824, Taiwan
| | - Aslia Asif
- Department of Earth and Environmental Sciences, National Chung Cheng University, Chiayi 621, Taiwan
- Doctoral Program in Science, Technology, Environment and Mathematics, National Chung Cheng University, Chiayi 621, Taiwan
| | - Cheng-Wei Fan
- Department of Earth and Environmental Sciences, National Chung Cheng University, Chiayi 621, Taiwan
| | - Bing-Mu Hsu
- Department of Earth and Environmental Sciences, National Chung Cheng University, Chiayi 621, Taiwan
| |
Collapse
|
30
|
Dowley A, Sweeney T, Conway E, Vigors S, Ryan MT, Yadav S, Wilson J, O'Doherty JV. The effects of dietary supplementation with mushroom or selenium enriched mushroom powders on the growth performance and intestinal health of post-weaned pigs. J Anim Sci Biotechnol 2023; 14:12. [PMID: 36631908 PMCID: PMC9832780 DOI: 10.1186/s40104-022-00808-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 11/23/2022] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND There is an urgent need to identify natural bioactive compounds that can enhance gastrointestinal health and promote pig growth performance in the absence of pharmacological levels of zinc oxide (ZnO). The objectives of this study were to: 1) compare the effects of mushroom powder supplemented with inorganic selenium (inSeMP) to mushroom powder enriched with organic selenium (orgSeMP) to pharmacological levels of ZnO on growth performance and faecal scores (FS) for the first 21 d post-weaning (Period 1); and 2) compare the molecular and microbial effects of inSeMP and orgSeMP in these pigs on d 39 post-weaning (Period 2). METHODS In Period 1, pigs (3 pigs/pen; 8 pens/treatment) were assigned to: (1) basal diet (control); (2) basal diet + zinc oxide (ZnO) (3100 mg/kg d 1-14, 1550 mg/kg d 15-21); (3) basal diet + mushroom powder supplemented with inorganic selenium (inSeMP) containing selenium (selenite) content of 0.3 mg/kg feed; (4) basal diet + mushroom powder enriched with organic selenium (orgSeMP) containing selenium (selenocysteine) content of 0.3 mg/kg feed. Mushroom powders were included at 6.5 g/kg of feed. RESULTS In Period 1, there was no effect of diets on average daily gain (ADG) and gain:feed (G:F) ratio (P > 0.05). The orgSeMP supplemented pigs had a lower average daily feed intake (ADFI) compared to all other groups (P < 0.05). The ZnO supplemented pigs had reduced FS compared to the basal and mushroom group, while the orgSeMP supplemented pigs had lower FS compared to the basal group during the 21 d experimental period (P < 0.05). In Period 2, there was no effect of diets on ADFI, ADG and G:F ratio (P > 0.05). The orgSeMP supplementation increased the caecal abundance of bacterial members of the Firmicutes and Bacteroidetes phylum, including Lactobacillus, Agathobacter, Roseburia, and Prevotella and decreased the abundance of Sporobacter compared to the basal group, while inSeMP increased the caecal abundance of Prevotella and decreased the caecal abundance of Sporobacter compared to the basal group (P < 0.05). Dietary supplementation with inSeMP increased expression of TLR4 and anti-inflammatory cytokine gene IL10 and decreased nutrient transporter gene FABP2 compared to the orgSeMP group (P < 0.05). CONCLUSION OrgSeMP is a novel and sustainable way to incorporate selenium and β-glucans into the diet of weaned pigs whilst improving FS and modulating the caecal microbiota.
Collapse
Affiliation(s)
- Alison Dowley
- School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Torres Sweeney
- School of Veterinary Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Eadaoin Conway
- School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Stafford Vigors
- School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Marion T Ryan
- School of Veterinary Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Supriya Yadav
- Mbio, Monaghan Mushroom Group, Tyholland, Co.Monaghan, Ireland
| | - Jude Wilson
- Mbio, Monaghan Mushroom Group, Tyholland, Co.Monaghan, Ireland
| | - John V O'Doherty
- School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
31
|
Jiang H, Deng S, Zhang J, Chen J, Li B, Zhu W, Zhang M, Zhang C, Meng Z. Acupuncture treatment for post-stroke depression: Intestinal microbiota and its role. Front Neurosci 2023; 17:1146946. [PMID: 37025378 PMCID: PMC10070763 DOI: 10.3389/fnins.2023.1146946] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 03/06/2023] [Indexed: 04/08/2023] Open
Abstract
Stroke-induced depression is a common complication and an important risk factor for disability. Besides psychiatric symptoms, depressed patients may also exhibit a variety of gastrointestinal symptoms, and even take gastrointestinal symptoms as the primary reason for medical treatment. It is well documented that stress may disrupt the balance of the gut microbiome in patients suffering from post-stroke depression (PSD), and that disruption of the gut microbiome is closely related to the severity of the condition in depressed patients. Therefore, maintaining the balance of intestinal microbiota can be the focus of research on the mechanism of acupuncture in the treatment of PSD. Furthermore, stroke can be effectively treated with acupuncture at all stages and it may act as a special microecological regulator by regulating intestinal microbiota as well. In this article, we reviewed the studies on changing intestinal microbiota after acupuncture treatment and examined the existing problems and development prospects of acupuncture, microbiome, and poststroke depression, in order to provide new ideas for future acupuncture research.
