1
|
Midya V, Nagdeo K, Lane JM, Torres-Olascoaga LA, Martínez GG, Horton MK, McRae N, Lopez I, Landero J, Gennings C, Téllez-Rojo MM, Wright RO, Arora M, Eggers S. Akkermansia muciniphila attenuates association between specific metal exposures during pregnancy and depressive symptoms in late childhood. iScience 2024; 27:111335. [PMID: 39640590 PMCID: PMC11617302 DOI: 10.1016/j.isci.2024.111335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 08/23/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024] Open
Abstract
Emerging research suggests that exposures to metals during pregnancy and consequent disruptions in gut microbiome (GM) are associated with depressive disorders in childhood. Akkermansia muciniphila, a GM bacteria, has been studied for its potential antidepressant effects. However, its role in influencing the association between prenatal metal exposures and depressive symptoms during childhood is unknown. Leveraging a well-characterized pediatric birth cohort and its microbiome substudy (n = 112), we investigated whether a certain subgroup of children at 9-11-year-of-age (characterized by a specific pattern of prenatal exposure to groups of metals or metal-clique) had worsened depressive symptoms and if the presence of A.muciniphila in GM modifies this association. A subgroup of children characterized by the prenatal metal-clique signature of zinc-chromium-cobalt had significantly increased depression scores; however, within that subgroup, children with A.muciniphila had much lower depression scores than those without A.muciniphila in the GM. Our analysis provides exploratory evidence hypothesizing A.muciniphila as an intervention attenuating the effect of prenatal metal-exposures-associated depressive disorders in late childhood.
Collapse
Affiliation(s)
- Vishal Midya
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kiran Nagdeo
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jamil M. Lane
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Libni A. Torres-Olascoaga
- Center for Research on Nutrition and Health, National Institute of Public Health, Cuernavaca, Mexico
| | - Gabriela Gil Martínez
- Center for Research on Nutrition and Health, National Institute of Public Health, Cuernavaca, Mexico
| | - Megan K. Horton
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nia McRae
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Inessa Lopez
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Julio Landero
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chris Gennings
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Martha Maria Téllez-Rojo
- Center for Research on Nutrition and Health, National Institute of Public Health, Cuernavaca, Mexico
| | - Robert O. Wright
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Manish Arora
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Linus Biotechnology, Inc., North Brunswick Township, NJ, USA
| | - Shoshannah Eggers
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Epidemiology, University of Iowa College of Public Health, Iowa City, IA, USA
| |
Collapse
|
2
|
Shi YC, Wu SC, Lin YC, Zheng YJ, Huang CH, Lee BH. Development of fermented Atemoya (Annona cherimola × Annona squamosa)-Amazake increased intestinal next-generation probiotics. Food Chem 2024; 459:140373. [PMID: 38986198 DOI: 10.1016/j.foodchem.2024.140373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 06/26/2024] [Accepted: 07/04/2024] [Indexed: 07/12/2024]
Abstract
Akkermansia muciniphila and Faecalibacterium prausnitzii are next-generation probiotics, which has been reported to protect disease and effectively utilize various carbohydrates (starch and pectin) as nutrients for growth. Atemoya exhibiting fruity flavor, which is suitable for enhancing aroma and attenuating unpleasant taste caused by the koji metabolites. Results indicated that malic acid was increased (from 42.4 to 70.1 mg/100 g) in fermented Atemoya-Amazake. In addition, fermented Atemoya-Amazake elevated growthes in A. muciniphila and F. prausnitzii. Similarly, the populations of Parabacteroides (5.7 fold) and Akkermansia (1.66 fold) were elevated by fermented Atemoya-Amazake treatment in an in vitro simulated gastrointestinal system compared to the control group. Results revealed that fermented Atemoya-Amazake modulated the intestinal microbiota through increasing the production of short-chain fatty acids (exhibiting anti-pathogenic activity) for 2.1, 2.5, 2.6, and 2.1 folds in acetic acid, propionic acid, isobutyric acid, and butyric acid, respectively; suggesting this fermented Atemoya-Amazake could be applied in intestinal protection.
Collapse
Affiliation(s)
- Yeu-Ching Shi
- Department of Food Sciences, National Chiayi University, Chiayi, Taiwan
| | - She-Ching Wu
- Department of Food Sciences, National Chiayi University, Chiayi, Taiwan.
| | - Yi-Ching Lin
- Department of Horticultural Science, National Chiayi University, Chiayi, Taiwan
| | - Yu-Juan Zheng
- Department of Horticultural Science, National Chiayi University, Chiayi, Taiwan
| | - Cheng-Hao Huang
- Department of Food Safety/Hygiene and Risk Management, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Bao-Hong Lee
- Department of Horticultural Science, National Chiayi University, Chiayi, Taiwan.
| |
Collapse
|
3
|
Ta LP, Corrigan S, Horniblow RD. Novel pectin-carboxymethylcellulose-based double-layered mucin/chitosan microcomposites successfully protect the next-generation probiotic Akkermansia muciniphila through simulated gastrointestinal transit and alter microbial communities within colonic ex vivo bioreactors. Int J Pharm 2024; 665:124670. [PMID: 39244071 DOI: 10.1016/j.ijpharm.2024.124670] [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: 05/16/2024] [Revised: 09/03/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
The rapid acceleration of microbiome research has identified many potential Next Generation Probiotics (NGPs). Conventional formulation processing methods are non-compatible, leading to reduced viability and unconfirmed incorporation into intestinal microbial communities; consequently, demand for more bespoke formulation strategies of such NGPs is apparent. In this study, Akkermansia muciniphila (A.muciniphila) as a candidate NGP was investigated for its growth and metabolism properties, based on which a novel microcomposite-based oral formulation was formed. Initially, a chitosan-based microcomposite was coated with mucin to establish a surface culture of A.muciniphila. This was followed by 'double encapsulation' with pectin (PEC) using a novel Entrapment Deposition by Prilling method to create core-shell double-encapsulated microcapsules. The formulation of A.muciniphila was verified to require no oxygen-restriction properties, and additionally, biopolymers were selected, including carboxymethylcellulose (CMC), that support and enhance its growth; consequently, a high viability (6 log CFU/g) of A.muciniphila microencapsulated in PEC-CMC double-encapsulates was obtained. Subsequently, the high stability of the PEC-CMC double-encapsulates was verified in simulated gastric fluid, successfully protecting and then releasing the A.muciniphila under intestinal conditions. Finally, employing a model of gastrointestinal transit and faecal-inoculated colonic bioreactors, significant alterations in microbial communities following administration and successful establishment of A.muciniphila were demonstrated.
Collapse
Affiliation(s)
- Linh Phuong Ta
- Department of Biomedical Sciences, School of Infection, Inflammation, and Immunology, College of Medicine and Health, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Sarah Corrigan
- Department of Biomedical Sciences, School of Infection, Inflammation, and Immunology, College of Medicine and Health, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Richard D Horniblow
- Department of Biomedical Sciences, School of Infection, Inflammation, and Immunology, College of Medicine and Health, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
| |
Collapse
|
4
|
Xiang Q, Yu M, Cai Q, Hu M, Rao B, Liang X, Liu Z, Xie Y, Cen K, Zhang R, Xu H, Liu Y. Multi-omics insights into the microbiota-gut-brain axis and cognitive improvement post-bariatric surgery. J Transl Med 2024; 22:945. [PMID: 39420319 PMCID: PMC11484437 DOI: 10.1186/s12967-024-05757-9] [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: 06/03/2024] [Accepted: 10/08/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Although numerous studies have shown that bariatric surgery results in sustained weight loss and modifications in gut microbiota composition and cognitive function, the exact underlying mechanisms are unclear. This study aimed to investigate the effects of bariatric surgery on cognitive function through the microbiota-gut-brain axis (MGBA). METHODS Demographic data, serum samples, fecal samples, cognitive assessment scales, and resting-state functional connectivity magnetic resonance imaging (rs-fMRI) scans were obtained from 39 obese patients before and after (6 months) laparoscopic sleeve gastrectomy (LSG). PCA analysis, OPLS-DA analysis, and permutation tests were used to conduct fecal 16 S microbiota profiling, serum metabolomics, and neuroimaging analyses, and a bariatric surgery-specific rs-fMRI brain functional connectivity network was constructed. Spearman correlation analysis and Co-inertia analysis were employed to correlate significant alterations in cognitive assessment scales and resting-state functional connectivity difference networks with differential serum metabolites and 16 S microbiota data to identify key gut microbiota and serum metabolic factors. RESULTS LSG significantly reduced the weight of obese patients, with reductions of up to 28%. Furthermore, cognitive assessment scale measurements revealed that LSG enhanced cognitive functions, including memory (HVLT, p = 0.000) and executive function (SCWT, p = 0.008). Also, LSG significantly altered gut microbiota composition (p = 0.001), with increased microbial abundance and diversity (p < 0.05). Moreover, serum metabolite levels were significantly altered, revealing intergroup differences in 229 metabolites mapped to 72 metabolic pathways (p < 0.05, VIP > 1). Spearman correlation analysis among cognitive assessment scales, gut microbiota species, and serum metabolites revealed correlations with 68 gut microbiota species and 138 serum metabolites (p < 0.05). Furthermore, pairwise correlations were detected between gut microbiota and serum metabolites (p < 0.05). Functional neuroimaging analysis revealed that LSG increased functional connectivity in cognitive-related frontotemporal networks (FPN, p < 0.01). Additionally, normalization of the default mode network (DMN) and salience network (SN) connectivity was observed after LSG (p < 0.001). Further canonical correlation and correlation analysis suggested that the cognitive-related brain network changes induced by LSG were associated with key gut microbiota species (Akkermansia, Blautia, Collinsella, Phascolarctobacterium, and Ruminococcus, p < 0.05) and neuroactive metabolites (Glycine, L-Serine, DL-Dopa, SM (d18:1/24:1(15Z), p < 0.05). CONCLUSION These findings indicate the pathophysiological role of the microbiota-gut-brain axis in enhancing cognitive function after bariatric surgery, and the study provides a basis for clinical dietary adjustments, probiotic supplementation, and guidance for bariatric surgery, but further research is still needed. TRIAL REGISTRATION Chinese Clinical Trial Registry, ChiCTR2100049403. Registered 02 August 2021, https://www.chictr.org.cn/ .
Collapse
Affiliation(s)
- Qiaoyuan Xiang
- Dapartment of Neurology, Zhongnan Hospital of Wuhan University, No.169 Donghu Street, Wuhan, Hubei, 430000, China
| | - Minhua Yu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qi Cai
- Dapartment of Neurology, Zhongnan Hospital of Wuhan University, No.169 Donghu Street, Wuhan, Hubei, 430000, China
| | - Mengjie Hu
- Department of Hepatobiliary, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Bo Rao
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xin Liang
- Dapartment of Neurology, Zhongnan Hospital of Wuhan University, No.169 Donghu Street, Wuhan, Hubei, 430000, China
| | - Zhenxing Liu
- Department of Neurology, Yiling Hospital of Yichang City, Yichang, Hubei, China
| | - Yu Xie
- Dapartment of Neurology, Zhongnan Hospital of Wuhan University, No.169 Donghu Street, Wuhan, Hubei, 430000, China
| | - Kuan Cen
- Dapartment of Neurology, Zhongnan Hospital of Wuhan University, No.169 Donghu Street, Wuhan, Hubei, 430000, China
| | - Renwei Zhang
- Dapartment of Neurology, Zhongnan Hospital of Wuhan University, No.169 Donghu Street, Wuhan, Hubei, 430000, China
| | - Haibo Xu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Yumin Liu
- Dapartment of Neurology, Zhongnan Hospital of Wuhan University, No.169 Donghu Street, Wuhan, Hubei, 430000, China.
| |
Collapse
|
5
|
Pepoyan A. Gut Akkermansia muciniphila, Prevotellaceae, and Enterobacteriaceae spp. as Possible Markers in Women-Related Nutritional and Clinical Trials: Familial Mediterranean Fever Disease. WOMEN'S HEALTH REPORTS (NEW ROCHELLE, N.Y.) 2024; 5:785-793. [PMID: 39439767 PMCID: PMC11491588 DOI: 10.1089/whr.2024.0076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 08/28/2024] [Indexed: 10/25/2024]
Abstract
Background Studies have shown that the gut microbiota of healthy men and men with familial Mediterranean fever (FMF) disease respond differently to placebo. Given the fact that the composition of the gut microbiota is different in men and women, this study aimed to describe in detail the placebo response of the gut microbiota in healthy and FMF women. Materials and Methods The bacterial response to placebo was fully evaluated on a previous PhyloChip™ DNA microarray-based assay (GEO Series; accession number GSE111835). Results The change in the total number of operational taxonomic units in healthy women exposed to placebo is more than that of healthy men, in contrast to FMF people (704 vs. 140 and 409 vs. 7560, respectively [p < 0.05]). Gut Firmicutes diversities are more sensitive to placebo, whereas Akkermansia muciniphila remained unchanged after the placebo administration for both healthy and FMF people. Gut Prevotellaceae and Enterobacteriaceae diversities of healthy subjects and FMF women are also almost unchanged from placebo. Meanwhile, only 56.35% of gut Enterobacteriaceae diversities in FMF men were placebo resistant. Conclusion The response to a placebo varies depending on a person's gender and health status. Healthy and FMF women's placebo study groups could be avoided by excluding placebo-sensitive 704 of 18,725 and 409 of 18,725 bacterial diversities, respectively. Because the placebo causes changes in all gut bacterial phyla in healthy and FMF women, and only the representatives of Enterobacteriaceae and Prevotellaceae families and A. muciniphila spp. are not affected by placebo, these bacteria can be considered as possible markers in women-related nutritional/clinical trials. Data on the response of the gut microbiota in healthy women to placebo might be used in studies of diseases other than FMF. The response of gut bacteria from different taxonomic affiliations to placebo may provide a basis for uncovering the role of these bacteria in the gut-brain axis.
