1
|
Mohsen E, Haffez H, Ahmed S, Hamed S, El-Mahdy TS. Multiple Sclerosis: A Story of the Interaction Between Gut Microbiome and Components of the Immune System. Mol Neurobiol 2025; 62:7762-7775. [PMID: 39934561 PMCID: PMC12078361 DOI: 10.1007/s12035-025-04728-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 01/27/2025] [Indexed: 02/13/2025]
Abstract
Multiple sclerosis (MS) is defined as an inflammatory disorder that chronically affects the central nervous system of young people mostly and is distributed globally. It is associated with degeneration and demyelination of the myelin sheath around the nerves, resulting in multiple neurological disability symptoms ranging from mild to severe cases that end with paralysis sometimes. MS is one of the rising diseases globally that is unfortunately associated with reduced quality of life and adding national economic burdens. The definite MS mechanism is not clearly defined; however, all the previous researches confirm the role of the immune system as the master contributor in the pathogenesis. Innate and adaptive immune cells are activated peripherally then attracted toward the central nervous system (CNS) due to the breakdown of the blood-brain barrier. Recently, the gut-brain axis was shown to depend on gut metabolites that are produced by different microorganisms in the colon. The difference in microbiota composition between individuals is responsible for diversity in secreted metabolites that affect immune responses locally in the gut or systemically when reach blood circulation to the brain. It may enhance or suppress immune responses in the central nervous system (CNS) (repeated short forms); consequently, it may exacerbate or ameliorate MS symptoms. Recent data showed that some metabolites can be used as adjuvant therapy in MS and other inflammatory diseases. This review sheds light on the nature of MS and the possible interaction between gut microbiota and immune system regulation through the gut-brain axis, hence contributing to MS pathogenesis.
Collapse
Affiliation(s)
- Esraa Mohsen
- Department of Microbiology and Immunology, Faculty of Pharmacy, Helwan University, PO Box 11795, Cairo, Egypt
| | - Hesham Haffez
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Helwan University, PO Box 11795, Cairo, Egypt
- Center of Scientific Excellence "Helwan Structural Biology Research (HSBR), Helwan University, Cairo, 11795, Egypt
| | - Sandra Ahmed
- Department of Neurology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Selwan Hamed
- Department of Microbiology and Immunology, Faculty of Pharmacy, Helwan University, PO Box 11795, Cairo, Egypt.
| | - Taghrid S El-Mahdy
- Department of Microbiology and Immunology, Faculty of Pharmacy, Helwan University, PO Box 11795, Cairo, Egypt
- Department of Microbiology and Immunology, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Cairo, Egypt
| |
Collapse
|
2
|
Livingston DBH, Sweet A, Chowdary M, Demissie MS, Rodrigue A, Gedara KP, Kishore L, Mahmoodianfard S, Power KA. Diet alters the effects of lipopolysaccharide on intestinal health and cecal microbiota composition in C57Bl/6 male mice. J Nutr Biochem 2025:109951. [PMID: 40345399 DOI: 10.1016/j.jnutbio.2025.109951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/13/2025] [Accepted: 05/02/2025] [Indexed: 05/11/2025]
Abstract
Lipopolysaccharide (LPS), a component of the gram-negative bacteria cell well, is established to induce an acute and transient inflammatory cascade upon exogenous administration in mice, while negatively impacting aspects of the microbiota gut-brain axis (mGBA). Dietary supplementation with flaxseed (FS) has been shown to partially attenuate these impairments across the mGBA, although the importance of intestinal and hepatic health has not been established in this context. In this study, we furthered our investigation of the impact of dietary supplementation with FS and FS oil (FO) in mitigating LPS-induced perturbations to intestinal health (i.e. cecal microbiota, barrier integrity, inflammation, short-chain fatty acid (SCFA) production), hepatic inflammation, and their relationships to LPS-induced systemic and neuroinflammation. We noted distinct, diet-dependent shifts in the microbiota composition twenty-four hours post-LPS injection, that may be partially driven by the LPS-induced anorexia. BD-fed mice challenged with LPS showed reduced Muribaculaceae, while FS-LPS mice had elevated Akkermansia, and both the FS-LPS and FO-LPS mice had reduced Bacteroides, each with unique correlations to diet intake. These LPS-driven shifts in the microbiota were coupled with elevated ileal mucous content in the FS- and FO-fed mice, and intestinal inflammation which was partially attenuated in the FS-fed mice. Collectively, we highlight that FS modulates aspects of the intestinal microenvironment, which may be related to the anti-inflammatory effects across the mGBA.
Collapse
Affiliation(s)
- Dawson B H Livingston
- Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada, K1H 8L1
| | - Allison Sweet
- Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada, K1H 8L1
| | - Maryam Chowdary
- Faculty of Science, Department of Biology, University of Ottawa, Ottawa, Canada, K1N 9A7
| | - Meron Samuel Demissie
- Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada, K1H 8L1
| | - Alexane Rodrigue
- Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada, K1H 8L1
| | - Kasuni Pillagawa Gedara
- Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada, K1H 8L1
| | - Lalit Kishore
- Faculty of Health Science, School of Nutrition Sciences, University of Ottawa, Ottawa, Canada, K1N 6N5
| | - Salma Mahmoodianfard
- Faculty of Health Science, School of Nutrition Sciences, University of Ottawa, Ottawa, Canada, K1N 6N5
| | - Krista A Power
- Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada, K1H 8L1; Faculty of Health Science, School of Nutrition Sciences, University of Ottawa, Ottawa, Canada, K1N 6N5.
| |
Collapse
|
3
|
Xue C, Lu M, Qin Y, Zhao X, Yang J, Yuan S, Wang Z, Cho N, Jiang C. Polygonati Rhizoma polysaccharide suppresses microglial activation and promotes functional recovery of spinal cord via improving intestinal microbiota. Int J Biol Macromol 2025:143934. [PMID: 40345303 DOI: 10.1016/j.ijbiomac.2025.143934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 04/16/2025] [Accepted: 05/03/2025] [Indexed: 05/11/2025]
Abstract
Spinal cord injury (SCI) can disrupt the gut microbiome and metabolites, impacting prognosis, substantially impairing quality of life, and leading to high socioeconomic costs (Jing et al., 2021). Recent studies indicate that plant polysaccharides exhibit antimicrobial, anti-inflammatory, and immune-enhancing properties, contributing positively to various neurological disorders; however, their role in SCI remains uncertain (Hou et al., 2020 [2]; Aswini et al., 2021 [3]; Dou et al., 2019 [4]). Polygonati Rhizoma polysaccharide (PRP) has shown anti-inflammatory and immunoregulatory effects, but its role in SCI remains unexplored. This study investigated whether PRP facilitates functional recovery after SCI by modulating the gut microbiota and SCFA levels. PRP was administered to SCI mice, and motor function was assessed using DeepLabCut-based behavioral analysis. Histological evaluations were performed with H&E, Nissl, and immunofluorescence staining. Microglial activation and inflammation were analyzed by Western blot and immunofluorescence. Gut microbiota composition and SCFAs were examined through 16S rRNA sequencing and targeted metabolite profiling. PRP treatment significantly improved motor function, reduced microglial activation, and attenuated neuroinflammation. These effects were associated with restoration of microbial diversity and elevated butyrate levels. This study highlights the role of the gut-brain axis in neural regeneration and suggests PRP as a promising therapeutic candidate for SCI.
Collapse
Affiliation(s)
- Chang Xue
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea; Jiuhuashan Polygonati Rhizoma Research Institute, Chizhou 247100, China
| | - Mengqi Lu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Jiuhuashan Polygonati Rhizoma Research Institute, Chizhou 247100, China.
| | - Yuwen Qin
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Jiuhuashan Polygonati Rhizoma Research Institute, Chizhou 247100, China.
| | - Xiaoli Zhao
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Jinfeng Yang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Shutong Yuan
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Zhouguang Wang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| | - Namki Cho
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Chengxi Jiang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Jiuhuashan Polygonati Rhizoma Research Institute, Chizhou 247100, China.
| |
Collapse
|
4
|
Situ Y, Zhang P, Zhang C, Jiang A, Zhang N, Zhu L, Mou W, Liu Z, Wong HZH, Zhang J, Cheng Q, Lin A, Luo P. The metabolic dialogue between intratumoural microbes and cancer: implications for immunotherapy. EBioMedicine 2025; 115:105708. [PMID: 40267755 PMCID: PMC12052696 DOI: 10.1016/j.ebiom.2025.105708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 04/02/2025] [Accepted: 04/03/2025] [Indexed: 04/25/2025] Open
Abstract
The tumour microenvironment (TME) exerts a profound influence on cancer progression and treatment outcomes. Recent investigations have elucidated the crucial role of intratumoural microbiota and their metabolites in shaping the TME and modulating anti-tumour immunity. This review critically assesses the influence of intratumoural microbial metabolites on the TME and cancer immunotherapy. We systematically analyse how microbial-derived glucose, amino acid, and lipid metabolites modulate immune cell function, cytokine secretion, and tumour growth. The roles of specific metabolites, including lactate, short-chain fatty acids, bile acids, and tryptophan derivatives, are comprehensively examined in regulating immune responses and tumour progression. Furthermore, we investigate the potential of these metabolites to augment the efficacy of cancer immunotherapies, with particular emphasis on immune checkpoint inhibitors. By delineating the mechanisms through which microbial metabolites influence the TME, this review provides insights into novel microbiome-based therapeutic strategies, thereby highlighting a promising frontier in personalised cancer medicine.
Collapse
Affiliation(s)
- Yingheng Situ
- Donghai County People's Hospital - Jiangnan University Smart Healthcare Joint Laboratory, Donghai County People's Hospital (Affiliated Kangda College of Nanjing Medical University); Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Pengpeng Zhang
- Department of Lung Cancer, Tianjin Lung Cancer Center, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Cangang Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Aimin Jiang
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Nan Zhang
- College of Life Science and Technology, Huazhong University of Science and Technology, China
| | - Lingxuan Zhu
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Weiming Mou
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zaoqu Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Hank Z H Wong
- Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Hunan, China.
| | - Anqi Lin
- Donghai County People's Hospital - Jiangnan University Smart Healthcare Joint Laboratory, Donghai County People's Hospital (Affiliated Kangda College of Nanjing Medical University); Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China.
| | - Peng Luo
- Donghai County People's Hospital - Jiangnan University Smart Healthcare Joint Laboratory, Donghai County People's Hospital (Affiliated Kangda College of Nanjing Medical University); Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China.
| |
Collapse
|
5
|
Lee SH, Han C, Shin C. IUPHAR Review: Microbiota-Gut-Brain Axis and its role in Neuropsychiatric Disorders. Pharmacol Res 2025; 216:107749. [PMID: 40306604 DOI: 10.1016/j.phrs.2025.107749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2025] [Revised: 04/20/2025] [Accepted: 04/25/2025] [Indexed: 05/02/2025]
Abstract
The human gut microbiome, composed of a vast array of microorganisms that have co-evolved with humans, is crucial for the development and function of brain systems. Research has consistently shown bidirectional communication between the gut and the brain through neuronal, endocrine, and immunological, and chemical pathways. Recent neuroscience studies have linked changes in the microbiome and microbial metabolites to various neuropsychiatric disorders such as autism, depression, anxiety, schizophrenia, eating disorders, and neurocognitive disorders. Novel metagenome-wide association studies have confirmed these microbiome variations in large samples and expanded our understanding of the interactions between human genes and the gut microbiome. The causal relationship between gut microbiota and neuropsychiatric disorders is being elucidated through the establishment of large cohort studies incorporating microbiome data and advanced statistical techniques. Ongoing animal and human studies focused on the microbiota-gut-brain axis are promising for developing new prevention and treatment strategies for neuropsychiatric conditions. The scope of these studies has broadened from microbiome-modulating therapies including prebiotics, probiotics, synbiotics and postbiotics to more extensive approaches such as fecal microbiota transplantation. Recent systematic reviews and meta-analyses have strengthened the evidence base for these innovative treatments. Despite extensive research over the past decade, many intriguing aspects still need to be elucidated regarding the role and therapeutic interventions of the microbiota-gut-brain axis in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Seung-Hoon Lee
- Department of Psychiatry, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Changsu Han
- Department of Psychiatry, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Cheolmin Shin
- Department of Psychiatry, Korea University Ansan Hospital, Korea University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
6
|
Mancino S, Boraso M, Galmozzi A, Serafini MM, De Fabiani E, Crestani M, Viviani B. Dose-dependent dual effects of HDAC inhibitors on glial inflammatory response. Sci Rep 2025; 15:12262. [PMID: 40211035 PMCID: PMC11986048 DOI: 10.1038/s41598-025-96241-x] [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: 06/05/2024] [Accepted: 03/26/2025] [Indexed: 04/12/2025] Open
Abstract
Neuroinflammation is defined as a process that includes cellular responses designed to protect the central nervous system from external influences, and it initiates in cases of extreme deviations from homeostasis. While it serves a protective role, excessive immune activation can lead to the release of neurotoxic factors, worsening disease progression. Histone deacetylases (HDACs) have been shown to modulate the expression of inflammatory genes by remodeling chromatin through the process of histone deacetylation. HDAC inhibitors (HDACi) alter histone acetylation and affect the transcription of genes involved in inflammatory pathways, making them promising therapeutic tools for the modulation of a variety of inflammatory diseases. However, their use is limited due to non-specific targeting and contradictory results. This study aimed to reconcile conflicting results and share insights on relevant HDACi in the inflammatory response induced by lipopolysaccharide (LPS), considering different exposure scenarios, cellular models, and associated molecular pathways. Specifically, the study evaluated the dose-dependent effects of two broad-spectrum HDACi, Trichostatin A (TSA) and Suberoylanilide Hydroxamic Acid (SAHA, Vorinostat), alongside selective inhibitors-MS-275 (Entinostat, class I), and MC1568 (class II)-on the expression and release of pro- and anti-inflammatory cytokines. Broad-spectrum HDAC inhibitors TSA and SAHA exhibited dose-dependent modulation of LPS-induced cytokine release. Co-treatment with TSA and LPS enhanced pro-inflammatory cytokines (TNF-α, IL-1β) and decreased IL10 in a dose-dependent manner at lower doses (≤ 10 nM), while high concentrations (100 nM) induced the anti-inflammatory IL-10. Pre-treatment with TSA led to a reduction in TNF-α levels induced by LPS, without affecting IL-1β or IL-10 levels. In contrast, the presence of TSA in LPS-triggered alveolar macrophages resulted in a decline in the production of both pro- and anti-inflammatory cytokine, irrespective of the TSA concentration. SAHA exhibited dual effects, enhancing TNF-α and IL-1β at nanomolar levels but suppressing TNF-α at micromolar doses in co-treated glial cells with LPS. Class-selective inhibitors highlighted distinct HDAC roles on LPS modulation: MS-275 reduced, while MC1568 enhanced, TNF-α release, alongside varied IL-1β and IL-10 modulation. To better understand the dual effects of SAHA, transcriptomic analysis of glial cells was conducted in the presence of LPS and low and high SAHA concentrations (100 nM or 5 µM). This analysis revealed a dose-dependent alteration in gene expression and pathway enrichment associated with cytokine signaling and immune regulation (e.g., JAK-STAT). Altogether, these findings reveal insights on the subtle, dose- and context-dependent role of HDACi in modulating glia inflammation.
Collapse
Affiliation(s)
- Samantha Mancino
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi Di Milano, Milan, Italy.
- Departamento de Bioengenharia E Instituto de Bioengenharia E Biociências, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal.
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal.
| | - Mariaserena Boraso
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi Di Milano, Milan, Italy
| | - Andrea Galmozzi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi Di Milano, Milan, Italy
- Department of Biomolecular Chemistry School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Melania Maria Serafini
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi Di Milano, Milan, Italy
| | - Emma De Fabiani
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi Di Milano, Milan, Italy
| | - Maurizio Crestani
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi Di Milano, Milan, Italy
| | - Barbara Viviani
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi Di Milano, Milan, Italy.
| |
Collapse
|
7
|
Chen X, Wei J, Zhang L, Wang H, Zhang Y, Li Z, Wang X, Liu L, Zhang Y, Zhang T. Association between plasma short-chain fatty acids and inflammation in human immunodeficiency virus-associated neurocognitive disorder: a pilot study. Lipids Health Dis 2025; 24:66. [PMID: 39984934 PMCID: PMC11846350 DOI: 10.1186/s12944-025-02477-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: 01/06/2025] [Accepted: 02/10/2025] [Indexed: 02/23/2025] Open
Abstract
BACKGROUND AND AIMS Short-chain fatty acids (SCFAs), key metabolites produced by gut microbiota, have neuroprotective effects in neurodegenerative diseases by modulating immune responses. However, their role in human immunodeficiency virus (HIV)-associated neurocognitive disorder (HAND) remains largely unexplored. METHODS We recruited HAND patients, HIV Control, and healthy controls (HC). Plasma SCFAs and SCFA-producing gut microbiota were quantified via gas chromatography-mass spectrometry and fecal metagenomic analysis. Inflammatory cytokine levels were measured using liquid chromatography. Receiver operating characteristic (ROC) curves were generated to evaluate the predictive accuracy of SCFAs for HAND. RESULTS Plasma SCFAs were significantly reduced in HAND patients, correlating with a decrease in SCFA-producing gut bacteria, such as Prevotella and its related species. Reduced SCFAs were positively correlated with pro-inflammatory cytokines and cognitive impairment, while being negatively correlated with anti-inflammatory cytokines. ROC curve analysis demonstrated that several SCFAs exhibited strong predictive accuracy for HAND status. CONCLUSIONS SCFAs may influence cognitive function by modulating inflammatory responses, and identifies plasma SCFAs as potential biomarkers and therapeutic targets for HAND. Further investigation is needed to delineate the mechanisms that SCFAs influence HAND pathology.