Collapse
Affiliation(s)
- Hailun Jiang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shizhe Deng
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jieying Zhang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Junjie Chen
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Boxuan Li
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Weiming Zhu
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Menglong Zhang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Chao Zhang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Chao Zhang,
| | - Zhihong Meng
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- *Correspondence: Zhihong Meng,
| |
Collapse
|
32
|
Ji Q, Li SJ, Zhao JB, Xiong Y, Du XH, Wang CX, Lu LM, Tan JY, Zhu ZR. Genetic and neural mechanisms of sleep disorders in children with autism spectrum disorder: a review. Front Psychiatry 2023; 14:1079683. [PMID: 37200906 PMCID: PMC10185750 DOI: 10.3389/fpsyt.2023.1079683] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 04/13/2023] [Indexed: 05/20/2023] Open
Abstract
Background The incidence of sleep disorders in children with autism spectrum disorder (ASD) is very high. Sleep disorders can exacerbate the development of ASD and impose a heavy burden on families and society. The pathological mechanism of sleep disorders in autism is complex, but gene mutations and neural abnormalities may be involved. Methods In this review, we examined literature addressing the genetic and neural mechanisms of sleep disorders in children with ASD. The databases PubMed and Scopus were searched for eligible studies published between 2013 and 2023. Results Prolonged awakenings of children with ASD may be caused by the following processes. Mutations in the MECP2, VGAT and SLC6A1 genes can decrease GABA inhibition on neurons in the locus coeruleus, leading to hyperactivity of noradrenergic neurons and prolonged awakenings in children with ASD. Mutations in the HRH1, HRH2, and HRH3 genes heighten the expression of histamine receptors in the posterior hypothalamus, potentially intensifying histamine's ability to promote arousal. Mutations in the KCNQ3 and PCDH10 genes cause atypical modulation of amygdala impact on orexinergic neurons, potentially causing hyperexcitability of the hypothalamic orexin system. Mutations in the AHI1, ARHGEF10, UBE3A, and SLC6A3 genes affect dopamine synthesis, catabolism, and reuptake processes, which can elevate dopamine concentrations in the midbrain. Secondly, non-rapid eye movement sleep disorder is closely related to the lack of butyric acid, iron deficiency and dysfunction of the thalamic reticular nucleus induced by PTCHD1 gene alterations. Thirdly, mutations in the HTR2A, SLC6A4, MAOA, MAOB, TPH2, VMATs, SHANK3, and CADPS2 genes induce structural and functional abnormalities of the dorsal raphe nucleus (DRN) and amygdala, which may disturb REM sleep. In addition, the decrease in melatonin levels caused by ASMT, MTNR1A, and MTNR1B gene mutations, along with functional abnormalities of basal forebrain cholinergic neurons, may lead to abnormal sleep-wake rhythm transitions. Conclusion Our review revealed that the functional and structural abnormalities of sleep-wake related neural circuits induced by gene mutations are strongly correlated with sleep disorders in children with ASD. Exploring the neural mechanisms of sleep disorders and the underlying genetic pathology in children with ASD is significant for further studies of therapy.
Collapse
Affiliation(s)
- Qi Ji
- Department of Psychology, Army Medical University, Chongqing, China
- College of Basic Medicine, Army Medical University, Chongqing, China
| | - Si-Jia Li
- Department of Psychology, Army Medical University, Chongqing, China
- College of Basic Medicine, Army Medical University, Chongqing, China
| | - Jun-Bo Zhao
- Department of Psychology, Army Medical University, Chongqing, China
- College of Basic Medicine, Army Medical University, Chongqing, China
| | - Yun Xiong
- Department of Psychology, Army Medical University, Chongqing, China
- College of Basic Medicine, Army Medical University, Chongqing, China
| | - Xiao-Hui Du
- Department of Psychology, Army Medical University, Chongqing, China
| | - Chun-Xiang Wang
- Department of Psychology, Army Medical University, Chongqing, China
| | - Li-Ming Lu
- College of Educational Sciences, Chongqing Normal University, Chongqing, China
| | - Jing-Yao Tan
- College of Educational Sciences, Chongqing Normal University, Chongqing, China
| | - Zhi-Ru Zhu
- Department of Psychology, Army Medical University, Chongqing, China
- *Correspondence: Zhi-Ru Zhu,
| |
Collapse
|
33
|
Plaza-Diaz J, Radar AM, Baig AT, Leyba MF, Costabel MM, Zavala-Crichton JP, Sanchez-Martinez J, MacKenzie AE, Solis-Urra P. Physical Activity, Gut Microbiota, and Genetic Background for Children and Adolescents with Autism Spectrum Disorder. CHILDREN (BASEL, SWITZERLAND) 2022; 9:1834. [PMID: 36553278 PMCID: PMC9777368 DOI: 10.3390/children9121834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/19/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022]
Abstract
It is estimated that one in 100 children worldwide has been diagnosed with autism spectrum disorder (ASD). Children with ASD frequently suffer from gut dysbiosis and gastrointestinal issues, findings which possibly play a role in the pathogenesis and/or severity of their condition. Physical activity may have a positive effect on the composition of the intestinal microbiota of healthy adults. However, the effect of exercise both on the gastrointestinal problems and intestinal microbiota (and thus possibly on ASD) itself in affected children is unknown. In terms of understanding the physiopathology and manifestations of ASD, analysis of the gut-brain axis holds some promise. Here, we discuss the physiopathology of ASD in terms of genetics and microbiota composition, and how physical activity may be a promising non-pharmaceutical approach to improve ASD-related symptoms.
