Skip to main content
Neural Regeneration Research logoLink to Neural Regeneration Research
. 2025 Apr 29;21(3):938–956. doi: 10.4103/NRR.NRR-D-24-01265

Short-chain fatty acids mediate enteric and central nervous system homeostasis in Parkinson’s disease: Innovative therapies and their translation

Shimin Pang 1, Zhili Ren 2,*, Hui Ding 2, Piu Chan 1,2,3
PMCID: PMC12296502  PMID: 40313087

Abstract

Short-chain fatty acids, metabolites produced by the fermentation of dietary fiber by gut microbiota, have garnered significant attention due to their correlation with neurodegenerative diseases, particularly Parkinson’s disease. In this review, we summarize the changes in short-chain fatty acid levels and the abundance of short-chain fatty acid-producing bacteria in various samples from patients with Parkinson’s disease, highlighting the critical role of gut homeostasis imbalance in the pathogenesis and progression of the disease. Focusing on the nervous system, we discuss the molecular mechanisms by which short-chain fatty acids influence the homeostasis of both the enteric nervous system and the central nervous system. We identify key processes, including the activation of G protein-coupled receptors and the inhibition of histone deacetylases by short-chain fatty acids. Importantly, structural or functional disruptions in the enteric nervous system mediated by these fatty acids may lead to abnormal α-synuclein expression and gastrointestinal dysmotility, which could serve as an initiating event in Parkinson’s disease. Furthermore, we propose that short-chain fatty acids help establish communication between the enteric nervous system and the central nervous system via the vagal nerve, immune circulation, and endocrine signaling. This communication may shed light on their potential role in the transmission of α-synuclein from the gut to the brain. Finally, we elucidate novel treatment strategies for Parkinson’s disease that target short-chain fatty acids and examine the challenges associated with translating short-chain fatty acid-based therapies into clinical practice. In conclusion, this review emphasizes the pivotal role of short-chain fatty acids in regulating gut–brain axis integrity and their significance in the pathogenesis of Parkinson’s disease from the perspective of the nervous system. Moreover, it highlights the potential value of short-chain fatty acids in early intervention for Parkinson’s disease. Future research into the molecular mechanisms of short-chain fatty acids and their synergistic interactions with other gut metabolites is likely to advance the clinical translation of innovative short-chain fatty acid-based therapies for Parkinson’s disease.

Keywords: alpha-synuclein, blood–brain barrier, blood circulation, central nervous system, endocrine, enteric nervous system, glial cell, gut–brain axis, gut microbiota, intestinal barrier, neuron, Parkinson’s disease, short chain fatty acids, vagus nerve

Introduction

Parkinson’s disease (PD) is primarily characterized by motor dysfunctions such as tremors and postural imbalance, resulting from dopaminergic neuron loss and α-synuclein (α-syn) deposition in the midbrain neurons (Morris et al., 2024; Jiao et al., 2025; Ni, 2025). Approximately 80% of patients with PD experience gastrointestinal symptoms, including excessive salivation, dysphagia, delayed gastric emptying, nausea, constipation, and changes in bowel habits, prior to the onset of classical motor symptoms (Kim and Sung, 2015; Zhang et al., 2023a). Notably, α-syn can accumulate in the enteric nervous system (ENS) before affecting central dopaminergic neurons. This finding suggests that PD involves both central and gastrointestinal pathology (Braak et al., 2006; Del Tredici and Duda, 2011; Gelpi et al., 2014).

The ENS primarily consists of two main nerve plexuses: the myenteric plexus, which regulates gastrointestinal motility, and the submucosal plexus, which controls mucosal electrolyte flow, hormone secretion, and intestinal epithelial cell (IEC) permeability (Sharkey and Mawe, 2023). The gut-brain axis theory proposed by Braak suggests that α-syn aggregation and misfolding in PD may begin in the ENS due to infection and inflammation, later spreading to the central nervous system (CNS) and causing central neuropathy (Braak et al., 2006; Bonaz, 2024). Studies show that α-syn injected into the intestinal muscular layer translocates to the brain via vagus nerve fibers in a time-dependent manner, supporting the hypothesis that PD may originate in the gastrointestinal tract (Holmqvist et al., 2014; Kim et al., 2019). Therefore, disrupting the structural and functional integrity of the ENS may initially trigger PD development.

Short-chain fatty acids (SCFAs) are organic acids produced via dietary fiber fermentation by gut microbiota in the host intestine (Cai et al., 2025). SCFAs primarily include acetate (60%), propionate (25%), and butyrate (15%), which regulate nervous system function and modulate various neurophysiological processes. In particular, SCFAs promote neuroprotection by regulating neuronal proliferation and differentiation (Yang et al., 2020a), oxidative stress (Saikachain et al., 2023), electrophysiological activity (Jessup et al., 2023), and neurotrophic factor secretion (Church et al., 2023), all essential for nervous system survival and functioning. They also promote neuroregeneration by facilitating neuronal repair (Jing et al., 2023). Therefore, SCFAs play a crucial role in ENS and CNS homeostasis, offering insights into PD pathology and potential therapeutic strategies.

Several studies have emphasized that SCFAs influence PD development and progression through their impact on ENS/CNS homeostasis (Paiva et al., 2017; Dalile et al., 2019; Hou et al., 2021b). While their protective effects in PD are well-documented, a comprehensive summary of their actions on neurons and glial cells is still lacking. This review aims to integrate existing literature and explore the role of SCFAs in PD, emphasizing their regulatory effects on ENS and CNS homeostasis within the gut–brain axis. It discusses their neuroprotective effects in PD and the potential to promote neuroregeneration, providing new insights into their clinical applications for PD treatment.

Retrieval Strategy

This review retrieved studies on the impact of SCFAs on the homeostasis of the ENS and CNS in PD. The search was conducted using the Web of Science, Google Scholar, and PubMed databases. Keywords included both Medical Subject Headings and free-text terms. Initial selection of articles for evaluation was based on the following keyword combinations: Parkinson’s disease, short-chain fatty acids, gut–brain axis, enteric nervous system, central nervous system homeostasis, neuron, glial cell, and alpha-synuclein. Most of the selected studies were published between 2019 and 2025. The search results were manually screened to include experimental and clinical studies, as well as relevant review articles that aligned with the research topic. Additionally, reference lists of included studies were reviewed to ensure comprehensiveness and accuracy.

Alteration of Short-Chain Fatty Acids in Patients With Parkinson’s Disease

Composition characteristics of short-chain fatty acids in patients with Parkinson’s disease

SCFAs are the most studied gut microbiota metabolites, primarily comprising acetate, propionate, and butyrate, which account for 95% of total SCFAs. Smaller amounts of valerate, iso-valerate, valproate, caproate, isocaproate, succinate, iso-butyrate, and hexanoate are also present (O’Riordan et al., 2022). SCFAs are key mediators of gut-brain communication, supporting various physiological processes within the body, including intestinal function, immune regulation, and neural development. This review examines the SCFA composition in different samples from patients with PD and its relationship with PD-related symptoms, highlighting the protective role of SCFAs in PD (Table 1).

Table 1.

Composition characteristics of SCFAs in different samples of patients with PD

Sample sources Participants Composition changes Clinical characteristics References
Fecal samples 14 PD, 14 HC BA↓ NM Liu et al., 2024
77 PD, 113 HC PA ↓, BA↓ BA (–) colonic transit Augustin et al., 2023
37 PD, 35 HC AA↓, PA↓, BA↓ CA (+) MDS-UPDRS total Huang et al., 2023
95 PD, 33 HC AA↓, PA↓, BA↓, IBA↓ AA, BA, IBA (–) Wexner score /H-Y stage/MDS-UPDRS I/II/total;
AA, BA (–) Disease duration;
BA, IBA (–) MDS-UPDRS III
Yang et al., 2022
96 PD, 85 HC AA↓, PA↓, BA↓ AA, PA, BA (–) UPDRS Part III score;
BA (+) MMSE score
Chen et al., 2022
35 PD, 50 HC AA↓, PA↓, BA↓, VA↓ NM De Pablo-Fernandez et al., 2022
104 PD, 96 HC BA↓ BA (+) cognitive score;
BA (–) postural instability-gait disorder score;
BA (–)/constipation severity
Tan et al., 2021
55 PD, 56 HC PA ↓, BA↓ BA (+) age NMS onset/age MS onset; Aho et al., 2021
17 PD, 17 HC AA↓, PA↓, BA↓ NM Huang et al., 2021
34 PD, 34 HC AA↓, PA↓, BA↓ BA (–) the use of COMT inhibitor Unger et al., 2016
Blood samples 44 PD, 42 HC AA↑, PA↓ NM Qi et al., 2023a
50 PD, 50 HC PA↓, BA↓, CA↓, HA↑ PA (–) UPDRs part III score / MMSE score;
PA (+) HAMD score / trihexyphenidyl /tizanidine
Wu et al., 2022a
95 PD, 33 HC AA↑, PA↑ AA, PA (+) α1-AT in feces / Wexner score / MDS-UPDRS I/II/total;
PA (+) H-Y stage / MDS-UPDRS III;
BA (–) MMSE score
Yang et al., 2022
96 PD, 85 HC PA↑, BA↑, VA↑ PA (–) UPDRS Part III score;
BA (–) MMSE score
Chen et al., 2022
46 PD, 46 HC PA↑ NM He et al., 2021
38 PD, 33 HC AA↑ AA (+) age;
PA (–) UPDRS part III score / the use of COMT inhibitor;
BA (+) use of monoamine oxidase inhibitors / anticholinergic drugs
Shin et al., 2020
19 PD, 21 HC AA↑ NM Toczylowska et al., 2020
Saliva samples 76 PD, 37 HC AA↑, PA↑ NM Kumari et al., 2020a
Urine samples 100 PD, 50 HC BA↑ NM Kumari et al., 2020b

This table summarizes the changes in SCFAs in feces, blood, saliva, and urine from patients with PD compared to healthy individuals. It also highlights the correlations between SCFAs and various clinical features. (+) Positive correlation; (–) negative correlation. α1-AT: α1-Antitrypsin; AA: acetic acid; BA: butyric acid; CA: caproic acid; COMT: catechol-O-methyltransferase; HA: heptanoic acid; HC: healthy controls; H–Y stage: Hoehn–Yahr stage; IBA: isobutyric acid; MDS-UPDRS: Movement Disorder Society-Unified Parkinson’s Disease Rating Scale; MMSE: Mini-Mental State Examination; MS: motor symptoms; NM: not mentioned; NMS: non-motor symptoms; PA: propanoic acid; PD: Parkinson’s disease; SCFAs: short-chain fatty acids; VA: valeric acid.

Unger et al. (2016) first analyzed SCFA concentrations in fecal samples, reporting significantly lower acetic, propionic, and butyric acid levels in the feces of patients with PD than in age-matched controls. These findings are consistent with those of subsequent studies (Huang et al., 2021, 2023; Chen et al., 2022). In another study, Unger et al. (2016) found no significant differences in fecal valeric acid, isovaleric acid, or isobutyric acid levels between patients with PD and controls, whereas De Pablo-Fernandez report significant reductions in isobutyric acid and valeric acid levels in the feces of patients with PD (De Pablo-Fernandez et al., 2022; Yang et al., 2022). Although the conclusions of these studies vary, they collectively indicate significantly reduced SCFA levels in the feces of patients with PD. Further analysis reveals a negative correlation between fecal butyric acid levels and disease duration (Yang et al., 2022), motor performance (Chen et al., 2022), postural instability-gait disorder, and constipation severity (Tan et al., 2021). Conversely, fecal butyric acid levels positively correlate with the age of onset of non-motor and motor symptoms (Aho et al., 2021). These findings highlight the significant role of butyric acid in PD onset and progression.

Notably, SCFA changes in peripheral blood samples do not correspond with those in fecal samples. Current studies consistently show significantly higher plasma SCFA concentrations in patients with PD than in healthy individuals (Shin et al., 2020; Toczylowska et al., 2020; He et al., 2021; Chen et al., 2022; Yang et al., 2022). Most SCFAs produced in the colon are absorbed and utilized by colon cells, with only a small fraction entering the peripheral circulation (Dalile et al., 2019). While fecal SCFA levels in patients with PD continuously decrease, their levels in the blood increase. This may be attributed to increased intestinal permeability, which allows SCFAs to enter systemic circulation, indicating intestinal epithelial barrier (IEB) dysfunction. One study measured SCFA levels in both feces and plasma of patients with PD while evaluating changes in α1-antitrypsin, a marker of intestinal-blood barrier permeability. Patients with PD exhibited higher fecal α1-antitrypsin levels than healthy controls. Moreover, plasma concentrations of acetic and propionic acids positively correlated with fecal α1-antitrypsin levels (Yang et al., 2022), suggesting that the abnormally high blood SCFA levels in these patients may result from SCFA translocation due to IEB damage. Additionally, Wu et al. (2022a) first measured SCFA levels in serum samples, revealing lower propionic, butyric, and caproic acid levels but higher heptanoic acid levels in patients with PD than in controls. These discrepancies may stem from differences in sample treatment methods, warranting further investigation.

In addition to fecal and blood analyses, some studies have measured SCFA levels in saliva and urine, finding elevated salivary acetic and propionic acid (Kumari et al., 2020a) and urinary butyric acid (Kumari et al., 2020b) in patients with PD. Since SCFAs cross the blood–brain barrier (BBB) and are present in cerebrospinal fluid (O’Riordan et al., 2022), assessing SCFA levels in the cerebrospinal fluid of patients with PD could provide valuable insights into their direct effects on the CNS.

Short-chain fatty acids-producing bacteria in patients with Parkinson’s disease

Studies show a significant reduction in SCFA-producing bacteria in patients with PD compared to healthy controls, primarily affecting Firmicutes, the dominant SCFA-producing phylum in the human gut (Table 2). A large meta-analysis by Romano et al. (2021) reveals lower abundances of Roseburia, Blautia, Fusicatenibacter, and Faecalibacterium in the gut of patients with PD than in controls. Additionally, bacteria such as Agathobacter, Lachnospira, Lachnospiraceae ND3007_group, Lachnospiraceae UCG-004, Oscillospira, and Butyricicoccus were reduced in patients with PD. Spearman correlation analysis further revealed a significant negative correlation between levodopa dosage and SCFA-producing bacteria levels (Wallen et al., 2020). Hill-Burns et al. (2017) found that PD medications independently affect gut microbiota composition. Compared to controls, the abundance of SCFA-producing bacteria such as Blautia, Faecalibacterium, and Roseburia was significantly reduced in patients with PD. However, after excluding individuals using catechol-O-methyltransferase inhibitors and anticholinergic drugs, the differences in the abundance of Blautia and Faecalibacterium between the control and PD groups were no longer significant (Hill-Burns et al., 2017). Given the gastrointestinal side effects of these drugs (Kaakkola, 2000; Ness et al., 2006), gut microbiota composition may provide valuable insights into their efficacy and side effects.

Table 2.