Collapse
Affiliation(s)
- Xue Chen
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory for HIV/AIDS Research, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Youan Hospital, Beijing Institute of Hepatology, Capital Medical University, Beijing, China
| | - Jiaqi Wei
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory for HIV/AIDS Research, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Ling Zhang
- Beijing Youan Hospital, Beijing Institute of Hepatology, Capital Medical University, Beijing, China
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Hu Wang
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory for HIV/AIDS Research, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yang Zhang
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory for HIV/AIDS Research, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Zhen Li
- Beijing Key Laboratory for HIV/AIDS Research, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Xia Wang
- Beijing Key Laboratory for HIV/AIDS Research, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Lifeng Liu
- Beijing Key Laboratory for HIV/AIDS Research, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yulin Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China.
| | - Tong Zhang
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China.
- Beijing Key Laboratory for HIV/AIDS Research, Beijing Youan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
8
|
Krawczyk A, Sladowska GE, Strzalka-Mrozik B. The Role of the Gut Microbiota in Modulating Signaling Pathways and Oxidative Stress in Glioma Therapies. Cancers (Basel) 2025; 17:719. [PMID: 40075568 PMCID: PMC11899293 DOI: 10.3390/cancers17050719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 02/16/2025] [Accepted: 02/18/2025] [Indexed: 03/14/2025] Open
Abstract
Tumors of the central nervous system (CNS), especially gliomas, pose a significant clinical challenge due to their aggressive nature and limited therapeutic options. Emerging research highlights the critical role of the gut microbiota in regulating CNS health and disease. The composition of the gut microbiota is essential for maintaining CNS homeostasis, as it modulates immune responses, oxidative status, and neuroinflammation. The microbiota-gut-brain axis, a bidirectional communication network, plays a pivotal role in cancer and CNS disease treatment, exerting its influence through neural, endocrine, immunological, and metabolic pathways. Recent studies suggest that the gut microbiota influences the solidification of the tumor microenvironment and that dysbiosis may promote glioma development by modulating systemic inflammation and oxidative stress, which contributes to tumorigenesis and CNS tumor progression. This review interrogates the impact of the gut microbiota on glioma, focusing on critical pathways such as NF-κB, MAPK, PI3K/Akt/mTOR, and Kynurenine/AhR that drive tumor proliferation, immune evasion, and therapy resistance. Furthermore, we explore emerging therapeutic strategies, including probiotics and microbiota-based interventions, which show potential in modulating these pathways and enhancing immunotherapies such as checkpoint inhibitors. By focusing on the multifaceted interactions between the gut microbiota, oxidative stress, and CNS tumors, this review highlights the potential of microbiota-targeted therapies and their manipulation to complement and enhance current treatments.
Collapse
Affiliation(s)
| | | | - Barbara Strzalka-Mrozik
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland; (A.K.); (G.E.S.)
| |
Collapse
|
9
|
Jiao F, Zhou L, Wu Z. The microbiota-gut-brain axis: a potential target in the small-molecule compounds and gene therapeutic strategies for Parkinson's disease. Neurol Sci 2025; 46:561-578. [PMID: 39546084 PMCID: PMC11772541 DOI: 10.1007/s10072-024-07878-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: 08/02/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUNDS Parkinson's disease (PD) is a common neurodegenerative disorder characterized by motor symptoms and non-motor symptoms. It has been found that intestinal issues usually precede motor symptoms. Microorganisms in the gastrointestinal tract can affect central nervous system through the microbiota-gut-brain axis. Accumulating evidence has shown that disturbances in the microbiota-gut-brain axis are linked with PD. Thus, this pathway appears to be a promising therapeutic target for treatment of PD. OBJECTIVES In this review, we mainly described gut dysbiosis in PD and their underlying mechanisms for mediating neuroinflammation and peripheral immune response in PD pathology and futher discussed the potential small-molecule compounds and genic therapeutic strategies targeting the microbiota-gut-brain axis and their applications in PD. CONCLUSIONS Studies have found that some small molecule compounds and alterations of inflammation-related genes can improve the motor and non-motor symptoms of PD by improving the microbiota-gut-brain axis, which may provide potentially beneficial drugs and molecular targets for the therapies of PD.
Collapse
Affiliation(s)
- Fengjuan Jiao
- School of Mental Health, Jining Medical University, No. 45, Jianshe South Road, Jining City, Shandong Province, 272067, P. R. China.
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, Shandong, 272067, P. R. China.
| | - Lincong Zhou
- School of Clinical Medicine, Jining Medical University, Jining, Shandong, 272067, PR China
| | - Zaixin Wu
- School of Clinical Medicine, Jining Medical University, Jining, Shandong, 272067, PR China
| |
Collapse
|
10
|
Shiraz MG, Nielsen J, Widmann J, Chung KHK, Davis TP, Rasmussen C, Scavenius C, Enghild JJ, Martin-Gallausiaux C, Singh Y, Javed I, Otzen DE. Young rat microbiota extracts strongly inhibit fibrillation of α-synuclein and protect neuroblastoma cells and zebrafish against α-synuclein toxicity. Mol Cells 2025; 48:100161. [PMID: 39603509 PMCID: PMC11699742 DOI: 10.1016/j.mocell.2024.100161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/12/2024] [Accepted: 11/19/2024] [Indexed: 11/29/2024] Open
Abstract
The clinical manifestations of Parkinson's disease (PD) are driven by aggregation of α-Synuclein (α-Syn) in the brain. However, there is increasing evidence that PD may be initiated in the gut and thence spread to the brain, eg, via the vagus nerve. Many studies link PD to changes in the gut microbiome, and bacterial amyloid has been shown to stimulate α-Syn aggregation. Yet, we are not aware of any studies reporting on a direct connection between microbiome components and α-Syn aggregation. Here, we report that soluble extract from the gut microbiome of the rats, particularly young rats transgenic for PD, shows a remarkably strong ability to inhibit in vitro α-Syn aggregation and keep it natively unfolded and monomeric. The active component(s) are heat-labile molecule(s) of around 30- to 100-kDa size, which are neither nucleic acid nor lipid. Proteomic analysis identified several proteins whose concentrations in different rat samples correlated with the samples' anti-inhibitory activity, while a subsequent pull-down assay linked the protein chaperone DnaK with the inhibitory activity of young rat's microbiome, confirmed in subsequent in vitro assays. Remarkably, the microbiome extracts also protected neuroblastoma SH-SY5Y cells and zebrafish embryos against α-Syn toxicity. Our study sheds new light on the gut microbiome as a potential source of protection against PD and opens up for new microbiome-based therapeutic strategies.
Collapse
Affiliation(s)
- Mohaddeseh Ghorbani Shiraz
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus Centrum, Denmark
| | - Janni Nielsen
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus Centrum, Denmark
| | - Jeremias Widmann
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus Centrum, Denmark
| | - Ka Hang Karen Chung
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Thomas Paul Davis
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Casper Rasmussen
- Department of Molecular Biology and Genetics, Aarhus University, Universitetsbyen 81, 8000 Aarhus Centrum, Denmark
| | - Carsten Scavenius
- Department of Molecular Biology and Genetics, Aarhus University, Universitetsbyen 81, 8000 Aarhus Centrum, Denmark
| | - Jan J Enghild
- Department of Molecular Biology and Genetics, Aarhus University, Universitetsbyen 81, 8000 Aarhus Centrum, Denmark
| | - Camille Martin-Gallausiaux
- Evolutionary Biology of the Microbial Cell - Biologie Evolutive de la Cellule Microbienne Institut Pasteur, 28 Rue du Docteur Roux, Paris 75724 Cedex 15, France
| | - Yogesh Singh
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Calwerstraße 7, 72076 Tübingen, Germany; NGS Competence Centre Tübingen (NCCT), University of Tübingen, Calwerstraße 7, 72076 Tübingen, Germany
| | - Ibrahim Javed
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Daniel E Otzen
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus Centrum, Denmark; Department of Molecular Biology and Genetics, Aarhus University, Universitetsbyen 81, 8000 Aarhus Centrum, Denmark.
| |
Collapse
|
11
|
Yang S, Wang Y, Wu Z, Wang D, Zhang X, Hu S, Zhang Q, Bu Y, Liu C, Huang C, Yang C. Increased levels of Escherichia-Shigella and Klebsiella in the gut contribute to the responsivity of placebo analgesia. Neuropharmacology 2024; 261:110168. [PMID: 39332670 DOI: 10.1016/j.neuropharm.2024.110168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 07/30/2024] [Accepted: 09/24/2024] [Indexed: 09/29/2024]
Abstract
Placebo analgesia is observed in both humans and animals. Given the complexity of placebo analgesia involving a variety of neurobiological, psychological, and psychosocial processes, further investigation into its underlying mechanisms is essential. Gut microbiota has been implicated in the responsivity of placebo analgesia, but its precise role remains unknown and warrants further investigations. Here, we conducted a conditioning training model with chronic inflammatory pain induced by complete Freund's adjuvant (CFA) in mice, associating parecoxib with different cues. Hierarchical clustering analysis of placebo analgesia behaviors was employed to classify mice into responders and non-responders phenotypes. Approximately 40% of CFA mice undergoing conditioning training exhibited placebo analgesia. Notably, placebo analgesia responders displayed reduced anxiety-like behaviors. 16S rRNA results revealed a distinct composition of gut microbiota composition among the control, placebo analgesia non-responders and responders groups. Notably, levels of Escherichia Shigella and Klebsiella in the gut were increased considerably in the placebo analgesia responders as compared to both control and non-responders groups. In conclusion, placebo analgesia responders demonstrated marked analgesia, reduced anxiety-like behaviors, and increased levels of Escherichia-Shigella and Klebsiella, implying a potential linkage between gut microbiota and placebo analgesia.
Collapse
Affiliation(s)
- Siqi Yang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yuanyuan Wang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Zifeng Wu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Di Wang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Xinying Zhang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Suwan Hu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Qi Zhang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yuchen Bu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Cunming Liu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Chaoli Huang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Chun Yang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
12
|
Jørgensen KS, Pedersen SS, Hjorth SA, Billestrup N, Prause M. Protection of beta cells against cytokine-induced apoptosis by the gut microbial metabolite butyrate. FEBS J 2024. [PMID: 39569473 DOI: 10.1111/febs.17334] [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: 07/04/2024] [Revised: 08/28/2024] [Accepted: 11/12/2024] [Indexed: 11/22/2024]
Abstract
Type 1 diabetes (T1D) is characterized by immune cell infiltration in the islets of Langerhans, leading to the destruction of insulin-producing beta cells. This destruction is driven by secreted cytokines and cytotoxic T cells inducing apoptosis in beta cells. Butyrate, a metabolite produced by the gut microbiota, has been shown to have various health benefits, including anti-inflammatory and anti-diabetic effects. In this study, we investigated the potential protective effects of butyrate on cytokine-induced apoptosis in beta cells and explored the underlying mechanisms. Insulin-secreting INS-1E cells and isolated mouse islets were treated with interleukin-1beta (IL-1β) or a combination of IL-1β and interferon-gamma (IFN-γ) in the presence or absence of butyrate. We analyzed apoptosis, nitric oxide (NO) levels, expression of stress-related genes, and immune cell migration. Our results demonstrated that butyrate significantly attenuated cytokine-induced apoptosis in both INS-1E cells and mouse islets, accompanied by a reduction in NO levels. Butyrate also decreased the expression of endoplasmic reticulum (ER) stress markers such as Chop, phosphorylated eIF2α and Atf4, as well as some pro-apoptotic genes including Dp5 and Puma. Butyrate reduced the cytokine-induced expression of the chemokine genes Cxcl1 and Cxcl10 in mouse islets, as well as the chemotactic activity of THP-1 monocytes toward conditioned media from IL-1β-exposed islets. In conclusion, these findings indicate that butyrate protects beta cells from cytokine-induced apoptosis and ER stress, suggesting its potential as a therapeutic agent to prevent beta cell destruction in T1D.
Collapse
Affiliation(s)
- Kasper Suhr Jørgensen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Signe Schultz Pedersen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Siv Annegrethe Hjorth
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Nils Billestrup
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Michala Prause
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| |
Collapse
|
13
|
Gold A, Kaye S, Gao J, Zhu J. Propionate Decreases Microglial Activation but Impairs Phagocytic Capacity in Response to Aggregated Fibrillar Amyloid Beta Protein. ACS Chem Neurosci 2024; 15:4010-4020. [PMID: 39394077 DOI: 10.1021/acschemneuro.4c00370] [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] [Indexed: 10/13/2024] Open
Abstract
Microglia, the innate immune cell of the brain, are a principal player in Alzheimer's disease (AD) pathogenesis. Their surveillance of the brain leads to interaction with the protein aggregates that drive AD pathogenesis, most notably Amyloid Beta (Aβ). Microglia attempt to clear and degrade Aβ using phagocytic machinery, spurring damaging neuroinflammation in the process. Thus, modulation of the microglial response to Aβ is crucial in mitigating AD pathophysiology. SCFAs, microbial byproducts of dietary fiber fermentation, are blood-brain barrier permeable molecules that have recently been shown to modulate microglial function. It is unclear whether propionate, one representative SCFA, has beneficial or detrimental effects on microglia in AD. Thus, we investigated its impact on microglial Aβ response in vitro. Using a multiomics approach, we characterized the transcriptomic, metabolomic, and lipidomic responses of immortalized murine microglia following 1 h of Aβ stimulation, as well as characterizing Aβ phagocytosis and secretion of reactive nitrogen species. Propionate blunted the early inflammatory response driven by Aβ, downregulating the expression of many Aβ-stimulated immune genes, including those regulating inflammation, the immune complement system, and chemotaxis. Further, it reduced the expression of Apoe and inflammation-promoting Aβ-binding scavenger receptors such as Cd36 and Msr1 in favor of inflammation-dampening Lpl, although this led to impaired phagocytosis. Finally, propionate shifted microglial metabolism, altering phospholipid composition and diverting arginine metabolism, resulting in decreased nitric oxide production. Altogether, our data demonstrate a modulatory role of propionate on microglia that may dampen immune activation in response to Aβ, although at the expense of phagocytic capacity.
Collapse
Affiliation(s)
- Andrew Gold
- Human Nutrition Program and James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
| | - Sarah Kaye
- Department of Neuroscience, The Ohio State University, Columbus, Ohio 43210, United States
| | - Jie Gao
- Department of Neuroscience, The Ohio State University, Columbus, Ohio 43210, United States
| | - Jiangjiang Zhu
- Human Nutrition Program and James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
14
|
Ding L, Wang J, Qiu S, Ren Z, Li Y, An P. Bioinformatics Approach to Identify the Pathogenetic Link of Gut Microbiota-Derived Short-Chain Fatty Acids and Ischemic Stroke. Mol Neurobiol 2024; 61:9478-9490. [PMID: 38649659 PMCID: PMC11496340 DOI: 10.1007/s12035-024-04176-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 04/11/2024] [Indexed: 04/25/2024]
Abstract
Stroke is a life-threatening condition that impairs the arteries and causes neurological impairment. The incidence of stroke is increasing year by year with the arrival of the aging population. Thus, there is an urgent need for early stroke diagnosis. Short-chain fatty acids (SCFAs) can modulate the central nervous system and directly and indirectly impact behavioral and cognitive functions. This study aimed to investigate the connection between SCFA metabolism and stroke development via bioinformatic analysis. Initially, the Gene Set Enrichment Analysis (GSEA) and immune cell infiltration analysis were performed based on RNA data from stroke patients to comprehend the mechanisms governing stroke pathogenesis. The functional analysis, including Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Protein-Protein Interaction (PPI), was performed based on the Differentially Expressed Gene (DEG) selected by the limma package. 1220 SCFA metabolism-related genes screened from Genecards databases were intersected with 242 genes in main modules determined by Weighted Gene Co-Expression Network Analysis (WGCNA), and the final 10 SCFA key genes were obtained. GO analysis revealed that these genes were involved in immune response processes. Through lasso regression analyses, we established a stroke early diagnosis model and selected 6 genes with diagnostic value. The genes were validated by the area under curve (AUC) values and had a relatively good diagnostic performance. Finally, 4 potential therapeutic drugs targeting these genes were predicted using the Drug Signatures Database (DSigDB) via Enrichr. In conclusion, this paper analyzes the involvement of SCFAs in the complex gut-brain axis mechanism, which contributes to developing new targets for treating central nervous system diseases and provides new ideas for early ischemic stroke diagnosis.