Collapse
Affiliation(s)
- Julio Plaza-Diaz
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain
| | - Ana Mei Radar
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Aiman Tariq Baig
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Marcos Federico Leyba
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Maria Macarena Costabel
- Children’s Hospital of Eastern Ontario, Division of Urology, Department of Surgery, University of Ottawa, Ottawa, ON K1H 8L6, Canada
| | | | - Javier Sanchez-Martinez
- Escuela de Kinesiología, Facultad de Salud, Universidad Santo Tomás, Viña del Mar 2520298, Chile
| | - Alex E. MacKenzie
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Patricio Solis-Urra
- Faculty of Education and Social Sciences, Universidad Andres Bello, Viña del Mar 2531015, Chile
- PROFITH “PROmoting FITness and Health through Physical Activity” Research Group, Sport and Health University Research Institute (iMUDS), Department of Physical Education and Sports, Faculty of Sport Sciences, University of Granada, 18071 Granada, Spain
- Servicio de Medicina Nuclear, Hospital Universitario Virgen de las Nieves, 18014 Granada, Spain
| |
Collapse
|
34
|
Deng W, Wang S, Li F, Wang F, Xing YP, Li Y, Lv Y, Ke H, Li Z, Lv PJ, Hao H, Chen Y, Xiao X. Gastrointestinal symptoms have a minor impact on autism spectrum disorder and associations with gut microbiota and short-chain fatty acids. Front Microbiol 2022; 13:1000419. [PMID: 36274684 PMCID: PMC9585932 DOI: 10.3389/fmicb.2022.1000419] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
Children with autism spectrum disorder (ASD) experience gastrointestinal (GI) issues more frequently and severely than children who are typically developing (TD). The connections between gastrointestinal problems, microbiota, and short-chain fatty acids (SCFAs) in ASD are still being debated. We enrolled 90 children, 45 of whom were diagnosed with ASD, and examined the impact of GI disorders on ASD. The six-item GI Severity Index questionnaire was used to evaluate gastrointestinal symptoms, while the Social Responsiveness Scale was used to evaluate autism symptoms. Further, the Children’s Sleep Habits Questionnaire and the Children’s Eating Behavior Questionnaire are used to assess sleep and eating disorders in children. We assessed fecal microbiota by 16S rRNA gene sequencing, and SCFA concentrations by gas chromatography/mass spectrometry. The results revealed that children with ASD exhibited a high rate of gastrointestinal issues (78%), as well as higher rates of social impairment and poor sleeping habits, compared to TD children. However, GI disturbances have a minor impact on autism. In addition, the levels of propionic acid, butyric acid, and valeric acid were significantly higher in the ASD group. Besides, the ASD, TD, and GI subgroups possessed distinct microbiome profiles. These findings suggest that gastrointestinal disturbances have no discernible effect on the core symptoms of autism. Although autism may not cause an increase in GI symptoms directly, alterations in metabolites, such as SCFAs, may cause GI symptoms.
Collapse
Affiliation(s)
- Wenlin Deng
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- *Correspondence: Wenlin Deng,
| | - Siqi Wang
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fang Li
- Department of Gastroenterology, Gastroenterology Endoscopy Center, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Fang Wang
- Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yi Pei Xing
- Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yongchun Li
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ying Lv
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Haoran Ke
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zitong Li
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Pin Jing Lv
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hu Hao
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ye Chen
- Nanfang Hospital, Southern Medical University, Guangzhou, China
- Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Xin Xiao
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
35
|
Lee MJ, Lai HC, Kuo YL, Chen VCH. Association between Gut Microbiota and Emotional-Behavioral Symptoms in Children with Attention-Deficit/Hyperactivity Disorder. J Pers Med 2022; 12:jpm12101634. [PMID: 36294773 PMCID: PMC9605220 DOI: 10.3390/jpm12101634] [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: 06/04/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 11/13/2022] Open
Abstract
Previous studies have explored the role of the microbiome in attention-deficit/hyperactivity disorder (ADHD). However, whether the microbiome is correlated with emotional-behavioral disturbances, the most common comorbid symptom of ADHD, remains unclear. We established a cross-sectional study in which 6- to 18-year-old children with ADHD who were receiving no medication and a healthy control group of children without ADHD were recruited to analyze their microbiome composition. Microbiota of fecal samples were collected and analyzed using a 16s rRNA gene sequencing approach. In comparison with the healthy control group, the gut microbiota in children with ADHD exhibited significantly lower beta diversity. The abundance of the phylum Proteobacteria and the genera Agathobacter, Phascolarctobacterium, Prevotella_2, Acidaminococcus, Roseburia, and Ruminococcus gnavus group was increased in the ADHD group compared with the healthy group. Linear discriminant effect size (LEfSe) analysis was used to highlight specific bacteria phylotypes that were differentially altered between the ADHD and control groups. A regression analysis was performed to investigate the association between microbiota and emotional-behavioral symptoms in children with ADHD. A significant association was noted between withdrawal and depression symptoms and Agathobacter (p = 0.044), and between rule-breaking behavior and the Ruminococcus gnavus group (p = 0.046) after adjusting for sex, age, and the ADHD core symptoms score. This study advances the knowledge of how gut microbiota composition may contribute to emotional-behavioral symptoms in children with ADHD. The detailed mechanisms underlying the role of the gut microbiota in ADHD pathophysiology still require further investigation.
Collapse
Affiliation(s)
- Min-Jing Lee
- Department of Psychiatry, Chang Gung Memorial Hospital, Chiayi Branch, Chiayi 613, Taiwan
- School of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Hsin-Chih Lai
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Microbiota Research Center and Emerging Viral Infections Research Center, Chang Gung University, Taoyuan 333, Taiwan
- Central Research Laboratory, Xiamen Chang Gung Hospital, Xiamen 361, China
- Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
- Research Center for Chinese Herbal Medicine and Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan
| | - Yu-Lun Kuo
- Biotools Co., Ltd., New Taipei City 221, Taiwan
| | - Vincent Chin-Hung Chen
- Department of Psychiatry, Chang Gung Memorial Hospital, Chiayi Branch, Chiayi 613, Taiwan
- School of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Correspondence: ; Tel.: +886-5-3621000 (ext. 2315); Fax: +886-5-3623002
| |
Collapse
|
36
|
Qiu L, Gong F, Wu J, You D, Zhao Y, Xu L, Cao X, Bao F. Exercise Interventions Improved Sleep Quality through Regulating Intestinal Microbiota Composition. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph191912385. [PMID: 36231686 PMCID: PMC9564517 DOI: 10.3390/ijerph191912385] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/18/2022] [Accepted: 09/26/2022] [Indexed: 05/08/2023]
Abstract
(1) Background: Sleep quality is closely related to the physical and mental health of college students. The objectives of this study were to obtain data on the sleep quality of university students and to investigate the relationship between intestinal flora and the improvement in sleep quality through exercise intervention. (2) Methods: Here, 11 university students with a body mass index (BMI) ≤ 18 and Pittsburgh Sleep Quality Index (PSQI) ≥ 7 were selected as experimental subjects, and another 11 healthy people were recruited as control subjects. The experimental group and control group were each intervened with exercise for 8 weeks. We used 16SrDNA sequencing technology to analyze the variations of the intestinal flora and the relation of the variations and sleep quality improvement between the experimental group and the control group before and after the exercise intervention. (3) Results: The differences in gut flora composition between people with sleep disorders and healthy people were statistically significant (p < 0.05). Before and after the exercise intervention, the differences were also statistically significant (p < 0.05) in people with sleep disorders. The sleep-disordered population had a larger proportion compared with the healthy population (p < 0.05). Blautia and Eubacterium hallii were microbe markers in the sleep-disordered population before and after the exercise intervention, while there was no microbe marker found in the healthy population. (4) Conclusions: The increase in Blautia and Eubacterium hallii, and the decrease in Agathobacter are associated with healthy sleep. Gut flora may be related to sleep disorders. Exercise intervention can improve sleep quality while changing the diversity of the gut flora, and exercise intervention targeting the gut flora is a new concept for preventing and treating sleep disorders.