Composition characteristics of SCFAs-producing bacteria in patients with PD

Participants Taxonomic level
Abundance of bacteria in PD Clinical characteristics References
SCFA-producing bacteria (Family) SCFA-producing bacteria (Genus)
14 PD, 14 CON Lachnospiraceae Blautia The abundance of these genera is negatively correlated with UPDRS part II/ III score and H–Y staging. Liu et al., 2024
199PD,131CON Lachnospiraceae Agathobacter These genera mediate PD-associated underweight. Shih et al., 2024
Lachnospiraceae Fusicatenibacter
Lachnospiraceae Roseburia
Ruminococcaceae Ruminococcaceae_UCG_013
224 PD, 112 CON Lachnospiraceae Fusicatenibacter The decrease in the abundance of these genera is a sign of the early rapid progression of PD. These genera decrease with the progression of α-syn pathology. Nishiwaki et al., 2022
Ruminococcaceae Faecalibacterium
Lachnospiraceae Blautia
199 PD, 131 CON Lachnospiraceae Anaerostipes These genera mediate PD-associated constipation. Fu et al., 2022
Lachnospiraceae Fusicatenibacter
Lachnospiraceae Coprococcus
Lachnospiraceae Dorea
Lachnospiraceae Blautia
Ruminococcaceae Faecalibacterium
Ruminococcaceae Ruminococcaceae_UCG_013
Lachnospiraceae Lachnospiraceae_ND3007_group
Lachnospiraceae Lachnospiraceae_UCG_004
680 PD, 523 CON Lachnospiraceae Roseburia NM Romano et al., 2021
Lachnospiraceae Blautia
Lachnospiraceae Fusicatenibacter
Ruminococcaceae Faecalibacterium
306 PD, 177 Con Lachnospiraceae Agathobacter The abundance of these genera is negatively correlated with Levodopa dose. Wallen et al., 2020
Lachnospiraceae Blautia
Lachnospiraceae Roseburia
Lachnospiraceae Fusicatenibacter
Lachnospiraceae Lachnospira
Lachnospiraceae Lachnospiraceae_ND3007_group
Lachnospiraceae Lachnospiraceae_UCG-004
Ruminococcaceae Faecalibacterium
Ruminococcaceae Oscillospira
Ruminococcaceae Butyricicoccus
223 PD, 137 CON Lachnospiraceae Roseburia NM Nishiwaki et al., 2020a
Ruminococcaceae Faecalibacterium
Lachnospiraceae Lachnospiraceae_ND3007_group
64 PD, 51 CON Lachnospiraceae Blautia NM Vascellari et al., 2020
Lachnospiraceae Lachnospira
64 PD, 64 CON Lachnospiraceae Roseburia NM Aho et al., 2019
80 PD, 72 CON Lachnospiraceae Roseburia The abundance of Lachnospiraceae is negatively correlated with UPDRS part III score and H–Y staging. Pietrucci et al., 2019
193 PD, 113 NC Lactobacillaceae Unclass. Lachnospiraceae These genera mediate PD-associated gait disturbances and postural instability. Barichella et al., 2019
Lachnospiraceae Roseburia NM
Ruminococcaceae Oscillospira
Ruminococcaceae Oscillospira
24 PD, 14 CON Lachnospiraceae Blautia The abundance of these genera is negatively correlated with PD duration and disease severity. Li et al., 2017
Ruminococcaceae Ruminococcus
Ruminococcaceae Faecalibacterium
89 PD, 66 CON Ruminococcaceae Faecalibacterium NM Petrov et al., 2017
197 PD, 130 CON Lachnospiraceae Blautia NM Hill-Burns et al., 2017
Ruminococcaceae Faecalibacterium
Lachnospiraceae Roseburia
38 PD, 34 CON Lachnospiraceae Blautia The abundance of Lachnospiraceae is negatively correlated with PD duration. Keshavarzian et al., 2015
Lachnospiraceae Coprococcus
Lachnospiraceae Roseburia

This table shows that the level of SCFAs-producing bacteria in the intestines of patients with PD is significantly reduced. CON: Controls; H–Y stage: Hoehn–Yahr stage; NM: not mentioned; PD: Parkinson’s disease; SCFAs: short-chain fatty acids; α-syn: alpha-synuclein.

Gut microbiota composition is linked to various clinical features of PD. Several SCFA-producing bacteria correlate with disease duration, severity, and α-syn pathology (Keshavarzian et al., 2015; Li et al., 2017; Pietrucci et al., 2019; Nishiwaki et al., 2022). Nishiwaki et al. (2022) used bacterial and clinical data to predict the association between gut dysbiosis and PD progression over 2 years. They applied the area under the receiver operating characteristic curve of the random forest method and assessed model performance through nested cross-validation with recursive feature elimination. The resulting area under the receiver operating characteristic curves (AUROCs) for Hoehn–Yahr (H–Y) stages 1–3 were 0.799, 0.705, and 0.509, indicating the reliability of these microbiota-based models in detecting early PD stages. Furthermore, patients with reduced levels of SCFA-producing genera, specifically Fusicatenibacter, Faecalibacterium, and Blautia, exhibited accelerated disease progression. Notably, Fusicatenibacter emerged as a potential predictive biomarker for early PD progression, achieving an AUROC of 0.861 in the random forest model for predicting H–Y stage progression. A 2-year follow-up of patients with PD indicates that the initially reduced levels of Fusicatenibacter, Faecalibacterium, and Blautia remained unchanged, suggesting that the reduction of these bacteria is not a consequence of PD progression but rather a potential contributing factor to the disease’s advancement (Nishiwaki et al., 2022). Notably, individuals with idiopathic rapid eye movement sleep behavior disorder showed no significant differences in these bacteria levels (Nishiwaki et al., 2020b). Therefore, the decline in SCFA-producing bacteria may serve as both a biomarker for the rapid PD progression and a predictor for the transition from idiopathic rapid eye movement sleep behavior disorder to PD. Additionally, Blautia, Ruminococcus, and Faecalibacterium at the genus level showed a significant negative correlation with disease duration and the Unified Parkinson’s Disease Rating Scale (UPDRS) score. However, Keshavarzian et al. (2015) found no such correlations with disease duration or the UPDRS score at the species level. Only Lachnospiraceae at the family level was negatively correlated with disease duration. Another study indicated that disease duration significantly influenced overall microbiota composition, although no significant changes were observed at the family and genus levels (Pietrucci et al., 2019). Specifically, decreased Lachnospiraceae levels in PD correlated inversely with H–Y staging and the MDS-UPDRS Part III score (Pietrucci et al., 2019).

Short-Chain Fatty Acids-Mediated Enteric Nervous System Homeostasis in Parkinson’s Disease

Intestinal pathological features in Parkinson’s disease

Gastrointestinal dysfunction is a key non-motor symptom of PD and can manifest several years before the onset of motor symptoms. Patient and animal model data indicates that PD affects both neuronal and glial cells within the ENS, implicating it in PD pathogenesis. α-syn pathology in the myenteric and submucosal plexus neurons has been consistently reported in patients with PD (Braak et al., 2006; Beach et al., 2010, 2016; Ohlsson and Englund, 2019; Tanei et al., 2021). Similar α-syn pathology has been observed in the intestines of transgenic mice expressing mutant α-syn (either A53T or A30P), where α-syn aggregation in the ENS precedes CNS involvement (Kuo et al., 2010). Immunohistochemical staining shows significantly fewer dopaminergic neurons in the myenteric plexus of patients with PD than in controls (Singaram et al., 1995). In 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated mice, dopaminergic marker alterations in the intestines appeared before lesions in the substantia nigra, suggesting that PD pathology may begin in the ENS (Tian et al., 2008). Enteric glial cells (EGCs) support ENS neuron survival and function while regulating gastrointestinal activities such as intestinal motility and epithelial integrity through complex interactions with neurons, immune cells, and IECs (Seguella et al., 2020). In response to injury, EGCs upregulate glial fibrillary acidic protein (GFAP) and S100B expression via toll-like receptors and MHC II on their surface, releasing numerous pro-inflammatory factors (such as interleukin [IL]-1β, IL-6, tumor necrosis factor-α [TNF-α], and inducible nitric oxide synthase). This process activates EGCs, thereby enhancing the pro-inflammatory environment, causing IEB damage, and inducing neurochemical changes (Seguella et al., 2020). Reactive intestinal EGC proliferation and pro-inflammatory cytokine upregulation are key intestinal features of PD in both humans and mice (Devos et al., 2013; Clairembault et al., 2014, 2015a; Perez-Pardo et al., 2019; Pellegrini et al., 2020; Thomasi et al., 2022). EGC-driven neuroinflammation in the intestines triggers early α-syn misfolding, aggregation, and gastrointestinal motility disorders in PD. The deposited α-syn may further exacerbate intestinal neuroinflammation, accelerating PD pathology (Seguella et al., 2020). Consequently, the “pathological loop” between EGCs and α-syn strongly indicates ENS involvement in PD pathogenesis.

Beyond ENS involvement, PD is also marked by functional and morphological changes in the IEB. Numerous reports indicate decreased expression and localization of tight junction proteins, such as zonula occludens-1 (ZO-1) and occludin, in colonic mucosal samples from patients with PD (Clairembault et al., 2015b; Perez-Pardo et al., 2019). This decrease correlates with increased intestinal permeability (Forsyth et al., 2011; Hasegawa et al., 2015) and chronic intestinal inflammation (Schwiertz et al., 2018; Dumitrescu et al., 2021), both of which appear early and persist as PD progresses. Similar changes in tight junction proteins and IEB function occur in neurotoxin-induced PD models, such as 6-hydroxydopamine (6-OHDA) (Pellegrini et al., 2020), rotenone (Ma et al., 2021; Fang et al., 2024), MPTP, and in mice overexpressing human α-syn (Gorecki et al., 2019). IEB dysfunction and increased intestinal permeability expose the ENS to the gut microbiome and its metabolites. Toxins from the intestinal lumen can enter the ENS through IEC breakdown, triggering neuroinflammation in EGCs and promoting neurotoxic α-syn aggregation in neurons. Thus, ENS and IEB involvement is closely linked to PD onset and progression, highlighting the potential role of the gastrointestinal tract in its pathogenesis.

Effects of short-chain fatty acids on enteric nervous system

Constipation, a common non-motor symptom in PD, significantly impacts patient quality of life and therapeutic outcomes. Studies involving patients with PD and animal models link constipation to ENS dysfunction and delayed intestinal transit (Wang et al., 2008; Hallett et al., 2012; Barbut et al., 2019; Rota et al., 2019; Camilleri, 2021). Environmental factors, especially luminal components such as glucose, ions, microorganisms, and their metabolites, influence ENS phenotype and function by regulating enteric neurons. Notably, SCFA levels negatively correlate with constipation severity in patients with PD (Yang et al., 2022). Fu et al. (2022) report a significant decrease in nine SCFA-producing bacteria genera, including Fusicatenibacter, Dorea, Blautia, and Faecalibacterium, in patients with PD. These bacterial changes accounted for 76.56% of the impact of constipation on PD (Fu et al., 2022). Therefore, understanding the relationship between SCFAs and the ENS will help elucidate the pathogenesis of constipation in patients with PD.

Distribution of short-chain fatty acid receptors and transporters in the gut

SCFAs are primarily produced in the lower gut by microbial fermentation of dietary fibers. Once generated in the intestinal lumen, SCFAs are rapidly transported into IECs via monocarboxylate transporters (MCTs) and sodium-coupled MCTs (SMCTs). They inhibit the activity of histone deacetylase (HDAC) (van der Hee and Wells, 2021), inducing histone hyperacetylation in gene promoter regions, which ultimately increases gene expression (Ediriweera, 2023). Additionally, SCFAs act as signaling molecules by binding to G protein-coupled receptors (GPCRs) such as GPR41 (also known as free fatty acid receptor 3 [FFAR3]), GPR43 (FFAR2), and GPR109A (Mann et al., 2024). This binding promotes IEC structure and function (Figure 1). FFAR2 and FFAR3 bind to acetate, propionate, and butyrate, while GPR109A primarily binds to butyrate for signal transduction (Ikeda et al., 2022). Goblet cells, essential to IECs, produce mucus as a first-line defense against microbial invasion. Research indicates that when butyrate is the sole energy source, it modulates mucin (MUC) gene expression in goblet cells, potentially through FFAR3, thereby influencing mucus production (Gaudier et al., 2004). Propionate activates GPR43 and STAT3, inhibits HDAC, and consequently increases IEC migration speed and persistence, promoting epithelial renewal and repair while preventing ulcers (Bilotta et al., 2021). In MPTP-induced PD mice, butyrate activates GPR109A, inhibits NF-κB signaling, and upregulates tight junction proteins such as occludin and claudin-1 in IECs, thereby restoring IEB integrity (Xu et al., 2022b).

Figure 1.

Figure 1

Expression of SCFA receptors and their transporters in the gut.

SCFAs can enter the cells through transporters such as MCT/SMCT, and can also bind FFAR2/FFAR3/GPR109A on IECs to regulate the intestinal barrier. In the lamina propria, a variety of immune cells express transporters or receptors for SCFAs, including FFAR2/FFAR3/GPR109A and MCT/SMCT in monocytes, macrophages and dendritic spines. FFAR2 is primarily expressed in neutrophils and Treg cells. In the enteric nervous system, nitrergic and cholinergic neurons express FFAR3. Created with Figdraw.com (copyright code: WUOPWf3f66). MCT: Monocarboxylate transporter; SCFA: short-chain fatty acid; SMCT: sodium conjugated monocarboxylate transporter; Treg cell: regulatory T cell.

Immune cells beneath IECs express MCTs and SMCTs, facilitating SCFA transport (Parada Venegas et al., 2019; Mann et al., 2024). SCFAs regulate intestinal immune balance via GPCR signaling. FFAR2 and FFAR3 are expressed in monocytes, macrophages, and dendritic cells (Priyadarshini et al., 2018). In neutrophils, acetate signaling through FFAR2 accelerates their recruitment to inflammation sites, enhancing inflammasome activation and IL-1β secretion, making them a first line of defense against toxins (Fachi et al., 2020; Yan et al., 2023). Additionally, FFAR2 is highly expressed in regulatory T (Treg) cells and group 3 innate lymphoid cells (ILC3s) (Smith et al., 2013; Chun et al., 2019). Docampo et al. (2022) first reported GPR109A expression in activated T cells, where it stabilizes T cell metabolism and reduces apoptosis. Butyrate binds to GPR109A on monocytes, macrophages, and dendritic cells, facilitating signal transduction that regulates NF-κB signaling and NLRP3 activation, thereby promoting the anti-inflammatory properties of immune cells (Singh et al., 2014; Wu et al., 2022b; Qi et al., 2023b; Mamedova et al., 2024).

SCFA transporters and receptors are also present in the ENS. Immunohistochemical analyses show that 62% of HuC/D-positive neurons (pan-neural markers) and 38% of choline acetyltransferase (ChAT)-positive neurons (cholinergic neural markers) co-localize with MCT2 in primary cultures of the ENS. After 24-hour butyrate (500 μmol/L) treatment, MCT2 expression in neurons significantly increases (Soret et al., 2010). In the ENS, intestinal neurons possess SCFA-responsive receptors. In FFAR3-mRFP reporter mice, FFAR3-mRFP was identified in the somas of PGP9.5-immunoreactive ganglia cells, a general marker for enteric neurons, particularly in the intestinal submucosa and muscle layers (Nohr et al., 2013). Subsequently, Hertati et al. (2020) identified FFAR3 expression in nerve-like fibrous structures within the intestinal lamina propria and muscular layer, indicating its presence in nerve fibers and cell bodies of intestinal neurons, especially intermuscular neurons. FFAR3 receptors co-localize with nitric oxide synthase (nNOS)-immunoreactive inhibitory motor neurons and ChAT-immunoreactive excitatory motor neurons in the ENS (Kaji et al., 2016, 2018; Caetano et al., 2023). In the myenteric plexus, FFAR3 is expressed in both cholinergic and nitrergic nerves, whereas in the mucosa and submucosal plexus, FFAR3 is predominantly expressed in cholinergic nerves (Kaji et al., 2016). While GFAP-positive EGCs do not co-localize with FFAR3, EGCs may possess other SCFA receptors, potentially interacting with SCFAs via MCTs (Caetano et al., 2023). Thus, the presence of multiple SCFA receptors in the ENS indicates the importance of SCFAs in regulating ENS homeostasis.

Direct interactions between short-chain fatty acids and enteric nervous system

In the ENS, SCFAs influence enteric neuron survival and stimulate neurogenesis. Vicentini et al. (2021) report that supplementation with a SCFA mixture (67.5 mM acetate, 25.9 mM propionate, and 40 mM butyrate) restored antibiotic-induced neuronal loss and improved neuronal survival rates. Wang et al. (2023) evaluated the effects of butyrate (0, 0.1, 0.5, 1, and 2.5 mM) on intestinal neuronal cell proliferation in vitro, identifying 0.5 mM butyrate as the optimal concentration. Notably, both deficient and excessive butyrate concentrations inhibited neuronal proliferation through distinct mechanisms. Butyrate deficiency caused cell cycle dysregulation and subsequent apoptosis, while excess butyrate induced cellular damage and cell cycle arrest by upregulating neuronal oxidative phosphorylation pathways (Wang et al., 2023). These findings suggest that intestinal neuron growth depends on maintaining SCFA concentrations within an optimal range.