Collapse
Affiliation(s)
- Liang Ding
- Department of Traditional Chinese Medicine, Qingdao Third People's Hospital, Qingdao City, Shandong Province, China
| | - Jianing Wang
- Neurology Department, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao City, Shandong Province, China
| | - Sha Qiu
- Department of Traditional Chinese Medicine, Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Hospital), Qingdao City, Shandong Province, China
| | - Zhizhen Ren
- Department of Traditional Chinese Medicine, Community Health Service Center of Shi'nan District in Qingdao, Qingdao City, Shandong Province, China
| | - Yuantao Li
- Acupuncture and Moxibustion Department, Qingdao Third People's Hospital, Qingdao City, Shandong Province, China
| | - Pengpeng An
- Emergency Internal Medicine Department, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao City, Shandong Province, China.
| |
Collapse
|
15
|
Kang JW, Vemuganti V, Kuehn JF, Ulland TK, Rey FE, Bendlin BB. Gut microbial metabolism in Alzheimer's disease and related dementias. Neurotherapeutics 2024; 21:e00470. [PMID: 39462700 PMCID: PMC11585892 DOI: 10.1016/j.neurot.2024.e00470] [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: 07/29/2024] [Revised: 09/30/2024] [Accepted: 10/04/2024] [Indexed: 10/29/2024] Open
Abstract
Multiple studies over the last decade have established that Alzheimer's disease and related dementias (ADRD) are associated with changes in the gut microbiome. These alterations in organismal composition result in changes in the abundances of functions encoded by the microbial community, including metabolic capabilities, which likely impact host disease mechanisms. Gut microbes access dietary components and other molecules made by the host and produce metabolites that can enter circulation and cross the blood-brain barrier (BBB). In recent years, several microbial metabolites have been associated with or have been shown to influence host pathways relevant to ADRD pathology. These include short chain fatty acids, secondary bile acids, tryptophan derivatives (such as kynurenine, serotonin, tryptamine, and indoles), and trimethylamine/trimethylamine N-oxide. Notably, some of these metabolites cross the BBB and can have various effects on the brain, including modulating the release of neurotransmitters and neuronal function, inducing oxidative stress and inflammation, and impacting synaptic function. Microbial metabolites can also impact the central nervous system through immune, enteroendocrine, and enteric nervous system pathways, these perturbations in turn impact the gut barrier function and peripheral immune responses, as well as the BBB integrity, neuronal homeostasis and neurogenesis, and glial cell maturation and activation. This review examines the evidence supporting the notion that ADRD is influenced by gut microbiota and its metabolites. The potential therapeutic advantages of microbial metabolites for preventing and treating ADRD are also discussed, highlighting their potential role in developing new treatments.
Collapse
Affiliation(s)
- Jea Woo Kang
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Vaibhav Vemuganti
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Jessamine F Kuehn
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Tyler K Ulland
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Federico E Rey
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Barbara B Bendlin
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
16
|
Vajdi M, Khorvash F, Askari G. A randomized, double-blind, placebo-controlled parallel trial to test the effect of inulin supplementation on migraine headache characteristics, quality of life and mental health symptoms in women with migraine. Food Funct 2024; 15:10088-10098. [PMID: 39291634 DOI: 10.1039/d4fo02796e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Migraine is a complex neurovascular disorder characterized by recurrent headache attacks that are often accompanied by symptoms such as vomiting, nausea, and sensitivity to sound or light. Preventing migraine attacks is highly important. Recent research has indicated that alterations in gut microbiota may influence the underlying mechanisms of migraines. This study aimed to investigate the effects of inulin supplementation on migraine headache characteristics, quality of life (QOL), and mental health symptoms in women with migraines. In a randomized double-blind placebo-controlled trial, 80 women with migraines aged 20 to 50 years were randomly assigned to receive 10 g day-1 of inulin or a placebo supplement for 12 weeks. Severity, frequency, and duration of migraine attacks, as well as depression, anxiety, stress, QOL, and headache impact test (HIT-6) scores, were examined at the start of the study and after 12 weeks of intervention. In this study, the primary outcome focused on the frequency of headache attacks, while secondary outcomes encompassed the duration and severity of headache attacks, QOL, and mental health. There was a significant reduction in severity (-1.95 vs. -0.84, P = 0.004), duration (-6.95 vs. -2.05, P = 0.023), frequency (-2.09 vs. -0.37, P < 0.001), and HIT-6 score (-10.30 vs. -6.52, P < 0.023) in the inulin group compared with the control. Inulin supplementation improved mental health symptoms, including depression (-4.47 vs. -1.45, P < 0.001), anxiety (-4.37 vs. -0.70, P < 0.001), and stress (-4.40 vs. -1.50, P < 0.001). However, no significant difference was observed between the two groups regarding changes in QOL score. This study provides evidence supporting the beneficial effects of inulin supplement on migraine symptoms and mental health status in women with migraines. Further studies are necessary to confirm these findings. Trial registration: Iranian Registry of Clinical Trials (https://www.irct.ir) (ID: IRCT20121216011763N58).
Collapse
Affiliation(s)
- Mahdi Vajdi
- Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Fariborz Khorvash
- Neurology Research Center, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Gholamreza Askari
- Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
17
|
Gao Y, Yao Q, Meng L, Wang J, Zheng N. Double-side role of short chain fatty acids on host health via the gut-organ axes. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 18:322-339. [PMID: 39290857 PMCID: PMC11406094 DOI: 10.1016/j.aninu.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 01/29/2024] [Accepted: 05/14/2024] [Indexed: 09/19/2024]
Abstract
Short chain fatty acids (SCFA) exist in dietary foods and are produced by the fermentation of gut microbiota, and are considered an important element for regulating host health. Through blood circulation, SCFA produced in the gut and obtained from foods have an impact on the intestinal health as well as vital organs of the host. It has been recognized that the gut is the "vital organ" in the host. As the gut microbial metabolites, SCFA could create an "axis" connecting the gut and to other organs. Therefore, the "gut-organ axes" have become a focus of research in recent years to analyze organism health. In this review, we summarized the sources, absorption properties, and the function of SCFA in both gut and other peripheral tissues (brain, kidney, liver, lung, bone and cardiovascular) in the way of "gut-organ axes". Short chain fatty acids exert both beneficial and pathological role in gut and other organs in various ways, in which the beneficial effects are more pronounced. In addition, the beneficial effects are reflected in both preventive and therapeutic effects. More importantly, the mechanisms behinds the gut and other tissues provided insight into the function of SCFA, assisting in the development of novel preventive and therapeutic strategies for maintaining the host health.
Collapse
Affiliation(s)
- Yanan Gao
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Milk and Milk Products Inspection Center of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Qianqian Yao
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Milk and Milk Products Inspection Center of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Department of Food Science, Faculty of Veterinary Medicine, University of Liège, Liège 4000, Belgium
| | - Lu Meng
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Milk and Milk Products Inspection Center of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Jiaqi Wang
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Milk and Milk Products Inspection Center of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Nan Zheng
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Milk and Milk Products Inspection Center of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| |
Collapse
|
18
|
Song L, Jiang W, Lin H, Yu J, Liu K, Zheng R. Post-translational modifications in sepsis-induced organ dysfunction: mechanisms and implications. Front Immunol 2024; 15:1461051. [PMID: 39234245 PMCID: PMC11371574 DOI: 10.3389/fimmu.2024.1461051] [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: 07/07/2024] [Accepted: 08/05/2024] [Indexed: 09/06/2024] Open
Abstract
As a grave and highly lethal clinical challenge, sepsis, along with its consequent multiorgan dysfunction, affects millions of people worldwide. Sepsis is a complex syndrome caused by a dysregulated host response to infection, leading to fatal organ dysfunction. An increasing body of evidence suggests that the pathogenesis of sepsis is both intricate and rapid and involves various cellular responses and signal transductions mediated by post-translational modifications (PTMs). Hence, a comprehensive understanding of the mechanisms and functions of PTMs within regulatory networks is imperative for understanding the pathological processes, diagnosis, progression, and treatment of sepsis. In this review, we provide an exhaustive and comprehensive summary of the relationship between PTMs and sepsis-induced organ dysfunction. Furthermore, we explored the potential applications of PTMs in the treatment of sepsis, offering a forward-looking perspective on the understanding of infectious diseases.
Collapse
Affiliation(s)
- Lin Song
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
- Intensive Care Unit, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Wei Jiang
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
- Intensive Care Unit, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Hua Lin
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
- Intensive Care Unit, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Jiangquan Yu
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
- Intensive Care Unit, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Ke Liu
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Ruiqiang Zheng
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
- Intensive Care Unit, Northern Jiangsu People's Hospital, Yangzhou, China
| |
Collapse
|
19
|
Ling Z, Lan Z, Cheng Y, Liu X, Li Z, Yu Y, Wang Y, Shao L, Zhu Z, Gao J, Lei W, Ding W, Liao R. Altered gut microbiota and systemic immunity in Chinese patients with schizophrenia comorbid with metabolic syndrome. J Transl Med 2024; 22:729. [PMID: 39103909 PMCID: PMC11302365 DOI: 10.1186/s12967-024-05533-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 07/23/2024] [Indexed: 08/07/2024] Open
Abstract
BACKGROUND Metabolic syndrome (MetS) is highly prevalent in individuals with schizophrenia (SZ), leading to negative consequences like premature mortality. Gut dysbiosis, which refers to an imbalance of the microbiota, and chronic inflammation are associated with both SZ and MetS. However, the relationship between gut dysbiosis, host immunological dysfunction, and SZ comorbid with MetS (SZ-MetS) remains unclear. This study aims to explore alterations in gut microbiota and their correlation with immune dysfunction in SZ-MetS, offering new insights into its pathogenesis. METHODS AND RESULTS We enrolled 114 Chinese patients with SZ-MetS and 111 age-matched healthy controls from Zhejiang, China, to investigate fecal microbiota using Illumina MiSeq sequencing targeting 16 S rRNA gene V3-V4 hypervariable regions. Host immune responses were assessed using the Bio-Plex Pro Human Cytokine 27-Plex Assay to examine cytokine profiles. In SZ-MetS, we observed decreased bacterial α-diversity and significant differences in β-diversity. LEfSe analysis identified enriched acetate-producing genera (Megamonas and Lactobacillus), and decreased butyrate-producing bacteria (Subdoligranulum, and Faecalibacterium) in SZ-MetS. These altered genera correlated with body mass index, the severity of symptoms (as measured by the Scale for Assessment of Positive Symptoms and Scale for Assessment of Negative Symptoms), and triglyceride levels. Altered bacterial metabolic pathways related to lipopolysaccharide biosynthesis, lipid metabolism, and various amino acid metabolism were also found. Additionally, SZ-MetS exhibited immunological dysfunction with increased pro-inflammatory cytokines, which correlated with the differential genera. CONCLUSION These findings suggested that gut microbiota dysbiosis and immune dysfunction play a vital role in SZ-MetS development, highlighting potential therapeutic approaches targeting the gut microbiota. While these therapies show promise, further mechanistic studies are needed to fully understand their efficacy and safety before clinical implementation.
Collapse
Affiliation(s)
- Zongxin Ling
- 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, Zhejiang, 310003, China.
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong, 250000, China.
| | - Zhiyong Lan
- Department of Psychiatry, Quzhou Third Hospital, Quzhou, Zhejiang, 324003, China
| | - Yiwen Cheng
- 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, Zhejiang, 310003, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong, 250000, China
| | - Xia Liu
- Department of Intensive Care Unit, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Zhimeng Li
- Department of Psychiatry, Quzhou Third Hospital, Quzhou, Zhejiang, 324003, China
| | - Ying Yu
- Department of Psychiatry, Quzhou Third Hospital, Quzhou, Zhejiang, 324003, China
| | - Yuwei Wang
- Department of Psychiatry, Quzhou Third Hospital, Quzhou, Zhejiang, 324003, China
| | - Li Shao
- School of Clinical Medicine, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, 310015, China
| | - Zhangcheng Zhu
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Jie Gao
- 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, Zhejiang, 310003, China
| | - Wenhui Lei
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong, 250000, China
- Department of Basic Medicine, Shandong First Medical University, Jinan, Shandong, 250000, China
| | - Wenwen Ding
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, China
| | - Rongxian Liao
- Department of Psychiatry, Quzhou Third Hospital, Quzhou, Zhejiang, 324003, China.
| |
Collapse
|
20
|
Sciarretta F, Zaccaria F, Ninni A, Ceci V, Turchi R, Apolloni S, Milani M, Della Valle I, Tiberi M, Chiurchiù V, D'Ambrosi N, Pedretti S, Mitro N, Volontè C, Amadio S, Aquilano K, Lettieri-Barbato D. Frataxin deficiency shifts metabolism to promote reactive microglia via glucose catabolism. Life Sci Alliance 2024; 7:e202402609. [PMID: 38631900 PMCID: PMC11024345 DOI: 10.26508/lsa.202402609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 04/05/2024] [Accepted: 04/08/2024] [Indexed: 04/19/2024] Open
Abstract
Immunometabolism investigates the intricate relationship between the immune system and cellular metabolism. This study delves into the consequences of mitochondrial frataxin (FXN) depletion, the primary cause of Friedreich's ataxia (FRDA), a debilitating neurodegenerative condition characterized by impaired coordination and muscle control. By using single-cell RNA sequencing, we have identified distinct cellular clusters within the cerebellum of an FRDA mouse model, emphasizing a significant loss in the homeostatic response of microglial cells lacking FXN. Remarkably, these microglia deficient in FXN display heightened reactive responses to inflammatory stimuli. Furthermore, our metabolomic analyses reveal a shift towards glycolysis and itaconate production in these cells. Remarkably, treatment with butyrate counteracts these immunometabolic changes, triggering an antioxidant response via the itaconate-Nrf2-GSH pathways and suppressing the expression of inflammatory genes. Furthermore, we identify Hcar2 (GPR109A) as a mediator involved in restoring the homeostasis of microglia without FXN. Motor function tests conducted on FRDA mice underscore the neuroprotective attributes of butyrate supplementation, enhancing neuromotor performance. In conclusion, our findings elucidate the role of disrupted homeostatic function in cerebellar microglia in the pathogenesis of FRDA. Moreover, they underscore the potential of butyrate to mitigate inflammatory gene expression, correct metabolic imbalances, and improve neuromotor capabilities in FRDA.
