1
|
Saadh MJ, Mustafa AN, Mustafa MA, S RJ, Dabis HK, Prasad GVS, Mohammad IJ, Adnan A, Idan AH. The role of gut-derived short-chain fatty acids in Parkinson's disease. Neurogenetics 2024; 25:307-336. [PMID: 39266892 DOI: 10.1007/s10048-024-00779-3] [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: 06/21/2024] [Accepted: 08/29/2024] [Indexed: 09/14/2024]
Abstract
The emerging function of short-chain fatty acids (SCFAs) in Parkinson's disease (PD) has been investigated in this article. SCFAs, which are generated via the fermentation of dietary fiber by gut microbiota, have been associated with dysfunction of the gut-brain axis and, neuroinflammation. These processes are integral to the development of PD. This article examines the potential therapeutic implications of SCFAs in the management of PD, encompassing their capacity to modulate gastrointestinal permeability, neuroinflammation, and neuronal survival, by conducting an extensive literature review. As a whole, this article emphasizes the potential therapeutic utility of SCFAs as targets for the management and treatment of PD.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan.
| | | | - Mohammed Ahmed Mustafa
- School of Pharmacy-Adarsh Vijendra Institute of Pharmaceutical Sciences, Shobhit University, Gangoh, Uttar Pradesh, 247341, India
- Department of Pharmacy, Arka Jain University, Jamshedpur, Jharkhand, 831001, India
| | - Renuka Jyothi S
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | | | - G V Siva Prasad
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra, Pradesh-531162, India
| | - Imad Jassim Mohammad
- College of Health and Medical Technology, National University of Science and Technology, Dhi Qar, 64001, Iraq
| | - Ahmed Adnan
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | | |
Collapse
|
2
|
Buoso C, Seifert M, Lang M, Griffith CM, Talavera Andújar B, Castelo Rueda MP, Fischer C, Doerrier C, Talasz H, Zanon A, Pramstaller PP, Schymanski EL, Pichler I, Weiss G. Dopamine‑iron homeostasis interaction rescues mitochondrial fitness in Parkinson's disease. Neurobiol Dis 2024; 196:106506. [PMID: 38648865 DOI: 10.1016/j.nbd.2024.106506] [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: 12/21/2023] [Revised: 04/17/2024] [Accepted: 04/17/2024] [Indexed: 04/25/2024] Open
Abstract
Imbalances of iron and dopamine metabolism along with mitochondrial dysfunction have been linked to the pathogenesis of Parkinson's disease (PD). We have previously suggested a direct link between iron homeostasis and dopamine metabolism, as dopamine can increase cellular uptake of iron into macrophages thereby promoting oxidative stress responses. In this study, we investigated the interplay between iron, dopamine, and mitochondrial activity in neuroblastoma SH-SY5Y cells and human induced pluripotent stem cell (hiPSC)-derived dopaminergic neurons differentiated from a healthy control and a PD patient with a mutation in the α-synuclein (SNCA) gene. In SH-SY5Y cells, dopamine treatment resulted in increased expression of the transmembrane iron transporters transferrin receptor 1 (TFR1), ferroportin (FPN), and mitoferrin2 (MFRN2) and intracellular iron accumulation, suggesting that dopamine may promote iron uptake. Furthermore, dopamine supplementation led to reduced mitochondrial fitness including decreased mitochondrial respiration, increased cytochrome c control efficiency, reduced mtDNA copy number and citrate synthase activity, increased oxidative stress and impaired aconitase activity. In dopaminergic neurons derived from a healthy control individual, dopamine showed comparable effects as observed in SH-SY5Y cells. The hiPSC-derived PD neurons harboring an endogenous SNCA mutation demonstrated altered mitochondrial iron homeostasis, reduced mitochondrial capacity along with increased oxidative stress and alterations of tricarboxylic acid cycle linked metabolic pathways compared with control neurons. Importantly, dopamine treatment of PD neurons promoted a rescue effect by increasing mitochondrial respiration, activating antioxidant stress response, and normalizing altered metabolite levels linked to mitochondrial function. These observations provide evidence that dopamine affects iron homeostasis, intracellular stress responses and mitochondrial function in healthy cells, while dopamine supplementation can restore the disturbed regulatory network in PD cells.