SCFAs regulate intestinal motility. A decreased abundance of butyrate-producing Fusobacterium is linked to functional constipation (Wang et al., 2023). Butyrate enhances colonic motility by inhibiting HDAC activity through the upregulation of the Src kinase signaling pathway, increasing H3K9 acetylation in enteric neurons. This process results in a higher proportion of excitatory ChAT-immunoreactive neurons within the myenteric plexus (Soret et al., 2010). In contrast, propionate reduces colonic motility (Hurst et al., 2014) but increases the number of vasoactive intestinal peptide (VIP) neurons in the submucosal plexus, promoting intestinal gluconeogenesis and regulating glucose and energy metabolism (De Vadder et al., 2015). These distinct effects of various SCFAs on intestinal motility warrant further investigation.

SCFAs protect against PD by directly acting on the ENS. In a 6-OHDA-induced PD mouse model, propionate and an FFAR3 agonist (the receptor for propionate in the ENS) alleviated motor deficits and dopaminergic neuronal loss. Furthermore, the neuroprotective effects of propionate in PD mice diminished when PGP9.5-positive enteric neurons were depleted via neurotoxicity (Hou et al., 2021b). These findings suggest that propionate helps prevent PD by targeting FFAR3 in enteric neurons. Additionally, SCFAs may help treat PD by regulating EGC function. Transcriptomic analysis revealed that butyrate significantly altered gene expression in the neurotrophic signaling pathway of rat EGCs. By inhibiting HDAC, butyrate enhances EGC secretion of nerve growth factor (Hou et al., 2021b). In PD, pramipexole combined with nerve growth factor improves cognitive function, enhances clinical efficacy, suppresses inflammation, and regulates brain neurotransmitters (Wang and Cheng, 2022).

Indirect interactions between short-chain fatty acids and the enteric nervous system

Crosstalk between short-chain fatty acids, intestinal endocrine and enteric nervous system

Enteroendocrine cells (EECs), primarily consisting of enterochromaffin (EC) and L cells, line the intestinal epithelium, directly facing the gut lumen. They respond to various chemical and mechanical stimuli from the lumen by releasing signaling molecules. Specifically, FFAR2 and FFAR3 receptors on EEC detect SCFAs, triggering the release of 5-hydroxytryptamine (5-HT), glucagon-like peptide-1 (GLP-1), and peptide YY (PYY) (Nøhr et al., 2013; Lu et al., 2018; Engevik et al., 2021; Tao et al., 2022; Masse and Lu, 2023). In addition to FFARs, colonic EECs express olfactory receptors such as Olfr78 (human ortholog: OR51E2) and Olfr558 (human ortholog: OR51E1), which also act as SCFA receptors (Bellono et al., 2017; Dinsart et al., 2024). These receptors facilitate EEC differentiation into ECs and regulate 5-HT secretion (Dinsart et al., 2024). Moreover, Olfr78 enhances protein kinase A signaling, increases cyclic adenosine monophosphate (cAMP) levels in EECs, and directly promotes PYY secretion (Nishida et al., 2021). EECs are anatomically and functionally linked to the ENS. EECs regulate neural development, gastrointestinal secretion, and motility through hormone and neurotransmitter release. This highlights the significant signaling interactions among SCFAs, EECs, and the ENS.

(1) 5-Hydroxytryptamine

Most 5-HT (serotonin) is produced in ECs via tryptophan hydroxylase 1 (TPH1), which converts tryptophan. Butyrate enhances TPH1 expression via the zinc finger transcription factor ZBP-89, thereby promoting 5-HT production and release (Nishida et al., 2021). Additionally, a smaller proportion of 5-HT is synthesized by tryptophan hydroxylase 2 (TPH2) in myenteric plexus neurons (Gershon, 2022; Zhu et al., 2022). Endogenous 5-HT is involved in regulating neurogenic gastrointestinal motility. Acetate, propionate, butyrate, and 5-HT can induce calcium (Ca2+) signaling in intestinal neurons to varying extents (Fung et al., 2021). 5-HT binds to its receptors on myenteric and submucosal cholinergic neurons in the gut, transmitting signals to enteric neurons. This signaling extends nerve fibers toward EECs, establishing direct contact and further stimulating calcium activity in cholinergic neurons. Consequently, acetylcholine secretion increases, thereby improving intestinal motility and secretion (Figure 2; Keating and Spencer, 2019; Ye et al., 2021; Martin et al., 2022; Zhai et al., 2023).

Figure 2.

Figure 2

Close link between SCFAs, intestinal endocrine system and ENS.

There are two primary subtypes of EECs in the gut: ECs, which secrete the majority of 5-HT (or serotonin), with a small portion produced by enteric neurons; and L cells, which specifically secrete GLP-1 and PYY. SCFAs-related receptors are present on the surface of both ECs and L-cells. SCFAs promote the expression of TPH1 via the zinc finger transcription factor ZBP-89. In ECs, tryptophan is converted to 5-HTP through the action of TPH1, while in enteric neurons, tryptophan is converted to 5-HTP by TPH2. Additionally, 5-HTP is converted to 5-HT by ADCC. Both ECs and neurons express SERT, which is responsible for the reuptake and recycling of 5-HT. SCFAs trigger PLC activity in L cells, increase IP3 levels, activate Ca2+ channels in the endoplasmic reticulum, and induce GLP-1 release. 5-HT and GLP-1 regulate the release of Ach in enteric neurons by binding to the 5-HT and GLP-1 receptors on the surface of these neurons, thus playing an important role in gastrointestinal motility. At the same time, they also contribute to the development and maturation of neurons. Created with figdraw.com (copyright code: WSPTAae3b6). 5-HT: 5-Hydroxytryptamine; 5-HTP: 5-hydroxytryptophan; 5-HTR: 5-HT receptor; Ach: acetylcholine; ADCC: aromatic L-amino acid decarboxylase; EC: enterochromaffin cell; EECs: enteroendocrine cells; ENS: enteric nervous system; ER: endoplasmic reticulum; GLP-1: glucagon-like peptide-1; IP3: inositol triphosphate; PLC: phospholipase C; PYY: peptide YY; SCFAs: short chain fatty acids; SERT: serotonin transporter; SERT: serotonin transporter; TPH: tryptophan hydroxylase; TPH2: tryptophan hydroxylase 2; Trp: tryptophan.

The role of 5-HT and its receptors in PD remains unclear. Intestinal biopsies from patients with PD show reduced 5-HT4 receptor expression (Barrenschee et al., 2017). Similarly, decreased 5-HT4 receptors in the colonic muscular layer of 6-OHDA rats is a key mechanism underlying colonic motility disorders and constipation (Zhang et al., 2015). In Thy1-α-syn PD mice, electroacupuncture-induced 5-HT release upregulates 5-HT4 receptor expression in the colon, activating the downstream cAMP/PKA signaling pathway. This activation promotes neurotransmitter secretion, including substance P and calcitonin gene-related peptide (CGRP), facilitating coordinated gastrointestinal smooth muscle contraction and relaxation. Consequently, this process accelerates intestinal transit and improves fecal excretion in the mice (Shen et al., 2022). Serotonergic neurons are among the first to develop in the ENS, influencing neurogenesis and driving the development and survival of later-born enteric neurons. Adding 5-HT to isolated enteric crest-derived cell cultures increases dopaminergic neuron number and survival rate and enhances neurite extension (Schertzer and Lam, 2021). Furthermore, TPH2 deficiency reduces dopaminergic neurons in the gut. Terminal immunoreactivity of the serotonin transporter surrounds dopaminergic neurons, suggesting serotonergic innervation (Li et al., 2011).

(2) Glucagon-like peptide-1

GLP-1, an incretin secreted by L cells, regulates glucose metabolism (Guo et al., 2024). SCFAs from gut microbiota stimulate GLP-1 secretion by activating FFAR2 and FFAR3 on L cells. This activation triggers phospholipase C activity, increases inositol triphosphate levels, and promotes calcium release from the endoplasmic reticulum, thereby inducing calcium responses in L cells (Figure 2; Ducastel et al., 2020; Peng et al., 2022). In the human L cell line NCI-H716, the olfactory receptor OR51E1 signaling upregulates intracellular cAMP and phosphorylated ERK, which enhances GLP-1 secretion (Han et al., 2018). These findings indicate the role of these receptors in mediating SCFA-regulated L-cell function within EECs.

In the ENS, GLP-1 receptors (GLP-1R) are expressed in ChAT- and nNOS)-positive nerve fibers within the myenteric plexus, extending from the gastric corpus to the distal colon, with the highest expression in the mid- and hindgut (Grunddal et al., 2022). This suggests a close relationship between GLP-1 and the ENS. While the exact mechanisms remain unclear, GLP-1 appears to activate neurons. Buckley et al. (2020) demonstrated that L cells detect bacterial signals in the intestinal lumen and act on colonic neurons expressing GLP-1R by secreting GLP-1. This secretion increases phosphoinositide 3-kinase expression and enhances calcium signaling, thereby boosting intestinal neuronal activity. GLP-1 significantly and dose-dependently enhances the survival of myenteric neurons in the rat small intestine. Notably, the neuroprotective effect of GLP-1 is reversed by the GLP-1R antagonist exendin (9-39) amide (Voss et al., 2012). In the gut, vagal sensory neurons expressing mechanosensory and chemosensory GLP-1R have been identified (Brierley and de Lartigue, 2022). GLP-1-positive cells, which express secretory cell markers such as SNAP25 and NEFM, communicate with vagal neurons in the lower intestinal plexus through a paracrine mechanism rather than direct synaptic contact (Cao et al., 2024). Notably, GLP-1 also regulates gastrointestinal motility. In response to the intestinal microbial environment, colonic L cells release GLP-1, which stimulates CGRP-positive neurons, accelerating gastrointestinal motility and promoting fecal excretion (Nakamori et al., 2024).

GLP-1 is closely associated with PD pathogenesis, and its analogs or receptor agonists are potential therapeutic strategies for this condition (Javed et al., 2024; Qi et al., 2025). These analogs reduce colonic inflammation, enhance intestinal permeability, and increase the levels of brain-derived neurotrophic factor, which supports neuronal survival, development, and differentiation. Notably, they effectively inhibit dopaminergic neuron loss and α-syn oligomer accumulation in the colon (Su et al., 2022). Studies show that probiotic Clostridium butyricum increases gut GLP-1 secretion and upregulates GLP-1R expression in brain tissue, significantly reversing motor deficits in PD mice (Sun et al., 2018, 2021). Additionally, sodium butyrate restores reduced colonic GLP-1 levels in PD mice, improving defecation rate and stool water content (Zhang et al., 2023b). Therefore, SCFA-mediated GLP-1 secretion is crucial for ENS homeostasis and plays a significant role in PD pathogenesis and treatment.

In addition to 5-HT and GLP-1, SCFAs directly stimulate EECs to produce neuropeptides such as PYY, neuropeptide Y, cholecystokinin, gastric inhibitory polypeptide, and substance P. These neuropeptides influence gastrointestinal functions and ENS development (Wang et al., 2010; Larraufie et al., 2018; Nishida et al., 2021; Geng et al., 2022). Therefore, SCFAs indirectly impact ENS homeostasis via the intestinal endocrine system.

Crosstalk between short-chain fatty acids, intestinal immune, and enteric nervous system

In mammals, the gastrointestinal tract is the largest immune system in the body. The immune system and ENS closely interact to regulate the gut neural network development, maturation, structure, and function. Gut microbiota activates resident immune cells, prompting them to release immune mediators that transmit signals to enteric neurons, thereby influencing ENS growth and function. Conversely, the ENS supports and enhances immune cell function by releasing neurotransmitters. As mentioned earlier, various gut immune cells express SCFA receptors, indicating that SCFAs may potentially maintain ENS integrity by regulating the gut immune system.

(1) Macrophages

Macrophages exhibit remarkable plasticity and adopt two main activation states depending on their environment: M1 or pro-inflammatory (classically activated) and M2 or anti-inflammatory (alternatively activated) macrophages. M1 macrophages release pro-inflammatory cytokines such as IL-1β, IL-6, and TNF-α, which trigger immune responses. In contrast, M2 macrophages release anti-inflammatory factors, including IL-10, IL-13, and TGF-β, promoting tissue repair and suppressing inflammation. In the gut, macrophages are primarily located in the lamina propria, with some in the muscle layer. Lamina propria macrophages typically exhibit the M1 phenotype and they are recruited from the bloodstream in response to microbial stimulation, contributing to immune defense against pathogens. Conversely, macrophages in the muscle layer exhibit the M2 phenotype and are involved in gut motility and inflammation repair (Figure 3A; Gabanyi et al., 2016). Macrophages and the ENS are closely located throughout the gut, and the macrophage state plays a crucial role in regulating ENS function. In vitro, Yao et al. (2021) observed that M1 macrophages secrete pro-inflammatory cytokines (including IL-1β, IL-6, and TNF-α), which activate the STAT3 and NF-κB pathways in enteric neurons and inhibit oxytocin signaling. This endogenous neuropeptide regulates secretion, motility, and immunity in the gastrointestinal tract. Conversely, M2 macrophages secrete TGF-β, activating Smad2/3 in enteric neurons and inhibiting Peg3, thereby promoting oxytocin signaling.

Figure 3.

Figure 3

Close link between SCFAs, intestinal immune system, and ENS.

(A) SCFAs promote the polarization of intestinal macrophages toward the M2 phenotype, which secretes IL-10 and IL-13, while inhibiting the polarization of M1 macrophages that secrete IL-6 and IL-1β. M1 macrophages can induce the death of intestinal neurons, whereas M2 macrophages help maintain neuronal survival. (B) CSF-1, secreted by enteric neurons, is essential for the development of MM. These MMs produce BMP2, which activates enteric neurons and influences muscular contractions. (C) Pathogen infection activates sympathetic neurons to release NE. When MM receives NE signals, it releases polyamines that protect intestinal neurons from death and prevent gastrointestinal motility disorders. (D) MM directly interacts with neurons containing p-α-syn in the myenteric plexus and can phagocytose p-α-syn, thus limiting the early spread of pathological alpha-synuclein in the gut. (E) SCFAs promote the differentiation of naïve intestinal T cells into Tregs cells while inhibiting their differentiation into Th17 cells. (F) IL-6 secreted by enteric neurons inhibits the differentiation of Tregs. (G) CD8+ T cells secrete perforin, which induces the death of intestinal neurons or EGCs through Fas signaling. (H) Intestinal CD4+ T cells can also induce the death of intestinal dopaminergic neurons, although the exact mechanism remains unclear. (I) SCFAs upregulate AKT/STAT3 or ERK/STAT3 signaling pathways through FFAR2, promoting the secretion of IL-22 by ILC3s to counteract intestinal inflammation. (J) IL-6 secreted by enteric neurons enhances the proliferation and secretion of IL-5, IL-9, and IL-13 by ILC2s. (K) VIP secreted by enteric neurons inhibits IL-22 release by ILC3s, which is detrimental to maintaining IEB integrity. (L) ILC3s increase IL-22 secretion in response to GFL secreted by EGCs through the RET receptor. Created with Figdraw.com (copyright code: TPOWU2eeb2). BMP2: Bone morphogenetic protein 2; CSF-1: colony-stimulating factor 1; EGCs: enteric glial cells; ENS: enteric nervous system; IEB: intestinal epithelial barrier; IL: interleukin; ILC3: group 3 innate lymphoid cells; MM: muscularis macrophages; NE: norepinephrine; NMU: neuropeptide neuromedin; p-α-syn: phosphorylated alpha-synuclein; SCFAs: short-chain fatty acids; VIP: vasoactive intestinal peptide.