Collapse
Affiliation(s)
- Francesca Sciarretta
- Department Biology, University of Rome Tor Vergata, Rome, Italy
- IRCCS Fondazione Bietti, Rome, Italy
| | - Fabio Zaccaria
- Department Biology, University of Rome Tor Vergata, Rome, Italy
- PhD Program in Evolutionary Biology and Ecology, University of Rome Tor Vergata, Rome, Italy
- IRCCS Fondazione Bietti, Rome, Italy
| | - Andrea Ninni
- Department Biology, University of Rome Tor Vergata, Rome, Italy
- PhD Program in Evolutionary Biology and Ecology, University of Rome Tor Vergata, Rome, Italy
- IRCCS Fondazione Bietti, Rome, Italy
| | - Veronica Ceci
- Department Biology, University of Rome Tor Vergata, Rome, Italy
- PhD Program in Evolutionary Biology and Ecology, University of Rome Tor Vergata, Rome, Italy
| | - Riccardo Turchi
- Department Biology, University of Rome Tor Vergata, Rome, Italy
| | - Savina Apolloni
- Department Biology, University of Rome Tor Vergata, Rome, Italy
| | - Martina Milani
- Department Biology, University of Rome Tor Vergata, Rome, Italy
- PhD Program in Cellular and Molecular Biology, University of Rome Tor Vergata, Rome, Italy
| | - Ilaria Della Valle
- Department Biology, University of Rome Tor Vergata, Rome, Italy
- PhD Program in Cellular and Molecular Biology, University of Rome Tor Vergata, Rome, Italy
| | - Marta Tiberi
- Laboratory of Resolution of Neuroinflammation, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Valerio Chiurchiù
- Laboratory of Resolution of Neuroinflammation, IRCCS Santa Lucia Foundation, Rome, Italy
- Institute of Translational Pharmacology, IFT-CNR, Rome, Italy
| | - Nadia D'Ambrosi
- Department Biology, University of Rome Tor Vergata, Rome, Italy
| | - Silvia Pedretti
- DiSFeB, Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Milano, Italy
| | - Nico Mitro
- DiSFeB, Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Milano, Italy
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Cinzia Volontè
- National Research Council, Institute for Systems Analysis and Computer Science "A. Ruberti", Rome, Italy
- Santa Lucia Foundation IRCCS, Experimental Neuroscience and Neurological Disease Models, Rome, Italy
| | - Susanna Amadio
- Santa Lucia Foundation IRCCS, Experimental Neuroscience and Neurological Disease Models, Rome, Italy
| | - Katia Aquilano
- Department Biology, University of Rome Tor Vergata, Rome, Italy
| | - Daniele Lettieri-Barbato
- Department Biology, University of Rome Tor Vergata, Rome, Italy
- IRCCS Fondazione Bietti, Rome, Italy
| |
Collapse
|
21
|
Guarnieri L, Bosco F, Leo A, Citraro R, Palma E, De Sarro G, Mollace V. Impact of micronutrients and nutraceuticals on cognitive function and performance in Alzheimer's disease. Ageing Res Rev 2024; 95:102210. [PMID: 38296163 DOI: 10.1016/j.arr.2024.102210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 01/10/2024] [Accepted: 01/25/2024] [Indexed: 02/13/2024]
Abstract
Alzheimer's disease (AD) is a major global health problem today and is the most common form of dementia. AD is characterized by the formation of β-amyloid (Aβ) plaques and neurofibrillary clusters, leading to decreased brain acetylcholine levels in the brain. Another mechanism underlying the pathogenesis of AD is the abnormal phosphorylation of tau protein that accumulates at the level of neurofibrillary aggregates, and the areas most affected by this pathological process are usually the cholinergic neurons in cortical, subcortical, and hippocampal areas. These effects result in decreased cognitive function, brain atrophy, and neuronal death. Malnutrition and weight loss are the most frequent manifestations of AD, and these are also associated with greater cognitive decline. Several studies have confirmed that a balanced low-calorie diet and proper nutritional intake may be considered important factors in counteracting or slowing the progression of AD, whereas a high-fat or hypercholesterolemic diet predisposes to an increased risk of developing AD. Especially, fruits, vegetables, antioxidants, vitamins, polyunsaturated fatty acids, and micronutrients supplementation exert positive effects on aging-related changes in the brain due to their antioxidant, anti-inflammatory, and radical scavenging properties. The purpose of this review is to summarize some possible nutritional factors that may contribute to the progression or prevention of AD, understand the role that nutrition plays in the formation of Aβ plaques typical of this neurodegenerative disease, to identify some potential therapeutic strategies that may involve some natural compounds, in delaying the progression of the disease.
Collapse
Affiliation(s)
- Lorenza Guarnieri
- Section of Pharmacology, Science of Health Department, School of Medicine, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy
| | - Francesca Bosco
- Section of Pharmacology, Science of Health Department, School of Medicine, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy.
| | - Antonio Leo
- Section of Pharmacology, Science of Health Department, School of Medicine, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy; Research Center FAS@UMG, Department of Health Science, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy
| | - Rita Citraro
- Section of Pharmacology, Science of Health Department, School of Medicine, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy; Research Center FAS@UMG, Department of Health Science, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy
| | - Ernesto Palma
- Department of Health Sciences, Institute of Research for Food Safety and Health (IRC-FSH), University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy
| | - Giovambattista De Sarro
- Section of Pharmacology, Science of Health Department, School of Medicine, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy; Research Center FAS@UMG, Department of Health Science, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy
| | - Vincenzo Mollace
- Department of Health Sciences, Institute of Research for Food Safety and Health (IRC-FSH), University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy
| |
Collapse
|
22
|
Xu Y, Wen L, Tang Y, Zhao Z, Xu M, Wang T, Chen Z. Sodium butyrate activates the K ATP channels to regulate the mechanism of Parkinson's disease microglia model inflammation. Immun Inflamm Dis 2024; 12:e1194. [PMID: 38501544 PMCID: PMC10949401 DOI: 10.1002/iid3.1194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 01/31/2024] [Accepted: 02/05/2024] [Indexed: 03/20/2024] Open
Abstract
BACKGROUND Parkinson's disease (PD) is a common neurodegenerative disorder. Microglia-mediated neuroinflammation has emerged as an involving mechanism at the initiation and development of PD. Activation of adenosine triphosphate (ATP)-sensitive potassium (KATP ) channels can protect dopaminergic neurons from damage. Sodium butyrate (NaB) shows anti-inflammatory and neuroprotective effects in some animal models of brain injury and regulates the KATP channels in islet β cells. In this study, we aimed to verify the anti-inflammatory effect of NaB on PD and further explored potential molecular mechanisms. METHODS We established an in vitro PD model in BV2 cells using 1-methyl-4-phenylpyridinium (MPP+ ). The effects of MPP+ and NaB on BV2 cell viability were detected by cell counting kit-8 assays. The morphology of BV2 cells with or without MPP+ treatment was imaged via an optical microscope. The expression of Iba-1 was examined by the immunofluorescence staining. The intracellular ATP content was estimated through the colorimetric method, and Griess assay was conducted to measure the nitric oxide production. The expression levels of pro-inflammatory cytokines and KATP channel subunits were evaluated by reverse transcription-quantitative polymerase chain reaction and western blot analysis. RESULTS NaB (5 mM) activated the KATP channels through elevating Kir6.1 and Kir6.1 expression in MPP+ -challenged BV2 cells. Both NaB and pinacidil (a KATP opener) suppressed the MPP+ -induced activation of BV2 cells and reduced the production of nitrite and pro-inflammatory cytokines in MPP+ -challenged BV2 cells. CONCLUSION NaB treatment alleviates the MPP+ -induced inflammatory responses in microglia via activation of KATP channels.
Collapse
Affiliation(s)
- Ye Xu
- Department of NeurologyThe First Affiliated Hospital of Hainan Medical UniversityHaikouHainanChina
| | - Laofu Wen
- Department of NeurologyThe First Affiliated Hospital of Hainan Medical UniversityHaikouHainanChina
| | - Yunyi Tang
- Department of NeurologyThe First Affiliated Hospital of Hainan Medical UniversityHaikouHainanChina
| | - Zhenqiang Zhao
- Department of NeurologyThe First Affiliated Hospital of Hainan Medical UniversityHaikouHainanChina
| | - Miaojing Xu
- Department of NeurologyThe First Affiliated Hospital of Hainan Medical UniversityHaikouHainanChina
- Department of Neurology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Tan Wang
- Department of NeurologyThe First Affiliated Hospital of Hainan Medical UniversityHaikouHainanChina
| | - Zhibin Chen
- Department of NeurologyThe First Affiliated Hospital of Hainan Medical UniversityHaikouHainanChina
| |
Collapse
|
23
|
Lan Z, Tang X, Lu M, Hu Z, Tang Z. The role of short-chain fatty acids in central nervous system diseases: A bibliometric and visualized analysis with future directions. Heliyon 2024; 10:e26377. [PMID: 38434086 PMCID: PMC10906301 DOI: 10.1016/j.heliyon.2024.e26377] [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: 11/06/2023] [Revised: 02/11/2024] [Accepted: 02/12/2024] [Indexed: 03/05/2024] Open
Abstract
Background Short-chain fatty acids (SCFAs) are thought to play a key role in the microbe-gut-brain axis and involve in the pathogenesis of a variety of neurological diseases. This study aimed to identify research hotspots and evolution trends in SCFAs in central nervous diseases (CNS) and examine current research trends. Methods The bibliometric analysis was performed using CiteSpace, and the results were visualized via network maps. Results From 2002 to 2022, 480 publications in the database met the criteria. On the country level, China produced the highest number of publications, while the United States had the highest centrality. On the institutional level, University College Cork contributed to the most publications, and John F. Cryan from this university was the key researcher with considerable academic influence. The article, the role of short-chain fatty acids in microbiota-gut-brain, written by Boushra Dalile et al., in 2019 was the most cited article. Furthermore, the journal Nutrients had the maximum number of publications, while Plos One was the most cited journal. "Gut microbiome", "SCFAs", and "central nervous system" were the three most frequent keywords. Among them, SCFAs had the highest centrality. "Animal model" was the keyword with the highest burst strength, with the latest burst keywords being "social behavior", "pathogenesis", and "insulin sensitive". In addition, the research topics on SCFAs in CNS diseases from 2002 to 2022 mainly focused on following aspects: SCFAs plays a key role in microbe-gut-brain crosstalk; The classification and definition of SCFAs in the field of CNS; Several CNS diseases that are closely related to SCFAs research; Mechanism and translational studies of SCFAs in the CNS diseases. And the hotspots over the past 5 years have gradually increased the attention to the therapeutic potential of SCFAs in the CNS diseases. Conclusion The research of SCFAs in CNS diseases is attracting growing attention. However, there is a lack of cooperation between countries and institutions, and additional measures are required to promote cooperation. The current evidence for an association between SCFAs and CNS diseases is preliminary and more work is needed to pinpoint the precise mechanism. Moreover, large-scale clinical trials are needed in the future to define the therapeutic potential of SCFAs in CNS diseases.
Collapse
Affiliation(s)
- Ziwei Lan
- Department of Neurology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Xiangqi Tang
- Department of Neurology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Ming Lu
- Hunan Provincial Key Laboratory of Neurorestoratology, The Second Affiliated Hospital, Hunan Normal University, Changsha, 410003, Hunan, China
| | - Zhiping Hu
- Department of Neurology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Zhenchu Tang
- Department of Neurology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| |
Collapse
|
24
|
Zaki RM, Ramasamy K, Ahmad Alwi NA, Mohd Yusoff R, Lim SM. Pediococcus pentosaceus LAB6- and Lactiplantibacillus plantarum LAB12-Derived Cell Free Supernatant Inhibited RhoA Activation and Reduced Amyloid-Β In Vitro. Probiotics Antimicrob Proteins 2024; 16:62-75. [PMID: 36443559 DOI: 10.1007/s12602-022-10009-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/14/2022] [Indexed: 11/30/2022]
Abstract
Alzheimer's disease (AD) is characterized by aggregation of amyloid beta (Aβ) plaque. RhoA may serve as a potential target for prevention against AD given its role in the amyloidogenic pathway. The recent emergence of the gut-brain axis has linked lactic acid bacteria (LAB) to neuroprotection against AD. This study assessed the importance of RhoA inhibition in mediating the neuroprotective potential of LAB. To this end, de Man, Rogosa and Sharpe (MRS) broth fermented by lactobacilli or pediococci were tested against SK-N-SH (a human neuroblastoma cell line) in the presence of RhoA activator II for 24 h after which the RhoA activity was measured using the G-LISA Kit. Fluorescence staining of f-actin stress fibres was performed to validate RhoA inhibition. SK-N-SH was transfected with plasmid expressing amyloid precursor protein (APP) gene. The Aβ concentration in transfected cells exposed to LAB-derived cell free supernatant (CFS) in the presence of RhoA activator II was measured using the ELISA kit. Furthermore, this study measured organic acids in LAB-derived CFS using the gas chromatography. It was found that LAB-derived CFS yielded strain-dependent inhibition of RhoA, with LAB6- and LAB12-derived CFS being the most potent Pediococcal- and Lactiplantibacillus-based RhoA inhibitor, respectively. Lesser stress fibres were formed under treatment with LAB-derived CFS. The LAB-derived CFS also significantly inhibited Aβ in SK-N-SH transfected with APP gene in the presence of RhoA activator II. The LAB-derived CFS was presented with increased lactic acid, acetic acid, butyric acid and propionic acid. The present findings warrant in-depth study using animal models.
Collapse
Affiliation(s)
- Ramli Muhammad Zaki
- Collaborative Drug Discovery Research (CDDR) Group, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Cawangan Selangor, Kampus Puncak Alam, 42300, Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia
- Faculty of Pharmacy and Health Sciences, Royal College of Medicine Perak, Universiti Kuala Lumpur , Ipoh, 30450, Perak, Malaysia
| | - Kalavathy Ramasamy
- Collaborative Drug Discovery Research (CDDR) Group, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Cawangan Selangor, Kampus Puncak Alam, 42300, Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia
| | - Nor Amalina Ahmad Alwi
- Collaborative Drug Discovery Research (CDDR) Group, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Cawangan Selangor, Kampus Puncak Alam, 42300, Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia
| | - Rosmadi Mohd Yusoff
- Collaborative Drug Discovery Research (CDDR) Group, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Cawangan Selangor, Kampus Puncak Alam, 42300, Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia
| | - Siong Meng Lim
- Collaborative Drug Discovery Research (CDDR) Group, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Cawangan Selangor, Kampus Puncak Alam, 42300, Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
25
|
Tang Y, Du J, Wu H, Wang M, Liu S, Tao F. Potential Therapeutic Effects of Short-Chain Fatty Acids on Chronic Pain. Curr Neuropharmacol 2024; 22:191-203. [PMID: 36173071 PMCID: PMC10788890 DOI: 10.2174/1570159x20666220927092016] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/03/2022] [Accepted: 09/13/2022] [Indexed: 11/22/2022] Open
Abstract
The intestinal homeostasis maintained by the gut microbiome and relevant metabolites is essential for health, and its disturbance leads to various intestinal or extraintestinal diseases. Recent studies suggest that gut microbiome-derived metabolites short-chain fatty acids (SCFAs) are involved in different neurological disorders (such as chronic pain). SCFAs are produced by bacterial fermentation of dietary fibers in the gut and contribute to multiple host processes, including gastrointestinal regulation, cardiovascular modulation, and neuroendocrine-immune homeostasis. Although SCFAs have been implicated in the modulation of chronic pain, the detailed mechanisms that underlie such roles of SCFAs remain to be further investigated. In this review, we summarize currently available research data regarding SCFAs as a potential therapeutic target for chronic pain treatment and discuss several possible mechanisms by which SCFAs modulate chronic pain.
Collapse
Affiliation(s)
- Yuanyuan Tang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China
- Key Laboratory for Molecular Neurology of Xinxiang, Xinxiang, Henan, China
| | - Juan Du
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China
| | - Hongfeng Wu
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China
| | - Mengyao Wang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China
| | - Sufang Liu
- Department of Biomedical Sciences, College of Dentistry, Texas A&M University Dallas, Texas, USA
| | - Feng Tao
- Department of Biomedical Sciences, College of Dentistry, Texas A&M University Dallas, Texas, USA
| |
Collapse
|
26
|
Meleady L, Towriss M, Kim J, Bacarac V, Dang V, Rowland ME, Ciernia AV. Histone deacetylase 3 regulates microglial function through histone deacetylation. Epigenetics 2023; 18:2241008. [PMID: 37506371 PMCID: PMC10392760 DOI: 10.1080/15592294.2023.2241008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
As the primary innate immune cells of the brain, microglia respond to damage and disease through pro-inflammatory release of cytokines and neuroinflammatory molecules. Histone acetylation is an activating transcriptional mark that regulates inflammatory gene expression. Inhibition of histone deacetylase 3 (Hdac3) has been utilized in pre-clinical models of depression, stroke, and spinal cord injury to improve recovery following injury, but the molecular mechanisms underlying Hdac3's regulation of inflammatory gene expression in microglia is not well understood. To address this lack of knowledge, we examined how pharmacological inhibition of Hdac3 in an immortalized microglial cell line (BV2) impacted histone acetylation and gene expression of pro- and anti-inflammatory genes in response to immune challenge with lipopolysaccharide (LPS). Flow cytometry and cleavage under tags & release using nuclease (CUT & RUN) revealed that Hdac3 inhibition increases global and promoter-specific histone acetylation, resulting in the release of gene repression at baseline and enhanced responses to LPS. Hdac3 inhibition enhanced neuroprotective functions of microglia in response to LPS through reduced nitric oxide release and increased phagocytosis. The findings suggest Hdac3 serves as a regulator of microglial inflammation, and that inhibition of Hdac3 facilitates the microglial response to inflammation and its subsequent clearing of debris or damaged cells. Together, this work provides new mechanistic insights into therapeutic applications of Hdac3 inhibition which mediate reduced neuroinflammatory insults through microglial response.