Collapse
Affiliation(s)
- Chiara Buoso
- Institute for Biomedicine, Eurac Research, 39100 Bolzano, Italy; Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Markus Seifert
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria; Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Martin Lang
- Institute for Biomedicine, Eurac Research, 39100 Bolzano, Italy
| | - Corey M Griffith
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Begoña Talavera Andújar
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | | | - Christine Fischer
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | | | - Heribert Talasz
- Institute of Medical Biochemistry, Protein Core Facility, Biocenter Innsbruck, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | | | | | - Emma L Schymanski
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Irene Pichler
- Institute for Biomedicine, Eurac Research, 39100 Bolzano, Italy.
| | - Guenter Weiss
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria; Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, 6020 Innsbruck, Austria.
| |
Collapse
|
3
|
Duan WX, Wang F, Liu JY, Liu CF. Relationship Between Short-chain Fatty Acids and Parkinson's Disease: A Review from Pathology to Clinic. Neurosci Bull 2024; 40:500-516. [PMID: 37755674 PMCID: PMC11003953 DOI: 10.1007/s12264-023-01123-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 05/15/2023] [Indexed: 09/28/2023] Open
Abstract
Parkinson's disease (PD) is a complicated neurodegenerative disease, characterized by the accumulation of α-synuclein (α-syn) in Lewy bodies and neurites, and massive loss of midbrain dopamine neurons. Increasing evidence suggests that gut microbiota and microbial metabolites are involved in the development of PD. Among these, short-chain fatty acids (SCFAs), the most abundant microbial metabolites, have been proven to play a key role in brain-gut communication. In this review, we analyze the role of SCFAs in the pathology of PD from multiple dimensions and summarize the alterations of SCFAs in PD patients as well as their correlation with motor and non-motor symptoms. Future research should focus on further elucidating the role of SCFAs in neuroinflammation, as well as developing novel strategies employing SCFAs and their derivatives to treat PD.
Collapse
Affiliation(s)
- Wen-Xiang Duan
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Fen Wang
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Jun-Yi Liu
- Department of Neurology, Dushu Lake Hospital affiliated to Soochow University, Suzhou, 215125, China.
| | - Chun-Feng Liu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China.
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
4
|
Chalova P, Tazky A, Skultety L, Minichova L, Chovanec M, Ciernikova S, Mikus P, Piestansky J. Determination of short-chain fatty acids as putative biomarkers of cancer diseases by modern analytical strategies and tools: a review. Front Oncol 2023; 13:1110235. [PMID: 37441422 PMCID: PMC10334191 DOI: 10.3389/fonc.2023.1110235] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 06/13/2023] [Indexed: 07/15/2023] Open
Abstract
Short-chain fatty acids (SCFAs) are the main metabolites produced by bacterial fermentation of non-digestible carbohydrates in the gastrointestinal tract. They can be seen as the major flow of carbon from the diet, through the microbiome to the host. SCFAs have been reported as important molecules responsible for the regulation of intestinal homeostasis. Moreover, these molecules have a significant impact on the immune system and are able to affect inflammation, cardiovascular diseases, diabetes type II, or oncological diseases. For this purpose, SCFAs could be used as putative biomarkers of various diseases, including cancer. A potential diagnostic value may be offered by analyzing SCFAs with the use of advanced analytical approaches such as gas chromatography (GC), liquid chromatography (LC), or capillary electrophoresis (CE) coupled with mass spectrometry (MS). The presented review summarizes the importance of analyzing SCFAs from clinical and analytical perspective. Current advances in the analysis of SCFAs focused on sample pretreatment, separation strategy, and detection methods are highlighted. Additionally, it also shows potential areas for the development of future diagnostic tools in oncology and other varieties of diseases based on targeted metabolite profiling.