Muscularis macrophages (MMs), located between the circular and longitudinal muscles, MMs play a crucial role in helping maintain immune homeostasis and regulate gut motility and secretion by interacting with the ENS under gut microbiota influence (Pendse et al., 2023). Recently, their close communication has garnered significant attention. MMs secrete bone morphogenetic protein 2, activating enteric neurons with corresponding receptors, thereby altering smooth muscle contraction patterns. MM depletion disrupts muscle coordination, prolonging colon transit time. Conversely, enteric neurons secrete colony-stimulating factor 1, a growth factor essential for macrophage development (Figure 3B; Muller et al., 2014). During early development, MMs shape the ENS by pruning synapses and phagocytosing enteric neurons. Depleting MMs before weaning disrupts ENS development, leading to structural abnormalities and impaired gut transit. After weaning, ENS-derived transforming growth factor-β enables MMs to adopt a neurosupportive phenotype. A specific MM subtype, associated with neuronal cell bodies and fibers, is vital for adult neuron survival. This subtype upregulates the transcription factor Nrf2, which protects enteric neurons from oxidative stress. Its dedifferentiation or depletion leads to enteric neuron loss and delays gut transit time (Viola et al., 2023). MMs also protect neurons via noradrenaline signaling. During microbial infection, the NLRP6 inflammasome activates the caspase-11 cell death pathway, reducing excitatory VGLUT2+ neurons and impairing gastrointestinal motility. In response, MMs upregulate neuroprotective programs. Specifically, MMs express β2-adrenergic receptors, allowing them to receive noradrenaline signals from gut-projecting sympathetic neurons. This interaction upregulates Arg1, promoting polyamine release, thereby enhancing enteric neuron survival and mitigating infection-induced gastrointestinal motility disorders (Figure 3C; Gabanyi et al., 2016; Matheis et al., 2020). MMs also phagocytose dying neurons and actively clear neuronal debris (Kulkarni et al., 2017). Their depletion in adulthood causes enteric neurodegeneration and functional impairment (Becker et al., 2018). With aging, MMs shift from an anti-inflammatory “M2” phenotype to a pro-inflammatory “M1” phenotype, partly due to reduced expression of the longevity-associated transcription factor FOXO3. This shift reduces enteric neuron density, depletes gut neural stem cells, and delays gut transit (Becker et al., 2018).

Macrophages and the ENS communicate bidirectional, with the ENS influencing macrophage-related inflammation. Stakenborg et al. (2019) further investigated the interaction between MMs and enteric neurons. MMs are situated near cholinergic neuronal fibers, which inhibit their activation in response to ATP. Cholinergic neurons trigger MMs to express the α7 nicotinic acetylcholine receptor, thereby exerting anti-inflammatory effects through cholinergic signaling (Stakenborg et al., 2019). Cholinergic neuron activation in the ENS reduces pro-inflammatory cytokine secretion in lipopolysaccharide (LPS)-stimulated MMs, thus alleviating intestinal inflammation (Rahman et al., 2024). In addition, EGCs play a crucial role in communicating with intestinal macrophages. They detect inflammation within the muscularis layer and, in their early stages, recruit monocytes via CCL2 to promote tissue repair. These monocytes differentiate into pro-resolving CD206+ and Timp2+ MMs through colony-stimulating factor 1. This differentiation promotes gut motility restoration after tissue damage. In turn, these MMs stimulate EGC proliferation, thereby serving a neurotrophic role (Stakenborg et al., 2022).

The interaction between macrophages and the ENS plays a complex role in intestinal α-syn pathology. MMs directly engage with myenteric neurons that produce pathological α-syn. Utilizing a complement C1q-dependent mechanism, MMs upregulate synaptic pruning-related genes, enabling them to phagocytose α-syn within neurons and limiting its early spread in the gut (Figure 3D; Mackie et al., 2023). However, prolonged pathological α-syn phagocytosis induces endolysosomal stress in MMs, depleting their synaptic clearance capacity and facilitating its spread within the ENS (Mackie et al., 2023). This study underscores the dynamic nature of macrophage responses to α-syn in the ENS, emphasizing their role in clearing toxic protein aggregates. A recent morphological study identified α-syn aggregates within macrophages, with phosphorylated S129-α-syn co-localizing with the pan-macrophage marker F4/80 during trinitrobenzenesulfonic acid-induced colitis. Furthermore, α-syn expression increased in cholinergic and VIP neurons, while tyrosine hydroxylase (TH) expression decreased (Casini et al., 2024). These findings further illustrate the intricate relationship between macrophages, the ENS, and α-syn.

SCFAs promote macrophage polarization toward the M2 phenotype. Recent studies show that SCFA-producing strains promote M2 macrophage polarization in the gut by inactivating the JAK/STAT3/FOXO3 axis, which alleviates inflammation (Zhao et al., 2024). Butyrate, a prominent SCFA, reprograms intestinal macrophage metabolism to achieve M2 polarization by upregulating genes associated with oxidative phosphorylation and lipid metabolism (Scott et al., 2018). As an HDAC inhibitor, butyrate increases H3K9 acetylation and reduces the production of pro-inflammatory mediators such as nitric oxide, IL-6, and IL-12 in intestinal macrophages (Chang et al., 2014). This enhances macrophage antibacterial activity and limits bacterial translocation (Schulthess et al., 2019). Additionally, butyrate enhances macrophage anti-inflammatory properties via GPCR signaling, enabling them to induce Treg and IL-10-producing T cell differentiation (Singh et al., 2014). In a colitis mouse model, the SCFA receptor GPR41 was expressed alongside increased CGRP-positive nerve fibers—neurotransmitters of intestinal cholinergic neurons—in the colonic lamina propria, with a large number of F4/80+ macrophages surrounding these nerve fibers (Hertati et al., 2020). This suggests that SCFA-sensitive gut neurons and macrophages interact during the pathological development of intestinal inflammation, highlighting the link between SCFAs, macrophages, and the ENS. Therefore, SCFAs may regulate ENS homeostasis through macrophages, warranting further study.

(2) T cells

The ability of SCFAs to regulate gastrointestinal T-cell homeostasis is widely documented. SCFAs influence the size and function of the colonic Treg cell pool and promote Foxp3+ IL-10+ Tregs production in an FFAR2-dependent manner to prevent colitis in mice (Smith et al., 2013). Additionally, SCFAs suppress IL-17 production by intestinal T cells through a histone deacetylase-dependent mechanism to counteract the development of intestinal inflammation (Figure 3E; Dupraz et al., 2021; Wang et al., 2021). These findings highlight the significance of gut microbiota-mediated interactions between the ENS and T cells in maintaining gut health.

T cells are categorized by surface markers into CD4+ T cells (helper T cells) and CD8+ T cells (cytotoxic T cells). CD4+ T cells further differentiate into various subsets, primarily including Th1, Th2, Th17, and Treg cells, which directly interact with the ENS to maintain neuroimmune stability under physiological conditions. Anatomically, Treg cells are found near CGRP+ and NOS1+ nerve fibers in the colonic lamina propria, mainly associated with nitrergic inhibitory motor neurons originating from the myenteric plexus. Functionally, enteric neurons release IL-6 to inhibit microbiota-induced Treg cell differentiation, thereby regulating immune cells (Figure 3F; Yan et al., 2021). Microbiota-mediated IL-17A signaling in Th17 cells and neurons in the gut promotes myenteric neurogenesis, evidenced by increased nestin expression. This process contributes to gut motility normalization (Marques de Souza et al., 2025).

Under pathological conditions, T cells disrupt the ENS integrity and cause functional dysregulation. Activated T lymphocytes increasingly adhere to enteric neurons or EGCs through the intercellular adhesion molecule-1/lymphocyte function-associated antigen-1 pathway, which promotes plexitis development (Pabois et al., 2020, 2024). In Crohn’s disease, an inflammatory bowel disease linked to increased risk of PD, early pathology includes EGC network destruction. This destruction involves cytotoxic autoimmune responses from CD8+ T cells that target EGCs. This immune response induces submucosal edema and vascular inflammation, ultimately causing severe intestinal inflammation and hemorrhagic necrosis (Cornet et al., 2001). CD8+ T cells mediate enteric neuron damage and impair colonic motility via perforin, TNF-α, and Fas signaling (Figure 3G; Brun et al., 2021; Sanchez-Ruiz et al., 2021; Janova et al., 2024). A recent study identified CD4+ T cells as key drivers of dopaminergic neuron damage in the gut (Garretti et al., 2023), although the molecular mechanisms underlying this process remain unclear (Figure 3H). These findings highlight the role of T cells in regulating ENS homeostasis and their participation in early PD-related gut pathology.

(3) Innate lymphoid cells

Innate lymphoid cells (ILCs) help maintain intestinal immune homeostasis by defending against infections, regulating IEB function, and interacting with the gut microbiota. Despite their lymphoid morphology, ILCs lack T cell receptors, classifying them as “innate” immune cells. Similar to T cells, they produce cytokines and play key roles in immune responses. Based on cytokine secretion and function, ILCs are classified into three major subsets: ILC1s, which produce IFN-γ; ILC2s, which mainly secrete IL-5, IL-13, and IL-9; and ILC3s, which include LTi ILC3s that secrete IL-17 and NCR+ ILC3s that secrete IL-22. ILCs are closely associated with the ENS.

SCFAs regulate ILC differentiation and function. In the gut, ILC3s predominantly express FFAR2 and FFAR3, with FFAR2 being the most abundant. Sepahi et al. (2021) report that soluble dietary fiber increases the number of ILC3s in both the small and large intestines. Acetate and propionate enhance ILC3 abundance and promote IL-22 secretion via FFAR2, utilizing the AKT/STAT3 or ERK/STAT3 signaling pathway (Figure 3I). FFAR2deletion in ILC3s reduces proliferation and IL-22 secretion, which impairs intestinal epithelial function. This impairment reduces mucin-associated proteins and antimicrobial peptides, increasing susceptibility to bacterial infections (Chun et al., 2019). Additionally, butyric acid shifts the ILC3 subset balance from Lti ILC3s to NCR+ ILC3s, enhancing IL-22 production, which regulates IEB function and limits intestinal inflammation (Chen et al., 2023). Therefore, SCFAs may indirectly regulate ENS development and function by modulating ILC composition and cytokine secretion.

In the gastrointestinal tract, ILC2s (IL-5, IL-9, and IL-13) co-localize with cholinergic neurons expressing neuromedin U, which rapidly activates their proliferation and secretion (Figure 3J), accelerating worm expulsion (Klose et al., 2017). Conversely, CGRP secreted by cholinergic neurons suppresses ILC2 expansion while increasing IL-5 expression (Xu et al., 2019). In the lamina propria, ILC3s selectively expressing the vasoactive intestinal peptide receptor 2 (also called VPAC2) are located near VIP-expressing neuronal projections. VPAC2 activation inhibits IL-22 production in ILC3s, potentially contributing to IEB collapse during infection (Figure 3K; Talbot et al., 2020). ILC3s are also found near EGCs expressing neurotrophic factors. Through the neuroregulatory receptor RET, ILC3s detect glial-derived neurotrophic factor ligands, activating STAT3 to release downstream IL-22 (Figure 3L). RET ablation in ILC3s reduces IL-22 production, impairs epithelial responses, disrupts microbiota balance, and increases susceptibility to intestinal inflammation and infection (Ibiza et al., 2016).

Short-Chain Fatty Acids–Mediated Central Nervous System Homeostasis in Parkinson’s Disease

Short-chain fatty acids and blood–brain barrier

The BBB is a tight junction formed between brain microvascular endothelial cells, astrocytes, pericytes, and the basal membrane, facilitated by tight junction proteins such as claudin, occludin, and ZO-1. This highly selective barrier prevents harmful and inflammatory substances from entering the brain. SCFAs can cross the BBB, as demonstrated through the uptake of 14C-labeled organic acids (Oldendorf, 1973). This process is associated with MCT1 or FFAR3 expression in cerebrovascular endothelial cells (Mac and Nalecz, 2003; Uhernik et al., 2011; Hoyles et al., 2018). SCFAs also regulate BBB integrity. In germ-free mice, increased BBB permeability was reversed by SCFA-producing bacteria and sodium butyrate, which upregulate tight junction proteins (Braniste et al., 2014). Additionally, SCFAs regulate GPR41-mediated levels of ZO-1, which is essential for the epithelial barrier in bovine rumen epithelial cells (Zhan et al., 2019). By binding and activating GPRs, SCFAs increase tight junction protein expression, maintaining BBB integrity. Furthermore, epigenetic factors such as increased HDAC expression and histone hypoacetylation contribute to BBB injury (Stamatovic et al., 2019). As known HDAC inhibitors, SCFAs reduce HDAC expression and increase histone acetylation, thereby restoring BBB function through epigenetic pathway regulation (Wang et al., 2011; Yu et al., 2013). SCFAs also inhibit systemic inflammation, reducing inflammatory cytokine and chemokine infiltration in circulation (Eslick et al., 2022b). They also diminish the excessive activation of glial cells (Caetano-Silva et al., 2023), indirectly protecting the BBB integrity.

Immunohistochemistry, cerebrospinal fluid analysis, and positron emission tomography reveal increased BBB permeability in patients with PD during disease progression (Kortekaas et al., 2005; Pisani et al., 2012; Gray and Woulfe, 2015; Al-Bachari et al., 2020). However, the role of SCFAs in BBB regulation remains unclear. In the MPTP model, sodium butyrate upregulates occludin and ZO-1, thereby restoring BBB integrity (Liu et al., 2017). Dong et al. (2020) found that polymanuronic acid inhibits peripheral inflammation in the gut, brain, and systemic circulation, improves BBB integrity, and restores dopaminergic neuron density in the substantia nigra through gut microbiota-derived SCFAs. Conversely, while SCFAs have protective effects in PD, Wang et al. (2024) recently observed that MPTP mice had significantly higher fecal SCFA levels than controls. Furthermore, chitosan administration alleviated motor symptoms and PD pathology by reducing acetate levels and restoring intestinal and brain barrier integrity rather than merely inhibiting MPTP-induced SCFA levels. Therefore, future studies must validate whether SCFA supplementation protects the BBB and clarify the underlying mechanisms in various PD models.

Mechanisms of short-chain fatty acids affecting the central nervous system

Effects of short-chain fatty acids on dopaminergic neurons

The degeneration and loss of dopaminergic neurons in the substantia nigra are key neuropathological features of PD. Reducing α-syn deposition and restoring dopaminergic neuron function are critical goals in PD treatment. Recent studies show that SCFAs promote dopaminergic neuron survival. Physiological doses of acetate, propionate, and butyrate promote the growth of human neural progenitor cells, suggesting that SCFAs regulate early neuronal development (Yang et al., 2020a). Histopathological analyses reveal that histone deacetylase 6 is enriched in neural Lewy bodies within the brains of patients with PD and colocalizes with α-syn (Miki et al., 2011; Mazzetti et al., 2020). Francelle et al. (2020) found that HDAC6 inhibitors protect dopaminergic neurons from α-syn toxicity. Sodium butyrate, an HDAC inhibitor (HDACi), mitigates α-syn-induced DNA damage in dopaminergic neurons by upregulating DNA repair genes and increasing acetylated histone 3 levels (Paiva et al., 2017). Furthermore, sodium butyrate inhibits apoptosis in dopaminergic neurons by enhancing histone acetylation (Kidd and Schneider, 2010). Thus, SCFAs act as HDAC inhibitors, promoting histone acetylation in dopaminergic neurons and protecting against α-syn neurotoxicity. SCFAs also regulate the TH gene expression (Dalile et al., 2019; Maisterrena et al., 2022), influencing dopaminergic neuron function. For instance, propionic acid increases the number of TH-positive dopaminergic neurons and promotes neuronal overgrowth in rotenone-treated rat midbrain neurons (Ostendorf et al., 2020). Additionally, valproic acid, though present at lower levels in the intestine, also promotes dopaminergic neuron growth. It restores the mammalian target of rapamycin (mTOR) signaling and autophagy pathways (Jayaraj et al., 2020), reduces intracellular reactive oxygen species (Hsu et al., 2020), and inhibits apoptosis in dopaminergic neurons. In summary, SCFAs and valproic acid show therapeutic potential for protecting and promoting dopaminergic neuron survival in PD.