Collapse
Affiliation(s)
- Laura Meleady
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Morgan Towriss
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Jennifer Kim
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, Canada
| | - Vince Bacarac
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Vivien Dang
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Megan E. Rowland
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Annie Vogel Ciernia
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| |
Collapse
|
27
|
de Souza PB, de Araujo Borba L, Castro de Jesus L, Valverde AP, Gil-Mohapel J, Rodrigues ALS. Major Depressive Disorder and Gut Microbiota: Role of Physical Exercise. Int J Mol Sci 2023; 24:16870. [PMID: 38069198 PMCID: PMC10706777 DOI: 10.3390/ijms242316870] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 11/24/2023] [Accepted: 11/25/2023] [Indexed: 12/18/2023] Open
Abstract
Major depressive disorder (MDD) has a high prevalence and is a major contributor to the global burden of disease. This psychiatric disorder results from a complex interaction between environmental and genetic factors. In recent years, the role of the gut microbiota in brain health has received particular attention, and compelling evidence has shown that patients suffering from depression have gut dysbiosis. Several studies have reported that gut dysbiosis-induced inflammation may cause and/or contribute to the development of depression through dysregulation of the gut-brain axis. Indeed, as a consequence of gut dysbiosis, neuroinflammatory alterations caused by microglial activation together with impairments in neuroplasticity may contribute to the development of depressive symptoms. The modulation of the gut microbiota has been recognized as a potential therapeutic strategy for the management of MMD. In this regard, physical exercise has been shown to positively change microbiota composition and diversity, and this can underlie, at least in part, its antidepressant effects. Given this, the present review will explore the relationship between physical exercise, gut microbiota and depression, with an emphasis on the potential of physical exercise as a non-invasive strategy for modulating the gut microbiota and, through this, regulating the gut-brain axis and alleviating MDD-related symptoms.
Collapse
Affiliation(s)
- Pedro Borges de Souza
- Center of Biological Sciences, Department of Biochemistry, Universidade Federal de Santa Catarina, Florianópolis 88037-000, SC, Brazil; (P.B.d.S.); (L.d.A.B.); (L.C.d.J.); (A.P.V.)
| | - Laura de Araujo Borba
- Center of Biological Sciences, Department of Biochemistry, Universidade Federal de Santa Catarina, Florianópolis 88037-000, SC, Brazil; (P.B.d.S.); (L.d.A.B.); (L.C.d.J.); (A.P.V.)
| | - Louise Castro de Jesus
- Center of Biological Sciences, Department of Biochemistry, Universidade Federal de Santa Catarina, Florianópolis 88037-000, SC, Brazil; (P.B.d.S.); (L.d.A.B.); (L.C.d.J.); (A.P.V.)
| | - Ana Paula Valverde
- Center of Biological Sciences, Department of Biochemistry, Universidade Federal de Santa Catarina, Florianópolis 88037-000, SC, Brazil; (P.B.d.S.); (L.d.A.B.); (L.C.d.J.); (A.P.V.)
| | - Joana Gil-Mohapel
- Island Medical Program, Faculty of Medicine, University of British Columbia, Victoria, BC V8P 5C2, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Ana Lúcia S. Rodrigues
- Center of Biological Sciences, Department of Biochemistry, Universidade Federal de Santa Catarina, Florianópolis 88037-000, SC, Brazil; (P.B.d.S.); (L.d.A.B.); (L.C.d.J.); (A.P.V.)
| |
Collapse
|
28
|
Pavan S, Gorthi SP, Prabhu AN, Das B, Mutreja A, Vasudevan K, Shetty V, Ramamurthy T, Ballal M. Dysbiosis of the Beneficial Gut Bacteria in Patients with Parkinson's Disease from India. Ann Indian Acad Neurol 2023; 26:908-916. [PMID: 38229613 PMCID: PMC10789430 DOI: 10.4103/aian.aian_460_23] [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: 05/24/2023] [Revised: 08/21/2023] [Accepted: 08/24/2023] [Indexed: 01/18/2024] Open
Abstract
Objectives Recent advancement in understanding neurological disorders has revealed the involvement of dysbiosis of the gut microbiota in the pathophysiology of Parkinson's disease (PD). We sequenced microbial DNA using fecal samples collected from PD cases and healthy controls (HCs) to evaluate the role of gut microbiota. Methods Full-length bacterial 16S rRNA gene sequencing of fecal samples was performed using amplified polymerase chain reaction (PCR) products on the GridION Nanopore sequencer. Sequenced data were analyzed using web-based tools BugSeq and MicrobiomeAnalyst. Results We found that certain bacterial families like Clostridia UCG 014, Cristensenellaceae, and Oscillospiraceae are higher in abundance, and Lachinospiracea, Coriobacteriaceae and genera associated with short-chain fatty acid production, Faecalibacterium, Fusicatenibacter, Roseburia and Blautia, are lower in abundance among PD cases when compared with the HC. Genus Akkermansia, Dialister, Bacteroides, and Lachnospiraceae NK4A136 group positively correlated with constipation in PD. Conclusion Observations from this study support the other global research on the PD gut microbiome background and provide fresh insight into the gut microbial composition of PD patients from a south Indian population. We report a higher abundance of Clostridia UCG 014 group, previously not linked to PD.
Collapse
Affiliation(s)
- Sujith Pavan
- Enteric Diseases Division, Department of Microbiology, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Sankar Prasad Gorthi
- Department of Neurology, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, India
- Department of Neurology, Bharati Vidyapeeth Medical College and Hospital, Pune, Maharashtra, India
| | - Arvind N. Prabhu
- Department of Neurology, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Bhabatosh Das
- Molecular Genetics Laboratory, Centre for Human Microbial Ecology, Translational Health Sciences and Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Ankur Mutreja
- Enteric Diseases Division, Department of Microbiology, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, India
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Karthick Vasudevan
- Department of Biotechnology, School of Applied Sciences, Reva University, Bengaluru, Karnataka, India
| | - Vignesh Shetty
- Enteric Diseases Division, Department of Microbiology, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, India
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Thandavarayan Ramamurthy
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, West Bengal, India
| | - Mamatha Ballal
- Enteric Diseases Division, Department of Microbiology, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
29
|
Khezri MR, Esmaeili A, Ghasemnejad-Berenji M. Role of Bmal1 and Gut Microbiota in Alzheimer's Disease and Parkinson's Disease Pathophysiology: The Probable Effect of Melatonin on Their Association. ACS Chem Neurosci 2023; 14:3883-3893. [PMID: 37823531 DOI: 10.1021/acschemneuro.3c00418] [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: 10/13/2023] Open
Abstract
In recent years, the role of new factors in the pathophysiology of neurodegenerative diseases has been investigated. Alzheimer's disease (AD) and Parkinson's disease (PD) are the most common neurodegenerative diseases worldwide. Although pathological changes such as the accumulation of aggregated proteins in the brain and inflammatory responses are known as the main factors involved in the development of these diseases, new studies show the role of gut microbiota and circadian rhythm in the occurrence of these changes. However, the association between circadian rhythm and gut microbiota in AD and PD has not yet been investigated. Recent results propose that alterations in circadian rhythm regulators, mainly Bmal1, may regulate the abundance of gut microbiota. This correlation has been linked to the regulation of the expression of immune-related genes and Bmal-1 mediated oscillation of IgA and hydrogen peroxide production. These data seem to provide new insight into the molecular mechanism of melatonin inhibiting the progression of AD and PD. Therefore, this manuscript aims to review the role of the gut microbiota and circadian rhythm in health and AD and PD and also presents a hypothesis on the effect of melatonin on their communication.
Collapse
Affiliation(s)
- Mohammad Rafi Khezri
- Faculty of Pharmacy. Urmia University of Medical Sciences, Urmia 571478334, Iran
| | - Ayda Esmaeili
- Department of Clinical Pharmacy, School of Pharmacy, Urmia University of Medical Sciences, Urmia 5715799313, Iran
| | - Morteza Ghasemnejad-Berenji
- Department of Pharmacology and Toxicology, School of Pharmacy, Urmia University of Medical Sciences, Urmia 5715799313, Iran
| |
Collapse
|
30
|
Wang Y, Xu Q, Meng M, Chang G, Ma N, Shen X. Butyrate Protects against γ-d-Glutamyl- meso-diaminopimelic Acid-Induced Inflammatory Response and Tight Junction Disruption through Histone Deacetylase 3 Inhibition in Bovine Mammary Epithelial Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:14638-14648. [PMID: 37767922 DOI: 10.1021/acs.jafc.3c04417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/29/2023]
Abstract
The present study was conducted to evaluate the regulatory actions and underlying mechanisms of butyrate on the inflammatory response and tight junction (TJ) disruption in bovine mammary epithelial cells (BMECs). Results showed that butyrate declined histone deacetylase 3 (HDAC3) expression, blocked NF-κB activation, and thus suppressed inflammatory cytokine production in γ-d-glutamyl-meso-diaminopimelic acid (iE-DAP)-triggered BMECs. Butyrate also depressed the protein abundance of myosin light chain kinase (MLCK), elevated the expression of TJ proteins, and restored the cellular distribution of TJ proteins and the barrier function of epithelial cells. HDAC3 overexpression abolished the protective effects of butyrate. In conclusion, butyrate alleviated the iE-DAP-induced inflammatory response and TJ injury by blocking NF-κB activation and decreasing inflammatory cytokine production and MLCK expression in a HDAC3-dependent manner. Our finding provides a mechanistic basis for further exploring the regulatory effects of butyrate on the mammary inflammatory response.
Collapse
Affiliation(s)
- Yan Wang
- College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Qianqian Xu
- College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Meijuan Meng
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Guangjun Chang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Nana Ma
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiangzhen Shen
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
31
|
Liou CW, Cheng SJ, Yao TH, Lai TT, Tsai YH, Chien CW, Kuo YL, Chou SH, Hsu CC, Wu WL. Microbial metabolites regulate social novelty via CaMKII neurons in the BNST. Brain Behav Immun 2023; 113:104-123. [PMID: 37393058 DOI: 10.1016/j.bbi.2023.06.029] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 06/28/2023] [Accepted: 06/28/2023] [Indexed: 07/03/2023] Open
Abstract
Social novelty is a cognitive process that is essential for animals to interact strategically with conspecifics based on their prior experiences. The commensal microbiome in the gut modulates social behavior through various routes, including microbe-derived metabolite signaling. Short-chain fatty acids (SCFAs), metabolites derived from bacterial fermentation in the gastrointestinal tract, have been previously shown to impact host behavior. Herein, we demonstrate that the delivery of SCFAs directly into the brain disrupts social novelty through distinct neuronal populations. We are the first to observe that infusion of SCFAs into the lateral ventricle disrupted social novelty in microbiome-depleted mice without affecting brain inflammatory responses. The deficit in social novelty can be recapitulated by activating calcium/calmodulin-dependent protein kinase II (CaMKII)-labeled neurons in the bed nucleus of the stria terminalis (BNST). Conversely, chemogenetic silencing of the CaMKII-labeled neurons and pharmacological inhibition of fatty acid oxidation in the BNST reversed the SCFAs-induced deficit in social novelty. Our findings suggest that microbial metabolites impact social novelty through a distinct neuron population in the BNST.
Collapse
Affiliation(s)
- Chia-Wei Liou
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, 1 University Rd, Tainan 70101, Taiwan; Department of Physiology, College of Medicine, National Cheng Kung University, 1 University Rd, Tainan 70101, Taiwan.
| | - Sin-Jhong Cheng
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan.
| | - Tzu-Hsuan Yao
- Department of Physiology, College of Medicine, National Cheng Kung University, 1 University Rd, Tainan 70101, Taiwan
| | - Tzu-Ting Lai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, 1 University Rd, Tainan 70101, Taiwan; Department of Physiology, College of Medicine, National Cheng Kung University, 1 University Rd, Tainan 70101, Taiwan
| | - Yu-Hsuan Tsai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, 1 University Rd, Tainan 70101, Taiwan; Department of Physiology, College of Medicine, National Cheng Kung University, 1 University Rd, Tainan 70101, Taiwan
| | - Che-Wei Chien
- Leeuwenhoek Laboratories Co. Ltd, Taipei 10672, Taiwan
| | - Yu-Lun Kuo
- Biotools Co. Ltd, New Taipei City 22175, Taiwan
| | - Shih-Hsuan Chou
- Biotools Co. Ltd, New Taipei City 22175, Taiwan; Graduate Institute of Biomedical and Pharmaceutical Science, Fu-Jen Catholic University, New Taipei City 24205, Taiwan
| | - Cheng-Chih Hsu
- Leeuwenhoek Laboratories Co. Ltd, Taipei 10672, Taiwan; Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan.
| | - Wei-Li Wu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, 1 University Rd, Tainan 70101, Taiwan; Department of Physiology, College of Medicine, National Cheng Kung University, 1 University Rd, Tainan 70101, Taiwan.
| |
Collapse
|
32
|
Wang L, Wang Z, Lan Y, Tuo Y, Ma S, Liu X. Inulin Attenuates Blood-Brain Barrier Permeability and Alleviates Behavioral Disorders by Modulating the TLR4/MyD88/NF-κB Pathway in Mice with Chronic Stress. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:13325-13337. [PMID: 37642581 DOI: 10.1021/acs.jafc.3c03568] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Depression and vulnerability to chronic stress are associated with inflammatory responses and the loss of blood-brain barrier (BBB) integrity. Dietary fiber and its short-chain fatty acid (SCFAs) metabolites have been reported to affect neuropsychiatric disorders. Here, a 9-week treatment course of inulin (0.037 g of inulin/kcal) exhibited in chronic unpredictable mild stress (CUMS) mice led to antidepressant and anxiolytic effects, as well as improved neurogenesis and synaptic plasticity by enhancing CREB/BDNF signaling. Importantly, inulin inhibited CUMS-induced decreased BBB permeability, reduced lipopolysaccharide (LPS) brain penetration, and modulated TLR4/MyD88/NF-κB signaling to alleviate neuroinflammatory responses. Furthermore, inulin protected the gut barrier integrity and led to the increased formation of SCFAs. Enhanced SCFAs formation was strongly positively correlated with behavioral improvements, BBB integrity, and neuroinflammatory responses. We speculate that dietary fiber may be a promising nutritional intervention to reverse the effects of chronic stress by regulating metabolites and protecting the BBB integrity.
Collapse
Affiliation(s)
- Lei Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Zhuo Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Yongli Lan
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xi'ning 810016, China
| | - Yanliang Tuo
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Shaobo Ma
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Xuebo Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| |
Collapse
|
33
|
Arishi RA, Lai CT, Geddes DT, Stinson LF. Impact of breastfeeding and other early-life factors on the development of the oral microbiome. Front Microbiol 2023; 14:1236601. [PMID: 37744908 PMCID: PMC10513450 DOI: 10.3389/fmicb.2023.1236601] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 08/25/2023] [Indexed: 09/26/2023] Open
Abstract
The oral cavity is home to the second most diverse microbiome in the human body. This community contributes to both oral and systemic health. Acquisition and development of the oral microbiome is a dynamic process that occurs over early life; however, data regarding longitudinal assembly of the infant oral microbiome is scarce. While numerous factors have been associated with the composition of the infant oral microbiome, early feeding practices (breastfeeding and the introduction of solids) appear to be the strongest determinants of the infant oral microbiome. In the present review, we draw together data on the maternal, infant, and environmental factors linked to the composition of the infant oral microbiome, with a focus on early nutrition. Given evidence that breastfeeding powerfully shapes the infant oral microbiome, the review explores potential mechanisms through which human milk components, including microbes, metabolites, oligosaccharides, and antimicrobial proteins, may interact with and shape the infant oral microbiome. Infancy is a unique period for the oral microbiome. By enhancing our understanding of oral microbiome assembly in early life, we may better support both oral and systemic health throughout the lifespan.
Collapse
Affiliation(s)
- Roaa A. Arishi
- School of Molecular Sciences, The University of Western Australia, Perth, WA, Australia
- Ministry of Health, Riyadh, Saudi Arabia
| | - Ching T. Lai
- School of Molecular Sciences, The University of Western Australia, Perth, WA, Australia
| | - Donna T. Geddes
- School of Molecular Sciences, The University of Western Australia, Perth, WA, Australia
| | - Lisa F. Stinson
- School of Molecular Sciences, The University of Western Australia, Perth, WA, Australia
| |
Collapse
|
34
|
Xie J, Bruggeman A, De Nolf C, Vandendriessche C, Van Imschoot G, Van Wonterghem E, Vereecke L, Vandenbroucke RE. Gut microbiota regulates blood-cerebrospinal fluid barrier function and Aβ pathology. EMBO J 2023; 42:e111515. [PMID: 37427561 PMCID: PMC10476279 DOI: 10.15252/embj.2022111515] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 05/20/2023] [Accepted: 06/02/2023] [Indexed: 07/11/2023] Open
Abstract
Accumulating evidence indicates that gut microbiota dysbiosis is associated with increased blood-brain barrier (BBB) permeability and contributes to Alzheimer's disease (AD) pathogenesis. In contrast, the influence of gut microbiota on the blood-cerebrospinal fluid (CSF) barrier has not yet been studied. Here, we report that mice lacking gut microbiota display increased blood-CSF barrier permeability associated with disorganized tight junctions (TJs), which can be rescued by recolonization with gut microbiota or supplementation with short-chain fatty acids (SCFAs). Our data reveal that gut microbiota is important not only for the establishment but also for the maintenance of a tight barrier. Also, we report that the vagus nerve plays an important role in this process and that SCFAs can independently tighten the barrier. Administration of SCFAs in AppNL-G-F mice improved the subcellular localization of TJs at the blood-CSF barrier, reduced the β-amyloid (Aβ) burden, and affected microglial phenotype. Altogether, our results suggest that modulating the microbiota and administering SCFAs might have therapeutic potential in AD via blood-CSF barrier tightening and maintaining microglial activity and Aβ clearance.