Collapse
Affiliation(s)
- Liangwu Qiu
- Department of Physical Education, Kunming Medical University, Kunming 650500, China
| | - Fuhong Gong
- Faculty of Basic Medical Science, Kunming Medical University, Kunming 650500, China
| | - Jiang Wu
- Department of Physical Education, Kunming Medical University, Kunming 650500, China
| | - Dingyun You
- Biomedical Engineering Research Center, Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Kunming Medical University, Kunming 650500, China
| | - Yinzhou Zhao
- Faculty of Basic Medical Science, Kunming Medical University, Kunming 650500, China
| | - Lianwu Xu
- Department of Physical Education, Kunming Medical University, Kunming 650500, China
- Correspondence: (L.X.); (X.C.); (F.B.)
| | - Xue Cao
- Department of Laboratory Animal Science, Kunming Medical University, Kunming 650500, China
- Correspondence: (L.X.); (X.C.); (F.B.)
| | - Fukai Bao
- Faculty of Basic Medical Science, Kunming Medical University, Kunming 650500, China
- Correspondence: (L.X.); (X.C.); (F.B.)
| |
Collapse
|
37
|
Zhao D, Liu H, Zhang H, Liu K, Zhang X, Liu Q, Wu Y, Zhang T, Zhang Q. Dietary supplementation with Cyberlindnera jadinii improved growth performance, serum biochemical Indices, antioxidant status, and intestinal health in growing raccoon dogs (Nyctereutes procyonoides). Front Microbiol 2022; 13:973384. [PMID: 36212816 PMCID: PMC9532689 DOI: 10.3389/fmicb.2022.973384] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
This study was conducted to investigate whether different dietary Cyberlindnera jadinii levels affect growth performance, serum immunity, antioxidant capacity, and intestinal microbiota in growing raccoon dogs. Forty-five healthy male raccoon dogs were randomly assigned to three treatment groups, with 15 raccoon dogs per group. Each raccoon dog was housed in an individual cage. The raccoon dogs in the three groups were fed diets supplemented with Cyberlindnera jadinii at dosages of 0 (N group), 1 × 109 (L group) and 5 × 109 CFU/g (H group). A 7-day pretest period preceded a formal test period of 30 days. The results showed that Cyberlindnera jadinii in the L and H groups improved average daily gain (ADG) (P < 0.05) and decreased the ratio of feed to weight (F/G) (P < 0.05). Serum immunoglobulins A and G levels were increased in the L and H groups compared to the N group (P < 0.05). Cyberlindnera jadinii in the L and H groups increased serum superoxide dismutase activity (P < 0.05), and serum glutathione peroxidase activity was increased in the L group compared to the N group (P < 0.05). The relative abundance of Firmicutes and Actinobacteriota were increased, and the relative abundance of Bacteroidota was decreased in the L and H groups compared to the N group (P < 0.05). The relative abundance of Proteobacteria and Cyanobacteria was increased in the H group compared to the other two groups (P < 0.05). The ratio of Firmicutes to Bacteroidetes in the Cyberlindnera jadinii supplementation groups increased compared with the N group (P < 0.05). The relative abundance of Megasphaera and Bifidobacterium were increased, and the relative abundance of Prevotella was decreased in the L and H groups compared to the N group (P < 0.05). The relative abundance of Dialister was increased, while the relative abundance of Blautia was decreased in the H group compared to the other two groups (P < 0.05). The relative abundance of Agathobacter was decreased in the H group compared to the N group (P < 0.05). In conclusion, dietary supplementation with Cyberlindnera jadinii increased growth performance, serum immunity, antioxidant capacity, and improved intestinal microbiota in growing raccoon dogs. Cyberlindnera jadinii can therefore be used as a growth promoter in raccoon dogs.
Collapse
|
38
|
Rustanti N, Murdiati A, Juffrie M, Rahayu ES. Effect of Probiotic Lactobacillus plantarum Dad-13 on Metabolic Profiles and Gut Microbiota in Type 2 Diabetic Women: A Randomized Double-Blind Controlled Trial. Microorganisms 2022; 10:microorganisms10091806. [PMID: 36144408 PMCID: PMC9502685 DOI: 10.3390/microorganisms10091806] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 11/30/2022] Open
Abstract
Several pathways link type 2 diabetes (T2D) mellitus to the gut microbiome. By modifying the gut microbiota (GM), probiotics may be useful in the treatment of T2D. Lactobacillus plantarum Dad-13 is an indigenous Indonesian probiotic strain that has colonized the digestive tracts of healthy Indonesian adults. Furthermore, the GM of Indonesians is dominated by L. plantarum. The probiotic L. plantarum Dad-13 is likely suitable for Indonesians. This study aimed to assess the effect of the probiotic L. plantarum Dad-13 on metabolic profiles and GM of women with T2D in Yogyakarta, Indonesia. Twenty women from each group of forty T2D patients received either a probiotic or a placebo. The probiotic group consumed 1 g skim milk powder containing 1010 CFU/g L. plantarum daily for 11 weeks. The placebo group received 1 g skim milk powder only daily for 11 weeks. At the start and end of the experiment, anthropometric measures, dietary intake surveys, blood samples, and fecal samples were obtained. The GM analysis of all samples was performed using polymerase chain reaction, and Illumina Novaseq was applied to the selected samples from each group at the beginning and end of the trial. Short-chain fatty acids (SCFAs) were analyzed with gas chromatography. The level of HbA1c in the probiotic group (n:10) significantly decreased from 9.34 ± 2.79% to 8.32 ± 2.04%. However, in comparison with the placebo (n:8), L. plantarum Dad-13 supplementation did not significantly decrease the HbA1c level. No significant change was observed in the fasting blood sugar and total cholesterol levels in either group. The GM analysis showed that L. plantarum Dad-13 supplementation resulted in a considerable increase in the L. plantarum number. No significant changes were observed in the Bifidobacterium and Prevotella populations. In addition, no significant change was observed in the fecal pH and SCFA (e.g., acetic acid, propionate, butyrate, and total SCFA) after supplementation with L. plantarum Dad-13.