Collapse
Affiliation(s)
- Astghik Pepoyan
- Food Safety and Biotechnology Department, Scientific Research Institute of Food Science and Biotechnology, Armenian National Agrarian University, Yerevan, Armenia
- International Association for Human and Animals Health Improvement, Yerevan, Armenia
| |
Collapse
|
6
|
Nanda S, Lamot B, Guarino N, Usler E, Chugani DC, Dutta A, Chow HM. Atypical gut microbiota composition in a mouse model of developmental stuttering. Sci Rep 2024; 14:23457. [PMID: 39379558 PMCID: PMC11461706 DOI: 10.1038/s41598-024-74766-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 09/30/2024] [Indexed: 10/10/2024] Open
Abstract
Developmental stuttering is a complex neurodevelopmental disorder characterized by disfluent speech. It has been associated with mutations in genes involved in lysosomal enzyme trafficking. Mice with mutations in one such gene, Gnptab, exhibit atypical vocalizations analogous to stuttering in humans. This mouse model has enabled the study of various molecular mechanisms related to the disorder. Simultaneously, an increasing number of reports have suggested the role of gut microbiota in altered brain function and development in neurological disorders. In this study, we compared gut microbiota profiles from Gnptab mutant mice to wildtype control mice. Microbiome analysis demonstrated a distinct microbiota profile in Gnptab mutant mice. The most significant alteration was an increased relative abundance of Akkermansia, a genus of mucin degrading bacteria, which has previously been associated with multiple neurological disorders. Moreover, the altered microbiota profile of these mice was predicted to result in differences in abundance of several metabolic pathways, including short chain fatty acid and lipopolysaccharide synthesis. These pathways may play a role in the onset, progression and persistence of developmental stuttering. This is the first study to show a potential link between developmental stuttering and changes in the gut microbiota, laying the groundwork for a new research direction.
Collapse
Affiliation(s)
- Sayan Nanda
- Department of Communication Sciences and Disorders, University of Delaware, Newark, DE, 19716, USA.
| | - Bryan Lamot
- Department of Animal and Food Sciences, University of Delaware, Newark, DE, 19716, USA
| | - Nicole Guarino
- Department of Communication Sciences and Disorders, University of Delaware, Newark, DE, 19716, USA
| | - Evan Usler
- Department of Communication Sciences and Disorders, University of Delaware, Newark, DE, 19716, USA
| | - Diane C Chugani
- Department of Communication Sciences and Disorders, University of Delaware, Newark, DE, 19716, USA
| | - Aditya Dutta
- Departments of Animal and Food Sciences, Biological Sciences, Medical and Molecular Sciences, Microbiology Graduate Program, University of Delaware, Newark, DE, 19716, USA.
| | - Ho Ming Chow
- Department of Communication Sciences and Disorders, University of Delaware, Newark, DE, 19716, USA.
| |
Collapse
|
7
|
Fu Y, Cheng HW. The Influence of Cecal Microbiota Transplantation on Chicken Injurious Behavior: Perspective in Human Neuropsychiatric Research. Biomolecules 2024; 14:1017. [PMID: 39199404 PMCID: PMC11352350 DOI: 10.3390/biom14081017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/26/2024] [Accepted: 08/13/2024] [Indexed: 09/01/2024] Open
Abstract
Numerous studies have evidenced that neuropsychiatric disorders (mental illness and emotional disturbances) with aggression (or violence) pose a significant challenge to public health and contribute to a substantial economic burden worldwide. Especially, social disorganization (or social inequality) associated with childhood adversity has long-lasting effects on mental health, increasing the risk of developing neuropsychiatric disorders. Intestinal bacteria, functionally as an endocrine organ and a second brain, release various immunomodulators and bioactive compounds directly or indirectly regulating a host's physiological and behavioral homeostasis. Under various social challenges, stress-induced dysbiosis increases gut permeability causes serial reactions: releasing neurotoxic compounds, leading to neuroinflammation and neuronal injury, and eventually neuropsychiatric disorders associated with aggressive, violent, or impulsive behavior in humans and various animals via a complex bidirectional communication of the microbiota-gut-brain (MGB) axis. The dysregulation of the MGB axis has also been recognized as one of the reasons for the prevalence of social stress-induced injurious behaviors (feather pecking, aggression, and cannibalistic pecking) in chickens. However, existing knowledge of preventing and treating these disorders in both humans and chickens is not well understood. In previous studies, we developed a non-mammal model in an abnormal behavioral investigation by rationalizing the effects of gut microbiota on injurious behaviors in chickens. Based on our earlier success, the perspective article outlines the possibility of reducing stress-induced injurious behaviors in chickens through modifying gut microbiota via cecal microbiota transplantation, with the potential for providing a biotherapeutic rationale for preventing injurious behaviors among individuals with mental disorders via restoring gut microbiota diversity and function.
Collapse
Affiliation(s)
- Yuechi Fu
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA;
| | - Heng-Wei Cheng
- Livestock Behavior Research Unit, USDA-ARS, West Lafayette, IN 47907, USA
| |
Collapse
|
8
|
Li K, Ding W, Li X, Gao H, Wang S, Li T, Zhao H, Zhang S. Intestinal Akkermansia muciniphila is Beneficial to Functional Recovery Following Ischemic Stroke. J Neuroimmune Pharmacol 2024; 19:43. [PMID: 39141019 DOI: 10.1007/s11481-024-10146-6] [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] [Accepted: 08/05/2024] [Indexed: 08/15/2024]
Abstract
Recent studies have demonstrated the interaction between gut microbiota and brain on ischemic stroke, but the roles of gut microbiota in the pathophysiology of ischemic stroke remain largely unclear. In this study, we detected a significant increase of intestinal Akkermansia muciniphila (AKK) following ischemic stroke by a rose bengal photothrombosis model. To investigate the function and mechanism of AKK on ischemic stroke, we performed the AKK administration prior to stroke surgery. The results showed that mice treated with AKK gained significantly higher body weight and behaved better than those in PBS group at 3 days after ischemic stroke. Consistently, AKK administration remarkably decreased the infarct volumes as well as the density of degenerating neurons and apoptotic cells after ischemic stroke. Notably, AKK is a potential therapeutic target in immune-related disorders connected to the microbiota, and inflammation is crucially involved in the pathophysiological process of ischemic stroke. For the determination of underlying mechanisms of this protective effect, we investigated whether there are associations between AKK and neuroinflammation following ischemic stroke. The results suggested that AKK administration significantly reduced the activation of astrocytes and microglia but up-regulated multiple anti-inflammatory factors following ischemic stroke. Therefore, our study highlighted the beneficial roles of intestinal AKK on ischemic stroke and provided a new perspective for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Kemin Li
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, Gansu Province, 730000, China
| | - Wancong Ding
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, Gansu Province, 730000, China
| | - Xinrui Li
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, Gansu Province, 730000, China
| | - Hao Gao
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, Gansu Province, 730000, China
| | - Shuang Wang
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, Gansu Province, 730000, China
| | - Ting Li
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, Gansu Province, 730000, China
| | - Haiyu Zhao
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, Gansu Province, 730000, China.
| | - Shengxiang Zhang
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, Gansu Province, 730000, China.
| |
Collapse
|
9
|
Hart DW, Sherman MA, Kim M, Pelzel R, Brown JL, Lesné SE. Standard diet and animal source influence hippocampal spatial reference learning and memory in congenic C57BL/6J mice. RESEARCH SQUARE 2024:rs.3.rs-4582616. [PMID: 39070656 PMCID: PMC11276007 DOI: 10.21203/rs.3.rs-4582616/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Background Assessing learning and memory has become critical to evaluate brain function in health, aging or neurological disease. The hippocampus is crucially involved in these processes as illustrated by H.M.'s remarkable case and by the well-established early symptoms of Alzheimer's disease. Numerous studies have reported the impact of gut microbiota on hippocampal structure and function using pro-, pre- and antibiotics, diet manipulations, germ-free conditions or fecal transfer. However, most diet manipulations have relied on Western diet paradigms (high fat, high energy, high carbohydrates). Here, we compared the impact of two standard diets, 5K52 and 2918 (6% fat, 18% protein, 3.1kcal/g), and how they influenced hippocampal learning and memory in adult 6-month-old congenic C57BL/6J mice from two sources. Results Using a hippocampal-dependent task, we found that 5K52-fed mice performed consistently better than 2918-fed animals in the Barnes circular maze. These behavioral differences were accompanied with marked changes in microbiota, which correlated with spatial memory retention performance. We next tested whether 2918-induced alterations in behavior and microbiome could be rescued by 5K52 diet for 3 months. Changing the 2918 diet to 5K52 diet mid-life improved spatial learning and memory in mice. Shotgun sequencing and principal component analyses revealed significant differences at both phylum and species levels. Multivariate analyses identified Akkermansia muciniphila or Bacteroidales bacterium M11 and Faecalibaculum rodentium as the strongest correlates to spatial memory retention in mice depending on the animal source. In both settings, the observed behavioral differences only affected hippocampal-dependent performance as mice fed with either diet did similarly well on the non-spatial variant of the Y-maze. Conclusions In summary, these findings demonstrate the diverging effects of seemingly equivalent standard diets on hippocampal memory. Based on these results, we strongly recommend the mandatory inclusion of the diet and source of animals used in rodent behavioral studies.
Collapse
|
10
|
Sung C, Park CG, Maienschein-Cline M, Chlipala G, Green S, Doorenbos A, Fink A, Bronas U, Lockwood M. Associations Between Gut Microbial Features and Sickness Symptoms in Kidney Transplant Recipients. Biol Res Nurs 2024; 26:368-379. [PMID: 38231673 DOI: 10.1177/10998004241227560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
PURPOSE The study investigated the relationship of gut microbiome features and sickness symptoms in kidney transplant recipients. METHODS Employing a prospective, longitudinal design, we collected data from 19 participants who had undergone living-donor kidney transplant at three timepoints (pre-transplant and 1 week and 3 months post-transplant). Sickness symptom data and fecal specimens were collected at each timepoint. Participants were grouped either as high or low sickness symptom severity at baseline. Shotgun metagenomics sequencing characterized gut microbial structure and functional gene content. Fecal microbial features, including alpha (evenness and richness within samples) and beta (dissimilarities between samples) diversity and relative abundances, were analyzed using R statistical packages. Cross-sectional and longitudinal analyses examined relationships between gut microbial features and sickness symptoms. RESULTS Although our exploratory findings revealed no significant differences in alpha and beta diversity between groups, the high-severity group showed lower microbial richness and evenness than the low-severity group. The high-severity group had enriched relative abundance of bacteria from the genera Citrobacter and Enterobacter and reduced relative abundance of bacteria from the genus Akkermansia across timepoints. No functional genes differed significantly between groups or timepoints. CONCLUSIONS Kidney transplant recipients with high symptom burden displayed increased putative proinflammatory bacteria and decreased beneficial bacteria. This study provides an effect size that future large cohort studies can employ to confirm associations between gut microbial features and sickness symptom experiences in the kidney transplant population. The study findings also have implications for future interventional studies aiming to alleviate the sickness symptom burden in this population.