Collapse
Affiliation(s)
- Petra Chalova
- Department of Pharmaceutical Analysis and Nuclear Pharmacy, Faculty of Pharmacy, Comenius University, Bratislava, Slovakia
- Biomedical Research Center of the Slovak Academy of Sciences, Institute of Virology, Bratislava, Slovakia
| | - Anton Tazky
- Department of Pharmaceutical Analysis and Nuclear Pharmacy, Faculty of Pharmacy, Comenius University, Bratislava, Slovakia
- Toxicological and Antidoping Center, Faculty of Pharmacy, Comenius University, Bratislava, Slovakia
| | - Ludovit Skultety
- Biomedical Research Center of the Slovak Academy of Sciences, Institute of Virology, Bratislava, Slovakia
- Institute of Microbiology, Academy of Sciences of the Czech Republic, Prague, Czechia
| | - Lenka Minichova
- Biomedical Research Center of the Slovak Academy of Sciences, Institute of Virology, Bratislava, Slovakia
| | - Michal Chovanec
- 2nd Department of Oncology, Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
| | - Sona Ciernikova
- Biomedical Research Center of the Slovak Academy of Sciences, Cancer Research Institute, Bratislava, Slovakia
| | - Peter Mikus
- Department of Pharmaceutical Analysis and Nuclear Pharmacy, Faculty of Pharmacy, Comenius University, Bratislava, Slovakia
- Toxicological and Antidoping Center, Faculty of Pharmacy, Comenius University, Bratislava, Slovakia
| | - Juraj Piestansky
- Toxicological and Antidoping Center, Faculty of Pharmacy, Comenius University, Bratislava, Slovakia
- Department of Galenic Pharmacy, Faculty of Pharmacy, Comenius University, Bratislava, Slovakia
| |
Collapse
|
5
|
Qi A, Liu L, Zhang J, Chen S, Xu S, Chen Y, Zhang L, Cai C. Plasma Metabolic Analysis Reveals the Dysregulation of Short-Chain Fatty Acid Metabolism in Parkinson's Disease. Mol Neurobiol 2023; 60:2619-2631. [PMID: 36690885 DOI: 10.1007/s12035-022-03157-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 12/01/2022] [Indexed: 01/25/2023]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder, characterized by high morbidity, high disability rate, and slow course of disease. The clinical diagnostic method of PD is complex and time-consuming, and there is no clear biomarker for clinical use. We aimed to investigate the plasma metabolites in PD and find out potential biomarkers with diagnostic ability. In the analysis of more than 40 metabolites including short-chain fatty acids, long-chain fatty acids, amino acids, and carbohydrates, the difference of short-chain fatty acids was observed. Acetic acid concentration was higher in PD than in healthy controls, and propanoic acid and 2,3,4-trihydroxybutyric acid were lower in PD. Compared with the early stage of PD, acetic acid increased significantly in the advanced stage of PD. Propanoic acid increased significantly in medicated PD compared with drug naïve PD. ROC analysis revealed acetic acid discriminated PD from healthy controls with 100% sensitivity, 88.9% specificity, and an area under the curve (AUC) of 0.981, and propanoic acid discriminated PD from healthy controls with an AUC of 0.981, 100% sensitivity, and 94.4% specificity. Acetic acid and propanoic acid may be a potential biomarker for differentiating PD from health, and the propanoic acid was evaluated as the most potential diagnostic marker because of its extremely high sensitivity and specificity.
Collapse
Affiliation(s)
- Ao Qi
- Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang, 524023, Guangdong, China
| | - Lulu Liu
- The First DongGuan Affiliated Hospital of Guangdong Medical University, Dongguan, 523475, Guangdong, China
| | - Junjie Zhang
- Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang, 524023, Guangdong, China
- School of Pharmacy, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Simei Chen
- Neurology Department of Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Simin Xu
- Neurology Department of Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Yusen Chen
- Neurology Department of Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China.
| | - Lijiang Zhang
- Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang, 524023, Guangdong, China.
- School of Pharmacy, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China.
| | - Chun Cai
- Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang, 524023, Guangdong, China.
- School of Pharmacy, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China.