Collapse
Affiliation(s)
- Ninik Rustanti
- Department of Food and Agricultural Product Technology, Faculty of Agricultural Technology, Universitas Gadjah Mada, Jl. Flora No 1 Bulaksumur, Yogyakarta 55281, Indonesia
- Department of Nutrition Science, Faculty of Medicine, Universitas Diponegoro, Jl. Prof Soedarto, Tembalang Semarang 50275, Indonesia
| | - Agnes Murdiati
- Department of Food and Agricultural Product Technology, Faculty of Agricultural Technology, Universitas Gadjah Mada, Jl. Flora No 1 Bulaksumur, Yogyakarta 55281, Indonesia
| | - Mohammad Juffrie
- Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Jl. Farmako Sekip Utara, Yogyakarta 55281, Indonesia
| | - Endang Sutriswati Rahayu
- Department of Food and Agricultural Product Technology, Faculty of Agricultural Technology, Universitas Gadjah Mada, Jl. Flora No 1 Bulaksumur, Yogyakarta 55281, Indonesia
- Center for Food and Nutrition Studies, Universitas Gadjah Mada, Jl. Teknika Utara Barek, Yogyakarta 55281, Indonesia
- Center of Excellence for Probiotics, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
- Correspondence:
| |
Collapse
|
39
|
The microbiota-gut-brain axis in sleep disorders. Sleep Med Rev 2022; 65:101691. [DOI: 10.1016/j.smrv.2022.101691] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 08/04/2022] [Accepted: 08/19/2022] [Indexed: 12/25/2022]
|
40
|
Huang P, Yu L, Tian F, Zhao J, Zhang H, Chen W, Zhai Q. Untargeted metabolomics revealed the key metabolites in milk fermented with starter cultures containing Lactobacillus plantarum CCFM8610. Lebensm Wiss Technol 2022. [DOI: 10.1016/j.lwt.2022.113768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
41
|
Effects of Traumatic Brain Injury on the Gut Microbiota Composition and Serum Amino Acid Profile in Rats. Cells 2022; 11:cells11091409. [PMID: 35563713 PMCID: PMC9102408 DOI: 10.3390/cells11091409] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/14/2022] [Accepted: 04/19/2022] [Indexed: 02/04/2023] Open
Abstract
Traumatic brain injury (TBI) heavily impacts the body: it damages the brain tissue and the peripheral nervous system and shifts homeostasis in many types of tissue. An acute brain injury compromises the “brain–gut-microbiome axis”, a well-balanced network formed by the brain, gastrointestinal tract, and gut microbiome, which has a complex effect: damage to the brain alters the composition of the microbiome; the altered microbiome affects TBI severity, neuroplasticity, and metabolic pathways through various bacterial metabolites. We modeled TBI in rats. Using a bioinformatics approach, we sought to identify correlations between the gut microbiome composition, TBI severity, the rate of neurological function recovery, and blood metabolome. We found that the TBI caused changes in the abundance of 26 bacterial genera. The most dramatic change was observed in the abundance of Agathobacter species. The TBI also altered concentrations of several metabolites, specifically citrulline and tryptophan. We found no significant correlations between TBI severity and the pre-existing gut microbiota composition or blood metabolites. However, we discovered some differences between the two groups of subjects that showed high and low rates of neurological function recovery, respectively. The present study highlights the role of the brain–gut-microbiome axis in TBI.
Collapse
|
42
|
Analysis of Faecal Microbiota and Small ncRNAs in Autism: Detection of miRNAs and piRNAs with Possible Implications in Host-Gut Microbiota Cross-Talk. Nutrients 2022; 14:nu14071340. [PMID: 35405953 PMCID: PMC9000903 DOI: 10.3390/nu14071340] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/07/2022] [Accepted: 03/21/2022] [Indexed: 02/01/2023] Open
Abstract
Intestinal microorganisms impact health by maintaining gut homeostasis and shaping the host immunity, while gut dysbiosis associates with many conditions, including autism, a complex neurodevelopmental disorder with multifactorial aetiology. In autism, gut dysbiosis correlates with symptom severity and is characterised by a reduced bacterial variability and a diminished beneficial commensal relationship. Microbiota can influence the expression of host microRNAs that, in turn, regulate the growth of intestinal bacteria by means of bidirectional host-gut microbiota cross-talk. We investigated possible interactions among intestinal microbes and between them and host transcriptional modulators in autism. To this purpose, we analysed, by "omics" technologies, faecal microbiome, mycobiome, and small non-coding-RNAs (particularly miRNAs and piRNAs) of children with autism and neurotypical development. Patients displayed gut dysbiosis related to a reduction of healthy gut micro- and mycobiota as well as up-regulated transcriptional modulators. The targets of dysregulated non-coding-RNAs are involved in intestinal permeability, inflammation, and autism. Furthermore, microbial families, underrepresented in patients, participate in the production of human essential metabolites negatively influencing the health condition. Here, we propose a novel approach to analyse faeces as a whole, and for the first time, we detected miRNAs and piRNAs in faecal samples of patients with autism.