Collapse
Affiliation(s)
- Choa Sung
- Department of Biobehavioral Nursing Science, College of Nursing, University of Illinois Chicago, Chicago, IL, USA
| | - Chang Gi Park
- Department of Population Health Nursing Science, College of Nursing, University of Illinois Chicago, Chicago, IL, USA
| | | | - George Chlipala
- Associate Director of Research Informatics Core, University of Illinois at Chicago, Chicago, IL, USA
| | - Stefan Green
- Department of Internal Medicine, Division of Infectious Disease, Rush University Medical Center, Chicago, IL, USA
| | - Ardith Doorenbos
- Department of Biobehavioral Nursing Science, College of Nursing, University of Illinois Chicago, Chicago, IL, USA
| | - Anne Fink
- Biobehavioral Science in Nursing and Rehabilitation & Regenerative Medicine, Columbia University, New York, NY, USA
| | - Ulf Bronas
- Department of Biobehavioral Nursing Science, College of Nursing, University of Illinois Chicago, Chicago, IL, USA
| | - Mark Lockwood
- Department of Biobehavioral Nursing Science, College of Nursing, University of Illinois Chicago, Chicago, IL, USA
| |
Collapse
|
11
|
Xu D, Ren L, Zhang W, Wu S, Yu M, He X, Wei Z. Therapeutic effects and mechanisms of fecal microbiota transplantation on EAE partly through HPA axis-mediated neuroendocrine regulation. Heliyon 2024; 10:e33214. [PMID: 39021924 PMCID: PMC11252752 DOI: 10.1016/j.heliyon.2024.e33214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/16/2024] [Accepted: 06/17/2024] [Indexed: 07/20/2024] Open
Abstract
Background The pathogenesis of multiple sclerosis (MS) may be closely related to immune regulation and inflammatory cytokines induced by specific flora. Repairing the intestinal flora may alter the immune response in MS patients, thus opening up novel approaches for the treatment of MS. Objective We aimed to test the therapeutic effect of fecal microbiota transplantation (FMT) on experimental autoimmune encephalomyelitis (EAE) and the characteristics of intestinal microbiota composition changes, explore the potential mechanisms of FMT treatment. Methods EAE animals were treated with FMT, with the therapeutic effects were evaluated by observing neurological scores and measuring serum levels of cortisol, IL-17, and TLR-2. Fecal microbiome 16S rRNA sequencing was used to profile changes in microbiota composition, and adrenalectomy pretreatment was used to test whether FMT effects were dependent on HPA axis function. Results FMT improved neurological function and reduced serum IL-17 to levels that were close to the control group. FMT reestablished intestinal homeostasis by altering the structure of the intestinal flora, increasing the abundance of beneficial flora, and regulating intestinal metabolites. We found that the therapeutic effects of FMT depended partly on the efferent function of the HPA axis; surgical disruption of the HPA axis altered the abundance and diversity of the intestinal flora. Conclusion FMT showed a neuroprotective effect on EAE by increasing the abundance of the beneficial flora, rebuilding intestinal homeostasis, reducing IL-17 and cortisol serum levels, and promoting serum TLR-2; the therapeutic effect of FMT on EAE is partly dependent on the HPA axis.
Collapse
Affiliation(s)
- Danhong Xu
- Department of Critical Care Medicine, First School of Clinical Medicine, The First Affiliated Hospital of Guangdong Pharmaceutical University, 19 Nonglinxia Road, Yuexiu District, Guangzhou, 510080, China
| | - Linxiang Ren
- Department of Neurology, Neurological Research Institute of Integrated Traditional Chinese and Western Medicine, First School of Clinical Medicine, The First Affiliated Hospital of Guangdong Pharmaceutical University, 19 Nonglinxia Road, Yuexiu District, Guangzhou, 510080, China
| | - Wenbin Zhang
- Department of Neurology, Shenzhen Guangming District People's Hospital, Shenzhen, Guangdong, 518106, China
| | - Shaohua Wu
- Department of Neurology, Neurological Research Institute of Integrated Traditional Chinese and Western Medicine, First School of Clinical Medicine, The First Affiliated Hospital of Guangdong Pharmaceutical University, 19 Nonglinxia Road, Yuexiu District, Guangzhou, 510080, China
| | - Minling Yu
- Department of Neurology, Neurological Research Institute of Integrated Traditional Chinese and Western Medicine, First School of Clinical Medicine, The First Affiliated Hospital of Guangdong Pharmaceutical University, 19 Nonglinxia Road, Yuexiu District, Guangzhou, 510080, China
| | - Xingxiang He
- Department of Gastroenterology, First School of Clinical Medicine, The First Affiliated Hospital of Guangdong Pharmaceutical University, 19 Nonglinxia Road, Yuexiu District, Guangzhou, 510080, China
| | - Zhisheng Wei
- Department of Neurology, Neurological Research Institute of Integrated Traditional Chinese and Western Medicine, First School of Clinical Medicine, The First Affiliated Hospital of Guangdong Pharmaceutical University, 19 Nonglinxia Road, Yuexiu District, Guangzhou, 510080, China
| |
Collapse
|
12
|
Mruk-Mazurkiewicz H, Kulaszyńska M, Czarnecka W, Podkówka A, Ekstedt N, Zawodny P, Wierzbicka-Woś A, Marlicz W, Skupin B, Stachowska E, Łoniewski I, Skonieczna-Żydecka K. Insights into the Mechanisms of Action of Akkermansia muciniphila in the Treatment of Non-Communicable Diseases. Nutrients 2024; 16:1695. [PMID: 38892628 PMCID: PMC11174979 DOI: 10.3390/nu16111695] [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: 05/08/2024] [Revised: 05/27/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
This comprehensive review delineates the extensive roles of Akkermansia muciniphila in various health domains, spanning from metabolic and inflammatory diseases to neurodegenerative disorders. A. muciniphila, known for its ability to reside in the mucous layer of the intestine, plays a pivotal role in maintaining gut integrity and interacting with host metabolic processes. Its influence extends to modulating immune responses and potentially easing symptoms across several non-communicable diseases, including obesity, diabetes, inflammatory bowel disease, and cancer. Recent studies highlight its capacity to interact with the gut-brain axis, suggesting a possible impact on neuropsychiatric conditions. Despite the promising therapeutic potential of A. muciniphila highlighted in animal and preliminary human studies, challenges remain in its practical application due to stability and cultivation issues. However, the development of pasteurized forms and synthetic mediums offers new avenues for its use in clinical settings, as recognized by regulatory bodies like the European Food Safety Authority. This narrative review serves as a crucial resource for understanding the broad implications of A. muciniphila across different health conditions and its potential integration into therapeutic strategies.
Collapse
Affiliation(s)
- Honorata Mruk-Mazurkiewicz
- Department of Biochemical Science, Pomeranian Medical University in Szczecin, Broniewskiego 24, 71-460 Szczecin, Poland (N.E.); (I.Ł.)
| | - Monika Kulaszyńska
- Department of Biochemical Science, Pomeranian Medical University in Szczecin, Broniewskiego 24, 71-460 Szczecin, Poland (N.E.); (I.Ł.)
| | - Wiktoria Czarnecka
- Department of Biochemical Science, Pomeranian Medical University in Szczecin, Broniewskiego 24, 71-460 Szczecin, Poland (N.E.); (I.Ł.)
| | - Albert Podkówka
- Department of Biochemical Science, Pomeranian Medical University in Szczecin, Broniewskiego 24, 71-460 Szczecin, Poland (N.E.); (I.Ł.)
| | - Natalia Ekstedt
- Department of Biochemical Science, Pomeranian Medical University in Szczecin, Broniewskiego 24, 71-460 Szczecin, Poland (N.E.); (I.Ł.)
| | - Piotr Zawodny
- Medical Center Zawodny Clinic, Ku Słońcu 58, 71-047 Szczecin, Poland;
| | | | - Wojciech Marlicz
- Department of Gastroenterology, Pomeranian Medical University in Szczecin, Unii Lubelskiej, 71-252 Szczecin, Poland
| | - Błażej Skupin
- Department of Biochemical Science, Pomeranian Medical University in Szczecin, Broniewskiego 24, 71-460 Szczecin, Poland (N.E.); (I.Ł.)
| | - Ewa Stachowska
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, Broniewskiego 24, 71-460 Szczecin, Poland
| | - Igor Łoniewski
- Department of Biochemical Science, Pomeranian Medical University in Szczecin, Broniewskiego 24, 71-460 Szczecin, Poland (N.E.); (I.Ł.)
| | - Karolina Skonieczna-Żydecka
- Department of Biochemical Science, Pomeranian Medical University in Szczecin, Broniewskiego 24, 71-460 Szczecin, Poland (N.E.); (I.Ł.)
| |
Collapse
|
13
|
Liu S, Wang Y, Zhang Y, Zeng L, Ling L, Luo Y, Liu W. The gut microbiota and post-traumatic major depression disorder: insights from bidirectional two-sample Mendelian randomization. Front Psychiatry 2024; 15:1383664. [PMID: 38807688 PMCID: PMC11130430 DOI: 10.3389/fpsyt.2024.1383664] [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: 02/07/2024] [Accepted: 04/29/2024] [Indexed: 05/30/2024] Open
Abstract
Background Exposure to trauma is often associated with an increased incidence of Major Depressive Disorder (MDD), yet the mechanisms underlying MDD development post-trauma remain elusive. The microbiota-gut-brain axis has been implicated in neuropsychiatric disorders, suggesting its potential role in post-traumatic MDD (PTMDD) development. Our study aimed to assess the significance of the gut microbiome-brain interaction in PTMDD. Methods We conducted a bidirectional two-sample Mendelian Randomization (MR) analysis to investigate the causal relationship between the gut microbiota and both PTMDD and trauma exposure in MDD. Genome-wide association study (GWAS) summary datasets for PTMDD and trauma exposure in MDD, both derived from the UK Biobank. The PTMDD dataset included 24,090 individuals (13,393 cases and 10,701 controls), while the dataset for trauma exposure in MDD comprised 22,880 participants (13,393 cases and 9,487 controls). Additionally, gut microbiota data from the MiBioGen consortium included 14,306 European individuals across 18 diverse cohorts. Results Our research identified a significant negative association between the phylum Verrucomicrobia (odds ratio (OR) [95% confidence interval (CI)] =0.799 [0.684-0.933], P=0.005) and the risk of developing PTMDD, suggesting a protective role for Verrucomicrobia against PTMDD. Conversely, our findings indicate no causal effects of the gut microbiota on trauma exposure in MDD. However, reverse analysis revealed that both PTMDD and MDD influence certain bacterial traits, affecting 5 and 9 bacterial traits, respectively. Moreover, Verrucomicrobia (OR [95% CI] = 1.166 [1.051 - 1.294], P=0.004) was found to be positively impacted by trauma exposure in MDD. Conclusion Our findings provide a cause-and-effect relationship between the gut microbiota and PTMDD, contributing to our understanding of the microbiota-gut-brain axis and its role in neuropsychiatric disorder development after trauma. This information provides an opportunity for new treatment and prevention methods which are aimed at the gut-brain interaction.
Collapse
Affiliation(s)
- Shan Liu
- The Second Affiliated Hospital, Department of Emergency, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yu Wang
- The Second Affiliated Hospital, Department of Emergency, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yonghu Zhang
- The Second Affiliated Hospital, Department of Emergency, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Liang Zeng
- The Second Affiliated Hospital, Department of Emergency, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Lin Ling
- The Second Affiliated Hospital, Department of Emergency, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yong Luo
- The Second Affiliated Hospital, Department of Emergency, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Wenjie Liu
- The Second Affiliated Hospital, Department of Anesthesiology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
14
|
Qu L, Li Y, Liu F, Fang Y, He J, Ma J, Xu T, Wang L, Lei P, Dong H, Jin L, Yang Q, Wu W, Sun D. Microbiota-Gut-Brain Axis Dysregulation in Alzheimer's Disease: Multi-Pathway Effects and Therapeutic Potential. Aging Dis 2024; 15:1108-1131. [PMID: 37728579 PMCID: PMC11081173 DOI: 10.14336/ad.2023.0823-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 08/23/2023] [Indexed: 09/21/2023] Open
Abstract
An essential regulator of neurodegenerative conditions like Alzheimer's disease (AD) is the gut microbiota. Alterations in intestinal permeability brought on by gut microbiota dysregulation encourage neuroinflammation, central immune dysregulation, and peripheral immunological dysregulation in AD, as well as hasten aberrant protein aggregation and neuronal death in the brain. However, it is unclear how the gut microbiota transmits information to the brain and how it influences brain cognition and function. In this review, we summarized the multiple pathways involved in the gut microbiome in AD and provided detailed treatment strategies based on the gut microbiome. Based on these observations, this review also discusses the problems, challenges, and strategies to address current therapeutic strategies.