Collapse
Affiliation(s)
- Junhua Xie
- VIB Center for Inflammation ResearchGhentBelgium
- Department of Biomedical Molecular BiologyGhent UniversityGhentBelgium
| | - Arnout Bruggeman
- VIB Center for Inflammation ResearchGhentBelgium
- Department of Biomedical Molecular BiologyGhent UniversityGhentBelgium
- Department of NeurologyGhent University HospitalGhentBelgium
| | - Clint De Nolf
- VIB Center for Inflammation ResearchGhentBelgium
- Department of Biomedical Molecular BiologyGhent UniversityGhentBelgium
- Department of Internal Medicine and PediatricsGhent UniversityGhentBelgium
| | - Charysse Vandendriessche
- VIB Center for Inflammation ResearchGhentBelgium
- Department of Biomedical Molecular BiologyGhent UniversityGhentBelgium
| | - Griet Van Imschoot
- VIB Center for Inflammation ResearchGhentBelgium
- Department of Biomedical Molecular BiologyGhent UniversityGhentBelgium
| | - Elien Van Wonterghem
- VIB Center for Inflammation ResearchGhentBelgium
- Department of Biomedical Molecular BiologyGhent UniversityGhentBelgium
| | - Lars Vereecke
- VIB Center for Inflammation ResearchGhentBelgium
- Department of Internal Medicine and PediatricsGhent UniversityGhentBelgium
- Ghent Gut Inflammation Group (GGIG)Ghent UniversityGhentBelgium
| | - Roosmarijn E Vandenbroucke
- VIB Center for Inflammation ResearchGhentBelgium
- Department of Biomedical Molecular BiologyGhent UniversityGhentBelgium
| |
Collapse
|
35
|
Ma L, Jiang X, Huang Q, Chen W, Zhang H, Pei H, Cao Y, Wang H, Li H. Traditional Chinese medicine for the treatment of Alzheimer's disease: A focus on the microbiota-gut-brain axis. Biomed Pharmacother 2023; 165:115244. [PMID: 37516021 DOI: 10.1016/j.biopha.2023.115244] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/11/2023] [Accepted: 07/25/2023] [Indexed: 07/31/2023] Open
Abstract
Alzheimer's disease (AD), the most frequent cause of dementia, is a neurodegenerative disorder characterised by a progressive decline in cognitive function that is associated with the formation of amyloid beta plaques and neurofibrillary tangles. Gut microbiota comprises of a complex community of microorganisms residing in the gastrointestinal ecosystem. These microorganisms can participate in gut-brain axis activities, thereby affecting cognitive function and associated behaviours. Increasing evidence has indicated that gut dysbiosis can jeopardise host immune responses and promote inflammation, which may be an initiating factor for the onset and evolution of AD. Traditional Chinese medicine (TCM) is a promising resource which encompasses immense chemical diversity and multiple-target characteristics for the treatment of AD. Many TCMs regulate the gut microbiota during treatment of diseases, indicating that gut microbiota may be an important target for TCM efficacy. In this review, we summarised the role of the microbiota-gut-brain axis in the development of AD and the effects of TCM in treating AD by regulating the gut microbiota. We anticipate that this review will provide novel perspectives and strategies for future AD research and treatments.
Collapse
Affiliation(s)
- Lina Ma
- Institute of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, PR China
| | - Xuefan Jiang
- Institute of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, PR China
| | - Qiaoyi Huang
- Institute of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, PR China
| | - Wenxuan Chen
- Institute of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, PR China
| | - Huiqin Zhang
- Institute of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, PR China
| | - Hui Pei
- Institute of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, PR China
| | - Yu Cao
- Institute of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, PR China
| | - Huichan Wang
- Institute of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, PR China
| | - Hao Li
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing 100102, PR China.
| |
Collapse
|
36
|
Duan H, Wang L, Huangfu M, Li H. The impact of microbiota-derived short-chain fatty acids on macrophage activities in disease: Mechanisms and therapeutic potentials. Biomed Pharmacother 2023; 165:115276. [PMID: 37542852 DOI: 10.1016/j.biopha.2023.115276] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/28/2023] [Accepted: 07/31/2023] [Indexed: 08/07/2023] Open
Abstract
Short-chain fatty acids (SCFAs) derived from the fermentation of carbohydrates by gut microbiota play a crucial role in regulating host physiology. Among them, acetate, propionate, and butyrate are key players in various biological processes. Recent research has revealed their significant functions in immune and inflammatory responses. For instance, butyrate reduces the development of interferon-gamma (IFN-γ) generating cells while promoting the development of regulatory T (Treg) cells. Propionate inhibits the initiation of a Th2 immune response by dendritic cells (DCs). Notably, SCFAs have an inhibitory impact on the polarization of M2 macrophages, emphasizing their immunomodulatory properties and potential for therapeutics. In animal models of asthma, both butyrate and propionate suppress the M2 polarization pathway, thus reducing allergic airway inflammation. Moreover, dysbiosis of gut microbiota leading to altered SCFA production has been implicated in prostate cancer progression. SCFAs trigger autophagy in cancer cells and promote M2 polarization in macrophages, accelerating tumor advancement. Manipulating microbiota- producing SCFAs holds promise for cancer treatment. Additionally, SCFAs enhance the expression of hypoxia-inducible factor 1 (HIF-1) by blocking histone deacetylase, resulting in increased production of antibacterial effectors and improved macrophage-mediated elimination of microorganisms. This highlights the antimicrobial potential of SCFAs and their role in host defense mechanisms. This comprehensive review provides an in-depth analysis of the latest research on the functional aspects and underlying mechanisms of SCFAs in relation to macrophage activities in a wide range of diseases, including infectious diseases and cancers. By elucidating the intricate interplay between SCFAs and macrophage functions, this review aims to contribute to the understanding of their therapeutic potential and pave the way for future interventions targeting SCFAs in disease management.
Collapse
Affiliation(s)
- Hongliang Duan
- Department of Thyroid Surgery, the Second Hospital of Jilin University, Changchun 130000, China
| | - LiJuan Wang
- Department of Endocrinology, the Second Hospital of Jilin University, Changchun 130000, China.
| | - Mingmei Huangfu
- Department of Thyroid Surgery, the Second Hospital of Jilin University, Changchun 130000, China
| | - Hanyang Li
- Department of Endocrinology, the Second Hospital of Jilin University, Changchun 130000, China
| |
Collapse
|
37
|
Schmitt V, Masanetz RK, Weidenfeller M, Ebbinghaus LS, Süß P, Rosshart SP, von Hörsten S, Zunke F, Winkler J, Xiang W. Gut-to-brain spreading of pathology in synucleinopathies: A focus on molecular signalling mediators. Behav Brain Res 2023; 452:114574. [PMID: 37423320 DOI: 10.1016/j.bbr.2023.114574] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 07/03/2023] [Accepted: 07/06/2023] [Indexed: 07/11/2023]
Abstract
Synucleinopathies are a group of neurodegenerative disorders, classically characterized by the accumulation of aggregated alpha synuclein (aSyn) in the central nervous system. Parkinson's disease (PD) and multiple system atrophy (MSA) are the two prominent members of this family. Current treatment options mainly focus on the motor symptoms of these diseases. However, non-motor symptoms, including gastrointestinal (GI) symptoms, have recently gained particular attention, as they are frequently associated with synucleinopathies and often arise before motor symptoms. The gut-origin hypothesis has been proposed based on evidence of an ascending spreading pattern of aggregated aSyn from the gut to the brain, as well as the comorbidity of inflammatory bowel disease and synucleinopathies. Recent advances have shed light on the mechanisms underlying the progression of synucleinopathies along the gut-brain axis. Given the rapidly expanding pace of research in the field, this review presents a summary of the latest findings on the gut-to-brain spreading of pathology and potential pathology-reinforcing mediators in synucleinopathies. Here, we focus on 1) gut-to-brain communication pathways, including neuronal pathways and blood circulation, and 2) potential molecular signalling mediators, including bacterial amyloid proteins, microbiota dysbiosis-induced alterations in gut metabolites, as well as host-derived effectors, including gut-derived peptides and hormones. We highlight the clinical relevance and implications of these molecular mediators and their possible mechanisms in synucleinopathies. Moreover, we discuss their potential as diagnostic markers in distinguishing the subtypes of synucleinopathies and other neurodegenerative diseases, as well as for developing novel individualized therapeutic options for synucleinopathies.
Collapse
Affiliation(s)
- Verena Schmitt
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Germany
| | - Rebecca Katharina Masanetz
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Germany
| | - Martin Weidenfeller
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Germany
| | - Lara Savannah Ebbinghaus
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Germany
| | - Patrick Süß
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Germany
| | - Stephan P Rosshart
- Department of Microbiome Research, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Germany
| | - Stephan von Hörsten
- Department for Experimental Therapy, University Hospital Erlangen, Preclinical Experimental Center, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Germany
| | - Friederike Zunke
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Germany
| | - Jürgen Winkler
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Germany
| | - Wei Xiang
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Germany.
| |
Collapse
|
38
|
Singh Y, Trautwein C, Romani J, Salker MS, Neckel PH, Fraccaroli I, Abeditashi M, Woerner N, Admard J, Dhariwal A, Dueholm MKD, Schäfer KH, Lang F, Otzen DE, Lashuel HA, Riess O, Casadei N. Overexpression of human alpha-Synuclein leads to dysregulated microbiome/metabolites with ageing in a rat model of Parkinson disease. Mol Neurodegener 2023; 18:44. [PMID: 37403161 DOI: 10.1186/s13024-023-00628-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 05/24/2023] [Indexed: 07/06/2023] Open
Abstract
BACKGROUND Braak's hypothesis states that sporadic Parkinson's disease (PD) follows a specific progression of pathology from the peripheral to the central nervous system, and this progression can be monitored by detecting the accumulation of alpha-Synuclein (α-Syn) protein. Consequently, there is growing interest in understanding how the gut (commensal) microbiome can regulate α-Syn accumulation, as this could potentially lead to PD. METHODS We used 16S rRNA and shotgun sequencing to characterise microbial diversity. 1H-NMR was employed to understand the metabolite production and intestinal inflammation estimated using ELISA and RNA-sequencing from feces and the intestinal epithelial layer respectively. The Na+ channel current and gut permeability were measured using an Ussing chamber. Immunohistochemistry and immunofluorescence imaging were applied to detect the α-Syn protein. LC-MS/MS was used for characterization of proteins from metabolite treated neuronal cells. Finally, Metascape and Ingenuity Pathway Analysis (IPA) bioinformatics tools were used for identification of dysregulated pathways. RESULTS We studied a transgenic (TG) rat model overexpressing the human SNCA gene and found that a progressive gut microbial composition alteration characterized by the reduction of Firmicutes to Bacteroidetes ratio could be detected in the young TG rats. Interestingly, this ratio then increased with ageing. The dynamics of Lactobacillus and Alistipes were monitored and reduced Lactobacillus and increased Alistipes abundance was discerned in ageing TG rats. Additionally, the SNCA gene overexpression resulted in gut α-Syn protein expression and increased with advanced age. Further, older TG animals had increased intestinal inflammation, decreased Na+ current and a robust alteration in metabolite production characterized by the increase of succinate levels in feces and serum. Manipulation of the gut bacteria by short-term antibiotic cocktail treatment revealed a complete loss of short-chain fatty acids and a reduction in succinate levels. Although antibiotic cocktail treatment did not change α-Syn expression in the enteric nervous system of the colon, however, reduced α-Syn expression was detected in the olfactory bulbs (forebrain) of the TG rats. CONCLUSION Our data emphasize that the gut microbiome dysbiosis synchronous with ageing leads to a specific alteration of gut metabolites and can be modulated by antibiotics which may affect PD pathology.
Collapse
Affiliation(s)
- Yogesh Singh
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Calwerstaße 7, 72076, Tübingen, Germany.
- NGS Competence Centre Tübingen (NCCT), University of Tübingen, Calwerstaße 7, 72076, Tübingen, Germany.
- Research Institute of Women's Health, University of Tübingen, Calwerstaße 7/6, 72076, Tübingen, Germany.
| | - Christoph Trautwein
- Werner Siemens Imaging Centre (WSIC), Department of Preclinical Imaging and Radiopharmacy, University of Tübingen, Röntgenweg 13, 72076, Tübingen, Germany
| | - Joan Romani
- School of Life Sciences, Institute of Bioengineering, Laboratory of Molecular and Chemical Biology of Neurodegeneration, École Polytechnique Fédérale de Lausanne (EPFL), SV LMNN Station 19, 1015 CH, Lausanne, Switzerland
| | - Madhuri S Salker
- Research Institute of Women's Health, University of Tübingen, Calwerstaße 7/6, 72076, Tübingen, Germany
| | - Peter H Neckel
- Institute of Clinical Anatomy and Cell Analysis, University of Tübingen, Österbergstraße 3, 72074, Tübingen, Germany
| | - Isabel Fraccaroli
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Calwerstaße 7, 72076, Tübingen, Germany
| | - Mahkameh Abeditashi
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Calwerstaße 7, 72076, Tübingen, Germany
| | - Nils Woerner
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Calwerstaße 7, 72076, Tübingen, Germany
| | - Jakob Admard
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Calwerstaße 7, 72076, Tübingen, Germany
| | - Achal Dhariwal
- Institute of Oral Biology, University of Oslo, Sognsvannsveien 10, 0316, Oslo, Norway
| | - Morten K D Dueholm
- Department of Chemistry and Bioscience, Aalborg University, Fredrik Bajers Vej 7H, 9220, Aalborg, Denmark
| | - Karl-Herbert Schäfer
- Enteric Nervous System Working Group, University of Applied Sciences Kaiserslautern, Zweibrücken Campus, Amerikastrasse 1, 66482, Zweibrücken, Germany
| | - Florian Lang
- Institute of Vegetative Physiology, University of Tübingen, Wilhelmstaße 56, 72074, Tübingen, Germany
| | - Daniel E Otzen
- Interdisciplinary Naonscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus C, Denmark
| | - Hilal A Lashuel
- School of Life Sciences, Institute of Bioengineering, Laboratory of Molecular and Chemical Biology of Neurodegeneration, École Polytechnique Fédérale de Lausanne (EPFL), SV LMNN Station 19, 1015 CH, Lausanne, Switzerland
| | - Olaf Riess
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Calwerstaße 7, 72076, Tübingen, Germany.
- NGS Competence Centre Tübingen (NCCT), University of Tübingen, Calwerstaße 7, 72076, Tübingen, Germany.
| | - Nicolas Casadei
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Calwerstaße 7, 72076, Tübingen, Germany.
- NGS Competence Centre Tübingen (NCCT), University of Tübingen, Calwerstaße 7, 72076, Tübingen, Germany.
| |
Collapse
|
39
|
Nawarathna G, Fakhruddin KS, Shorbagi AISA, Samaranayake LP. The gut microbiota-neuroimmune crosstalk and neuropathic pain: a scoping review. GUT MICROBIOME (CAMBRIDGE, ENGLAND) 2023; 4:e10. [PMID: 39295900 PMCID: PMC11406377 DOI: 10.1017/gmb.2023.7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 03/01/2023] [Accepted: 05/23/2023] [Indexed: 09/21/2024]
Abstract
Environmental stressors can disrupt the gut-brain relationship and alter the gut microbial composition, potentially leading to chronic pain, including neuropathic pain (NP). To understand this complex relationship, we conducted a systematic scoping review to examine the gut microbial-neuroimmune connection to NP and the potential therapeutic targets. The review includes English-language manuscripts in databases such as MEDLINE, Cochrane, and DOAJ between January 2000 and April 2022. Out of the 48 full texts examined, only 15 articles met the inclusion criteria. These included a randomised controlled trial involving 327 individuals, an in vitro, and 13 animal model studies. The findings suggest that the gut flora plays a role in the immunological, neurological, and metabolic signalling pathways associated with NP. Animal studies have been the primary focus in this area, indicating that an imbalanced-gut microbiome and subsequent activation of biochemical and neuro-immunologic pathways may influence the development of NP. This review provides a comprehensive summary of the gut microbiome-immune-NP axis and identifies potential therapeutic targets. However, since most of the evidence comes from animal studies, future research should include clinical trials to gain a better understanding of the role of gut microbiota in NP and discover new therapeutic strategies.