| |
Collapse
|
6
|
Weng H, Song W, Fu K, Guan Y, Cai G, Huang E, Chen X, Zou H, Ye Q. Proteomic profiling reveals the potential mechanisms and regulatory targets of sirtuin 4 in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced Parkinson's mouse model. Front Neurosci 2023; 16:1035444. [PMID: 36760798 PMCID: PMC9905825 DOI: 10.3389/fnins.2022.1035444] [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: 09/02/2022] [Accepted: 12/06/2022] [Indexed: 01/26/2023] Open
Abstract
Introduction Parkinson's disease (PD), as a common neurodegenerative disease, currently has no effective therapeutic approaches to delay or stop its progression. There is an urgent need to further define its pathogenesis and develop new therapeutic targets. An increasing number of studies have shown that members of the sirtuin (SIRT) family are differentially involved in neurodegenerative diseases, indicating their potential to serve as targets in therapeutic strategies. Mitochondrial SIRT4 possesses multiple enzymatic activities, such as deacetylase, ADP ribosyltransferase, lipoamidase, and deacylase activities, and exhibits different enzymatic activities and target substrates in different tissues and cells; thus, mitochondrial SIRT4 plays an integral role in regulating metabolism. However, the role and mechanism of SIRT4 in PD are not fully understood. This study aimed to investigate the potential mechanism and possible regulatory targets of SIRT4 in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mice. Methods The expression of the SIRT4 protein in the MPTP-induced PD mouse mice or key familial Parkinson disease protein 7 knockout (DJ-1 KO) rat was compared against the control group by western blot assay. Afterwards, quantitative proteomics and bioinformatics analyses were performed to identify altered proteins in the vitro model and reveal the possible functional role of SIRT4. The most promising molecular target of SIRT4 were screened and validated by viral transfection, western blot assay and reverse transcription quantitative PCR (RT-qPCR) assays. Results The expression of the SIRT4 protein was found to be altered both in the MPTP-induced PD mouse mice and DJ-1KO rats. Following the viral transfection of SIRT4, a quantitative proteomics analysis identified 5,094 altered proteins in the vitro model, including 213 significantly upregulated proteins and 222 significantly downregulated proteins. The results from bioinformatics analyses indicated that SIRT4 mainly affected the ribosomal pathway, propionate metabolism pathway, peroxisome proliferator-activated receptor (PPAR) signaling pathway and peroxisome pathway in cells, and we screened 25 potential molecular targets. Finally, only fatty acid binding protein 4 (FABP4) in the PPAR signaling pathway was regulated by SIRT4 among the 25 molecules. Importantly, the alterations in FABP4 and PPARγ were verified in the MPTP-induced PD mouse model. Discussion Our results indicated that FABP4 in the PPAR signaling pathway is the most promising molecular target of SIRT4 in an MPTP-induced mouse model and revealed the possible functional role of SIRT4. This study provides a reference for future drug development and mechanism research with SIRT4 as a target or biomarker.
Collapse
Affiliation(s)
- Huidan Weng
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China,Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, Fuzhou, China
| | - Wenjing Song
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China,Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, Fuzhou, China
| | - Kangyue Fu
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yunqian Guan
- Cell Therapy Center, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Guoen Cai
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China,Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, Fuzhou, China
| | - En Huang
- The School of Basic Medical Sciences, Fujian Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Medical University, Fuzhou, China
| | - Xiaochun Chen
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China,Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, Fuzhou, China
| | - Haiqiang Zou
- Department of Neurosurgery, General Hospital of Southern Theatre Command, PLA, Guangzhou, Guangdong, China,Haiqiang Zou,
| | - Qinyong Ye
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China,Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, Fuzhou, China,*Correspondence: Qinyong Ye,
| |
Collapse
|
7
|
Xie NN, Wu CY, Ge Q, Zhou J, Long F, Mao Q, Li SL, Shen H. Structure-specific antitumor effects and potential gut microbiota-involved mechanisms of ginseng polysaccharides on B16F10 melanoma-bearing mice. Food Funct 2023; 14:796-809. [PMID: 36607268 DOI: 10.1039/d2fo03383f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Ginseng polysaccharides (GPs) have shown gut microbiota-related antitumor effects. However, the relation between their structures and antitumor functions remains unknown. Here, crude polysaccharide (GP-c) and its fractions neutral polysaccharide (GP-n) and pectin (GP-a) were prepared for structure characterization and anti-B16F10 melanoma effect evaluation, and their influence on gut microbiota diversities and short-chain fatty acids (SCFAs) were also analyzed. Spearman correlations among the altered gut microbiota, SCFAs, and antitumor effects were conducted to elucidate the structure-function relationships. It was shown that the structures of GP-c, GP-n, and GP-a varied in monosaccharide composition and molecular weight distribution. GP-n and GP-c showed anti-melanoma effects, whereas GP-a promoted its growth slightly. GP-n and GP-c restored SCFAs levels such as acetic acid and butyric acid; moreover, it improved the gut microbiota ecosystem by upregulating the abundance of Allobaculum and Bifidobacterium. However, the restoration effect of GP-a was weak, or even worse. In addition, these two bacteria were negatively correlated with the tumor weight and related with the altered SCFAs. In conclusion, GP-n is essential for the anti-melanoma effects of GP, and the potential mechanisms might be related with its specific regulation of Allobaculum and Bifidobacterium abundance, and tumor-associated SCFAs levels. The outcomes highlighted here enable a deeper insight into the structure-function relationship of GP and propose new opinions on its antitumor effect.