Collapse
Affiliation(s)
- Linkai Qu
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325000, China.
- College of Veterinary Medicine, Jilin University, Changchun 130118, China.
| | - Yanwei Li
- Core Facilities, Zhejiang University School of Medicine, Hangzhou 310058, China.
| | - Fan Liu
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325000, China.
| | - Yimeng Fang
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325000, China.
| | - Jiaxuan He
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325000, China.
| | - Jiahui Ma
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325000, China.
| | - Ting Xu
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325000, China.
| | - Lei Wang
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325000, China.
| | - Pengyu Lei
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325000, China.
| | - Hao Dong
- College of Life Sciences, Jilin Agricultural University, Changchun 130118, China.
| | - Libo Jin
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325000, China.
| | - Qinsi Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China.
| | - Wei Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Da Sun
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325000, China.
| |
Collapse
|
15
|
Zhou C, Guo S, Gong P, Ba Q, Yao W. Nano-Selenium Alleviates Cd-Induced Chronic Colitis through Intestinal Flora. Nutrients 2024; 16:1330. [PMID: 38732577 PMCID: PMC11085897 DOI: 10.3390/nu16091330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/21/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024] Open
Abstract
BACKGROUND Cadmium (Cd) is an environmental contaminant that poses risks to human and animal health. Selenium (Se), a beneficial element, alleviates the detrimental consequences of colitis and Cd toxicity. Se is found in food products as both inorganic Se (sodium selenite) and organic Se (typically Se-enriched yeast). Nano-selenium (nano-Se; a novel form of Se produced through the bioreduction of Se species) has recently garnered considerable interest, although its effects against Cd-induced enterotoxicity are poorly understood. The aim of this study was to investigate the impact of nano-selenium on mitigating cadmium toxicity and safeguarding the integrity of the intestinal barrier. METHODS For a total of two cycles, we subjected 6-week-old C57 mice to chronic colitis by exposing them to Cd and nano-selenium for two weeks, followed by DSS water for one week. RESULTS The application of nano-selenium mitigated the intensity of colitis and alleviated inflammation in the colon. Nano-selenium enhanced the diversity of the intestinal flora, elevated the concentration of short-chain fatty acids (SCFAs) in feces, and improved the integrity of the intestinal barrier. CONCLUSIONS In summary, nano-Se may reduce intestinal inflammation by regulating the growth of intestinal microorganisms and protecting the intestinal barrier.
Collapse
Affiliation(s)
- Chengdong Zhou
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi’an 710021, China; (C.Z.); (S.G.); (P.G.)
| | - Shengliang Guo
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi’an 710021, China; (C.Z.); (S.G.); (P.G.)
| | - Pin Gong
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi’an 710021, China; (C.Z.); (S.G.); (P.G.)
| | - Qian Ba
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Zhijiang Middle Road, Shanghai 200071, China
| | - Wenbo Yao
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi’an 710021, China; (C.Z.); (S.G.); (P.G.)
- Department of Pediatrics, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Zhijiang Middle Road, Shanghai 200071, China
| |
Collapse
|
16
|
Zhang N, Gao X, Li D, Xu L, Zhou G, Xu M, Peng L, Sun G, Pan F, Li Y, Ren R, Huang R, Yang Y, Wang Z. Sleep deprivation-induced anxiety-like behaviors are associated with alterations in the gut microbiota and metabolites. Microbiol Spectr 2024; 12:e0143723. [PMID: 38421192 PMCID: PMC10986621 DOI: 10.1128/spectrum.01437-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 02/03/2024] [Indexed: 03/02/2024] Open
Abstract
The present study aimed to characterize the gut microbiota and serum metabolome changes associated with sleep deprivation (SD) as well as to explore the potential benefits of multi-probiotic supplementation in alleviating SD-related mental health disorders. Rats were subjected to 7 days of SD, followed by 14 days of multi-probiotics or saline administration. Open-field tests were conducted at baseline, end of SD (day 7), and after 14 days of saline or multi-probiotic gavage (day 21). Metagenomic sequencing was conducted on fecal samples, and serum metabolites were measured by untargeted liquid chromatography tandem-mass spectrometry. At day 7, anxiety-like behaviors, including significant decreases in total movement distance (P = 0.0002) and staying time in the central zone (P = 0.021), were observed. In addition, increased levels of lipopolysaccharide (LPS; P = 0.028) and decreased levels of uridine (P = 0.018) and tryptophan (P = 0.01) were detected in rats after 7 days of SD. After SD, the richness of the gut bacterial community increased, and the levels of Akkermansia muciniphila, Muribaculum intestinale, and Bacteroides caecimuris decreased. The changes in the host metabolism and gut microbiota composition were strongly associated with the anxiety-like behaviors caused by SD. In addition, multi-probiotic supplementation for 14 days modestly improved the anxiety-like behaviors in SD rats but significantly reduced the serum level of LPS (P = 0.045). In conclusion, SD induces changes in the gut microbiota and serum metabolites, which may contribute to the development of chronic inflammatory responses and affect the gut-brain axis, causing anxiety-like behaviors. Probiotic supplementation significantly reduces serum LPS, which may alleviate the influence of chronic inflammation. IMPORTANCE The disturbance in the gut microbiome and serum metabolome induced by SD may be involved in anxiety-like behaviors. Probiotic supplementation decreases serum levels of LPS, but this reduction may be insufficient for alleviating SD-induced anxiety-like behaviors.
Collapse
Affiliation(s)
- Nana Zhang
- Medical School of Chinese PLA, Beijing, China
- Department of Gastroenterology and Hepatology, The First Centre of Chinese PLA General Hospital, Beijing, China
| | - Xuefeng Gao
- Shenzhen Key Laboratory of Gastrointestinal Microbiota and Disease, Integrative Microecology Clinical Center, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
- Shenzhen Clinical Research Center for Digestive Disease, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
- The Clinical Innovation & Research Center, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Donghao Li
- Department of Gastroenterology and Hepatology, The First Centre of Chinese PLA General Hospital, Beijing, China
| | - Lijuan Xu
- Department of Gastroenterology and Hepatology, The First Centre of Chinese PLA General Hospital, Beijing, China
| | - Guanzhou Zhou
- Department of Gastroenterology and Hepatology, The First Centre of Chinese PLA General Hospital, Beijing, China
| | - Mengqi Xu
- Medical School of Chinese PLA, Beijing, China
- Department of Gastroenterology and Hepatology, The First Centre of Chinese PLA General Hospital, Beijing, China
| | - Lihua Peng
- Medical School of Chinese PLA, Beijing, China
- Department of Gastroenterology and Hepatology, The First Centre of Chinese PLA General Hospital, Beijing, China
| | - Gang Sun
- Medical School of Chinese PLA, Beijing, China
- Department of Gastroenterology and Hepatology, The First Centre of Chinese PLA General Hospital, Beijing, China
| | - Fei Pan
- Medical School of Chinese PLA, Beijing, China
- Department of Gastroenterology and Hepatology, The First Centre of Chinese PLA General Hospital, Beijing, China
| | - Yan Li
- Medical School of Chinese PLA, Beijing, China
- Department of Gastroenterology and Hepatology, The First Centre of Chinese PLA General Hospital, Beijing, China
| | - Rongrong Ren
- Medical School of Chinese PLA, Beijing, China
- Department of Gastroenterology and Hepatology, The First Centre of Chinese PLA General Hospital, Beijing, China
| | - Ruolan Huang
- Department of Neurology, Shenzhen University Clinical Research Center for Neurological Diseases, Shenzhen University General Hospital, Shenzhen, China
| | - Yunsheng Yang
- Medical School of Chinese PLA, Beijing, China
- Department of Gastroenterology and Hepatology, The First Centre of Chinese PLA General Hospital, Beijing, China
| | - Zikai Wang
- Medical School of Chinese PLA, Beijing, China
- Department of Gastroenterology and Hepatology, The First Centre of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
17
|
Warren M, O'Connor C, Lee JE, Burton J, Walton D, Keathley J, Wammes M, Osuch E. Predispose, precipitate, perpetuate, and protect: how diet and the gut influence mental health in emerging adulthood. Front Nutr 2024; 11:1339269. [PMID: 38505265 PMCID: PMC10948435 DOI: 10.3389/fnut.2024.1339269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 02/02/2024] [Indexed: 03/21/2024] Open
Abstract
Medicine often employs the 4Ps of predisposing, precipitating, perpetuating, and protective factors to identify salient influences on illness states, and to help guide patient care. Mental illness is a significant cause of morbidity and mortality worldwide. Mental health is a complex combination of biological, psychological, environmental, and social factors. There is growing interest in the gut-brain-microbiome (GBM) axis and its impact on mental health. We use the medical model of the 4Ps to explore factors involving the connection between nutrition and the GBM axis and their associated risks with mental health problems in emerging adults (EAs), a life stage when mental illness onset is the most common. We review the impact of current dietary trends on the GBM and on mental health, and the role that gut microbiome-based interventions can have in modulating the GBM axis of EAs. We discuss the implications of gut health on the GBM and areas for clinical intervention.
Collapse
Affiliation(s)
- Michael Warren
- Department of Psychiatry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Colleen O'Connor
- School of Food and Nutritional Sciences, Brescia University College, London, ON, Canada
| | - Ju Eun Lee
- Geriatrics, Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Jeremy Burton
- Department of Surgery, Microbiology and Immunology, Lawson Health Research Institute, Western University, London, ON, Canada
| | - David Walton
- School of Physical Therapy, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Justine Keathley
- Department of Human Health and Nutritional Sciences, College of Biological Science, University of Guelph, Guelph, ON, Canada
| | - Michael Wammes
- Department of Psychiatry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Elizabeth Osuch
- Department of Psychiatry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
- London Health Sciences Centre, Lawson Health Research Institute, London, ON, Canada
- First Episode Mood and Anxiety Program, London Health Sciences Centre, London, ON, Canada
| |
Collapse
|
18
|
Xu W, Zhang S, Yang Y, Zhan J, Zang C, Yu H, Wu C. Therapeutic potential of dietary nutrients and medicinal foods against metabolic disorders: Targeting Akkermansia muciniphila. FOOD FRONTIERS 2024; 5:329-349. [DOI: 10.1002/fft2.341] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
Abstract
AbstractAs one of the most attractive next‐generation probiotics, mucin‐degrading Akkermansia muciniphila has emerged as an essential and integral factor in maintaining human health and affecting pathological outcomes. Its abundance is inversely associated with various metabolic diseases (e.g., obesity and type 2 diabetes), cardiovascular diseases, and intestinal inflammation. Supplementing A. muciniphila to restore the gut microbiota ecosystem is a promising approach for treating metabolic disorders. However, the direct utilization of this probiotic is limited by technological and regulatory hurdles, such as the in vitro bulk culture of A. muciniphila and the need for expensive animal‐derived materials. Therefore, enrichment of A. muciniphila using nutraceutical supplements is a feasible strategy. Dietary supplements, especially medicinal herbs, offer a vast and valuable resource as potential prebiotics for promoting the growth of A. muciniphila in the gut, ensuring reliable safety and efficacy. In this study, we first systemically reviewed the dietary substances and medicinal foods known to promote A. muciniphila from over 100 literature sources, aiming to establish a candidate basis for future exploration of prebiotics targeting A. muciniphila. Furthermore, we summarized and discussed the major regulatory factors and mechanisms responsible for the beneficial effect of A. muciniphila on metabolic disorders, hoping to open up exciting directions for in‐depth research on the pharmacological mechanism of A. muciniphila and pave the way for its clinical therapeutics.