Effects of short-chain fatty acids on glial cells

Under physiological conditions, microglia and astrocytes secrete neurotrophic factors and anti-inflammatory cytokines, supporting neuronal growth, synaptic pruning, metabolite clearance, and inflammation balance. However, prolonged exposure to danger signals triggers uncontrolled activation, leading to excessive pro-inflammatory substance secretion, neuronal damage, and chronic neuroinflammation (Wang et al., 2022). Clinical and molecular evidence strongly links glial-induced neuroinflammation to PD pathogenesis and progression. Neuroimaging studies on microglia-specific radiotracers show increased microglial activation in the early stages of PD. This activation negatively correlates with dopaminergic terminal density and positively correlates with motor symptoms (Ouchi et al., 2005). SCFAs derived from gut microbiota regulate microglia maturation and homeostasis. A decade ago, researchers discovered that germ-free (GF) mice exhibited abnormal microglia morphology and function, including altered cell proportions and an immature phenotype, impairing their innate immune response. Similar microglial immaturity was observed in FFAR2–/– mice (Erny et al., 2015). Adding SCFAs to the drinking water of GF mice reversed their defective microglial phenotype. However, studies on the relationship between SCFAs and microglia yield conflicting findings. For instance, Sampson et al. (2016) reported that α-syn overexpressing mice raised in a GF environment exhibited reduced microglia activation in the substantia nigra, along with significantly fewer α-syn aggregates and improved motor function compared to those under pathogen-free conditions. However, when SCFA administration increased microglial activation in GF-α-syn overexpressing mice, characterized by larger cell bodies, reduced branching, and more severe motor impairments compared to GF wild-type mice.

Studies on PD models suggest that SCFAs shift microglia from a pro-inflammatory to an anti-inflammatory state, enhance their phagocytic and clearance capabilities, and protect against α-syn-induced neurotoxicity. For instance, sodium butyrate activates GPRs, inhibits the NF-κB and mitogen-activated protein kinase (MAPK) signaling pathways, and decreases the number of microglia and pro-inflammatory cytokine expression, thereby preventing neuroinflammation and neurodegeneration (Hou et al., 2021a; Xu et al., 2022b; Guo et al., 2023). In BV2 cells treated with 1-methyl-4-phenylpyridinium, sodium butyrate facilitated KATP channel opening by increasing Kir6.1 and Kir6.2 expression. This reduced nitric oxide and pro-inflammatory cytokine production, effectively inhibiting the pro-inflammatory phenotype activation of microglia (Xu et al., 2024). Beyond their effects on microglia, SCFAs also influence astrocyte function. For example, VA decreased the number of activated astrocytes in response to rotenone (Jayaraj et al., 2020). Sodium butyrate and valproic acid have shown potential as neuroprotective agents, reversing manganese-induced reductions in glutamate aspartate transporter and glutamate transporter 1 mRNA and protein levels in astrocytes, thereby protecting the dopaminergic system (Johnson et al., 2018a, b). Although the precise mechanisms by which SCFAs affect glial function in the CNS remain unclear, their anti-neuroinflammatory potential in PD remains promising.

How Do Short-Chain Fatty Acids Connect the Enteric and Central Nervous System?

Vagus nerve pathway

Braak proposed that α-syn aggregates spread retrogradely from the intestine to the brain (Braak et al., 2006). Since then, the vagus nerve has been identified as a crucial conduit for transmitting information between the ENS and CNS. Holmqvist et al. (2014) demonstrated time-dependent transport of visualized α-syn from the intestinal myenteric plexus to the brain via the vagus nerve. Patients who underwent full truncal vagotomy exhibited a significantly lower risk of PD, particularly with follow-up beyond 20 years, supporting α-syn transmission through the vagus nerve (Svensson et al., 2015). Thus, the vagus nerve is the fastest and most direct communication pathway between the ENS and CNS. SCFAs can directly influence vagus nerve function. FFAR3 expression has been identified in vagal sensory neurons innervating the entire intestine (Cook et al., 2021). In anesthetized male rats, intraluminal sodium butyrate administration induced vagal afferent fiber discharge, an effect that was abolished after subphrenic vagotomy. Notably, this persisted even after pretreatment with L-type calcium channel antagonists, suggesting that SCFAs directly activate vagal afferent fibers independent of smooth muscle involvement (Lal et al., 2001). These findings suggest that SCFAs may directly induce vagus nerve signal transduction through FFAR3 and activate different CNS neuronal populations (Figure 4, middle panel).

Figure 4.

Figure 4

SCFAs connect pathological events in the gut and brain through the vagus nerve, blood circulation, and endocrine pathways in PD.

SCFAs, produced by the intestinal microbiota during the breakdown of dietary fiber, pass through IECs via MCT, SMCT, and FFARs. SCFAs can inhibit the transition of EGCs to a pro-inflammatory state, decrease abnormal alpha-synuclein (α-syn) expression in intestinal neurons, and consequently reduce the spread of α-syn from the ENS to the CNS (middle panel). Additionally, SCFAs are vital for maintaining the integrity of the IEB, which helps prevent the activation of intestinal immune cells and the release of pro-inflammatory cytokines triggered by microbial and toxin attacks. This, in turn, limits the polarization of immune cells toward a pro-inflammatory state in the bloodstream, indirectly protecting dopaminergic neurons and glial cells from inflammation (left panel). SCFAs also influence the function of EECs and stimulate the secretion of neuroprotective factors such as 5-HT, GLP-1, and PYY. These factors enter the brain via systemic circulation and play a crucial role in protecting against neuroinflammation and degeneration of dopaminergic neurons. Furthermore, SCFAs impact the expression of α-syn in EECs, thereby reducing the spread of α-syn from EECs to intestinal and brain neurons (right panel). Created with Figdraw.com (copyright code: TTTAY4b464). 5-HT: 5-Hydroxytryptamine; CNS: central nervous system; EECs: enteroendocrine cells; EGC: enteric glial cell; ENS: enteric nervous system; FFAR: free fatty acid receptor; GLP-1: glucagon like peptide-1; IEB: intestinal epithelial barrier; IECs: intestinal epithelial cells; MCT: monocarboxylate transporters; PD: Parkinson’s disease; PYY: peptide YY; SCFAs: short chain fatty acids; SMCT: sodium-coupled monocarboxylate transporters; α-syn: α-synuclein.

Blood circulation pathway

When IEB integrity is compromised, microorganisms, toxins, inflammatory cytokines, and α-syn enter the bloodstream, triggering systemic inflammation. Various blood immune cells become abnormally activated in response to these toxins and inflammatory factors, which subsequently activate pro-inflammatory signaling pathways and secrete additional inflammatory mediators. Prolonged systemic inflammation weakens the BBB, allowing inflammatory factors to penetrate the brain parenchyma, resulting in neuroinflammation and neurodegeneration. Upregulated pro-inflammatory factors and α-syn in the CNS can also enter the intestine via the bloodstream, further affecting the ENS. Consequently, blood circulation establishes a bidirectional connection between the intestine and brain, playing a key role in PD onset and progression (Chaudhry et al., 2023). In 2014, Grozdanov et al. discovered that circulating peripheral blood monocytes in patients with PD were altered in function and composition, displaying pathological hyperactivity in response to LPS, which correlated with disease severity. Conversely, Nissen et al. (2019) reported reduced monocyte viability and lower sensitivity to LPS and fibrillar α-syn, potentially affecting disease progression. Despite these differences, peripheral blood mononuclear cells clearly mediate central and peripheral inflammation in PD. Thus, maintaining immune cell balance in peripheral blood may help prevent or treat PD.

SCFAs are highly expressed in various immune cells and they play key roles in innate immunity. Butyrate alters the metabolic profile of human peripheral blood monocyte-derived macrophages, enhancing LC3-related antibacterial activity via mTOR by inhibiting HDAC3 (Schulthess et al., 2019). A mixture of SCFAs significantly reduces the release of cytotoxic molecules such as IL-1β, MCP-1, TNF-α, and reactive oxygen species in LPS-stimulated human THP-1 monocytic cells and differentiated human HL-60 myelomonocytic cells (Wenzel et al., 2020). SCFAs also regulate neutrophil migration to inflammatory sites in response to immune reactions by activating intracellular protein kinase P38 and coupling G proteins through the GPR43 receptor (Le Poul et al., 2003). Natural killer cells link innate and adaptive immunity by secreting pro-inflammatory cytokines that activate and polarize other immune cells. However, butyrate strongly inhibits NK cell activation by downregulating mTORC1 activity, c-Myc mRNA expression, and metabolic pathways (Zaiatz-Bittencourt et al., 2023). In adaptive immunity, butyrate inhibits HDAC activity, leading to epigenetic regulation of T cells by suppressing Th17-related RORγt and IL-17 expression while increasing Treg cell-related FoxP3 expression (McBride et al., 2023). In B cells, SCFAs elevate acetyl-CoA levels and enhance cellular metabolism, thereby supporting B cell activation, differentiation, and antibody production (Kim et al., 2016). These studies support the regulatory roles of SCFAs in innate and adaptive immunity within the circulatory system (Figure 4, left panel). Additionally, SCFAs inhibit pro-inflammatory cytokine expression in the serum of MPTP PA models, thereby reducing systemic inflammation and endotoxemia (Guo et al., 2023).

Endocrine pathway

EECs detect microbial structural components, such as LPS, through surface toll-like receptors and respond by releasing hormones and neurotransmitters into the bloodstream or CNS. GLP-1 and PYY, secreted by EECs, influence CNS activity by binding to functional receptors in different brain regions (Morimoto et al., 2008; Holzer et al., 2012; Katsurada and Yada, 2016). Enhancing GLP-1 signaling alleviates neuroinflammation, dopaminergic neuron damage, and motor disorders in patients with PD and animal models (Li et al., 2009; Athauda et al., 2017; Brauer et al., 2020; Sun et al., 2021).

SCFAs significantly influence endogenous GLP-1 secretion into the systemic circulation by binding to FFAR on EECs (Tolhurst et al., 2012; Psichas et al., 2015). In an MPTP-induced PD mouse model, oral sodium butyrate improved neurobehavioral disorders by enhancing EEC activity, specifically increasing GLP-1 levels in the colon and upregulating GLP-1 receptors in the brain (Liu et al., 2017). This finding was recently confirmed in a rotenone-induced PD mouse model (Zhang et al., 2023b).

Morphologically, EECs possess neurofilament-containing axon-like processes (neuropods) that transmit signals to intestinal neurons, EGCs, and vagal neurons (Bohorquez et al., 2014, 2015; Kaelberer et al., 2018). In human intestinal tissue, α-syn co-localizes with EECs (Chandra et al., 2017; Casini et al., 2021) and is situated near α-syn-immunoreactive enteric neurons (Chandra et al., 2017). This suggests that danger signals in the intestinal lumen may induce α-syn misfolding in EECs, facilitating its transmission to intestinal neurons. This process may contribute to ENS degeneration and promote a bottom-up spread of PD pathology, ultimately leading to CNS degeneration (Figure 4, right panel). Notably, sodium butyrate concentrations exceeding 10 mM inhibited the proliferation and survival of murine neuroendocrine STC-1 cells while increasing α-syn mRNA expression without affecting protein expression. Additionally, conditioned media from sodium butyrate-stimulated STC-1 cells induced pro-inflammatory cytokine release in SH-SY5Y cells (Qiao et al., 2020a).

Synergistic interactions between short chain fatty acids and other gut microbiota and microbial metabolites in Parkinson’s disease

We previously reviewed the effects of microbiota-derived SCFAs on the ENS and CNS. Beyond their independent functions, SCFAs interact with other gut microbiota components or metabolites to form a complex synergistic network in PD pathogenesis. SCFAs, particularly butyrate, help maintain intestinal homeostasis by connecting the ENS and CNS. Sodium butyrate is commonly used to evaluate the effects on gut microbiota in the MPTP mouse model. Results show that sodium butyrate restores gut microbiota diversity and composition balance by reducing the abundance of Proteobacteria, Bacteroides, and Akkermansia. Notably, Proteobacteria is a key biomarker of intestinal microbiota dysbiosis (Guo et al., 2023). Mishra et al. (2024) found that butyric acid levels decrease in aged mice due to reduced butyrate-producing bacteria within the gut microbiota. Furthermore, butyrate supplementation mitigates aging-related gut permeability and inflammation. These findings indicate that butyrate and butyrate-producing bacteria influence mucin formation and intestinal permeability, thereby affecting brain function.

Bile acids, key microbial metabolites, significantly impact gut microbiome structure and function. Generally, they regulate gut microbiota abundance, diversity, and metabolism. Li et al. (2021) found significant microbiota differences in the appendix tissues of patients with PD linked to bile acid disturbances. This suggests that the interaction between SCFAs and bile acids may play a crucial role in the complex relationship between gut microbiota and PD (Hasuike et al., 2020). Unlike SCFAs, which result from bacterial fermentation of dietary fiber, amino acids primarily stem from protein metabolism. Mi et al. (2023) report that dietary branched-chain amino acids increased serum levels of propionic, butyric, isobutyric, and acetic acids in a PD mouse model, potentially enhancing their pharmacological efficacy.

Short Chain Fatty Acids for Guiding Parkinson’s Disease Therapeutic Strategies

Targeting gut microbiota

Numerous studies suggest that SCFAs and their associated microorganisms may help identify novel therapeutic targets for PD. The Mediterranean diet (NCT06705517), probiotics, prebiotics, and fecal microbiota transplantation (FMT) have been widely researched in animal and cellular models (Zhang et al., 2023a). Clinical trials have begun to assess their safety, feasibility, and effectiveness (Cheng et al., 2023). In the Netherlands, researchers are conducting safety and feasibility studies on FMT for patients with PD, although these studies have not been completed (Vendrik et al., 2023). In China, institutions such as Huashan Hospital and the Third Military Medical University have investigated the effectiveness of microbial therapy for patients with PD (ChiCTR1900027055, ChiCTR1900021405). The ChiCTR1900021405 project reports that freeze-dried fecal microbiota capsules significantly improved autonomic and gastrointestinal symptoms in patients with PD while increasing gut microbiota diversity, thereby confirming the feasibility of oral FMT (Cheng et al., 2023). In addition to FMT, probiotics such as Lactobacillus casei and Bifidobacterium lactis M8 have been studied in patients with PD (ChiCTR1800016795, ChiCTR1800016977). A team from Ruijin Hospital found that Lactobacillus casei significantly reduced non-motor symptoms, such as constipation, in patients with PD (Yang et al., 2023). Furthermore, combining Bifidobacterium lactis M8 with PD medications enhanced their clinical efficacy, potentially by regulating lipid, SCFA, and neurotransmitter metabolism (Sun et al., 2022).

Short-chain fatty acids–related pharmacological action

SCFAs regulate homeostasis by activating GPCR signaling through various pathways (Ikeda et al., 2022). GPCRs, the most diverse transmembrane protein family, were first identified as SCFA receptors in 2003 (Brown et al., 2003). Functionally, they belong to the FFAR family, with SCFAs acting as natural ligands for activating GPCRs. Five major GPCRs—GPR43, GPR41, GPR109A, Olfr78, and Olfr558—have been identified (Ikeda et al., 2022). Yang et al. (2020b) demonstrated that GPR41 activation and HDAC inhibition are critical pathways for SCFAs, particularly butyrate. This activation promotes IL-22 production in ILCs and CD4+ T cells. Tetrastigma hemsleyanum polysaccharide restores butyrate levels and upregulates GPR41 and GPR43 expression in vivo and in vitro, thus protecting against intestinal inflammation and IEB impairment (Lin et al., 2023). These studies suggest that the SCFA family represents a novel target for maintaining intestinal homeostasis. Therefore, given the similar intestinal pathological alterations in PD, further research should explore whether SCFAs exert neuroprotective effects against PD through the GPCR pathway. Chinese researchers confirmed that butyrate and monomethyl fumarate (a GPR109A agonist) improve motor symptoms and pathological alterations in PD mouse models by activating GPR109A (Xu et al., 2022b). Additionally, Olfr78 and Olfr558, olfactory receptors expressed in the olfactory epithelium, colonic endocrine cells, autonomic ganglia, and vascular smooth muscle, are also considered SCFA receptors. Olfr78 regulates blood pressure and food intake (Nishida et al., 2021; Xu et al., 2022a); however, it is rarely studied in neurological diseases. Recent research indicates that Olfr78 overexpression inhibits calcium overload and reduces neuronal apoptosis in a rat model of middle cerebral artery occlusion/reperfusion by activating the cAMP/PKA-MAPK pathway (Kang et al., 2024). This provides new insights into potential therapeutic targets for other neurological diseases.