Collapse
Affiliation(s)
- Ni-Na Xie
- Department of Pharmaceutical Analysis, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, People's Republic of China.
| | - Cheng-Ying Wu
- Department of Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine and Jiangsu Branch of China Academy of Chinese Medical Sciences, Nanjing 210028, People's Republic of China.
| | - Qiong Ge
- Department of Pharmaceutical Analysis, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, People's Republic of China.
| | - Jing Zhou
- Department of Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine and Jiangsu Branch of China Academy of Chinese Medical Sciences, Nanjing 210028, People's Republic of China.
| | - Fang Long
- Department of Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine and Jiangsu Branch of China Academy of Chinese Medical Sciences, Nanjing 210028, People's Republic of China.
| | - Qian Mao
- Department of Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine and Jiangsu Branch of China Academy of Chinese Medical Sciences, Nanjing 210028, People's Republic of China.
| | - Song-Lin Li
- Department of Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine and Jiangsu Branch of China Academy of Chinese Medical Sciences, Nanjing 210028, People's Republic of China.
| | - Hong Shen
- Department of Pharmaceutical Analysis, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, People's Republic of China.
| |
Collapse
|
8
|
The Interplay between Gut Microbiota and Parkinson's Disease: Implications on Diagnosis and Treatment. Int J Mol Sci 2022; 23:ijms232012289. [PMID: 36293176 PMCID: PMC9603886 DOI: 10.3390/ijms232012289] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/05/2022] [Accepted: 10/12/2022] [Indexed: 11/05/2022] Open
Abstract
The bidirectional interaction between the gut microbiota (GM) and the Central Nervous System, the so-called gut microbiota brain axis (GMBA), deeply affects brain function and has an important impact on the development of neurodegenerative diseases. In Parkinson’s disease (PD), gastrointestinal symptoms often precede the onset of motor and non-motor manifestations, and alterations in the GM composition accompany disease pathogenesis. Several studies have been conducted to unravel the role of dysbiosis and intestinal permeability in PD onset and progression, but the therapeutic and diagnostic applications of GM modifying approaches remain to be fully elucidated. After a brief introduction on the involvement of GMBA in the disease, we present evidence for GM alterations and leaky gut in PD patients. According to these data, we then review the potential of GM-based signatures to serve as disease biomarkers and we highlight the emerging role of probiotics, prebiotics, antibiotics, dietary interventions, and fecal microbiota transplantation as supportive therapeutic approaches in PD. Finally, we analyze the mutual influence between commonly prescribed PD medications and gut-microbiota, and we offer insights on the involvement also of nasal and oral microbiota in PD pathology, thus providing a comprehensive and up-to-date overview on the role of microbial features in disease diagnosis and treatment.
Collapse
|
9
|
Tan AH, Lim SY, Lang AE. The microbiome-gut-brain axis in Parkinson disease - from basic research to the clinic. Nat Rev Neurol 2022; 18:476-495. [PMID: 35750883 DOI: 10.1038/s41582-022-00681-2] [Citation(s) in RCA: 133] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/27/2022] [Indexed: 12/12/2022]
Abstract
Evidence for a close bidirectional link between the brain and the gut has led to a paradigm shift in neurology, especially in the case of Parkinson disease (PD), in which gastrointestinal dysfunction is a prominent feature. Over the past decade, numerous high-quality preclinical and clinical publications have shed light on the highly complex relationship between the gut and the brain in PD, providing potential for the development of new biomarkers and therapeutics. With the advent of high-throughput sequencing, the role of the gut microbiome has been specifically highlighted. Here, we provide a critical review of the literature on the microbiome-gut-brain axis in PD and present perspectives that will be useful for clinical practice. We begin with an overview of the gut-brain axis in PD, including the potential roles and interrelationships of the vagus nerve, α-synuclein in the enteric nervous system, altered intestinal permeability and inflammation, and gut microbes and their metabolic activities. The sections that follow synthesize the proposed roles of gut-related factors in the development and progression of, in responses to PD treatment, and as therapeutic targets. Finally, we summarize current knowledge gaps and challenges and delineate future directions for the field.