Collapse
Affiliation(s)
- Wenyi Xu
- School of Chinese Materia Medica Tianjin University of Traditional Chinese Medicine Tianjin China
- Beijing QuantiHealth Technology Co., Ltd. Beijing China
| | - Shaozhuo Zhang
- School of Chinese Materia Medica Tianjin University of Traditional Chinese Medicine Tianjin China
| | - Yanan Yang
- School of Chinese Materia Medica Tianjin University of Traditional Chinese Medicine Tianjin China
| | - Jiaguo Zhan
- School of Chinese Materia Medica Tianjin University of Traditional Chinese Medicine Tianjin China
| | - Chenchen Zang
- Institute of Medicinal Plant Development Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
| | - Huifang Yu
- Institute of Medicinal Plant Development Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
| | - Chongming Wu
- School of Chinese Materia Medica Tianjin University of Traditional Chinese Medicine Tianjin China
- Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine Tianjin China
| |
Collapse
|
19
|
Gao X, Lin C, Feng Y, You Y, Jin Z, Li M, Zhou Y, Chen K. Akkermansia muciniphila-derived small extracellular vesicles attenuate intestinal ischemia-reperfusion-induced postoperative cognitive dysfunction by suppressing microglia activation via the TLR2/4 signaling. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119630. [PMID: 37967793 DOI: 10.1016/j.bbamcr.2023.119630] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 11/07/2023] [Accepted: 11/07/2023] [Indexed: 11/17/2023]
Abstract
Akkermansia muciniphila (AKK) bacteria improve the functions of theere intestinal and blood-brain barriers (BBB) via their extracellular vesicles (AmEvs). However, their role in postoperative cognitive dysfunction (POCD) and its underlying mechanisms remain unclear. To investigate, we used C57BL/6 J mice divided into five groups: Sham, POCD, POCD+Akk, POCD+Evs, and POCD+Evs + PLX5622. POCD was induced through intestinal ischemia-reperfusion (I/R). The mice's cognitive function was assessed using behavioral tests, and possible mechanisms were explored by examining gut and BBB permeability, inflammation, and microglial function. Toll-like receptor (TLR) 2/4 pathway-related proteins were also investigated both in vitro and in vivo. PLX5622 chow was employed to eliminate microglial cells. Our findings revealed a negative correlation between AKK abundance and POCD symptoms. Supplementation with either AKK or AmEvs improved cognitive function, improved the performance of the intestinal barrier and BBB, and decreased inflammation and microglial activation in POCD mice compared to controls. Moreover, AmEvs treatment inhibited TLR2/4 signaling in the brains of POCD mice and LPS-treated microglial cells. In microglial-ablated POCD mice, however, AmEvs failed to protect BBB integrity. Overall, AmEvs is a potential therapeutic strategy for managing POCD by enhancing gut and BBB integrity and inhibiting microglial-mediated TLR2/4 signaling.
Collapse
Affiliation(s)
- Xiang Gao
- Department of Anesthesiology, Fujian Maternity and Child Health Hospital, Fuzhou, 350000, China; College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350000, China
| | - Chuantao Lin
- Department of Anesthesiology, Fujian Maternity and Child Health Hospital, Fuzhou, 350000, China; College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350000, China
| | - Yebin Feng
- Department of Science and Education, Fujian Maternity and Child Health Hospital, Fuzhou, 350000, China; College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350000, China
| | - Yi You
- Department of Anesthesiology, Fujian Maternity and Child Health Hospital, Fuzhou, 350000, China; College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350000, China
| | - Zhe Jin
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Mengyun Li
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yufeng Zhou
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Kai Chen
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
20
|
Konstanti P, Ligthart K, Fryganas C, Constantinos P, Smidt H, de Vos WM, Belzer C. Physiology of γ-aminobutyric acid production by Akkermansia muciniphila. Appl Environ Microbiol 2024; 90:e0112123. [PMID: 38088552 PMCID: PMC10807452 DOI: 10.1128/aem.01121-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 11/06/2023] [Indexed: 01/25/2024] Open
Abstract
Gut bacteria hold the potential to produce a broad range of metabolites that can modulate human functions, including molecules with neuroactive potential. One such molecule is γ-aminobutyric acid (GABA), the main inhibitory neurotransmitter of the central nervous system in animals. Metagenomic analyses suggest that the genomes of many gut bacteria encode glutamate decarboxylase (GAD), the enzyme that catalyzes GABA production. The genome of Akkermansia muciniphila, a mucin specialist and potential next-generation probiotic from the human gut, is predicted to encode GAD, suggesting a contributing role in GABA production in the human gut. In this study, A. muciniphila was grown in batch cultures with and without pH control. In both experiments, A. muciniphila was found to produce GABA as a response to acid (pH <5.5), although only when GABA precursors, either glutamate or glutamine, were present in the medium. Proteomic analysis comparing A. muciniphila grown with and without precursors at pH 4 did not show a difference in GAD expression, suggesting that it is expressed regardless of the presence of GABA precursors. To further investigate the function of A. muciniphila GAD, we heterologously expressed the gad gene (encoded by locus tag Amuc_0372) with a His tag in Escherichia coli and purified the GAD protein. Enzyme assays showed GAD activity in a pH range between 4 and 6, with the highest specific activity at pH 5 of 144 ± 16 µM GABA/min/mg. Overall, our results demonstrate the ability of A. muciniphila to produce GABA as an acid response and unravel the conditions under which GABA production in A. muciniphila occurs.IMPORTANCEAkkermansia muciniphila is considered to be a beneficial bacterium from the human gut, but the exact mechanisms by which A. muciniphila influences its host are not yet fully understood. To this end, it is important to identify which metabolites are produced and consumed by A. muciniphila that may contribute to a healthy gut. In the present study, we demonstrate the ability of A. muciniphila to produce γ-aminobutyric acid (GABA) when grown in an acidic environment, which often occurs in the gut. GABA is the major inhibitory neurotransmitter in the central nervous system and is present in the human gut. For this reason, it is considered an important bacterial metabolite. Our finding that A. muciniphila produces GABA in acidic environments adds to the growing body of understanding of its relationship with host health and provides an explanation on how it can survive acid stress in the human gut.
Collapse
Affiliation(s)
- Prokopis Konstanti
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, the Netherlands
| | - Kate Ligthart
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, the Netherlands
| | - Christos Fryganas
- Food Quality and Design, Wageningen University & Research, Wageningen, the Netherlands
| | - Patinios Constantinos
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, the Netherlands
| | - Hauke Smidt
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, the Netherlands
| | - Willem M. de Vos
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, the Netherlands
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Clara Belzer
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, the Netherlands
| |
Collapse
|
21
|
Wu H, Qi S, Yang R, Pan Q, Lu Y, Yao C, He N, Huang S, Ling X. Strategies for high cell density cultivation of Akkermansia muciniphila and its potential metabolism. Microbiol Spectr 2024; 12:e0238623. [PMID: 38059626 PMCID: PMC10782997 DOI: 10.1128/spectrum.02386-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 11/08/2023] [Indexed: 12/08/2023] Open
Abstract
IMPORTANCE Currently, there is significant interest in Akkermansia muciniphila as a promising next-generation probiotic, making it a hot topic in scientific research. However, to achieve efficient industrial production, there is an urgent need to develop an in vitro culture method to achieve high biomass using low-cost carbon sources such as glucose. This study aims to explore the high-density fermentation strategy of A. muciniphila by optimizing the culture process. This study also employs techniques such as LC-MS and RNA-Seq to explain the possible regulatory mechanism of high-density cell growth and increased cell surface hydrophobicity facilitating cell colonization of the gut in vitro culture. Overall, this research sheds light on the potential of A. muciniphila as a probiotic and provides valuable insights for future industrial production.
Collapse
Affiliation(s)
- Haiting Wu
- Department of Chemical and Biochemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, People's Republic of China
| | - Shuhua Qi
- Department of Chemical and Biochemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, People's Republic of China
| | - Ruixiong Yang
- Department of Chemical and Biochemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, People's Republic of China
| | - Qihua Pan
- Department of Chemical and Biochemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, People's Republic of China
| | - Yinghua Lu
- Department of Chemical and Biochemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, People's Republic of China
- Xiamen Key Laboratory of Synthetic Biotechnology, Xiamen University, Xiamen, People's Republic of China
- The Key Laboratory for Chemical Biology of Fujian Province, Xiamen University, Xiamen, People's Republic of China
| | - Chuanyi Yao
- Department of Chemical and Biochemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, People's Republic of China
- Xiamen Key Laboratory of Synthetic Biotechnology, Xiamen University, Xiamen, People's Republic of China
| | - Ning He
- Department of Chemical and Biochemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, People's Republic of China
- Xiamen Key Laboratory of Synthetic Biotechnology, Xiamen University, Xiamen, People's Republic of China
| | - Song Huang
- Department of Microbiome and Health, Bluepha Co., Ltd, Shenzhen, People's Republic of China
| | - Xueping Ling
- Department of Chemical and Biochemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, People's Republic of China
- Xiamen Key Laboratory of Synthetic Biotechnology, Xiamen University, Xiamen, People's Republic of China
| |
Collapse
|
22
|
Pontifex MG, Connell E, Le Gall G, Lang L, Pourtau L, Gaudout D, Angeloni C, Zallocco L, Ronci M, Giusti L, Müller M, Vauzour D. A novel Mediterranean diet-inspired supplement ameliorates cognitive, microbial, and metabolic deficits in a mouse model of low-grade inflammation. Gut Microbes 2024; 16:2363011. [PMID: 38835220 PMCID: PMC11155709 DOI: 10.1080/19490976.2024.2363011] [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: 01/10/2024] [Accepted: 05/29/2024] [Indexed: 06/06/2024] Open
Abstract
The Mediterranean diet (MD) and its bioactive constituents have been advocated for their neuroprotective properties along with their capacity to affect gut microbiota speciation and metabolism. Mediated through the gut brain axis, this modulation of the microbiota may partly contribute to the neuroprotective properties of the MD. To explore this potential interaction, we evaluated the neuroprotective properties of a novel bioactive blend (Neurosyn240) resembling the Mediterranean diet in a rodent model of chronic low-grade inflammation. Behavioral tests of cognition, brain proteomic analysis, 16S rRNA sequencing, and 1H NMR metabolomic analyses were employed to develop an understanding of the gut-brain axis interactions involved. Recognition memory, as assessed by the novel object recognition task (NOR), decreased in response to LPS insult and was restored with Neurosyn240 supplementation. Although the open field task performance did not reach significance, it correlated with NOR performance indicating an element of anxiety related to this cognitive change. Behavioral changes associated with Neurosyn240 were accompanied by a shift in the microbiota composition which included the restoration of the Firmicutes: Bacteroidota ratio and an increase in Muribaculum, Rikenellaceae Alloprevotella, and most notably Akkermansia which significantly correlated with NOR performance. Akkermansia also correlated with the metabolites 5-aminovalerate, threonine, valine, uridine monophosphate, and adenosine monophosphate, which in turn significantly correlated with NOR performance. The proteomic profile within the brain was dramatically influenced by both interventions, with KEGG analysis highlighting oxidative phosphorylation and neurodegenerative disease-related pathways to be modulated. Intriguingly, a subset of these proteomic changes simultaneously correlated with Akkermansia abundance and predominantly related to oxidative phosphorylation, perhaps alluding to a protective gut-brain axis interaction. Collectively, our results suggest that the bioactive blend Neurosyn240 conferred cognitive and microbiota resilience in response to the deleterious effects of low-grade inflammation.
Collapse
Affiliation(s)
- Matthew G. Pontifex
- Norwich Medical School, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, UK
| | - Emily Connell
- Norwich Medical School, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, UK
| | - Gwenaelle Le Gall
- Norwich Medical School, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, UK
| | - Leonie Lang
- Norwich Medical School, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, UK
| | | | | | - Cristina Angeloni
- Department for Life Quality Studies, Alma Mater Studiorum, University of Bologna, Alma, Italy
| | - Lorenzo Zallocco
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Maurizio Ronci
- Department of Pharmacy, University G. d’Annunzio of Chieti-Pescara, Chieti, Italy
| | - Laura Giusti
- School of Pharmacy, University of Camerino, Camerino, Italy
| | - Michael Müller
- Norwich Medical School, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, UK
| | - David Vauzour
- Norwich Medical School, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, UK
| |
Collapse
|
23
|
Park G, Kadyan S, Hochuli N, Pollak J, Wang B, Salazar G, Chakrabarty P, Efron P, Sheffler J, Nagpal R. A modified Mediterranean-style diet enhances brain function via specific gut-microbiome-brain mechanisms. Gut Microbes 2024; 16:2323752. [PMID: 38444392 PMCID: PMC10936641 DOI: 10.1080/19490976.2024.2323752] [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: 09/08/2023] [Accepted: 02/22/2024] [Indexed: 03/07/2024] Open
Abstract
Alzheimer's disease (AD) is a debilitating brain disorder with rapidly mounting prevalence worldwide, yet no proven AD cure has been discovered. Using a multi-omics approach in a transgenic AD mouse model, the current study demonstrated the efficacy of a modified Mediterranean-ketogenic diet (MkD) on AD-related neurocognitive pathophysiology and underlying mechanisms related to the gut-microbiome-brain axis. The findings revealed that MkD induces profound shifts in the gut microbiome community and microbial metabolites. Most notably, MkD promoted growth of the Lactobacillus population, resulting in increased bacteria-derived lactate production. We discovered elevated levels of microbiome- and diet-derived metabolites in the serum as well, signaling their influence on the brain. Importantly, these changes in serum metabolites upregulated specific receptors that have neuroprotective effects and induced alternations in neuroinflammatory-associated pathway profiles in hippocampus. Additionally, these metabolites displayed strong favorable co-regulation relationship with gut-brain integrity and inflammatory markers, as well as neurobehavioral outcomes. The findings underscore the ameliorative effects of MkD on AD-related neurological function and the underlying gut-brain communication via modulation of the gut microbiome-metabolome arrays.