Collapse
Affiliation(s)
- Gayani Nawarathna
- Department of Basic Sciences, Faculty of Dental Sciences, University of Peradeniya, Peradeniya, Sri Lanka
| | - Kausar S. Fakhruddin
- Department of Preventive and Restorative Dentistry, University of Sharjah, Sharjah, UAE
| | - Ali I. S. A. Shorbagi
- Clinical Sciences Department, College of Medicine, University of Sharjah, Sharjah, UAE
| | | |
Collapse
|
40
|
Churchward MA, Michaud ER, Mullish BH, Miguens Blanco J, Garcia Perez I, Marchesi JR, Xu H, Kao D, Todd KG. Short-chain fatty and carboxylic acid changes associated with fecal microbiota transplant communally influence microglial inflammation. Heliyon 2023; 9:e16908. [PMID: 37484415 PMCID: PMC10360965 DOI: 10.1016/j.heliyon.2023.e16908] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 05/24/2023] [Accepted: 05/31/2023] [Indexed: 07/25/2023] Open
Abstract
The intestinal microbiota has been proposed to influence human mental health and cognition through the gut-brain axis. Individuals experiencing recurrent Clostridioides difficile infection (rCDI) frequently report depressive symptoms, which are improved after fecal microbiota transplantation (FMT); however, mechanisms underlying this association are poorly understood. Short-chain fatty acids and carboxylic acids (SCCA) produced by the intestinal microbiota cross the blood brain barrier and have been proposed to contribute to gut-brain communication. We hypothesized that changes in serum SCCA measured before and after successful FMT for rCDI influences the inflammatory response of microglia, the resident immune cells of the central nervous system. Serum SCCA were quantified using gas chromatography-mass spectroscopy from 38 patients who participated in a randomized trial comparing oral capsule-vs colonoscopy-delivered FMT for rCDI, and quality of life was assessed by SF-36 at baseline, 4, and 12 weeks after FMT treatment. Successful FMT was associated with improvements in mental and physical health, as well as significant changes in a number of circulating SCCA, including increased butyrate, 2-methylbutyrate, valerate, and isovalerate, and decreased 2-hydroxybutyrate. Primary cultured microglia were treated with SCCA and the response to a pro-inflammatory stimulus was measured. Treatment with a combination of SCCA based on the post-FMT serum profile, but not single SCCA species, resulted in significantly reduced inflammatory response including reduced cytokine release, reduced nitric oxide release, and accumulation of intracellular lipid droplets. This suggests that both levels and diversity of SCCA may be an important contributor to gut-brain communication.
Collapse
Affiliation(s)
- Matthew A. Churchward
- Department of Biological and Environmental Sciences, Concordia University of Edmonton, AB, T5B 4E4, Canada
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, AB, T6G 2R3, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, T6G 2R3, Canada
| | - Emily R. Michaud
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, AB, T6G 2R3, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, T6G 2R3, Canada
| | - Benjamin H. Mullish
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W2 1NY, UK
| | - Jesús Miguens Blanco
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W2 1NY, UK
| | - Isabel Garcia Perez
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W2 1NY, UK
| | - Julian R. Marchesi
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W2 1NY, UK
| | - Huiping Xu
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine Indianapolis, IN, USA, 46202
| | - Dina Kao
- Division of Gastroenterology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, T6G 2R3, Canada
| | - Kathryn G. Todd
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, AB, T6G 2R3, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, T6G 2R3, Canada
- Department of Biomedical Engineering, University of Alberta, Edmonton, AB, T6G 2R3, Canada
| |
Collapse
|
41
|
Cuervo-Zanatta D, Syeda T, Sánchez-Valle V, Irene-Fierro M, Torres-Aguilar P, Torres-Ramos MA, Shibayama-Salas M, Silva-Olivares A, Noriega LG, Torres N, Tovar AR, Ruminot I, Barros LF, García-Mena J, Perez-Cruz C. Dietary Fiber Modulates the Release of Gut Bacterial Products Preventing Cognitive Decline in an Alzheimer's Mouse Model. Cell Mol Neurobiol 2023; 43:1595-1618. [PMID: 35953741 PMCID: PMC11412426 DOI: 10.1007/s10571-022-01268-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 07/28/2022] [Indexed: 12/11/2022]
Abstract
Fiber intake is associated with a lower risk for Alzheimer´s disease (AD) in older adults. Intake of plant-based diets rich in soluble fiber promotes the production of short-chain fatty acids (SCFAs: butyrate, acetate, propionate) by gut bacteria. Butyrate administration has antiinflammatory actions, but propionate promotes neuroinflammation. In AD patients, gut microbiota dysbiosis is a common feature even in the prodromal stages of the disease. It is unclear whether the neuroprotective effects of fiber intake rely on gut microbiota modifications and specific actions of SCFAs in brain cells. Here, we show that restoration of the gut microbiota dysbiosis through the intake of soluble fiber resulted in lower propionate and higher butyrate production, reduced astrocyte activation and improved cognitive function in 6-month-old male APP/PS1 mice. The neuroprotective effects were lost in antibiotic-treated mice. Moreover, propionate promoted higher glycolysis and mitochondrial respiration in astrocytes, while butyrate induced a more quiescent metabolism. Therefore, fiber intake neuroprotective action depends on the modulation of butyrate/propionate production by gut bacteria. Our data further support and provide a mechanism to explain the beneficial effects of dietary interventions rich in soluble fiber to prevent dementia and AD. Fiber intake restored the concentration of propionate and butyrate by modulating the composition of gut microbiota in male transgenic (Tg) mice with Alzheimer´s disease. Gut dysbiosis was associated with intestinal damage and high propionate levels in control diet fed-Tg mice. Fiber-rich diet restored intestinal integrity and promoted the abundance of butyrate-producing bacteria. Butyrate concentration was associated with better cognitive performance in fiber-fed Tg mice. A fiber-rich diet may prevent the development of a dysbiotic microbiome and the related cognitive dysfunction in people at risk of developing Alzheimer´s disease.
Collapse
Affiliation(s)
- Daniel Cuervo-Zanatta
- Laboratorio de Neuroplasticidad y Neurodegeneración, Departamento de Farmacologia, Centro de Investigación y de Estudios Avanzados del I.P.N. (Cinvestav), Av. IPN 2508, Ciudad de Mexico, 07360, México
- Laboratorio de Referencia y Soporte Para Genomas, Transcriptomas y Caracterización de Microbiomas, Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del I.P.N. (Cinvestav), Av. IPN 2508, Ciudad de Mexico, 07360, México
| | - Tauqeerunnisa Syeda
- Laboratorio de Neuroplasticidad y Neurodegeneración, Departamento de Farmacologia, Centro de Investigación y de Estudios Avanzados del I.P.N. (Cinvestav), Av. IPN 2508, Ciudad de Mexico, 07360, México
| | - Vicente Sánchez-Valle
- Laboratorio de Neuroplasticidad y Neurodegeneración, Departamento de Farmacologia, Centro de Investigación y de Estudios Avanzados del I.P.N. (Cinvestav), Av. IPN 2508, Ciudad de Mexico, 07360, México
| | - Mariangel Irene-Fierro
- Laboratorio de Neuroplasticidad y Neurodegeneración, Departamento de Farmacologia, Centro de Investigación y de Estudios Avanzados del I.P.N. (Cinvestav), Av. IPN 2508, Ciudad de Mexico, 07360, México
| | - Pablo Torres-Aguilar
- Unidad Periférica de Neurociencias, Instituto de Neurología y Neurocirugía Manuel Velasco Suárez (INNNMVS), Ciudad de Mexico, 14269, México
| | - Mónica Adriana Torres-Ramos
- Unidad Periférica de Neurociencias, Instituto de Neurología y Neurocirugía Manuel Velasco Suárez (INNNMVS), Ciudad de Mexico, 14269, México
| | - Mineko Shibayama-Salas
- Departmento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del I.P.N. (Cinvestav), Av. IPN 2508, 07360, Ciudad de Mexico, Mexico
| | - Angélica Silva-Olivares
- Departmento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del I.P.N. (Cinvestav), Av. IPN 2508, 07360, Ciudad de Mexico, Mexico
| | - Lilia G Noriega
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y de la Nutrición "Salvador Zubiran" (INCMNSZ), 14080, Ciudad de México, Mexico
| | - Nimbe Torres
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y de la Nutrición "Salvador Zubiran" (INCMNSZ), 14080, Ciudad de México, Mexico
| | - Armando R Tovar
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y de la Nutrición "Salvador Zubiran" (INCMNSZ), 14080, Ciudad de México, Mexico
| | - Iván Ruminot
- Universidad San Sebastián, Facultad de Medicina y Ciencia, Centro de Estudios Científicos-CECs, Valdivia, Chile
| | - L Felipe Barros
- Universidad San Sebastián, Facultad de Medicina y Ciencia, Centro de Estudios Científicos-CECs, Valdivia, Chile
| | - Jaime García-Mena
- Laboratorio de Referencia y Soporte Para Genomas, Transcriptomas y Caracterización de Microbiomas, Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del I.P.N. (Cinvestav), Av. IPN 2508, Ciudad de Mexico, 07360, México.
| | - Claudia Perez-Cruz
- Laboratorio de Neuroplasticidad y Neurodegeneración, Departamento de Farmacologia, Centro de Investigación y de Estudios Avanzados del I.P.N. (Cinvestav), Av. IPN 2508, Ciudad de Mexico, 07360, México.
| |
Collapse
|
42
|
Al-Kaisey AM, Figgett W, Hawson J, Mackay F, Joseph SA, Kalman JM. Gut Microbiota and Atrial Fibrillation: Pathogenesis, Mechanisms and Therapies. Arrhythm Electrophysiol Rev 2023; 12:e14. [PMID: 37427301 PMCID: PMC10326663 DOI: 10.15420/aer.2022.33] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 01/23/2023] [Indexed: 07/11/2023] Open
Abstract
Over the past decade there has been an interest in understanding the role of gut microbiota in the pathogenesis of AF. A number of studies have linked the gut microbiota to the occurrence of traditional AF risk factors such as hypertension and obesity. However, it remains unclear whether gut dysbiosis has a direct effect on arrhythmogenesis in AF. This article describes the current understanding of the effect of gut dysbiosis and associated metabolites on AF. In addition, current therapeutic strategies and future directions are discussed.
Collapse
Affiliation(s)
- Ahmed M Al-Kaisey
- Department of Cardiology, Royal Melbourne Hospital, Melbourne, Australia
- Department of Medicine, University of Melbourne, Melbourne, Australia
| | - William Figgett
- Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Joshua Hawson
- Department of Cardiology, Royal Melbourne Hospital, Melbourne, Australia
- Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Fabienne Mackay
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Stephen A Joseph
- Department of Cardiology, Royal Melbourne Hospital, Melbourne, Australia
- Department of Cardiology, Western Health, Melbourne, Australia
| | - Jonathan M Kalman
- Department of Cardiology, Royal Melbourne Hospital, Melbourne, Australia
- Department of Medicine, University of Melbourne, Melbourne, Australia
| |
Collapse
|
43
|
Liu P, Liu M, Xi D, Bai Y, Ma R, Mo Y, Zeng G, Zong S. Short-chain fatty acids ameliorate spinal cord injury recovery by regulating the balance of regulatory T cells and effector IL-17 + γδ T cells. J Zhejiang Univ Sci B 2023; 24:312-325. [PMID: 37056207 PMCID: PMC10106403 DOI: 10.1631/jzus.b2200417] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 12/30/2022] [Indexed: 04/15/2023]
Abstract
Spinal cord injury (SCI) causes motor, sensory, and autonomic dysfunctions. The gut microbiome has an important role in SCI, while short-chain fatty acids (SCFAs) are one of the main bioactive mediators of microbiota. In the present study, we explored the effects of oral administration of exogenous SCFAs on the recovery of locomotor function and tissue repair in SCI. Allen's method was utilized to establish an SCI model in Sprague-Dawley (SD) rats. The animals received water containing a mixture of 150 mmol/L SCFAs after SCI. After 21 d of treatment, the Basso, Beattie, and Bresnahan (BBB) score increased, the regularity index improved, and the base of support (BOS) value declined. Spinal cord tissue inflammatory infiltration was alleviated, the spinal cord necrosis cavity was reduced, and the numbers of motor neurons and Nissl bodies were elevated. Enzyme-linked immunosorbent assay (ELISA), real-time quantitative polymerase chain reaction (qPCR), and immunohistochemistry assay revealed that the expression of interleukin (IL)-10 increased and that of IL-17 decreased in the spinal cord. SCFAs promoted gut homeostasis, induced intestinal T cells to shift toward an anti-inflammatory phenotype, and promoted regulatory T (Treg) cells to secrete IL-10, affecting Treg cells and IL-17+ γδ T cells in the spinal cord. Furthermore, we observed that Treg cells migrated from the gut to the spinal cord region after SCI. The above findings confirm that SCFAs can regulate Treg cells in the gut and affect the balance of Treg and IL-17+ γδ T cells in the spinal cord, which inhibits the inflammatory response and promotes the motor function in SCI rats. Our findings suggest that there is a relationship among gut, spinal cord, and immune cells, and the "gut-spinal cord-immune" axis may be one of the mechanisms regulating neural repair after SCI.
Collapse
Affiliation(s)
- Pan Liu
- Department of Spine Osteopathic, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Department of Orthopaedics, the Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453000, China
| | - Mingfu Liu
- Department of Spine Osteopathic, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Deshuang Xi
- Department of Spine Osteopathic, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Yiguang Bai
- Department of Spine Osteopathic, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Department of Orthopaedics, Nanchong Central Hosipital, the Second Clinical Institute of North Sichuan Medical College, Nanchong 637000, China
| | - Ruixin Ma
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning 530021, China
| | - Yaomin Mo
- Department of Spine Osteopathic, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Gaofeng Zeng
- College of Public Hygiene of Guangxi Medical University, Nanning 530021, China.
| | - Shaohui Zong
- Department of Spine Osteopathic, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China.
| |
Collapse
|
44
|
Iseli GC, Ulrich S, Schmidt A. Elucidating gut microbiota-hippocampus interactions in emerging psychosis: A new perspective for the development of early interventions for memory impairments. Front Psychiatry 2023; 14:1098019. [PMID: 37032923 PMCID: PMC10076719 DOI: 10.3389/fpsyt.2023.1098019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 02/28/2023] [Indexed: 04/11/2023] Open
Abstract
Hippocampal dysregulation might be a key pathophysiological factor for memory impairments in psychosis. Contemporary models particularly postulate that an imbalance of hippocampal glutamate and GABA leads to impaired memory and may thus serve as a therapeutic target to improve memory deficits. However, currently available interventions in early stages of psychosis do not explicitly target hippocampal pathology. A novel approach for manipulating hippocampus-dependent memory processes is provided via the gut microbiota. In this perspective article, we first recapitulate compelling evidence for emerging hippocampus pathology during the development of psychosis. The following sections emphasize the critical role of the gut microbiota in hippocampus plasticity and memory, and summarize existing evidence of gut microbiota alterations in different stages of psychosis. Finally, we propose a novel conceptual roadmap for future studies deciphering gut microbiota-hippocampus synergisms in emerging psychosis and argue that specific microbial supplementation might be promising for improving hippocampus-dependent memory deficits in early stages of psychosis.
Collapse
Affiliation(s)
| | | | - André Schmidt
- Department of Clinical Research (DKF), University Psychiatric Clinics (UPK), Translational Neurosciences, University of Basel, Basel, Switzerland
| |
Collapse
|
45
|
Ney LM, Wipplinger M, Grossmann M, Engert N, Wegner VD, Mosig AS. Short chain fatty acids: key regulators of the local and systemic immune response in inflammatory diseases and infections. Open Biol 2023; 13:230014. [PMID: 36977462 PMCID: PMC10049789 DOI: 10.1098/rsob.230014] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023] Open
Abstract
The human intestinal microbiome substantially affects human health and resistance to infections in its dynamic composition and varying release of microbial-derived metabolites. Short-chain fatty acids (SCFA) produced by commensal bacteria through fermentation of indigestible fibres are considered key regulators in orchestrating the host immune response to microbial colonization by regulating phagocytosis, chemokine and central signalling pathways of cell growth and apoptosis, thereby shaping the composition and functionality of the intestinal epithelial barrier. Although research of the last decades provided valuable insight into the pleiotropic functions of SCFAs and their capability to maintain human health, mechanistic details on how SCFAs act across different cell types and other organs are not fully understood. In this review, we provide an overview of the various functions of SCFAs in regulating cellular metabolism, emphasizing the orchestration of the immune response along the gut-brain, the gut-lung and the gut-liver axes. We discuss their potential pharmacological use in inflammatory diseases and infections and highlight new options of relevant human three-dimensional organ models to investigate and validate their biological functions in more detail.