Collapse
Affiliation(s)
- Ai Huey Tan
- Division of Neurology, Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia. .,Mah Pooi Soo & Tan Chin Nam Centre for Parkinson's & Related Disorders, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.
| | - Shen Yang Lim
- Division of Neurology, Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.,Mah Pooi Soo & Tan Chin Nam Centre for Parkinson's & Related Disorders, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Anthony E Lang
- Edmond J. Safra Program in Parkinson's Disease, Toronto Western Hospital, Toronto, Ontario, Canada.,Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, Ontario, Canada
| |
Collapse
|
10
|
Gut microbial metabolites in Parkinson's disease: Association with lifestyle, disease characteristics, and treatment status. Neurobiol Dis 2022; 170:105780. [PMID: 35654277 PMCID: PMC9241494 DOI: 10.1016/j.nbd.2022.105780] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 05/13/2022] [Accepted: 05/25/2022] [Indexed: 12/11/2022] Open
Abstract
There is growing appreciation of the importance of the intestinal microbiota in Parkinson’s disease (PD), and one potential mechanism by which the intestinal microbiota can communicate with the brain is via bacteria-derived metabolites. In this study, plasma levels of bacterial-derived metabolites including trimethylamine-N-oxide (TMAO), short chain fatty acids (SCFA), the branched chain fatty acid isovalerate, succinate, and lactate were evaluated in PD subjects (treatment naïve and treated) which were compared to (1) population controls, (2) spousal / household controls (similar lifestyle to PD subjects), and (3) subjects with multiple system atrophy (MSA). Analyses revealed an increase in the TMAO pathway in PD subjects which was independent of medication status, disease characteristics, and lifestyle. Lactic acid was decreased in treated PD subjects, succinic acid positively correlated with disease severity, and the ratio of pro-inflammatory TMAO to the putative anti-inflammatory metabolite butyric acid was significantly higher in PD subjects compared to controls indicating a pro-inflammatory shift in the metabolite profile in PD subjects. Finally, acetic and butyric acid were different between PD and MSA subjects indicating that metabolites may differentiate these synucleinopathies. In summary, (1) TMAO is elevated in PD subjects, a phenomenon independent of disease characteristics, treatment status, and lifestyle and (2) metabolites may differentiate PD and MSA subjects. Additional studies to understand the potential of TMAO and other bacterial metabolites to serve as a biomarker or therapeutic targets are warranted.
Collapse
|
11
|
Qu L, Dong Z, Ma S, Liu Y, Zhou W, Wang Z, Wu C, Ma R, Jiang X, Zu T, Cheng M, Wu Y. Gut Microbiome Signatures Are Predictive of Cognitive Impairment in Hypertension Patients—A Cohort Study. Front Microbiol 2022; 13:841614. [PMID: 35464979 PMCID: PMC9024414 DOI: 10.3389/fmicb.2022.841614] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/26/2022] [Indexed: 12/29/2022] Open
Abstract
Growing evidence has demonstrated that hypertension was associated with dysbiosis of intestinal flora. Since intestinal microbes could critically regulate neurofunction via the intestinal–brain axis, the study aimed to reveal the role and prediction value of intestinal flora alteration in hypertension-associated cognitive impairment. A cohort of 97 participants included 63 hypertension patients and 34 healthy controls. The structure of intestinal flora was analyzed by V3–V4 16S rRNA amplicon sequencing. The cognitive function was assessed using the Montreal Cognitive Assessment (MoCA) scale, and 31 patients were considered to have cognitive impairment (MoCA < 26). Patients with cognitive impairment had considerable alterations in intestinal flora structure, composition, and function compared with normal-cognitive patients. In particular, the abundance of LPS-containing taxa (Proteobacteria, Gammaproteobacteria, Enterobacterales, Enterobacteriaceae, and Escherichia–Shigella) and SCFA-producing taxon (Prevotella) significantly changed in cognition-impaired patients. Tax4Fun predication results showed downregulation of glycan biosynthesis and metabolism in hypertension patients with cognitive impairment. Additionally, the pathway was demonstrated to be significantly correlated with LPS-containing taxa (Proteobacteria, Gammaproteobacteria, Enterobacterales, Enterobacteriaceae, and Escherichia–Shigella) and SCFA-producing taxon Prevotella. Furthermore, the taxa-based multiple joint prediction model (9×) was demonstrated to have excellent diagnostic potential for cognitive impairment of hypertension patients (AUC = 0.944). The current study revealed the involvement of intestinal microbiota dysbiosis in cognition-impaired hypertension patients and provided an objective predictive index for this cognition disorder.