Collapse
Affiliation(s)
- Gwoncheol Park
- The Gut Biome Lab, Department of Health, Nutrition, and Food Sciences, College of Education, Health, and Human Science, Florida State University, Tallahassee, FL, USA
- Department of Health, Nutrition, and Food Sciences, College of Education, Health, and Human Science, Florida State University, Tallahassee, FL, USA
| | - Saurabh Kadyan
- The Gut Biome Lab, Department of Health, Nutrition, and Food Sciences, College of Education, Health, and Human Science, Florida State University, Tallahassee, FL, USA
- Department of Health, Nutrition, and Food Sciences, College of Education, Health, and Human Science, Florida State University, Tallahassee, FL, USA
| | - Nathaniel Hochuli
- The Gut Biome Lab, Department of Health, Nutrition, and Food Sciences, College of Education, Health, and Human Science, Florida State University, Tallahassee, FL, USA
- Department of Health, Nutrition, and Food Sciences, College of Education, Health, and Human Science, Florida State University, Tallahassee, FL, USA
| | - Julie Pollak
- Department of Chemistry and Chemical Engineering, Florida Institute of Technology, Melbourne, FL, USA
| | - Bo Wang
- Department of Chemistry and Chemical Engineering, Florida Institute of Technology, Melbourne, FL, USA
| | - Gloria Salazar
- Department of Health, Nutrition, and Food Sciences, College of Education, Health, and Human Science, Florida State University, Tallahassee, FL, USA
| | - Paramita Chakrabarty
- Center for Translational Research in Neurodegenerative Diseases, Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | - Philip Efron
- Sepsis and Critical Illness Research Center, Department of Surgery, University of Florida College of Medicine, Gainesville, FL, USA
| | - Julia Sheffler
- Center for Translational Behavioral Science, Department of Behavioral Sciences and Social Medicine, Florida State University College of Medicine, Tallahassee, FL, USA
| | - Ravinder Nagpal
- The Gut Biome Lab, Department of Health, Nutrition, and Food Sciences, College of Education, Health, and Human Science, Florida State University, Tallahassee, FL, USA
- Department of Health, Nutrition, and Food Sciences, College of Education, Health, and Human Science, Florida State University, Tallahassee, FL, USA
| |
Collapse
|
24
|
Zhong HJ, Wang SQ, Zhang RX, Zhuang YP, Li L, Yi SZ, Li Y, Wu L, Ding Y, Zhang J, Xie X, He XX, Wu Q. Supplementation with high-GABA-producing Lactobacillus plantarum L5 ameliorates essential tremor triggered by decreased gut bacteria-derived GABA. Transl Neurodegener 2023; 12:58. [PMID: 38093327 PMCID: PMC10717605 DOI: 10.1186/s40035-023-00391-9] [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: 08/14/2023] [Accepted: 11/19/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND The γ-aminobutyric acid (GABA) hypothesis posits a role of GABA deficiency in the central nervous system in the pathogenesis and progression of essential tremor (ET). However, the specific causative factor for GABA deficiency is not clear. The gut microbiota in mammals has recently been considered as a significant source of GABA. Furthermore, the GABA-based signals originating from the intestine can be transmitted to the brain through the "enteric nervous system-vagus nerve-brain" axis. However, the plausible contribution of gut microbiota to ET seems inspiring but remains obscure. METHODS Fecal samples from patients with ET and healthy controls were examined by metagenomic sequencing to compare the composition of gut microbiota and the expression of genes involved in GABA biosynthesis. The impact of gut microbiota on ET was explored through transplantation of fecal microbiota from patients with ET into the murine ET model. Lactic acid bacteria producing high amounts of GABA were identified through whole-genome sequencing and ultra-performance liquid chromatography-tandem mass spectrometry. Subsequently, mice were treated with the high-GABA-producing strain Lactobacillus plantarum L5. Tremor severity, behavioral tests, pro-inflammatory cytokines, GABA concentration, and gut microbiota composition were examined in these mice. RESULTS The gut microbiota of patients with ET demonstrated an impaired GABA-producing capacity and a reduced fecal GABA concentration. Transplantation of the gut microbiota from patients with ET induced an extension of tremor duration and impaired mobility in the murine model of ET. L5 exhibited an augmented GABA-producing capacity, with the De Man-Rogosa-Sharpe culture broth containing 262 mg/l of GABA. In addition, administration of L5 significantly decreased the tremor severity and enhanced the movement capability and grasping ability of ET mice. In vivo mechanistic experiments indicated that L5 reshaped the gut microbial composition, supplemented the mucosa-associated microbiota with GABA-producing capacity, increased the GABA concentrations in the cerebellum, and diminished inflammation in the central nervous system. CONCLUSIONS These findings highlight that deficiency of GABA-producing gut microbes plays an essential role in the pathogenesis of ET and that L5 is a promising candidate for treating ET.
Collapse
Affiliation(s)
- Hao-Jie Zhong
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510000, China
- Department of Gastroenterology, the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510000, China
| | - Si-Qi Wang
- Department of Gastroenterology, the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510000, China
| | - Ruo-Xin Zhang
- Department of Gastroenterology, the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510000, China
| | - Yu-Pei Zhuang
- Department of Gastroenterology, the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510000, China
| | - Longyan Li
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510000, China
| | - Shuo-Zhao Yi
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510000, China
| | - Ying Li
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510000, China
| | - Lei Wu
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510000, China
- Department of Gastroenterology, the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510000, China
| | - Yu Ding
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510000, China
| | - Jumei Zhang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510000, China
| | - Xinqiang Xie
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510000, China.
| | - Xing-Xiang He
- Department of Gastroenterology, the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510000, China.
| | - Qingping Wu
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510000, China.
| |
Collapse
|
25
|
Liu W, Jiang H, Liu X, Zheng Y, Liu Y, Pan F, Yu F, Li Z, Gu M, Du Q, Li X, Zhang H, Han D. Altered intestinal microbiota enhances adenoid hypertrophy by disrupting the immune balance. Front Immunol 2023; 14:1277351. [PMID: 38090578 PMCID: PMC10715246 DOI: 10.3389/fimmu.2023.1277351] [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: 08/14/2023] [Accepted: 11/06/2023] [Indexed: 12/18/2023] Open
Abstract
Introduction Adenoid hypertrophy (AH) is a common upper respiratory disorder in children. Disturbances of gut microbiota have been implicated in AH. However, the interplay of alteration of gut microbiome and enlarged adenoids remains elusive. Methods 119 AH children and 100 healthy controls were recruited, and microbiome profiling of fecal samples in participants was performed using 16S rRNA gene sequencing. Fecal microbiome transplantation (FMT) was conducted to verify the effects of gut microbiota on immune response in mice. Results In AH individuals, only a slight decrease of diversity in bacterial community was found, while significant changes of microbial composition were observed between these two groups. Compared with HCs, decreased abundances of Akkermansia, Oscillospiraceae and Eubacterium coprostanoligenes genera and increased abundances of Bacteroides, Faecalibacterium, Ruminococcus gnavus genera were revealed in AH patients. The abundance of Bacteroides remained stable with age in AH children. Notably, a microbial marker panel of 8 OTUs were identified, which discriminated AH from HC individuals with an area under the curve (AUC) of 0.9851 in the discovery set, and verified in the geographically different validation set, achieving an AUC of 0.9782. Furthermore, transfer of mice with fecal microbiota from AH patients dramatically reduced the proportion of Treg subsets within peripheral blood and nasal-associated lymphoid tissue (NALT) and promoted the expansion of Th2 cells in NALT. Conclusion These findings highlight the effect of the altered gut microbiota in the AH pathogenesis.
Collapse
Affiliation(s)
- Wenxin Liu
- Department of Clinical Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Huier Jiang
- Department of Clinical Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiling Liu
- Shanghai Key Laboratory of Forensic Medicine, Shanghai Forensic Service Platform, Academy of Forensic Science, Ministry of Justice, Shanghai, China
| | - Yue Zheng
- Department of Clinical Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yanan Liu
- Department of Clinical Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Fen Pan
- Department of Clinical Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Fangyuan Yu
- Department of Clinical Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhi Li
- Department of Pathology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Meizhen Gu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qingqing Du
- Department of Clinical Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoyan Li
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hong Zhang
- Department of Clinical Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Dingding Han
- Department of Clinical Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Medical School, Guangxi University, Nanning, China
| |
Collapse
|
26
|
Iyaswamy A, Lu K, Guan XJ, Kan Y, Su C, Liu J, Jaganathan R, Vasudevan K, Paul J, Thakur A, Li M. Impact and Advances in the Role of Bacterial Extracellular Vesicles in Neurodegenerative Disease and Its Therapeutics. Biomedicines 2023; 11:2056. [PMID: 37509695 PMCID: PMC10377521 DOI: 10.3390/biomedicines11072056] [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: 06/23/2023] [Revised: 07/16/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
Bacterial Extracellular Vesicles (BEVs) possess the capability of intracellular interactions with other cells, and, hence, can be utilized as an efficient cargo for worldwide delivery of therapeutic substances such as monoclonal antibodies, proteins, plasmids, siRNA, and small molecules for the treatment of neurodegenerative diseases (NDs). BEVs additionally possess a remarkable capacity for delivering these therapeutics across the blood-brain barrier to treat Alzheimer's disease (AD). This review summarizes the role and advancement of BEVs for NDs, AD, and their treatment. Additionally, it investigates the critical BEV networks in the microbiome-gut-brain axis, their defensive and offensive roles in NDs, and their interaction with NDs. Furthermore, the part of BEVs in the neuroimmune system and their interference with ND, as well as the risk factors made by BEVs in the autophagy-lysosomal pathway and their potential outcomes on ND, are all discussed. To conclude, this review aims to gain a better understanding of the credentials of BEVs in NDs and possibly discover new therapeutic strategies.
Collapse
Affiliation(s)
- Ashok Iyaswamy
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore 641021, India
| | - Kejia Lu
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Xin-Jie Guan
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Yuxuan Kan
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Chengfu Su
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Jia Liu
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Ravindran Jaganathan
- Preclinical Department, Faculty of Medicine, Royal College of Medicine Perak, Universiti Kuala Lumpur, Ipoh 30450, Malaysia
| | | | - Jeyakumari Paul
- Department of Physiology, Dr. ALM PG Institute of Basic Medical Sciences, University of Madras, Chennai 600005, India
| | - Abhimanyu Thakur
- Pritzker School of Molecular Engineering, Ben May Department for Cancer Research, University of Chicago, Chicago, IL 60637, USA
| | - Min Li
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| |
Collapse
|
27
|
Qu L, Liu F, Fang Y, Wang L, Chen H, Yang Q, Dong H, Jin L, Wu W, Sun D. Improvement in Zebrafish with Diabetes and Alzheimer's Disease Treated with Pasteurized Akkermansia muciniphila. Microbiol Spectr 2023; 11:e0084923. [PMID: 37191572 PMCID: PMC10269592 DOI: 10.1128/spectrum.00849-23] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 05/01/2023] [Indexed: 05/17/2023] Open
Abstract
Diabetes and Alzheimer's disease (AD) are associated with specific changes in the composition of the intestinal flora. Studies have shown that the supplementation with pasteurized Akkermansia muciniphila has therapeutic and preventive effects on diabetes. However, it is not clear whether there is any association with improvement in and prevention of Alzheimer's disease and diabetes with Alzheimer's disease. Here, we found that pasteurized Akkermansia muciniphila can significantly improve the blood glucose, body mass index, and diabetes indexes of zebrafish with diabetes mellitus complicated with Alzheimer's disease and also alleviate the related indexes of Alzheimer's disease. The memory, anxiety, aggression, and social preference behavior of zebrafish with combined type 2 diabetes mellitus (T2DM) and Alzheimer's disease (TA zebrafish) were significantly improved after pasteurized Akkermansia muciniphila treatment. Moreover, we examined the preventive effect of pasteurized Akkermansia muciniphila on diabetes mellitus complicated with Alzheimer's disease. The results showed that the zebrafish in the prevention group were better in terms of biochemical index and behavior than the zebrafish in the treatment group. These findings provide new ideas for the prevention and treatment of diabetes mellitus complicated with Alzheimer's disease. IMPORTANCE The interaction between intestinal microflora and host affects the progression of diabetes and Alzheimer's disease. As a recognized next-generation probiotic, Akkermansia muciniphila has been shown to play a key role in the progression of diabetes and Alzheimer's disease, but whether A. muciniphila can improve diabetes complicated with Alzheimer's disease and its potential mechanism are unclear. In this study, a new zebrafish model of diabetes mellitus complicated with Alzheimer's disease was established, and the effect of Akkermansia muciniphila on diabetes mellitus complicated with Alzheimer's disease is discussed. The results showed that Akkermansia muciniphila after pasteurization significantly improved and prevented diabetes mellitus complicated with Alzheimer's disease. Treatment with pasteurized Akkermansia muciniphila improved the memory, social preference, and aggressive and anxiety behavior of TA zebrafish and alleviated the pathological characteristics of T2DM and AD. These results provide a new prospect for probiotics in the treatment of diabetes and Alzheimer's disease.