Collapse
|
43
|
Jiang H, Zeng W, Zhang X, Pei Y, Zhang H, Li Y. The role of gut microbiota in patients with benign and malignant brain tumors: a pilot study. Bioengineered 2022; 13:7847-7859. [PMID: 35291914 PMCID: PMC9208447 DOI: 10.1080/21655979.2022.2049959] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Gut microbiota is associated with the growth of various tumors, including malignant gliomas, through the brain-gut axis. Moreover, the gut microbiota in patients with malignant tumors may considerably differ from those with benign tumors. However, the associations of gut microbiota with benign and malignant brain tumors remain unclear. Hence, in order to explore these underlying relationships, patients with benign meningioma (n = 32), malignant glioma (n = 27), and healthy individuals (n = 41) were selected to participate in this study. The results showed that the diversity of the microbial ecosystem in brain tumor patients were less than the healthy controls, while no significant differences were observed between the meningioma and glioma groups. The microbial composition also differed significantly between individuals with brain tumors and healthy participants. In meningioma group, pathogenic bacteria like Enterobacteriaceae were increased, whereas certain carcinogenic bacteria were overrepresented in the glioma group, including Fusobacterium and Akkermansia. Furthermore, benign and malignant brain tumor patients lacked SCFA-producing probiotics. Thus, a microbial biomarker panel including Fusobacterium, Akkermansia, Escherichia/Shigella, Lachnospira, Agathobacter, and Bifidobacterium was established. Diagnostic models confirmed that this panel could distinguish between brain tumor patients and healthy patients. Additionally, gut microbiota can affect the differentiation and proliferation of brain tumors via several metabolic pathways based on annotations from the Kyoto Encyclopedia of Genes and Genomes (KEGG). This is the first study designed to investigate whether gut microbiota differs between benign and malignant brain tumor patients, and our work concluded that intestinal flora is a valuable tool for the diagnosis and treatment of brain tumors.
Collapse
Affiliation(s)
- Haixiao Jiang
- Department of Clinical Medicine,School of Medicine, Yangzhou University, Yangzhou, Jiangsu, China.,Department of Neurosurgery, Clinical Medical College of Yangzhou University, Yangzhou, Jiangsu, China
| | - Wei Zeng
- Department of Clinical Medicine,School of Medicine, Yangzhou University, Yangzhou, Jiangsu, China.,Department of Neurosurgery, Clinical Medical College of Yangzhou University, Yangzhou, Jiangsu, China
| | - Xiaoli Zhang
- Department of Clinical Medicine,School of Medicine, Yangzhou University, Yangzhou, Jiangsu, China.,Department of Medical Imaging, The Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu, China
| | - Yunlong Pei
- Department of Neurosurgery, Clinical Medical College of Yangzhou University, Yangzhou, Jiangsu, China
| | - Hengzhu Zhang
- Department of Clinical Medicine,School of Medicine, Yangzhou University, Yangzhou, Jiangsu, China.,Department of Neurosurgery, Clinical Medical College of Yangzhou University, Yangzhou, Jiangsu, China
| | - Yuping Li
- Department of Clinical Medicine,School of Medicine, Yangzhou University, Yangzhou, Jiangsu, China.,Department of Neurosurgery, Clinical Medical College of Yangzhou University, Yangzhou, Jiangsu, China
| |
Collapse
|
44
|
Gut microbiota and plasma cytokine levels in patients with attention-deficit/hyperactivity disorder. Transl Psychiatry 2022; 12:76. [PMID: 35197458 PMCID: PMC8866486 DOI: 10.1038/s41398-022-01844-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 02/02/2022] [Accepted: 02/07/2022] [Indexed: 12/12/2022] Open
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a common childhood mental disorder with undetermined pathophysiological mechanisms. The gut microbiota and immunological dysfunction may influence brain functions and social behaviours. In the current study, we aimed to explore the correlation of gut microbiome imbalance and inflammation in the pathophysiology of ADHD. Forty-one children with ADHD and thirty-nine healthy-control (HC) individuals were recruited. Faecal samples from all participants were collected and submitted for 16 S rRNA V3-V4 amplicon microbiome sequencing analysis. The plasma levels of 10 cytokines, including TNF-α, IL-6, IL-1β, IL-2, IL-10, IL-13, IL-17A, IFN-α2, IFN-γ, and MCP-1, were determined using a custom-made sandwich enzyme-linked immunosorbent assay (ELISA) developed by Luminex Flowmetrix. There was no significant difference between the ADHD and HC groups in species diversity in the faeces, as determined with α-diversity and β-diversity analysis. In the ADHD group, three differentially abundant taxonomic clades at the genus level were observed, namely Agathobacter, Anaerostipes, and Lachnospiraceae. Top differentially abundant bacteria and representative biological pathways were identified in children with ADHD using linear discriminant analysis (LDA) effect size (LEfSe), and the phylogenetic investigation of communities by reconstruction of unobserved states (PICRUSt) analysis, respectively. The plasma levels of TNF-α were significantly lower in children with ADHD than in HCs. Within the ADHD group, the levels of TNF-α were negatively correlated with ADHD symptoms and diversity of the gut microbiome. Our study provides new insights into the association between gut microbiome dysbiosis and immune dysregulation, which may contribute to the pathophysiology of ADHD.