Histone deacetylase inhibitor–related pharmacological action

Histone acetylation becomes imbalanced with aging and in age-related neurodegenerative conditions, including PD. A recent study on HDACi show their benefits in both in vivo and in vitro PD models (Zhang et al., 2024). Butyrate, a widely studied HDACi and SCFA discovered over a decade ago, improves motor impairment and survival in a rotenone-induced Drosophila PD model. Specifically, a loss-of-function mutation in Sin3A, which reduces HDAC activity, conferred resistance to rotenone-induced locomotor impairment and early mortality in flies. Furthermore, butyrate supplementation in these mutant flies improves locomotor function (St Laurent et al., 2013). In a 6-OHDA rat model with lesions, butyrate administration increases striatal dopamine levels, brain-derived neurotrophic factor, and histone acetylation, demonstrating its neuroprotective effects against PD (Sharma et al., 2015). The pharmacological HDACi valproic acid, a well-known anti-epileptic drug, protects against PD-like behavior and neuropathological alterations in various PD models (Zhang et al., 2024). Newly synthesized pan-HDACi, such as cinnamyl sulphonamide hydroxamate derivatives (NMJ-2 and NMJ-3), attenuates MPTP-induced abnormal behavioral changes in motor and non-motor functions by preventing inflammation and restoring dopamine levels (Meka et al., 2023). While these new compounds have been extensively designed, they are not discussed here (Toledano-Pinedo et al., 2024). Overall, SCFAs as natural HDACi, combined with synthetic compounds, show promise for treating PD, supported by pharmacological evidence. Their potential mechanisms include activating neuronal survival genes, such as brain-derived and glial cell-derived neurotrophic factors, and restorating histone acetylation homeostasis (Sharma and Taliyan, 2015; Zhang et al., 2024). Although HDACi has primarily been studied for cancer, clinical trials for other conditions, including PD, are expected.

Limitations

Translating SCFA-based therapies into clinical practice faces several significant challenges: 1) Limited bioavailability and metabolism: SCFAs are predominantly produced in the gut through microbial fermentation, absorbed by the intestine, and rapidly metabolized in the liver. As a result, the bioavailability of exogenously administered SCFAs is often limited, particularly with oral supplementation. A substantial proportion of orally administered SCFAs undergo first-pass metabolism in the liver, which reduces their systemic concentration and effectiveness. The rapid metabolism of SCFAs in vivo complicates efforts to achieve efficient absorption and maintain therapeutic levels. 2) Unclear optimal dosage and duration: The optimal dose and duration of SCFA supplementation remain undefined, presenting challenges for long-term safety and efficacy. Low doses may yield minimal therapeutic effects, while high concentrations could lead to adverse reactions such as gut irritation, acid-base imbalances, or disruption of the microbiota. The variability in individual responses, coupled with the absence of standardized treatment protocols, makes it difficult to establish clear therapeutic guidelines. 3) Challenges in targeted delivery: Developing targeted delivery systems or strategies to ensure that SCFAs reach the brain and exert beneficial effects is a significant hurdle. The therapeutic potential of SCFAs often relies on their local effects in the gut and their ability to modulate the gut–brain axis indirectly. Given that the blood-brain barrier restricts the passage of many compounds, including SCFAs, systemic effects may be insufficient for effectively treating neurological disorders without targeted delivery mechanisms. 4) Variable clinical responses: Variability in clinical responses presents another challenge. Patients with different genetic backgrounds, stages of disease progression, or comorbidities may respond differently to SCFA-based treatments. This heterogeneity complicates the prediction of SCFA efficacy across a broad patient population. Understanding the factors influencing patient response variability and developing personalized SCFA therapies will be essential for successful clinical translation.

Discussion

Numerous studies have reported significant differences in the total SCFA concentrations in blood or fecal samples from patients with PD compared to healthy controls. This highlights the close association between SCFAs and the pathogenesis and progression of PD. However, current research faces several limitations, leaving many questions unanswered. Since SCFAs are primarily produced in the colon and can enter the bloodstream through the IEB, most human studies have focused on fecal or blood samples as convenient detection targets. These samples better reflect the peripheral origin and effects of SCFAs. Nevertheless, the majority of existing studies have either analyzed SCFA concentrations from a single source (e.g., feces or blood) or assessed only the abundance of SCFA-producing bacteria. Relying on a single research metric provides limited insights into the role and prognostic value of SCFAs in the pathogenesis of PD. It is also worth noting that changes in SCFA levels or SCFA-producing bacteria are not unique to PD; similar reductions in fecal SCFAs have been observed in cardiovascular diseases, inflammatory bowel diseases, chronic kidney diseases, and obesity (Lau et al., 2021; Eslick et al., 2022a; Moleón et al., 2023; Ozturk et al., 2024). This suggests that alterations in SCFAs may broadly reflect host homeostatic imbalances rather than serving as specific markers of a particular disease. Therefore, when evaluating the potential of SCFAs as early diagnostic biomarkers for PD, it may be insufficient to monitor only SCFA concentrations or the abundance of SCFA-producing bacteria. A more comprehensive approach would involve combining SCFA measurements with the detection of PD-related pathological markers (such as α-syn) or other biomarkers (including inflammatory cytokines and barrier proteins) in intestinal tissues or blood, along with imaging techniques to assess CNS pathological changes. This multidimensional analytical strategy could provide critical insights into the role of SCFAs in PD pathogenesis and facilitate the development of early diagnostic and therapeutic strategies for the disease.

Most existing studies adopt a cross-sectional design, which is insufficient for establishing causal relationships between SCFAs and the progression of PD. Therefore, longitudinal follow-up studies are urgently needed. It can be hypothesized that in the early stages of PD, when gastrointestinal symptoms emerge, alterations in the gut microbiota have already occurred, characterized by a reduction in the abundance of SCFA-producing bacteria, leading to decreased SCFA levels in the gut. These changes may trigger an imbalance in inflammatory responses within the body, transmitting inflammatory signals to the CNS via the gut-brain axis and initiating neuropathological changes associated with PD. As the disease progresses, inflammatory signals originating in the CNS may propagate back to the gut, further disrupting intestinal homeostasis and creating a vicious cycle. This cycle could exacerbate the ecological imbalance of SCFA-producing bacteria, ultimately impairing SCFA synthesis. To determine whether changes in the composition of SCFA-producing bacteria are a cause or consequence of PD, future studies should prioritize large-scale prospective cohort studies and long-term follow-up investigations.

It is important to emphasize the dual role of SCFAs in neuroinflammation. While most studies suggest that SCFAs primarily exert neuroprotective effects, some findings present contradictory evidence. For instance, Qiao et al. (2020b) reported that sodium butyrate exacerbated MPTP-induced reductions in the neurotransmitters dopamine and 5-HT, promoted neuronal loss in the striatum, and induced intestinal inflammation. In contrast, Liu et al. (2017) demonstrated that sodium butyrate treatment improved cognitive behavior and motor coordination in MPTP-treated mice, prevented dopaminergic neuron degeneration, and reduced tyrosine hydroxylase expression in the striatum. A possible explanation for these inconsistencies could be differences in experimental models, dosing regimens, and treatment durations. In the former study, sodium butyrate was administered at a dose of 165 mg/kg for 1 week, while MPTP was given at 30 mg/kg for 5 consecutive days. In the latter study, sodium butyrate was administered at 200 mg/kg for 3 weeks, with MPTP given at the same dose for 7 consecutive days. These differences in recovery conditions following MPTP administration may alter the therapeutic effects of sodium butyrate. Future studies could employ dose-gradient experiments (e.g., low, medium, and high doses) to determine the effective threshold of SCFAs in different PD models. The dose-dependent neuroprotective effects of butyrate have been well demonstrated in vitro. For instance, in enteric neurons, optimal concentrations of butyrate support cell vitality, while either a deficiency or excessive concentrations inhibit neuronal proliferation (Wang et al., 2023). Similarly, high concentrations of sodium butyrate can suppress the proliferation and survival of neuroendocrine cells (Qiao et al., 2020a), potentially due to cell cycle arrest caused by upregulated oxidative phosphorylation pathways at excessive concentrations (Wang et al., 2023). Moreover, in germ-free conditions, SCFAs have been shown to activate microglia in α-syn overexpression mouse models, exacerbating motor impairments (Sampson et al., 2016). These findings suggest that the effects of SCFAs in the nervous system are regulated by multiple factors, including their type, concentration, treatment duration, experimental conditions, and the host environment. When treating neurodegenerative diseases such as PD, it is crucial to precisely control the dosage and duration of SCFAs according to the disease stage and individual patient variability.

Conclusions

In recent years, research has consistently unveiled the substantial impact of gut microbiota and their metabolites on human health. Since the gut–brain axis theory was proposed, a growing number of studies have focused on the effects of the intestinal microenvironment on CNS disorders. This review provides an overview of the composition of SCFAs and SCFA-producing bacteria in patients with PD. From a nervous system perspective, we summarize the potential molecular mechanisms by which SCFAs modulate ENS and CNS homeostasis in the development of PD. We propose that SCFAs may facilitate communication between the ENS and the CNS through the vagus nerve, blood circulation, and endocrine pathways, thereby enabling bidirectional communication between the gut and the brain (Figure 5). By focusing on the nervous system, our review highlights the pivotal role of SCFAs in mediating neural function regulation and regenerative capacity, offering significant protective effects in PD. Furthermore, we emphasize that SCFAs-mediated homeostasis of the ENS may serve as a potential early-targeting mechanism for the occurrence and intervention of PD.

Figure 5.

Figure 5

SCFAs are a crucial bridge in the gut-brain communication.

SCFAs play a crucial role in maintaining gut health by regulating microbial composition, preserving intestinal barrier integrity, and modulating the structure and function of the ENS. SCFAs also help protect the integrity of the blood-brain barrier and promote the development and maturation of the CNS. A decrease in the abundance of SCFA-producing bacteria in the gut can lead to microbial imbalance, allowing harmful bacteria and their metabolic byproducts to disrupt the intestinal epithelial barrier. Furthermore, environmental toxins and inflammatory factors can induce the abnormal expression and aggregation of α-synuclein in the enteric nervous plexus, triggering the initial signals of PD pathology. The ENS communicates directly with the CNS via the vagus nerve, transmitting the abnormal α-synuclein retrogradely to the CNS. This process leads to glial cell activation, central nervous inflammation, and the degeneration of dopaminergic neurons, ultimately resulting in PD-related motor dysfunction. Additionally, environmental toxins and inflammatory factors can circulate through the bloodstream, breaching the blood-brain barrier to attack the CNS. Therefore, maintaining gut homeostasis may be a prerequisite for the onset of PD and represents a critical therapeutic target for the disease. By preserving gut microbial balance and regulating the synthesis and action of SCFAs, new avenues for early intervention, prevention, and even treatment of PD may be explored. Created with figdraw.com (copyright code: ISURU7ba87). CNS: Central nervous system; ENS: enteric nervous system; PD: Parkinson’s disease; SCFAs: short chain fatty acids.

Funding Statement

Funding: This work was supported by the National Key R&D Program of China, No. 2021YFC2501200 (to PC).

Footnotes

Conflicts of interest: The authors declare no conflicts of interest.

C-Editor: Zhao M; S-Editors: Wang J, Li CH; L-Editor: Song LP; T-Editor: Jia Y

Data availability statement:

Not applicable.