Collapse
Affiliation(s)
- Lei Qu
- Department of Pathogenic Biology, Binzhou Medical University, Yantai, China
| | - Zhouyan Dong
- Department of Pathogenic Biology, Binzhou Medical University, Yantai, China
| | - Songcui Ma
- Yantai Yuhuangding Hospital, Yantai, China
| | - Yaping Liu
- Institute of Health and Disease Management, Binzhou Medical University, Yantai, China
| | - Wei Zhou
- Institute of Health and Disease Management, Binzhou Medical University, Yantai, China
| | - Zitong Wang
- Institute of Health and Disease Management, Binzhou Medical University, Yantai, China
| | - Chen Wu
- School of Nursing and Rehabilitation, Shandong University, Jinan, China
| | - Rui Ma
- Institute of Health and Disease Management, Binzhou Medical University, Yantai, China
| | - Xinze Jiang
- Institute of Health and Disease Management, Binzhou Medical University, Yantai, China
| | - Tingting Zu
- Clinical Medicine School, Binzhou Medical University, Yantai, China
| | - Mei Cheng
- Institute of Health and Disease Management, Binzhou Medical University, Yantai, China
- *Correspondence: Mei Cheng,
| | - Yulong Wu
- Department of Pathogenic Biology, Binzhou Medical University, Yantai, China
- Yulong Wu,
| |
Collapse
|
12
|
Linard M, Ravier A, Mougué L, Grgurina I, Boutillier AL, Foubert-Samier A, Blanc F, Helmer C. Infectious Agents as Potential Drivers of α-Synucleinopathies. Mov Disord 2022; 37:464-477. [PMID: 35040520 DOI: 10.1002/mds.28925] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 12/10/2021] [Accepted: 12/27/2021] [Indexed: 12/16/2022] Open
Abstract
α-synucleinopathies, encompassing Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy, are devastating neurodegenerative diseases for which available therapeutic options are scarce, mostly because of our limited understanding of their pathophysiology. Although these pathologies are attributed to an intracellular accumulation of the α-synuclein protein in the nervous system with subsequent neuronal loss, the trigger(s) of this accumulation is/are not clearly identified. Among the existing hypotheses, interest in the hypothesis advocating the involvement of infectious agents in the onset of these diseases is renewed. In this article, we aimed to review the ongoing relevant factors favoring and opposing this hypothesis, focusing on (1) the potential antimicrobial role of α-synuclein, (2) potential entry points of pathogens in regard to early symptoms of diverse α-synucleinopathies, (3) pre-existing literature reviews assessing potential associations between infectious agents and Parkinson's disease, (4) original studies assessing these associations for dementia with Lewy bodies and multiple system atrophy (identified through a systematic literature review), and finally (5) potential susceptibility factors modulating the effects of infectious agents on the nervous system. © 2022 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Morgane Linard
- University of Bordeaux, INSERM, Bordeaux Population Health Research Center, UMR U1219, Bordeaux, France
| | - Alix Ravier
- CM2R (Memory Resource and Research Centre), Geriatrics Department, University Hospitals of Strasbourg, Strasbourg, France
| | - Louisa Mougué
- Cognitive-Behavioral Unit and Memory Consultations, Hospital of Sens, Sens, France
| | - Iris Grgurina
- University of Strasbourg, UMR7364 CNRS, LNCA, Strasbourg, France
| | | | - Alexandra Foubert-Samier
- University of Bordeaux, INSERM, Bordeaux Population Health Research Center, UMR U1219, Bordeaux, France.,French Reference Centre for MSA, University Hospital of Bordeaux, Bordeaux, France
| | - Frédéric Blanc
- CM2R (Memory Resource and Research Centre), Geriatrics Department, University Hospitals of Strasbourg, Strasbourg, France.,ICube Laboratory and FMTS (Fédération de Médecine Translationnelle de Strasbourg), Team IMIS, University of Strasbourg, Strasbourg, France
| | - Catherine Helmer
- University of Bordeaux, INSERM, Bordeaux Population Health Research Center, UMR U1219, Bordeaux, France