Collapse
|
45
|
Ni JJ, Xu Q, Yan SS, Han BX, Zhang H, Wei XT, Feng GJ, Zhao M, Pei YF, Zhang L. Gut Microbiota and Psychiatric Disorders: A Two-Sample Mendelian Randomization Study. Front Microbiol 2022; 12:737197. [PMID: 35185808 PMCID: PMC8856606 DOI: 10.3389/fmicb.2021.737197] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 12/14/2021] [Indexed: 12/15/2022] Open
Abstract
Evidence supports the observational associations of gut microbiota with a variety of psychiatric disorders, but the causal nature of such associations remains obscure. Aiming to comprehensively investigate their causal relationship and to identify specific causal microbe taxa for psychiatric diseases, we conducted a two-sample Mendelian randomization (MR) analysis of gut microbiome with 15 psychiatric diseases. Specifically, the microbiome genome-wide association study (GWAS) in 18,473 individuals from the MiBioGen study was used as exposure sample, and the GWAS for 15 psychiatric diseases was used as outcome samples. One-hundred ninety bacterial taxa from six levels were available for analysis. At a multiple-testing corrected significance level (phylum P < 5.56 × 10–3, class P < 3.33 × 10–3, order P < 2.63 × 10–3, family P < 1.67 × 10–3, genus P < 4.90 × 10–4, and species P < 3.33 × 10–3), the following eight causal associations from seven bacterial features (one phylum + three classes + one order + one family + one species) were identified: family Prevotellaceae with autism spectrum disorder (P = 5.31 × 10–4), class Betaproteobacteria with bipolar disorder (P = 1.53 × 10–3), class Actinobacteria with schizophrenia (P = 1.33 × 10–3), class Bacteroidia and order Bacteroidales with Tourette syndrome (P = 2.51 × 10–3 and 2.51 × 10–3), phylum Actinobacteria and class Actinobacteria with extroversion (P = 8.22 × 10–4 and 1.09 × 10–3), and species Clostridium innocuum with neuroticism (P = 8.92 × 10–4). Sensitivity analysis showed no evidence of reverse causality, pleiotropy, and heterogeneity. Our findings offered novel insights into the gut microbiota–mediated development mechanism of psychiatric disorders.
Collapse
Affiliation(s)
- Jing-Jing Ni
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Qian Xu
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
- Department of Epidemiology and Biostatistics, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Shan-Shan Yan
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
- Department of Epidemiology and Biostatistics, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Bai-Xue Han
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
- Department of Epidemiology and Biostatistics, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Hong Zhang
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Xin-Tong Wei
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
- Department of Epidemiology and Biostatistics, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Gui-Juan Feng
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
- Department of Epidemiology and Biostatistics, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Min Zhao
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Yu-Fang Pei
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
- Department of Epidemiology and Biostatistics, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Lei Zhang
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, China
- *Correspondence: Lei Zhang,
| |
Collapse
|
46
|
Wen M, Dang X, Feng S, He Q, Li X, Liu T, He X. Integrated Analyses of Gut Microbiome and Host Metabolome in Children With Henoch-Schönlein Purpura. Front Cell Infect Microbiol 2022; 11:796410. [PMID: 35145922 PMCID: PMC8821812 DOI: 10.3389/fcimb.2021.796410] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 12/27/2021] [Indexed: 12/13/2022] Open
Abstract
Recent studies have shown that intestinal microbes and metabolites are involved in the pathogenesis of many diseases. However, whether and how they are related to Henoch–Schönlein purpura (HSP) has yet to be understood. This work is designed to detect gut microbes, intestinal and serum metabolites in children with HSP, trying to discover the etiology and pathogenesis of HSP. A total of 86 children were recruited in this study, namely, 58 children with HSP (HSP group) and 28 healthy children as control groups (CON group). 16S rDNA amplicon sequencing technology and UPLC-QTOF/MS non-targeted metabolomics analysis were used to detect the intestinal microbes and metabolites, and also multi-reaction monitoring technology for detecting serum arachidonic acid (AA) and its metabolites. Then, correlation analysis was performed to explore the possible interaction between the differential gut microbes and metabolites. As a result, at the microbiota family level, the CON group had an advantage of Coriobacteriaceae while the HSP group had a dominant Bacteroidaceae. Five kinds of bacteria in the HSP group were significantly enriched at the genus level, and seven kinds of bacteria were significantly enriched in the CON group. A total of 59 kinds of gut metabolites significantly differ between the two groups, in which most are lipids and peptides. Spearman correlation analysis showed that Bacteroides, Dialister, and Agathobacter were associated with unsaturated fatty acids, especially AA metabolism. Then, we tested the AA related metabolites in serum and found thromboxane B2, leukotriene B4, prostaglandin D2, 9S-hydroxyoctadecadienoic acid, and 13S-hydroxyoctadecadienoic acid significantly changed. In conclusion, children with HSP had dominant Bacteroidaceae and decreased Coriobacteriaceae in the family level of gut microbes, and also lipids and peptides changed most in the gut metabolites. Our data suggested that the biosynthesis and metabolism of unsaturated fatty acids, especially AA and its metabolites, might participate in the occurrence and development of HSP.
Collapse
Affiliation(s)
- Min Wen
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
- Laboratory of Pediatric Nephrology, Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiqiang Dang
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
- Laboratory of Pediatric Nephrology, Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Shipin Feng
- Department of Pediatric Nephrology, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Qingnan He
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
- Laboratory of Pediatric Nephrology, Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoyan Li
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
- Laboratory of Pediatric Nephrology, Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Taohua Liu
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiaojie He
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
- Laboratory of Pediatric Nephrology, Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Xiaojie He,
| |
Collapse
|
47
|
Tang Q, Xu E, Wang Z, Xiao M, Cao S, Hu S, Wu Q, Xiong Y, Jiang Z, Wang F, Yang G, Wang L, Yi H. Dietary Hermetia illucens Larvae Meal Improves Growth Performance and Intestinal Barrier Function of Weaned Pigs Under the Environment of Enterotoxigenic Escherichia coli K88. Front Nutr 2022; 8:812011. [PMID: 35118109 PMCID: PMC8805673 DOI: 10.3389/fnut.2021.812011] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 12/22/2021] [Indexed: 12/12/2022] Open
Abstract
The aim of this study was to evaluate the effect of Hermetia illucens larvae meal (HI) on the growth performance and intestinal barrier function of weaned pigs. To achieve this, 72 weaned pigs [28-day-old, 8.44 ± 0.04 kg body weight (BW)] were randomly assigned to three dietary treatments: basal diet (negative control, NC), zinc oxide-supplemented diet (positive control, PC), and HI-supplemented diet [100% replacement of fishmeal (FM), HI], for 28 days in the presence of enterotoxigenic Escherichia coli (ETEC). The results showed that HI and PC increased (p < 0.05) the average daily gain (ADG) and average daily feed intake (ADFI) of weaned pigs from day 1 to 14, and decreased diarrhea incidence from day 1 to 28. Additionally, HI increased (p < 0.05) claudin-1, occludin, mucin-1 (MUC-1), and MUC-2 expression, goblet cell number, and secretory immunoglobulin A (sIgA) concentration in the intestine of weaned pigs. Compared with NC, HI downregulated (p < 0.05) interleukin-1β (IL-1β) and IL-8 expression, and upregulated IL-10, transforming growth factor-β (TGF-β), antimicrobial peptide [porcine β defensin 1 (pBD1), pBD2, protegrin 1-5 (PG1-5)] expression in the jejunum or ileum. Moreover, HI decreased (p < 0.05) toll-like receptor 2 (TLR2), phosphorylated nuclear factor-κB (p-NF-κB), and phosphorylated mitogen-activated protein kinase (p-MAPK) expression, and increased sirtuin 1 (SIRT1) expression in the ileum. Additionally, HI increased histone deacetylase 3 (HDAC3) expression and acetylation of histone 3 lysine 27 (acH3k27) in the ileum. Furthermore, HI positively influenced the intestinal microbiota composition and diversity of weaned pigs and increased (p < 0.05) butyrate and valerate concentrations. Overall, dietary HI improved growth performance and intestinal barrier function, as well as regulated histone acetylation and TLR2-NF-κB/MAPK signaling pathways in weaned pigs.