References

  1. Aho VTE, Pereira PAB, Voutilainen S, Paulin L, Pekkonen E, Auvinen P, Scheperjans F. Gut microbiota in Parkinson’s disease: temporal stability and relations to disease progression. EBioMedicine. 2019;44:691–707. doi: 10.1016/j.ebiom.2019.05.064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Aho VTE, Houser MC, Pereira PAB, Chang J, Rudi K, Paulin L, Hertzberg V, Auvinen P, Tansey MG, Scheperjans F. Relationships of gut microbiota, short-chain fatty acids, inflammation, and the gut barrier in Parkinson’s disease. Mol Neurodegener. 2021;16:6. doi: 10.1186/s13024-021-00427-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Al-Bachari S, Naish JH, Parker GJM, Emsley HCA, Parkes LM. Blood-brain barrier leakage is increased in Parkinson’s disease. Front Physiol. 2020;11:593026. doi: 10.3389/fphys.2020.593026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Athauda D, Maclagan K, Skene SS, Bajwa-Joseph M, Letchford D, Chowdhury K, Hibbert S, Budnik N, Zampedri L, Dickson J, Li Y, Aviles-Olmos I, Warner TT, Limousin P, Lees AJ, Greig NH, Tebbs S, Foltynie T. Exenatide once weekly versus placebo in Parkinson’s disease: a randomised, double-blind, placebo-controlled trial. Lancet. 2017;390:1664–1675. doi: 10.1016/S0140-6736(17)31585-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Augustin A, et al. Faecal metabolite deficit, gut inflammation and diet in Parkinson’s disease: integrative analysis indicates inflammatory response syndrome. Clin Transl Med. 2023;13:e1152. doi: 10.1002/ctm2.1152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Barbut D, Stolzenberg E, Zasloff M. Gastrointestinal immunity and alpha-synuclein. J Parkinsons Dis. 2019;9:S313–322. doi: 10.3233/JPD-191702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Barichella M, Severgnini M, Cilia R, Cassani E, Bolliri C, Caronni S, Ferri V, Cancello R, Ceccarani C, Faierman S, Pinelli G, De Bellis G, Zecca L, Cereda E, Consolandi C, Pezzoli G. Unraveling gut microbiota in Parkinson’s disease and atypical parkinsonism. Mov Disord. 2019;34:396–405. doi: 10.1002/mds.27581. [DOI] [PubMed] [Google Scholar]
  8. Barrenschee M, Zorenkov D, Böttner M, Lange C, Cossais F, Scharf AB, Deuschl G, Schneider SA, Ellrichmann M, Fritscher-Ravens A, Wedel T. Distinct pattern of enteric phospho-alpha-synuclein aggregates and gene expression profiles in patients with Parkinson’s disease. Acta Neuropathol Commun. 2017;5:1. doi: 10.1186/s40478-016-0408-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Beach TG, Adler CH, Sue LI, Vedders L, Lue L, White Iii CL, Akiyama H, Caviness JN, Shill HA, Sabbagh MN, Walker DG, Arizona Parkinson’s Disease C. Multi-organ distribution of phosphorylated alpha-synuclein histopathology in subjects with Lewy body disorders. Acta Neuropathol. 2010;119:689–702. doi: 10.1007/s00401-010-0664-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Beach TG, Corbille AG, Letournel F, Kordower JH, Kremer T, Munoz DG, Intorcia A, Hentz J, Adler CH, Sue LI, Walker J, Serrano G, Derkinderen P. Multicenter assessment of immunohistochemical methods for Pathological alpha-synuclein in sigmoid colon of autopsied Parkinson’s disease and control subjects. J Parkinsons Dis. 2016;6:761–770. doi: 10.3233/JPD-160888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Becker L, Nguyen L, Gill J, Kulkarni S, Pasricha PJ, Habtezion A. Age-dependent shift in macrophage polarisation causes inflammation-mediated degeneration of enteric nervous system. Gut. 2018;67:827–836. doi: 10.1136/gutjnl-2016-312940. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Bellono NW, Bayrer JR, Leitch DB, Castro J, Zhang C, O’Donnell TA, Brierley SM, Ingraham HA, Julius D. Enterochromaffin cells are gut chemosensors that couple to sensory neural pathways. Cell. 2017;170:185–198.e116. doi: 10.1016/j.cell.2017.05.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Bilotta AJ, Ma C, Yang W, Yu Y, Yu Y, Zhao X, Zhou Z, Yao S, Dann SM, Cong Y. Propionate enhances cell speed and persistence to promote intestinal epithelial turnover and repair. Cell Mol Gastroenterol Hepatol. 2021;11:1023–1044. doi: 10.1016/j.jcmgh.2020.11.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Bohorquez DV, Samsa LA, Roholt A, Medicetty S, Chandra R, Liddle RA. An enteroendocrine cell-enteric glia connection revealed by 3D electron microscopy. PLoS One. 2014;9:e89881. doi: 10.1371/journal.pone.0089881. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Bohorquez DV, Shahid RA, Erdmann A, Kreger AM, Wang Y, Calakos N, Wang F, Liddle RA. Neuroepithelial circuit formed by innervation of sensory enteroendocrine cells. J Clin Invest. 2015;125:782–786. doi: 10.1172/JCI78361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Bonaz B. The gut-brain axis in Parkinson’s disease. Rev Neurol (Paris) 2024;180:65–78. doi: 10.1016/j.neurol.2023.11.004. [DOI] [PubMed] [Google Scholar]
  17. Braak H, de Vos RA, Bohl J, Del Tredici K. Gastric alpha-synuclein immunoreactive inclusions in meissner’s and auerbach’s plexuses in cases staged for Parkinson’s disease-related brain pathology. Neurosci Lett. 2006;396:67–72. doi: 10.1016/j.neulet.2005.11.012. [DOI] [PubMed] [Google Scholar]
  18. Braniste V, Al-Asmakh M, Kowal C, Anuar F, Abbaspour A, Toth M, Korecka A, Bakocevic N, Ng LG, Kundu P, Gulyas B, Halldin C, Hultenby K, Nilsson H, Hebert H, Volpe BT, Diamond B, Pettersson S. The gut microbiota influences blood-brain barrier permeability in mice. Sci Transl Med. 2014;6:263ra158. doi: 10.1126/scitranslmed.3009759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Brauer R, Wei L, Ma T, Athauda D, Girges C, Vijiaratnam N, Auld G, Whittlesea C, Wong I, Foltynie T. Diabetes medications and risk of Parkinson’s disease: a cohort study of patients with diabetes. Brain. 2020;143:3067–3076. doi: 10.1093/brain/awaa262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Brierley DI, de Lartigue G. Reappraising the role of the vagus nerve in GLP-1-mediated regulation of eating. Br J Pharmacol. 2022;179:584–599. doi: 10.1111/bph.15603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Brown AJ, et al. The Orphan G protein-coupled receptors GPR41 and GPR43 are activated by propionate and other short chain carboxylic acids. J Biol Chem. 2003;278:11312–11319. doi: 10.1074/jbc.M211609200. [DOI] [PubMed] [Google Scholar]
  22. Brun P, Conti J, Zatta V, Russo V, Scarpa M, Kotsafti A, Porzionato A, De Caro R, Scarpa M, Fassan M, Calistri A, Castagliuolo I. Persistent herpes simplex virus type 1 infection of enteric neurons triggers CD8(+) T cell response and gastrointestinal neuromuscular dysfunction. Front Cell Infect Microbiol. 2021;11:615350. doi: 10.3389/fcimb.2021.615350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Buckley MM, O’Brien R, Brosnan E, Ross RP, Stanton C, Buckley JM, O’Malley D. Glucagon-like peptide-1 secreting l-cells coupled to sensory nerves translate microbial signals to the host rat nervous system. Front Cell Neurosci. 2020;14:95. doi: 10.3389/fncel.2020.00095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Caetano-Silva ME, Rund L, Hutchinson NT, Woods JA, Steelman AJ, Johnson RW. Inhibition of inflammatory microglia by dietary fiber and short-chain fatty acids. Sci Rep. 2023;13:2819. doi: 10.1038/s41598-022-27086-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Caetano MAF, Magalhaes HIR, Duarte JRL, Conceicao LB, Castelucci P. Butyrate protects myenteric neurons loss in mice following experimental ulcerative colitis. Cells. 2023;12:1672. doi: 10.3390/cells12131672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Cai X, Cai X, Xie Q, Xiao X, Li T, Zhou T, Sun H. NLRP3 inflammasome and gut microbiota-brain axis: a new perspective on white matter injury after intracerebral hemorrhage. Neural Regen Res. 2025 doi: 10.4103/NRR.NRR-D-24-00917. doi: 10.4103/NRR.NRR-D-24-00917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Camilleri M. Gastrointestinal motility disorders in neurologic disease. J Clin Invest. 2021;131:e143771. doi: 10.1172/JCI143771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Cao N, Merchant W, Gautron L. Limited evidence for anatomical contacts between intestinal GLP-1 cells and vagal neurons in male mice. Sci Rep. 2024;14:23666. doi: 10.1038/s41598-024-74000-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Casini A, Mancinelli R, Mammola CL, Pannarale L, Chirletti P, Onori P, Vaccaro R. Distribution of alpha-synuclein in normal human jejunum and its relations with the chemosensory and neuroendocrine system. Eur J Histochem. 2021;65:3310. doi: 10.4081/ejh.2021.3310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Casini A, Vivacqua G, Ceci L, Leone S, Vaccaro R, Tagliafierro M, Bassi FM, Vitale S, Bocci E, Pannarale L, Carotti S, Franchitto A, Mancini P, Sferra R, Vetuschi A, Latella G, Onori P, Gaudio E, Mancinelli R. TNBS colitis induces architectural changes and alpha-synuclein overexpression in mouse distal colon: a morphological study. Cell Tissue Res. 2024;399:247–265. doi: 10.1007/s00441-024-03932-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Chandra R, Hiniker A, Kuo YM, Nussbaum RL, Liddle RA. Alpha-synuclein in gut endocrine cells and its implications for Parkinson’s disease. JCI Insight. 2017;2:e92295. doi: 10.1172/jci.insight.92295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Chang PV, Hao L, Offermanns S, Medzhitov R. The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition. Proc Natl Acad Sci U S A. 2014;111:2247–2252. doi: 10.1073/pnas.1322269111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Chaudhry TS, Senapati SG, Gadam S, Mannam H, Voruganti HV, Abbasi Z, Abhinav T, Challa AB, Pallipamu N, Bheemisetty N, Arunachalam SP. The impact of microbiota on the gut-brain axis: examining the complex interplay and implications. J Clin Med. 2023;12:5231. doi: 10.3390/jcm12165231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Chen MJ, Feng Y, Gao L, Lin MX, Wang SD, Tong ZQ. Composite sophora colon-soluble capsule ameliorates dss-induced ulcerative colitis in mice via gut microbiota-derived butyric acid and NCR(+) ILC3. Chin J Integr Med. 2023;29:424–433. doi: 10.1007/s11655-022-3317-1. [DOI] [PubMed] [Google Scholar]
  35. 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 patients with Parkinson disease. Neurology. 2022;98:e848–858. doi: 10.1212/WNL.0000000000013225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Cheng Y, Tan G, Zhu Q, Wang C, Ruan G, Ying S, Qie J, Hu X, Xiao Z, Xu F, Chen L, Chen M, Pei Y, Zhang H, Tian Y, Chen D, Liu X, Huang H, Wei Y. Efficacy of fecal microbiota transplantation in patients with Parkinson’s disease: clinical trial results from a randomized, placebo-controlled design. Gut Microbes. 2023;15:2284247. doi: 10.1080/19490976.2023.2284247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Chun E, Lavoie S, Fonseca-Pereira D, Bae S, Michaud M, Hoveyda HR, Fraser GL, Gallini Comeau CA, Glickman JN, Fuller MH, Layden BT, Garrett WS. Metabolite-sensing receptor ffar2 regulates colonic group 3 innate lymphoid cells and gut immunity. Immunity. 2019;51:871–884.e876. doi: 10.1016/j.immuni.2019.09.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Church JS, Bannish JAM, Adrian LA, Rojas Martinez K, Henshaw A, Schwartzer JJ. Serum short chain fatty acids mediate hippocampal BDNF and correlate with decreasing neuroinflammation following high pectin fiber diet in mice. Front Neurosci. 2023;17:1134080. doi: 10.3389/fnins.2023.1134080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Clairembault T, Leclair-Visonneau L, Neunlist M, Derkinderen P. Enteric glial cells: new players in Parkinson’s disease? Mov Disord. 2015;30:494–498. doi: 10.1002/mds.25979. [DOI] [PubMed] [Google Scholar]
  40. Clairembault T, Kamphuis W, Leclair-Visonneau L, Rolli-Derkinderen M, Coron E, Neunlist M, Hol EM, Derkinderen P. Enteric GFAP expression and phosphorylation in Parkinson’s disease. J Neurochem. 2014;130:805–815. doi: 10.1111/jnc.12742. [DOI] [PubMed] [Google Scholar]
  41. Clairembault T, Leclair-Visonneau L, Coron E, Bourreille A, Le Dily S, Vavasseur F, Heymann MF, Neunlist M, Derkinderen P. Structural alterations of the intestinal epithelial barrier in Parkinson’s disease. Acta Neuropathol Commun. 2015;3:12. doi: 10.1186/s40478-015-0196-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Cook TM, Gavini CK, Jesse J, Aubert G, Gornick E, Bonomo R, Gautron L, Layden BT, Mansuy-Aubert V. Vagal neuron expression of the microbiota-derived metabolite receptor, free fatty acid receptor (FFAR3), is necessary for normal feeding behavior. Mol Metab. 2021;54:101350. doi: 10.1016/j.molmet.2021.101350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Cornet A, Savidge TC, Cabarrocas J, Deng WL, Colombel JF, Lassmann H, Desreumaux P, Liblau RS. Enterocolitis induced by autoimmune targeting of enteric glial cells: a possible mechanism in Crohn’s disease? Proc Natl Acad Sci U S A. 2001;98:13306–13311. doi: 10.1073/pnas.231474098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Dalile B, Van Oudenhove L, Vervliet B, Verbeke K. The role of short-chain fatty acids in microbiota-gut-brain communication. Nat Rev Gastroenterol Hepatol. 2019;16:461–478. doi: 10.1038/s41575-019-0157-3. [DOI] [PubMed] [Google Scholar]
  45. De Pablo-Fernandez E, Gebeyehu GG, Flain L, Slater R, Frau A, Ijaz UZ, Warner T, Probert C. The faecal metabolome and mycobiome in Parkinson’s disease. Parkinsonism Relat Disord. 2022;95:65–69. doi: 10.1016/j.parkreldis.2022.01.005. [DOI] [PubMed] [Google Scholar]
  46. De Vadder F, Plessier F, Gautier-Stein A, Mithieux G. Vasoactive intestinal peptide is a local mediator in a gut-brain neural axis activating intestinal gluconeogenesis. Neurogastroenterol Motil. 2015;27:443–448. doi: 10.1111/nmo.12508. [DOI] [PubMed] [Google Scholar]
  47. Del Tredici K, Duda JE. Peripheral Lewy body pathology in Parkinson’s disease and incidental Lewy body disease: four cases. J Neurol Sci. 2011;310:100–106. doi: 10.1016/j.jns.2011.06.003. [DOI] [PubMed] [Google Scholar]
  48. Devos D, Lebouvier T, Lardeux B, Biraud M, Rouaud T, Pouclet H, Coron E, Bruley des Varannes S, Naveilhan P, Nguyen JM, Neunlist M, Derkinderen P. Colonic inflammation in Parkinson’s disease. Neurobiol Dis. 2013;50:42–48. doi: 10.1016/j.nbd.2012.09.007. [DOI] [PubMed] [Google Scholar]
  49. Dinsart G, Leprovots M, Lefort A, Libert F, Quesnel Y, Veithen A, Vassart G, Huysseune S, Parmentier M, Garcia MI. The olfactory receptor Olfr78 promotes differentiation of enterochromaffin cells in the mouse colon. EMBO Rep. 2024;25:304–333. doi: 10.1038/s44319-023-00013-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Docampo MD, et al. Alloreactive T cells deficient of the short-chain fatty acid receptor GPR109A induce less graft-versus-host disease. Blood. 2022;139:2392–2405. doi: 10.1182/blood.2021010719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Dong XL, Wang X, Liu F, Liu X, Du ZR, Li RW, Xue CH, Wong KH, Wong WT, Zhao Q, Tang QJ. Polymannuronic acid prevents dopaminergic neuronal loss via brain-gut-microbiota axis in Parkinson’s disease model. Int J Biol Macromol. 2020;164:994–1005. doi: 10.1016/j.ijbiomac.2020.07.180. [DOI] [PubMed] [Google Scholar]
  52. Ducastel S, Touche V, Trabelsi MS, Boulinguiez A, Butruille L, Nawrot M, Peschard S, Chávez-Talavera O, Dorchies E, Vallez E, Annicotte JS, Lancel S, Briand O, Bantubungi K, Caron S, Bindels LB, Delzenne NM, Tailleux A, Staels B, Lestavel S. The nuclear receptor FXR inhibits glucagon-like peptide-1 secretion in response to microbiota-derived short-chain fatty acids. Sci Rep. 2020;10:174. doi: 10.1038/s41598-019-56743-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Dumitrescu L, Marta D, Danau A, Lefter A, Tulba D, Cozma L, Manole E, Gherghiceanu M, Ceafalan LC, Popescu BO. Serum and fecal markers of intestinal inflammation and intestinal barrier permeability are elevated in Parkinson’s disease. Front Neurosci. 2021;15:689723. doi: 10.3389/fnins.2021.689723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Dupraz L, Magniez A, Rolhion N, Richard ML, Da Costa G, Touch S, Mayeur C, Planchais J, Agus A, Danne C, Michaudel C, Spatz M, Trottein F, Langella P, Sokol H, Michel ML. Gut microbiota-derived short-chain fatty acids regulate IL-17 production by mouse and human intestinal γδ T cells. Cell Rep. 2021;36:109332. doi: 10.1016/j.celrep.2021.109332. [DOI] [PubMed] [Google Scholar]