Collapse
Affiliation(s)
- Linkai Qu
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, China
- College of Life Sciences, Jilin Agricultural University, Changchun, China
| | - Fan Liu
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, China
| | - Yimeng Fang
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, China
| | - Lei Wang
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, China
| | - Haojie Chen
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, China
| | - Qinsi Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Hao Dong
- College of Life Sciences, Jilin Agricultural University, Changchun, China
| | - Libo Jin
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, China
| | - Wei Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Da Sun
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, China
| |
Collapse
|
28
|
Mandato C, Colucci A, Lanzillo R, Staiano A, Scarpato E, Schiavo L, Operto FF, Serra MR, Di Monaco C, Napoli JS, Massa G, Vajro P. Multiple Sclerosis-Related Dietary and Nutritional Issues: An Updated Scoping Review with a Focus on Pediatrics. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1022. [PMID: 37371254 PMCID: PMC10297186 DOI: 10.3390/children10061022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/03/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023]
Abstract
PURPOSE Lifestyle/dietetic habits play an important role in the development and progression of multiple sclerosis (MS) disease. Here, we examine the basic pathomechanisms underlying intestinal and brain barrier modifications in MS and consider diets and dietary supplementations proposed over time to complement pharmacological therapies for improving disease outcome both in adults and in children. METHODS Scoping literature search about evidence-based findings in MS-related gut-brain axis (GBA) pathophysiology and nutritional issues at all ages. FINDINGS Data show that (1) no universal best diet exists, (2) healthy/balanced diets are, however, necessary to safeguard the adequate intake of all essential nutrients, (3) diets with high intakes of fruits, vegetables, whole grains, and lean proteins that limit processed foods, sugar, and saturated fat appear beneficial for their antioxidant and anti-inflammatory properties and their ability to shape a gut microbiota that respects the gut and brain barriers, (4) obesity may trigger MS onset and/or its less favorable course, especially in pediatric-onset MS. Vitamin D and polyunsaturated fatty acids are the most studied supplements for reducing MS-associated inflammation. CONCLUSIONS Pending results from other and/or newer approaches targeting the GBA (e.g., pre- and probiotics, engineered probiotics, fecal-microbiota transplantation), accurate counseling in choosing adequate diet and maintaining physical activity remains recommended for MS prevention and management both in adults and children.
Collapse
Affiliation(s)
- Claudia Mandato
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, Pediatrics Section, University of Salerno, 84081 Baronissi, Salerno, Italy (P.V.)
| | - Angelo Colucci
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, Pediatrics Section, University of Salerno, 84081 Baronissi, Salerno, Italy (P.V.)
| | - Roberta Lanzillo
- Multiple Sclerosis Clinical Care and Research Centre, Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, 80138 Naples, Naples, Italy
| | - Annamaria Staiano
- Department of Translational Medical Science, Section of Pediatrics, University of Naples Federico II, 80138 Naples, Naples, Italy
| | - Elena Scarpato
- Department of Translational Medical Science, Section of Pediatrics, University of Naples Federico II, 80138 Naples, Naples, Italy
| | - Luigi Schiavo
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, Nutrition Section, University of Salerno, 84081 Baronissi, Salerno, Italy
| | - Francesca Felicia Operto
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, Pediatric Psychiatry Section, University of Salerno, 84081 Baronissi, Salerno, Italy
| | - Maria Rosaria Serra
- Department of Translational Medical Science, Section of Pediatrics, University of Naples Federico II, 80138 Naples, Naples, Italy
| | - Cristina Di Monaco
- Multiple Sclerosis Clinical Care and Research Centre, Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, 80138 Naples, Naples, Italy
| | - Julia Sara Napoli
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, Pediatrics Section, University of Salerno, 84081 Baronissi, Salerno, Italy (P.V.)
| | - Grazia Massa
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, Pediatrics Section, University of Salerno, 84081 Baronissi, Salerno, Italy (P.V.)
| | - Pietro Vajro
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, Pediatrics Section, University of Salerno, 84081 Baronissi, Salerno, Italy (P.V.)
| |
Collapse
|
29
|
Isnard S, Royston L, Scott SC, Mabanga T, Lin J, Fombuena B, Bu S, Berini CA, Goldberg MS, Finkelman M, Brouillette MJ, Fellows LK, Mayo NE, Routy JP. Translocation of bacterial LPS is associated with self-reported cognitive abilities in men living with HIV receiving antiretroviral therapy. AIDS Res Ther 2023; 20:30. [PMID: 37202809 DOI: 10.1186/s12981-023-00525-z] [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: 02/07/2023] [Accepted: 05/11/2023] [Indexed: 05/20/2023] Open
Abstract
BACKGROUND Gut damage allows translocation of bacterial lipopolysaccharide (LPS) and fungal β-D-glucan (BDG) into the blood. This microbial translocation contributes to systemic inflammation and risk of non-AIDS comorbidities in people living with HIV, including those receiving antiretroviral therapy (ART). We assessed whether markers of gut damage and microbial translocation were associated with cognition in ART-treated PLWH. METHODS Eighty ART-treated men living with HIV from the Positive Brain Health Now Canadian cohort were included. Brief cognitive ability measure (B-CAM) and 20-item patient deficit questionnaire (PDQ) were administered to all participants. Three groups were selected based on their B-CAM levels. We excluded participants who received proton pump inhibitors or antiacids in the past 3 months. Cannabis users were also excluded. Plasma levels of intestinal fatty acid binding protein (I-FABP), regenerating islet-derived protein 3 α (REG3α), and lipopolysaccharides (LPS = were quantified by ELISA, while 1-3-β-D-glucan BDG) levels were assessed using the Fungitell assay. Univariable, multivariable, and splines analyses were performed. RESULTS Plasma levels of I-FABP, REG3α, LPS and BDG were not different between groups of low, intermediate and high B-CAM levels. However, LPS and REG3α levels were higher in participants with PDQ higher than the median. Multivariable analyses showed that LPS association with PDQ, but not B-CAM, was independent of age and level of education. I-FABP, REG3α, and BDG levels were not associated with B-CAM nor PDQ levels in multivariable analyses. CONCLUSION In this well characterized cohort of ART-treated men living with HIV, bacterial but not fungal translocation was associated with presence of cognitive difficulties. These results need replication in larger samples.
Collapse
Affiliation(s)
- Stéphane Isnard
- Infectious Disease and Immunity in Global Health Program, Research Institute of McGill University Health Centre, 1001 Boulevard Décarie, Montreal, QC, H4A 3J1, Canada
- Chronic Viral Illness Service, McGill University Health Centre, Montreal, QC, Canada
- CIHR Canadian HIV Trials Network, Vancouver, BC, Canada
| | - Léna Royston
- Infectious Disease and Immunity in Global Health Program, Research Institute of McGill University Health Centre, 1001 Boulevard Décarie, Montreal, QC, H4A 3J1, Canada
- Chronic Viral Illness Service, McGill University Health Centre, Montreal, QC, Canada
- Division of Infectious Diseases, Geneva University Hospitals, Geneva, Switzerland
| | - Susan C Scott
- Division of Clinical Epidemiology, Center for Outcomes Research and Evaluation, McGill University Health Centre (MUHC), Montreal, Canada
| | - Tsoarello Mabanga
- Infectious Disease and Immunity in Global Health Program, Research Institute of McGill University Health Centre, 1001 Boulevard Décarie, Montreal, QC, H4A 3J1, Canada
- Chronic Viral Illness Service, McGill University Health Centre, Montreal, QC, Canada
| | - John Lin
- Infectious Disease and Immunity in Global Health Program, Research Institute of McGill University Health Centre, 1001 Boulevard Décarie, Montreal, QC, H4A 3J1, Canada
- Chronic Viral Illness Service, McGill University Health Centre, Montreal, QC, Canada
| | - Brandon Fombuena
- Infectious Disease and Immunity in Global Health Program, Research Institute of McGill University Health Centre, 1001 Boulevard Décarie, Montreal, QC, H4A 3J1, Canada
- Chronic Viral Illness Service, McGill University Health Centre, Montreal, QC, Canada
| | - Simeng Bu
- Infectious Disease and Immunity in Global Health Program, Research Institute of McGill University Health Centre, 1001 Boulevard Décarie, Montreal, QC, H4A 3J1, Canada
- Chronic Viral Illness Service, McGill University Health Centre, Montreal, QC, Canada
| | - Carolina A Berini
- Infectious Disease and Immunity in Global Health Program, Research Institute of McGill University Health Centre, 1001 Boulevard Décarie, Montreal, QC, H4A 3J1, Canada
- Chronic Viral Illness Service, McGill University Health Centre, Montreal, QC, Canada
| | | | | | - Marie-Josée Brouillette
- Infectious Disease and Immunity in Global Health Program, Research Institute of McGill University Health Centre, 1001 Boulevard Décarie, Montreal, QC, H4A 3J1, Canada
- Chronic Viral Illness Service, McGill University Health Centre, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, Canada
| | - Lesley K Fellows
- Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Nancy E Mayo
- Division of Clinical Epidemiology, Center for Outcomes Research and Evaluation, McGill University Health Centre (MUHC), Montreal, Canada
- School of Physical and Occupational Therapy, McGill University, Montreal, Canada
- Department of Medicine, Division of Geriatrics, McGill University, Montreal, Canada
| | - Jean-Pierre Routy
- Infectious Disease and Immunity in Global Health Program, Research Institute of McGill University Health Centre, 1001 Boulevard Décarie, Montreal, QC, H4A 3J1, Canada.
- Chronic Viral Illness Service, McGill University Health Centre, Montreal, QC, Canada.
- Division of Hematology, McGill University Health Centre, Montreal, QC, Canada.
| |
Collapse
|
30
|
Centner AM, Khalili L, Ukhanov V, Kadyan S, Nagpal R, Salazar G. The Role of Phytochemicals and Gut Microbiome in Atherosclerosis in Preclinical Mouse Models. Nutrients 2023; 15:1212. [PMID: 36904211 PMCID: PMC10005405 DOI: 10.3390/nu15051212] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/10/2023] [Accepted: 02/14/2023] [Indexed: 03/06/2023] Open
Abstract
Gut microbiome alterations have recently been linked to many chronic conditions including cardiovascular disease (CVD). There is an interplay between diet and the resident gut microbiome, where the food eaten affects populations of certain microbes. This is important, as different microbes are associated with various pathologies, as they can produce compounds that are disease-promoting or disease-protecting. The Western diet negatively affects the host gut microbiome, ultimately resulting in heightened arterial inflammation and cell phenotype changes as well as plaque accumulation in the arteries. Nutritional interventions including whole foods rich in fiber and phytochemicals as well as isolated compounds including polyphenols and traditional medicinal plants show promise in positively influencing the host gut microbiome to alleviate atherosclerosis. This review investigates the efficacy of a vast array of foods and phytochemicals on host gut microbes and atherosclerotic burden in mice. Reduction in plaque by interventions was associated with increases in bacterial diversity, reduction in the Firmicutes/Bacteroidetes (F/B) ratio, and upregulation of Akkermansia. Upregulation in CYP7 isoform in the liver, ABC transporters, bile acid excretion, and the level of acetic acid, propionic acid, and butyric acid were also noted in several studies reducing plaque. These changes were also associated with attenuated inflammation and oxidative stress. In conclusion, an increase in the abundance of Akkermansia with diets rich in polyphenols, fiber, and grains is likely to reduce plaque burden in patients suffering from CVD.