| |
Collapse
|
13
|
Chen SJ, Chen CC, Liao HY, Lin YT, Wu YW, Liou JM, Wu MS, Kuo CH, Lin CH. Association of Fecal and Plasma Levels of Short-Chain Fatty Acids With Gut Microbiota and Clinical Severity in Parkinson Disease Patients. Neurology 2022; 98:e848-e858. [PMID: 34996879 PMCID: PMC8883514 DOI: 10.1212/wnl.0000000000013225] [Citation(s) in RCA: 111] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 11/30/2021] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Short-chain fatty acids (SCFAs) are gut microbial metabolites that promote the disease process in a rodent model of Parkinson's disease (PD), but fecal levels of SCFAs in PD patients are reduced. Simultaneous assessments of fecal and plasma SCFA levels, and their inter-relationships with the PD disease process are scarce. We aimed to compare fecal and plasma levels of different SCFAs subtypes in PD patients and healthy controls to delineate their interrelations and link to gut microbiota changes and clinical severity of PD. METHODS A cohort of 96 PD patients and 85 controls were recruited from National Taiwan University Hospital. Fecal and plasma concentrations of SCFAs were measured using chromatography and mass spectrometry. Gut microbiota was analyzed using metagenomic shotgun sequencing. Body mass index and medical co-morbidities were evaluated, and dietary information was obtained using a food frequency questionnaire. To assess motor and cognitive impairment, we used the Movement Disorder Society-Unified Parkinson's Disease Rating Scale (MDS-UPDRS) and the Mini-Mental Status Examination (MMSE). RESULTS Compared with controls, PD patients had lower fecal but higher plasma concentrations of acetate, propionate, and butyrate. After adjustment for age, sex, disease duration, and anti-PD medication dosage, MDS-UPDRS part III motor scores correlated with reduced fecal levels of acetate (ρ = -0.37, p = 0.012), propionate (ρ = -0.32, p = 0.036), and butyrate (ρ = -0.40, p = 0.004) and with increased plasma propionate concentrations (ρ = 0.26, p = 0.042) in PD patients. MMSE scores negatively correlated with plasma levels of butyrate (ρ = -0.09, p = 0.027) and valerate (ρ = -0.032, p = 0.033) after adjustment for confounders. SCFAs-producing gut bacteria correlated positively with fecal levels of SCFAs in healthy controls but revealed no association in patients with PD. In the PD patient group, the abundance of pro-inflammatory microbes, such as Clostridiales bacterium NK3B98 and Ruminococcus sp. AM07-15, significantly correlated with decreased fecal levels and increased plasma levels of SCFAs, especially propionic acid. DISCUSSION Reductions in fecal SCFAs but increased plasma SCFAs were observed in PD patients and corelated to specific gut microbiota changes and the clinical severity of PD. CLASSIFICATION OF EVIDENCE This study provides Class III evidence that gut metabolite SCFAs distinguish between PD patients and controls, and are associated with disease severity in patients with PD.
Collapse
Affiliation(s)
- Szu-Ju Chen
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Neurology, National Taiwan University Hospital Bei-Hu Branch, Taipei, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chieh-Chang Chen
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.,Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsin-Yu Liao
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ya-Ting Lin
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Wei Wu
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Jyh-Ming Liou
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ming-Shiang Wu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ching-Hua Kuo
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan.,The Metabolomics Core Laboratory, NTU Centers of Genomic and Precision Medicine, National Taiwan University, Taipei, Taiwan.,Department of Pharmacy, National Taiwan University Hospital, Taipei, Taiwan
| | - Chin-Hsien Lin
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|