Collapse
Affiliation(s)
- Qingsong Tang
- State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Institute of Animal Nutrition and Feed Science, College of Animal Science, Ministry of Education, Guizhou University, Guiyang, China
| | - E. Xu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Institute of Animal Nutrition and Feed Science, College of Animal Science, Ministry of Education, Guizhou University, Guiyang, China
| | - Zhikang Wang
- State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Mingfei Xiao
- State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Institute of Animal Nutrition and Feed Science, College of Animal Science, Ministry of Education, Guizhou University, Guiyang, China
| | - Shuting Cao
- State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Shenglan Hu
- State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Qiwen Wu
- State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Yunxia Xiong
- State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Zongyong Jiang
- State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Fengying Wang
- Guangzhou AnRuiJie Environmental Protection Technology Co., Ltd., Guangzhou, China
| | - Geling Yang
- Guangzhou AnRuiJie Environmental Protection Technology Co., Ltd., Guangzhou, China
| | - Li Wang
- State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- Li Wang
| | - Hongbo Yi
- State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- *Correspondence: Hongbo Yi
| |
Collapse
|
48
|
The interplay between Sleep and Gut Microbiota. Brain Res Bull 2022; 180:131-146. [DOI: 10.1016/j.brainresbull.2021.12.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/27/2021] [Accepted: 12/30/2021] [Indexed: 02/06/2023]
|
49
|
Pan ZY, Zhong HJ, Huang DN, Wu LH, He XX. Beneficial Effects of Repeated Washed Microbiota Transplantation in Children With Autism. Front Pediatr 2022; 10:928785. [PMID: 35783298 PMCID: PMC9249087 DOI: 10.3389/fped.2022.928785] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 05/30/2022] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE While fecal microbiota transplantation is demonstrated to improve symptoms of autism spectrum disorder (ASD), it remains unclear whether additional treatment courses yield better results. This study sought to evaluate the efficacy of repeated washed microbiota transplantation (WMT) in children with ASD. METHODS Retrospective data from children who were serially treated with WMT, including ASD symptoms, sleep disorders, gastrointestinal (GI) symptoms, and white blood cell (WBC) and globulin levels were obtained. The effect of WMT on children with ASD and whether additional WMT courses led to a further improvement in symptoms were assessed. RESULTS Aberrant Behavior Checklist (ABC), Childhood Autism Rating Scale, and Sleep Disturbance Scale for Children (SDSC) scores, the proportion of children with constipation and abnormal fecal forms, and WBC and globulin levels were all significantly lower in ASD children after WMT. More WMT treatment courses led to significantly lower scores on the ABC and SDSC. CONCLUSION WMT significantly improved ASD and GI symptoms and sleep disorders in children with ASD, and reduced systemic inflammation. Additional WMT courses led to more obvious improvements in ASD symptoms within three treatment courses.
Collapse
Affiliation(s)
- Zhao-Yu Pan
- Department of Gastroenterology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Research Center for Engineering Techniques of Microbiota-Targeted Therapies of Guangdong Province, Guangzhou, China
| | - Hao-Jie Zhong
- Department of Gastroenterology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Research Center for Engineering Techniques of Microbiota-Targeted Therapies of Guangdong Province, Guangzhou, China
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
- *Correspondence: Hao-Jie Zhong,
| | - Dong-Ni Huang
- Department of Gastroenterology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Research Center for Engineering Techniques of Microbiota-Targeted Therapies of Guangdong Province, Guangzhou, China
| | - Li-Hao Wu
- Department of Gastroenterology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Research Center for Engineering Techniques of Microbiota-Targeted Therapies of Guangdong Province, Guangzhou, China
| | - Xing-Xiang He
- Department of Gastroenterology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Research Center for Engineering Techniques of Microbiota-Targeted Therapies of Guangdong Province, Guangzhou, China
- Xing-Xiang He,
| |
Collapse
|
50
|
Zhang X, Liu L, Luo J, Peng X. Anti-aging potency correlates with metabolites from in vitro fermentation of edible fungal polysaccharides using human fecal intestinal microflora. Food Funct 2022; 13:11592-11603. [DOI: 10.1039/d2fo01951e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Aging is a natural process in which the structural integrity of an organism declines over time.
Collapse
Affiliation(s)
- Xiaomei Zhang
- Department of Food Science and Engineering, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Liu Liu
- Department of Food Science and Engineering, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Jianming Luo
- Department of Food Science and Engineering, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Xichun Peng
- Department of Food Science and Engineering, Jinan University, Guangzhou, Guangdong, 510632, China
| |
Collapse
|