Collapse
Affiliation(s)
- Lisa-Marie Ney
- Institute of Biochemistry II, Jena University Hospital, Kastanienallee 1, 07747 Jena, Germany
| | - Maximilian Wipplinger
- Institute of Biochemistry II, Jena University Hospital, Kastanienallee 1, 07747 Jena, Germany
| | - Martha Grossmann
- Institute of Biochemistry II, Jena University Hospital, Kastanienallee 1, 07747 Jena, Germany
| | - Nicole Engert
- Institute of Biochemistry II, Jena University Hospital, Kastanienallee 1, 07747 Jena, Germany
| | - Valentin D Wegner
- Institute of Biochemistry II, Jena University Hospital, Kastanienallee 1, 07747 Jena, Germany
| | - Alexander S Mosig
- Institute of Biochemistry II, Jena University Hospital, Kastanienallee 1, 07747 Jena, Germany
- Center for Sepsis Control and Care, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| |
Collapse
|
46
|
Salminen A. Activation of aryl hydrocarbon receptor (AhR) in Alzheimer's disease: role of tryptophan metabolites generated by gut host-microbiota. J Mol Med (Berl) 2023; 101:201-222. [PMID: 36757399 PMCID: PMC10036442 DOI: 10.1007/s00109-023-02289-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 12/19/2022] [Accepted: 01/17/2023] [Indexed: 02/10/2023]
Abstract
Gut microbiota in interaction with intestinal host tissues influences many brain functions and microbial dysbiosis has been linked with brain disorders, such as neuropsychiatric conditions and Alzheimer's disease (AD). L-tryptophan metabolites and short-chained fatty acids (SCFA) are major messengers in the microbiota-brain axis. Aryl hydrocarbon receptors (AhR) are main targets of tryptophan metabolites in brain microvessels which possess an enriched expression of AhR protein. The Ah receptor is an evolutionarily conserved, ligand-activated transcription factor which is not only a sensor of xenobiotic toxins but also a pleiotropic regulator of both developmental processes and age-related tissue degeneration. Major microbiota-produced tryptophan metabolites involve indole derivatives, e.g., indole 3-pyruvic acid, indole 3-acetaldehyde, and indoxyl sulfate, whereas indoleamine and tryptophan 2,3-dioxygenases (IDO/TDO) of intestine host cells activate the kynurenine (KYN) pathway generating KYN metabolites, many of which are activators of AhR signaling. Chronic kidney disease (CKD) increases the serum level of indoxyl sulfate which promotes AD pathogenesis, e.g., it disrupts integrity of blood-brain barrier (BBB) and impairs cognitive functions. Activation of AhR signaling disturbs vascular homeostasis in brain; (i) it controls blood flow via the renin-angiotensin system, (ii) it inactivates endothelial nitric oxide synthase (eNOS), thus impairing NO production and vasodilatation, and (iii) it induces oxidative stress, stimulates inflammation, promotes cellular senescence, and enhances calcification of vascular walls. All these alterations are evident in cerebral amyloid angiopathy (CAA) in AD pathology. Moreover, AhR signaling can disturb circadian regulation and probably affect glymphatic flow. It seems plausible that dysbiosis of gut microbiota impairs the integrity of BBB via the activation of AhR signaling and thus aggravates AD pathology. KEY MESSAGES: Dysbiosis of gut microbiota is associated with dementia and Alzheimer's disease. Tryptophan metabolites are major messengers from the gut host-microbiota to brain. Tryptophan metabolites activate aryl hydrocarbon receptor (AhR) signaling in brain. The expression of AhR protein is enriched in brain microvessels and blood-brain barrier. Tryptophan metabolites disturb brain vascular integrity via AhR signaling. Dysbiosis of gut microbiota promotes inflammation and AD pathology via AhR signaling.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, Kuopio, 70211, Finland.
| |
Collapse
|
47
|
Fock E, Parnova R. Mechanisms of Blood-Brain Barrier Protection by Microbiota-Derived Short-Chain Fatty Acids. Cells 2023; 12:cells12040657. [PMID: 36831324 PMCID: PMC9954192 DOI: 10.3390/cells12040657] [Citation(s) in RCA: 104] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/13/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
Impairment of the blood-brain barrier (BBB) integrity is implicated in the numerous neurological disorders associated with neuroinflammation, neurodegeneration and aging. It is now evident that short-chain fatty acids (SCFAs), mainly acetate, butyrate and propionate, produced by anaerobic bacterial fermentation of the dietary fiber in the intestine, have a key role in the communication between the gastrointestinal tract and nervous system and are critically important for the preservation of the BBB integrity under different pathological conditions. The effect of SCFAs on the improvement of the compromised BBB is mainly based on the decrease in paracellular permeability via restoration of junctional complex proteins affecting their transcription, intercellular localization or proteolytic degradation. This review is focused on the revealed and putative underlying mechanisms of the direct and indirect effects of SCFAs on the improvement of the barrier function of brain endothelial cells. We consider G-protein-coupled receptor-mediated effects of SCFAs, SCFAs-stimulated acetylation of histone and non-histone proteins via inhibition of histone deacetylases, and crosstalk of these signaling pathways with transcriptional factors NF-κB and Nrf2 as mainstream mechanisms of SCFA's effect on the preservation of the BBB integrity.
Collapse
Affiliation(s)
| | - Rimma Parnova
- Correspondence: ; Tel.: +7-812-552-79-01; Fax: +7-812-552-30-12
| |
Collapse
|
48
|
Freijy TM, Cribb L, Oliver G, Metri NJ, Opie RS, Jacka FN, Hawrelak JA, Rucklidge JJ, Ng CH, Sarris J. Effects of a high-prebiotic diet versus probiotic supplements versus synbiotics on adult mental health: The "Gut Feelings" randomised controlled trial. Front Neurosci 2023; 16:1097278. [PMID: 36815026 PMCID: PMC9940791 DOI: 10.3389/fnins.2022.1097278] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 12/15/2022] [Indexed: 02/09/2023] Open
Abstract
Background Preliminary evidence supports the use of dietary interventions and gut microbiota-targeted interventions such as probiotic or prebiotic supplementation for improving mental health. We report on the first randomised controlled trial (RCT) to examine the effects of a high-prebiotic dietary intervention and probiotic supplements on mental health. Methods "Gut Feelings" was an 8-week, 2 × 2 factorial RCT of 119 adults with moderate psychological distress and low prebiotic food intake. Treatment arms: (1) probiotic supplement and diet-as-usual (probiotic group); (2) high-prebiotic diet and placebo supplement (prebiotic diet group); (3) probiotic supplement and high-prebiotic diet (synbiotic group); and (4) placebo supplement and diet-as-usual (placebo group). The primary outcome was assessment of total mood disturbance (TMD; Profile of Mood States Short Form) from baseline to 8 weeks. Secondary outcomes included anxiety, depression, stress, sleep, and wellbeing measures. Results A modified intention-to-treat analysis using linear mixed effects models revealed that the prebiotic diet reduced TMD relative to placebo at 8 weeks [Cohen's d = -0.60, 95% confidence interval (CI) = -1.18, -0.03; p = 0.039]. There was no evidence of symptom improvement from the probiotic (d = -0.19, 95% CI = -0.75, 0.38; p = 0.51) or synbiotic treatments (d = -0.03, 95% CI = -0.59, 0.53; p = 0.92). Improved anxiety, stress, and sleep were noted in response to the prebiotic diet while the probiotic tentatively improved wellbeing, relative to placebo. No benefit was found in response to the synbiotic intervention. All treatments were well tolerated with few adverse events. Conclusion A high-prebiotic dietary intervention may improve mood, anxiety, stress, and sleep in adults with moderate psychological distress and low prebiotic intake. A synbiotic combination of high-prebiotic diet and probiotic supplement does not appear to have a beneficial effect on mental health outcomes, though further evidence is required. Results are limited by the relatively small sample size. Clinical trial registration https://www.anzctr.org.au/Trial/Registration/TrialReview.aspx?id=372753, identifier ACTRN12617000795392.
Collapse
Affiliation(s)
- Tanya M. Freijy
- Professorial Unit, The Melbourne Clinic, Department of Psychiatry, The University of Melbourne, Melbourne, VIC, Australia,Faculty of Medicine, Dentistry and Health Sciences, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia
| | - Lachlan Cribb
- Professorial Unit, The Melbourne Clinic, Department of Psychiatry, The University of Melbourne, Melbourne, VIC, Australia
| | - Georgina Oliver
- Professorial Unit, The Melbourne Clinic, Department of Psychiatry, The University of Melbourne, Melbourne, VIC, Australia
| | - Najwa-Joelle Metri
- NICM Health Research Institute, Western Sydney University, Westmead, NSW, Australia
| | - Rachelle S. Opie
- IPAN, School of Exercise and Nutrition Sciences, Deakin University, Melbourne, VIC, Australia
| | - Felice N. Jacka
- School of Medicine, Food and Mood Centre, IMPACT Strategic Research Centre, Deakin University, Melbourne, VIC, Australia,Centre for Adolescent Health, Murdoch Children’s Research Institute, Melbourne, VIC, Australia,College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, OLD, Australia
| | - Jason A. Hawrelak
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart, TAS, Australia,Human Nutrition and Functional Medicine Department, University of Western States, Portland, OR, United States
| | - Julia J. Rucklidge
- School of Psychology, Speech and Hearing, University of Canterbury, Christchurch, New Zealand
| | - Chee H. Ng
- Professorial Unit, The Melbourne Clinic, Department of Psychiatry, The University of Melbourne, Melbourne, VIC, Australia
| | - Jerome Sarris
- Professorial Unit, The Melbourne Clinic, Department of Psychiatry, The University of Melbourne, Melbourne, VIC, Australia,Faculty of Medicine, Dentistry and Health Sciences, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia,NICM Health Research Institute, Western Sydney University, Westmead, NSW, Australia,*Correspondence: Jerome Sarris,
| |
Collapse
|
49
|
Molecular Landscape of Tourette's Disorder. Int J Mol Sci 2023; 24:ijms24021428. [PMID: 36674940 PMCID: PMC9865021 DOI: 10.3390/ijms24021428] [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: 11/28/2022] [Revised: 12/29/2022] [Accepted: 01/01/2023] [Indexed: 01/12/2023] Open
Abstract
Tourette's disorder (TD) is a highly heritable childhood-onset neurodevelopmental disorder and is caused by a complex interplay of multiple genetic and environmental factors. Yet, the molecular mechanisms underlying the disorder remain largely elusive. In this study, we used the available omics data to compile a list of TD candidate genes, and we subsequently conducted tissue/cell type specificity and functional enrichment analyses of this list. Using genomic data, we also investigated genetic sharing between TD and blood and cerebrospinal fluid (CSF) metabolite levels. Lastly, we built a molecular landscape of TD through integrating the results from these analyses with an extensive literature search to identify the interactions between the TD candidate genes/proteins and metabolites. We found evidence for an enriched expression of the TD candidate genes in four brain regions and the pituitary. The functional enrichment analyses implicated two pathways ('cAMP-mediated signaling' and 'Endocannabinoid Neuronal Synapse Pathway') and multiple biological functions related to brain development and synaptic transmission in TD etiology. Furthermore, we found genetic sharing between TD and the blood and CSF levels of 39 metabolites. The landscape of TD not only provides insights into the (altered) molecular processes that underlie the disease but, through the identification of potential drug targets (such as FLT3, NAALAD2, CX3CL1-CX3CR1, OPRM1, and HRH2), it also yields clues for developing novel TD treatments.
Collapse
|
50
|
Ma H, Yang L, Liu Y, Yan R, Wang R, Zhang P, Bai Z, Liu Y, Ren Y, Li Y, Jiang X, Wang T, Ma P, Zhang Q, Li A, Guo M, Zhang X, Jia S, Wang H. Butyrate suppresses atherosclerotic inflammation by regulating macrophages and polarization via GPR43/HDAC-miRNAs axis in ApoE-/- mice. PLoS One 2023; 18:e0282685. [PMID: 36888629 PMCID: PMC9994734 DOI: 10.1371/journal.pone.0282685] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 02/17/2023] [Indexed: 03/09/2023] Open
Abstract
Chronic low-grade inflammation is regarded to an important signature of atherosclerosis (AS). Macrophage (Mψ) and related polarization have been demonstrated to play a crucial role in the occurrence and development of AS inflammation. Butyrate, a bioactive molecule produced by the intestinal flora, has been increasingly demonstrated to exhibit a vital role for regulating the inflammation in chronic metabolic diseases. However, the effectiveness and multiple anti-inflammation mechanisms of butyrate on AS still need to be further understood. ApoE-/- mice fed with high-fat diet as AS model were administered with sodium butyrate (NaB) for 14 weeks of treatment. Our results showed that the atherosclerotic lesion in the AS group was dramatically reduced after NaB intervention. Moreover, deteriorated routine parameters of AS including body weights (BWs), low-density lipoprotein (LDL-C), triglyceride (TG), total cholesterol (TC) were significantly reversed by NaB administration. Abnormal elevated plasma and aorta pro-inflammatory indicators including interleukin (IL)-1β, IL-6, IL-17A, tumor necrosis factor (TNF)-α and lipopolysaccharide (LPS), as well as reduced anti-inflammatory IL-10 in plasma were respectively rectified after NaB administration. Consistently, accumulated Mψ and associated imbalance of polarization in the arota were attenuated with NaB treatment. Importantly, we demonstrated that the suppression of Mψ and associated polarization of NaB was dependent on binding G-protein coupled receptor (GPR) and inhibiting histone deacetylase HDAC3. Moreover, we found that intestinal butyrate-producing bacteria, anti-inflammatory bacteria and intestinal tight junction protein zonula occludens-1 (ZO)-1 may contribute to this effectiveness. Intriguingly, according to transcriptome sequencing of atherosclerotic aorta, 29 elevated and 24 reduced miRNAs were found after NaB treatment, especially miR-7a-5p, suggesting that non-coding RNA may possess a potential role in the protection of NaB against AS. Correlation analysis showed that there were close complicated interactions among gut microbiota, inflammation and differential miRNAs. Collectively, this study revealed that dietary NaB may ameliorate atherosclerotic inflammation by regulating Mψ polarization via GPR43/HDAC-miRNAs axis in ApoE-/- mice.
Collapse
Affiliation(s)
- Huiyan Ma
- Clinical Medical College, Ningxia Medical University, Yinchuan, China
- Heart Centre & Department of Cardiovascular Diseases, General Hospital of Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, China
| | - Libo Yang
- Heart Centre & Department of Cardiovascular Diseases, General Hospital of Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, China
| | - Yajuan Liu
- Clinical Medical College, Ningxia Medical University, Yinchuan, China
- Heart Centre & Department of Cardiovascular Diseases, General Hospital of Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, China
| | - Ru Yan
- Heart Centre & Department of Cardiovascular Diseases, General Hospital of Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, China
| | - Rui Wang
- Clinical Medical College, Ningxia Medical University, Yinchuan, China
| | - Peng Zhang
- Clinical Medical College, Ningxia Medical University, Yinchuan, China
| | - Zhixia Bai
- Clinical Medical College, Ningxia Medical University, Yinchuan, China
- Heart Centre & Department of Cardiovascular Diseases, General Hospital of Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, China
| | - Yuanyuan Liu
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Yi Ren
- Clinical Medical College, Ningxia Medical University, Yinchuan, China
| | - Yiwei Li
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Xin Jiang
- Clinical Medical College, Ningxia Medical University, Yinchuan, China
| | - Ting Wang
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Ping Ma
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Qining Zhang
- Clinical Medical College, Ningxia Medical University, Yinchuan, China
- Heart Centre & Department of Cardiovascular Diseases, General Hospital of Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, China
| | - Aifei Li
- Heart Centre & Department of Cardiovascular Diseases, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Mixue Guo
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Xiaoxia Zhang
- College of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan, China
- * E-mail: (XZ); (SJ); (HW)
| | - Shaobin Jia
- Heart Centre & Department of Cardiovascular Diseases, General Hospital of Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, China
- * E-mail: (XZ); (SJ); (HW)
| | - Hao Wang
- Clinical Medical College, Ningxia Medical University, Yinchuan, China
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
- * E-mail: (XZ); (SJ); (HW)
| |
Collapse
|