Collapse
Affiliation(s)
- Ann M. Centner
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL 32306, USA
| | - Leila Khalili
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, FL 32306, USA
| | - Vladimir Ukhanov
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, FL 32306, USA
| | - Saurabh Kadyan
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, FL 32306, USA
| | - Ravinder Nagpal
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, FL 32306, USA
| | - Gloria Salazar
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, FL 32306, USA
| |
Collapse
|
31
|
Lee HL, Kim JM, Moon JH, Kim MJ, Jeong HR, Go MJ, Kim HJ, Eo HJ, Lee U, Heo HJ. Anti-Amnesic Effect of Synbiotic Supplementation Containing Corni fructus and Limosilactobacillus reuteri in DSS-Induced Colitis Mice. Int J Mol Sci 2022; 24:ijms24010090. [PMID: 36613533 PMCID: PMC9820465 DOI: 10.3390/ijms24010090] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/12/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
This study was conducted to compare the synbiotic activity between Corni fructus (C. fructus) and Limosilactobacillus reuteri (L. reuteri) on dextran sulfate sodium (DSS)-induced colitis and cognitive dysfunction in C57BL/6 mice. C. fructus (as prebiotics, PRE), L. reuteri (as probiotics, PRO), and synbiotics (as a mixture of L. reuteri and C. fructus, SYN) were fed to mice for 3 weeks. Consumption of PRE, PRO, and SYN ameliorated colitis symptoms in body weight, large intestinal length, and serum albumin level. Moreover, SYN showed a synergistic effect on intestinal permeability and intestinal anti-inflammation response. Also, SYN significantly improved cognitive function as a result of measuring the Y-maze and passive avoidance tests in DSS-induced behavioral disorder mice. Especially, SYN also restored memory function by increasing the cholinergic system and reducing tau and amyloid β pathology. In addition, PRE, PRO, and SYN ameliorated dysbiosis by regulating the gut microbiota and the concentration of short-chain fatty acids (SCFAs) in feces. The bioactive compounds of C. fructus were identified with quinic acid, morroniside, loganin, and cornuside, using ultra-performance liquid chromatography-quadrupole time-of-flight tandem mass spectrometry (UPLC-Q-TOF-MS2). In conclusion, synbiotic supplementation alleviated DSS-induced colitis and cognitive dysfunction by modulating gut microbiota, proinflammatory cytokines, and SCFAs production.
Collapse
Affiliation(s)
- Hyo Lim Lee
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Jong Min Kim
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Jong Hyun Moon
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Min Ji Kim
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Hye Rin Jeong
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Min Ji Go
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Hyun-Jin Kim
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Hyun Ji Eo
- Division of Special Forest Resources, Department of Forest Bioresources, National Institute of Forest Science (NIFoS), Suwon 16631, Republic of Korea
| | - Uk Lee
- Division of Special Forest Resources, Department of Forest Bioresources, National Institute of Forest Science (NIFoS), Suwon 16631, Republic of Korea
| | - Ho Jin Heo
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
- Correspondence: ; Tel.: +82-(55)-7721907
| |
Collapse
|
32
|
Șchiopu CG, Ștefănescu C, Boloș A, Diaconescu S, Gilca-Blanariu GE, Ștefănescu G. Functional Gastrointestinal Disorders with Psychiatric Symptoms: Involvement of the Microbiome-Gut-Brain Axis in the Pathophysiology and Case Management. Microorganisms 2022; 10:2199. [PMID: 36363791 PMCID: PMC9694215 DOI: 10.3390/microorganisms10112199] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/03/2022] [Accepted: 11/05/2022] [Indexed: 11/09/2022] Open
Abstract
Functional Gastrointestinal Disorders have been an important cause of poor life quality in affected populations. The unclear etiology and pathophysiological mechanism alter the clinical evolution of the patient. Although a strong connection with psychological stress has been observed, it was not until recently that the gut-brain axis involvement has been revealed. Furthermore, the current literature not only promotes the gut-brain axis modulation as a therapeutical target for functional digestive disorders but also states that the gut microbiome has a main role in this bi-directional mechanism. Psychiatric symptoms are currently recognized as an equally important aspect of the clinical manifestation and modulation of both the digestive and central nervous systems and could be the best approach in restoring the balance. As such, this article proposes a detailed description of the physiology of the microbiome-gut-brain axis, the pathophysiology of the functional gastrointestinal disorders with psychiatric symptoms and current perspectives for therapeutical management, as revealed by the latest studies in the scientific literature.
Collapse
Affiliation(s)
- Cristina Gabriela Șchiopu
- Department of Psychiatry, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iași, Romania
| | - Cristinel Ștefănescu
- Department of Psychiatry, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iași, Romania
| | - Alexandra Boloș
- Department of Psychiatry, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iași, Romania
| | - Smaranda Diaconescu
- Medical-Surgical Department, Faculty of Medicine, University “Titu Maiorescu”, 040441 Bucuresti, Romania
| | | | - Gabriela Ștefănescu
- Department of Gastroentereology, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iași, Romania
| |
Collapse
|
33
|
Li T, Chu C, Yu L, Zhai Q, Wang S, Zhao J, Zhang H, Chen W, Tian F. Neuroprotective Effects of Bifidobacterium breve CCFM1067 in MPTP-Induced Mouse Models of Parkinson's Disease. Nutrients 2022; 14:4678. [PMID: 36364939 PMCID: PMC9655354 DOI: 10.3390/nu14214678] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/22/2022] [Accepted: 10/27/2022] [Indexed: 08/31/2023] Open
Abstract
There is mounting evidence that the microbiota-gut-brain axis (MGBA) is critical in the pathogenesis and progression of Parkinson's disease (PD), suggesting that probiotic therapy restoring gut microecology may slow down disease progression. In this study, we examined the disease-alleviating effects of Bifidobacterium breve CCFM1067, orally administered for 5 weeks in a PD mouse model. Our study shows that supplementation with the probiotic B. breve CCFM1067 protected dopaminergic neurons and suppressed glial cell hyperactivation and neuroinflammation in PD mice. In addition, the antioxidant capacity of the central nervous system was enhanced and oxidative stress was alleviated. Moreover, B. breve CCFM1067 protected the blood-brain and intestinal barriers from damage in the MPTP-induced mouse model. The results of fecal microbiota analysis showed that B. breve CCFM1067 intervention could act on the MPTP-induced microecological imbalance in the intestinal microbiota, suppressing the number of pathogenic bacteria (Escherichia-Shigella) while increasing the number of beneficial bacteria (Bifidobacterium and Akkermansia) in PD mice. In addition, the increase in short chain fatty acids (acetic and butyric acids) may explain the anti-inflammatory action of B. breve CCFM1067 in the gut or brain of the MPTP-induced PD mouse model. In conclusion, we demonstrated that the probiotic B. breve CCFM1067, which can prevent or treat PD by modulating the gut-brain axis, can be utilized as a possible new oral supplement for PD therapy.
Collapse
Affiliation(s)
- Tiantian Li
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Chuanqi Chu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Leilei Yu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Shunhe Wang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
- Department of Child Health Care, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi 214002, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Fengwei Tian
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
34
|
Lee DW, Ryu YK, Chang DH, Park HY, Go J, Maeng SY, Hwang DY, Kim BC, Lee CH, Kim KS. Agathobaculum butyriciproducens Shows Neuroprotective Effects in a 6-OHDA-Induced Mouse Model of Parkinson's Disease. J Microbiol Biotechnol 2022; 32:1168-1177. [PMID: 36168204 PMCID: PMC9628974 DOI: 10.4014/jmb.2205.05032] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 08/23/2022] [Accepted: 08/29/2022] [Indexed: 12/15/2022]
Abstract
Parkinson's disease (PD) is the second-most prevalent neurodegenerative disease and is characterized by dopaminergic neuronal death in the midbrain. Recently, the association between alterations in PD pathology and the gut microbiota has been explored. Microbiota-targeted interventions have been suggested as a novel therapeutic approach for PD. Agathobaculum butyriciproducens SR79T (SR79) is an anaerobic bacterium. Previously, we showed that SR79 treatment induced cognitive improvement and reduced Alzheimer's disease pathologies in a mouse model. In this study, we hypothesized that SR79 treatment may have beneficial effects on PD pathology. To investigate the therapeutic effects of SR79 on PD, 6-hydroxydopamine (6-OHDA)-induced mouse models were used. D-Amphetamine sulfate (d-AMPH)-induced behavioral rotations and dopaminergic cell death were analyzed in unilateral 6-OHDA-lesioned mice. Treatment with SR79 significantly decreased ipsilateral rotations induced by d-AMPH. Moreover, SR79 treatment markedly activated the AKT/GSK3β signaling pathway in the striatum. In addition, SR79 treatment affected the Nrf2/ARE signaling pathway and its downstream target genes in the striatum of 6-OHDA-lesioned mice. Our findings suggest a protective role of SR79 in 6-OHDA-induced toxicity by regulating the AKT/Nrf2/ARE signaling pathway and astrocyte activation. Thus, SR79 may be a potential microbe-based intervention and therapeutic strategy for PD.
Collapse
Affiliation(s)
- Da Woon Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea,Department of Biomaterials Science, College of Natural Resources and Life Science and Industry Convergence Research Institute, Pusan National University, Miryang 50463, Republic of Korea
| | - Young-Kyoung Ryu
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Dong-Ho Chang
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Hye-Yeon Park
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Jun Go
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - So-Young Maeng
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea,College of Biosciences and Biotechnology, Chung-Nam National University, Daejeon 34134, Republic of Korea
| | - Dae Youn Hwang
- Department of Biomaterials Science, College of Natural Resources and Life Science and Industry Convergence Research Institute, Pusan National University, Miryang 50463, Republic of Korea
| | - Byoung-Chan Kim
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea,HealthBiome, Inc., Daejeon 34141, Republic of Korea
| | - Chul-Ho Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea,Department of Biosystems and Bioengineering, University of Science and Technology (UST), Daejeon 34113, Republic of Korea,Corresponding authors C.H. Lee E-mail:
| | - Kyoung-Shim Kim
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea,
K.S. Kim Phone: 82-42-860-4634 Fax : 82-42-860-4609 E-mail:
| |
Collapse
|
35
|
Zhu M, Liu X, Ye Y, Yan X, Cheng Y, Zhao L, Chen F, Ling Z. Gut Microbiota: A Novel Therapeutic Target for Parkinson’s Disease. Front Immunol 2022; 13:937555. [PMID: 35812394 PMCID: PMC9263276 DOI: 10.3389/fimmu.2022.937555] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 05/26/2022] [Indexed: 12/16/2022] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disease characterized by motor dysfunction. Growing evidence has demonstrated that gut dysbiosis is involved in the occurrence, development and progression of PD. Numerous clinical trials have identified the characteristics of the changed gut microbiota profiles, and preclinical studies in PD animal models have indicated that gut dysbiosis can influence the progression and onset of PD via increasing intestinal permeability, aggravating neuroinflammation, aggregating abnormal levels of α-synuclein fibrils, increasing oxidative stress, and decreasing neurotransmitter production. The gut microbiota can be considered promising diagnostic and therapeutic targets for PD, which can be regulated by probiotics, psychobiotics, prebiotics, synbiotics, postbiotics, fecal microbiota transplantation, diet modifications, and Chinese medicine. This review summarizes the recent studies in PD-associated gut microbiota profiles and functions, the potential roles, and mechanisms of gut microbiota in PD, and gut microbiota-targeted interventions for PD. Deciphering the underlying roles and mechanisms of the PD-associated gut microbiota will help interpret the pathogenesis of PD from new perspectives and elucidate novel therapeutic strategies for PD.
Collapse
Affiliation(s)
- Manlian Zhu
- Department of Geriatrics, Lishui Second People’s Hospital, Lishui, China
| | - Xia Liu
- Department of Intensive Care Unit, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yiru Ye
- Department of Respiratory Medicine, Lishui Central Hospital, Lishui, China
| | - Xiumei Yan
- Department of Laboratory Medicine, Lishui Second People’s Hospital, Lishui, China
| | - Yiwen Cheng
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Longyou Zhao
- Department of Laboratory Medicine, Lishui Second People’s Hospital, Lishui, China
| | - Feng Chen
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- *Correspondence: Zongxin Ling, ; ; Feng Chen,
| | - Zongxin Ling
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- *Correspondence: Zongxin Ling, ; ; Feng Chen,
| |
Collapse
|