  55. Ediriweera MK. Fatty acids as histone deacetylase inhibitors: old biochemistry tales in a new life sciences town. Drug Discov Today. 2023;28:103569. doi: 10.1016/j.drudis.2023.103569. [DOI] [PubMed] [Google Scholar]
  56. Engevik MA, Luck B, Visuthranukul C, Ihekweazu FD, Engevik AC, Shi Z, Danhof HA, Chang-Graham AL, Hall A, Endres BT, Haidacher SJ, Horvath TD, Haag AM, Devaraj S, Garey KW, Britton RA, Hyser JM, Shroyer NF, Versalovic J. Human-derived bifidobacterium dentium modulates the mammalian serotonergic system and gut-brain axis. Cell Mol Gastroenterol Hepatol. 2021;11:221–248. doi: 10.1016/j.jcmgh.2020.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Erny D, Hrabe de Angelis AL, Jaitin D, Wieghofer P, Staszewski O, David E, Keren-Shaul H, Mahlakoiv T, Jakobshagen K, Buch T, Schwierzeck V, Utermohlen O, Chun E, Garrett WS, McCoy KD, Diefenbach A, Staeheli P, Stecher B, Amit I, Prinz M. Host microbiota constantly control maturation and function of microglia in the CNS. Nat Neurosci. 2015;18:965–977. doi: 10.1038/nn.4030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Eslick S, Thompson C, Berthon B, Wood L. Short-chain fatty acids as anti-inflammatory agents in overweight and obesity: a systematic review and meta-analysis. Nutr Rev. 2022;80:838–856. doi: 10.1093/nutrit/nuab059. [DOI] [PubMed] [Google Scholar]
  59. Eslick S, Williams EJ, Berthon BS, Wright T, Karihaloo C, Gately M, Wood LG. Weight loss and short-chain fatty acids reduce systemic inflammation in monocytes and adipose tissue macrophages from obese subjects. Nutrients. 2022;14:765. doi: 10.3390/nu14040765. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Fachi JL, et al. Acetate coordinates neutrophil and ILC3 responses against C. difficile through FFAR2. J Exp Med. 2020;217:jem.20190489. doi: 10.1084/jem.20190489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Fang X, Liu S, Muhammad B, Zheng M, Ge X, Xu Y, Kan S, Zhang Y, Yu Y, Zheng K, Geng D, Liu CF. Gut microbiota dysbiosis contributes to alpha-synuclein-related pathology associated with C/EBPbeta/AEP signaling activation in a mouse model of Parkinson’s disease. Neural Regen Res. 2024;19:2081–2088. doi: 10.4103/1673-5374.391191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Forsyth CB, Shannon KM, Kordower JH, Voigt RM, Shaikh M, Jaglin JA, Estes JD, Dodiya HB, Keshavarzian A. Increased intestinal permeability correlates with sigmoid mucosa alpha-synuclein staining and endotoxin exposure markers in early Parkinson’s disease. PLoS One. 2011;6:e28032. doi: 10.1371/journal.pone.0028032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Francelle L, Outeiro TF, Rappold GA. Inhibition of HDAC6 activity protects dopaminergic neurons from alpha-synuclein toxicity. Sci Rep. 2020;10:6064. doi: 10.1038/s41598-020-62678-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Fu SC, Shih LC, Wu PH, Hsieh YC, Lee CH, Lin SH, Wang H. Exploring the causal effect of constipation on Parkinson’s disease through mediation analysis of microbial data. Front Cell Infect Microbiol. 2022;12:871710. doi: 10.3389/fcimb.2022.871710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Fung C, Cools B, Malagola S, Martens T, Tack J, Kazwiny Y, Vanden Berghe P. Luminal short-chain fatty acids and 5-HT acutely activate myenteric neurons in the mouse proximal colon. Neurogastroenterol Motil. 2021;33:e14186. doi: 10.1111/nmo.14186. [DOI] [PubMed] [Google Scholar]
  66. Gabanyi I, Muller PA, Feighery L, Oliveira TY, Costa-Pinto FA, Mucida D. Neuro-immune interactions drive tissue programming in intestinal macrophages. Cell. 2016;164:378–391. doi: 10.1016/j.cell.2015.12.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Garretti F, Monahan C, Sloan N, Bergen J, Shahriar S, Kim SW, Sette A, Cutforth T, Kanter E, Agalliu D, Sulzer D. Interaction of an α-synuclein epitope with HLA-DRB1(*)15:01 triggers enteric features in mice reminiscent of prodromal Parkinson’s disease. Neuron. 2023;111:3397–3413.e3395. doi: 10.1016/j.neuron.2023.07.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Gaudier E, Jarry A, Blottière HM, de Coppet P, Buisine MP, Aubert JP, Laboisse C, Cherbut C, Hoebler C. Butyrate specifically modulates MUC gene expression in intestinal epithelial goblet cells deprived of glucose. Am J Physiol Gastrointest Liver Physiol. 2004;287:G1168–1174. doi: 10.1152/ajpgi.00219.2004. [DOI] [PubMed] [Google Scholar]
  69. Gelpi E, Navarro-Otano J, Tolosa E, Gaig C, Compta Y, Rey MJ, Marti MJ, Hernandez I, Valldeoriola F, Rene R, Ribalta T. Multiple organ involvement by alpha-synuclein pathology in Lewy body disorders. Mov Disord. 2014;29:1010–1018. doi: 10.1002/mds.25776. [DOI] [PubMed] [Google Scholar]
  70. Geng ZH, Zhu Y, Li QL, Zhao C, Zhou PH. Enteric nervous system: the bridge between the gut microbiota and neurological disorders. Front Aging Neurosci. 2022;14:810483. doi: 10.3389/fnagi.2022.810483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Gershon MD. The shaggy dog story of enteric signaling: serotonin, a molecular megillah. Adv Exp Med Biol. 2022;1383:307–318. doi: 10.1007/978-3-031-05843-1_28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Gorecki AM, Preskey L, Bakeberg MC, Kenna JE, Gildenhuys C, MacDougall G, Dunlop SA, Mastaglia FL, Akkari PA, Koengten F, Anderton RS. Altered gut microbiome in parkinson’s disease and the influence of lipopolysaccharide in a human alpha-synuclein over-expressing mouse model. Front Neurosci. 2019;13:839. doi: 10.3389/fnins.2019.00839. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Gray MT, Woulfe JM. Striatal blood-brain barrier permeability in Parkinson’s disease. J Cereb Blood Flow Metab. 2015;35:747–750. doi: 10.1038/jcbfm.2015.32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Grozdanov V, Bliederhaeuser C, Ruf WP, Roth V, Fundel-Clemens K, Zondler L, Brenner D, Martin-Villalba A, Hengerer B, Kassubek J, Ludolph AC, Weishaupt JH, Danzer KM. Inflammatory dysregulation of blood monocytes in Parkinson’s disease patients. Acta Neuropathol. 2014;128:651–663. doi: 10.1007/s00401-014-1345-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Grunddal KV, Jensen EP, Ørskov C, Andersen DB, Windeløv JA, Poulsen SS, Rosenkilde MM, Knudsen LB, Pyke C, Holst JJ. Expression profile of the GLP-1 receptor in the gastrointestinal tract and pancreas in adult female mice. Endocrinology. 2022;163:bqab216. doi: 10.1210/endocr/bqab216. [DOI] [PubMed] [Google Scholar]
  76. Guo Q, Hou X, Cui Q, Li S, Shen G, Luo Q, Wu H, Chen H, Liu Y, Chen A, Zhang Z. Pectin mediates the mechanism of host blood glucose regulation through intestinal flora. Crit Rev Food Sci Nutr. 2024;64:6714–6736. doi: 10.1080/10408398.2023.2173719. [DOI] [PubMed] [Google Scholar]
  77. Guo TT, Zhang Z, Sun Y, Zhu RY, Wang FX, Ma LJ, Jiang L, Liu HD. Neuroprotective effects of sodium butyrate by restoring gut microbiota and inhibiting TLR4 signaling in mice with MPTP-induced Parkinson’s disease. Nutrients. 2023;15:930. doi: 10.3390/nu15040930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Hallett PJ, McLean JR, Kartunen A, Langston JW, Isacson O. alpha-Synuclein overexpressing transgenic mice show internal organ pathology and autonomic deficits. Neurobiol Dis. 2012;47:258–267. doi: 10.1016/j.nbd.2012.04.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Han YE, Kang CW, Oh JH, Park SH, Ku CR, Cho YH, Lee MK, Lee EJ. Olfactory receptor OR51E1 mediates GLP-1 secretion in human and rodent enteroendocrine l cells. J Endoc Soc. 2018;2:1251–1258. doi: 10.1210/js.2018-00165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Hasegawa S, Goto S, Tsuji H, Okuno T, Asahara T, Nomoto K, Shibata A, Fujisawa Y, Minato T, Okamoto A, Ohno K, Hirayama M. Intestinal dysbiosis and lowered serum lipopolysaccharide-binding protein in Parkinson’s disease. PLoS One. 2015;10:e0142164. doi: 10.1371/journal.pone.0142164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Hasuike Y, Endo T, Koroyasu M, Matsui M, Mori C, Yamadera M, Fujimura H, Sakoda S. Bile acid abnormality induced by intestinal dysbiosis might explain lipid metabolism in Parkinson’s disease. Med Hypotheses. 2020;134:109436. doi: 10.1016/j.mehy.2019.109436. [DOI] [PubMed] [Google Scholar]
  82. He X, Qian Y, Xu S, Zhang Y, Mo C, Guo W, Yang X, Xiao Q. Plasma short-chain fatty acids differences in multiple system atrophy from Parkinson’s disease. J Parkinsons Dis. 2021;11:1167–1176. doi: 10.3233/JPD-212604. [DOI] [PubMed] [Google Scholar]
  83. Hertati A, Hayashi S, Ogata H, Miyata K, Kato R, Yamamoto T, Kadowaki M. Morphological elucidation of short-chain fatty acid receptor GPR41-positive enteric sensory neurons in the colon of mice with dextran sulfate sodium-induced colitis. Heliyon. 2020;6:e05647. doi: 10.1016/j.heliyon.2020.e05647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Hill-Burns EM, Debelius JW, Morton JT, Wissemann WT, Lewis MR, Wallen ZD, Peddada SD, Factor SA, Molho E, Zabetian CP, Knight R, Payami H. Parkinson’s disease and Parkinson’s disease medications have distinct signatures of the gut microbiome. Mov Disord. 2017;32:739–749. doi: 10.1002/mds.26942. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Holmqvist S, Chutna O, Bousset L, Aldrin-Kirk P, Li W, Bjorklund T, Wang ZY, Roybon L, Melki R, Li JY. Direct evidence of Parkinson pathology spread from the gastrointestinal tract to the brain in rats. Acta Neuropathol. 2014;128:805–820. doi: 10.1007/s00401-014-1343-6. [DOI] [PubMed] [Google Scholar]
  86. Holzer P, Reichmann F, Farzi A. Neuropeptide Y, peptide YY and pancreatic polypeptide in the gut-brain axis. Neuropeptides. 2012;46:261–274. doi: 10.1016/j.npep.2012.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Hou Y, Li X, Liu C, Zhang M, Zhang X, Ge S, Zhao L. Neuroprotective effects of short-chain fatty acids in MPTP induced mice model of Parkinson’s disease. Exp Gerontol. 2021;150:111376. doi: 10.1016/j.exger.2021.111376. [DOI] [PubMed] [Google Scholar]
  88. Hou YF, Shan C, Zhuang SY, Zhuang QQ, Ghosh A, Zhu KC, Kong XK, Wang SM, Gong YL, Yang YY, Tao B, Sun LH, Zhao HY, Guo XZ, Wang WQ, Ning G, Gu YY, Li ST, Liu JM. Gut microbiota-derived propionate mediates the neuroprotective effect of osteocalcin in a mouse model of Parkinson’s disease. Microbiome. 2021;9:34. doi: 10.1186/s40168-020-00988-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Hoyles L, Snelling T, Umlai UK, Nicholson JK, Carding SR, Glen RC, McArthur S. Microbiome-host systems interactions: protective effects of propionate upon the blood-brain barrier. Microbiome. 2018;6:55. doi: 10.1186/s40168-018-0439-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Hsu SW, Hsu PC, Chang WS, Yu CC, Wang YC, Yang JS, Tsai FJ, Chen KY, Tsai CW, Bau DT. Protective effects of valproic acid on 6-hydroxydopamine-induced neuroinjury. Environ Toxicol. 2020;35:840–848. doi: 10.1002/tox.22920. [DOI] [PubMed] [Google Scholar]
  91. Huang P, Zhang P, Du J, Gao C, Liu J, Tan Y, Chen S. Association of fecal short-chain fatty acids with clinical severity and gut microbiota in essential tremor and its difference from Parkinson’s disease. NPJ Parkinsons Dis. 2023;9:115. doi: 10.1038/s41531-023-00554-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Huang T, Shi H, Xu Y, Ji L. The gut microbiota metabolite propionate ameliorates intestinal epithelial barrier dysfunction-mediated Parkinson’s disease via the AKT signaling pathway. Neuroreport. 2021;32:244–251. doi: 10.1097/WNR.0000000000001585. [DOI] [PubMed] [Google Scholar]
  93. Hurst NR, Kendig DM, Murthy KS, Grider JR. The short chain fatty acids, butyrate and propionate, have differential effects on the motility of the guinea pig colon. Neurogastroenterol Motil. 2014;26:1586–1596. doi: 10.1111/nmo.12425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Ibiza S, García-Cassani B, Ribeiro H, Carvalho T, Almeida L, Marques R, Misic AM, Bartow-McKenney C, Larson DM, Pavan WJ, Eberl G, Grice EA, Veiga-Fernandes H. Glial-cell-derived neuroregulators control type 3 innate lymphoid cells and gut defence. Nature. 2016;535:440–443. doi: 10.1038/nature18644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Ikeda T, Nishida A, Yamano M, Kimura I. Short-chain fatty acid receptors and gut microbiota as therapeutic targets in metabolic, immune, and neurological diseases. Pharmacol Ther. 2022;239:108273. doi: 10.1016/j.pharmthera.2022.108273. [DOI] [PubMed] [Google Scholar]
  96. Janova H, Zhao FR, Desai P, Mack M, Thackray LB, Stappenbeck TS, Diamond MS. West Nile virus triggers intestinal dysmotility via T cell-mediated enteric nervous system injury. J Clin Invest. 2024;134:e181421. doi: 10.1172/JCI181421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Javed D, Jajja FA, Javed A. GLP-1 receptor agonists: a revolution in the treatment of Parkinson’s disease? Eur Neuropsychopharmacol. 2024;86:11–12. doi: 10.1016/j.euroneuro.2024.06.003. [DOI] [PubMed] [Google Scholar]
  98. Jayaraj RL, Beiram R, Azimullah S, Mf NM, Ojha SK, Adem A, Jalal FY. Valeric acid protects dopaminergic neurons by suppressing oxidative stress, neuroinflammation and modulating autophagy pathways. Int J Mol Sci. 2020;21:7670. doi: 10.3390/ijms21207670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Jessup D, Woods K, Thakker S, Damaj MI, Akbarali HI. Short-chain fatty acid, butyrate prevents morphine-and paclitaxel-induced nociceptive hypersensitivity. Sci Rep. 2023;13:17805. doi: 10.1038/s41598-023-44857-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Jiao F, Meng L, Du K, Li X. The autophagy-lysosome pathway: a potential target in the chemical and gene therapeutic strategies for Parkinson’s disease. Neural Regen Res. 2025;20:139–158. doi: 10.4103/NRR.NRR-D-23-01195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Jing Y, Yang D, Bai F, Wang Q, Zhang C, Yan Y, Li Z, Li Y, Chen Z, Li J, Yu Y. Spinal cord injury-induced gut dysbiosis influences neurological recovery partly through short-chain fatty acids. NPJ Biofilms Microbiomes. 2023;9:99. doi: 10.1038/s41522-023-00466-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Johnson J, Jr., Pajarillo E, Karki P, Kim J, Son DS, Aschner M, Lee E. Valproic acid attenuates manganese-induced reduction in expression of GLT-1 and GLAST with concomitant changes in murine dopaminergic neurotoxicity. Neurotoxicology. 2018;67:112–120. doi: 10.1016/j.neuro.2018.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Johnson J, Jr., Pajarillo EAB, Taka E, Reams R, Son DS, Aschner M, Lee E. Valproate and sodium butyrate attenuate manganese-decreased locomotor activity and astrocytic glutamate transporters expression in mice. Neurotoxicology. 2018;64:230–239. doi: 10.1016/j.neuro.2017.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Kaakkola S. Clinical pharmacology, therapeutic use and potential of COMT inhibitors in Parkinson’s disease. Drugs. 2000;59:1233–1250. doi: 10.2165/00003495-200059060-00004. [DOI] [PubMed] [Google Scholar]
  105. Kaelberer MM, Buchanan KL, Klein ME, Barth BB, Montoya MM, Shen X, Bohorquez DV. A gut-brain neural circuit for nutrient sensory transduction. Science. 2018;361:eaat5236. doi: 10.1126/science.aat5236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Kaji I, Akiba Y, Konno K, Watanabe M, Kimura S, Iwanaga T, Kuri A, Iwamoto K, Kuwahara A, Kaunitz JD. Neural FFA3 activation inversely regulates anion secretion evoked by nicotinic ACh receptor activation in rat proximal colon. J Physiol. 2016;594:3339–3352. doi: 10.1113/JP271441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Kaji I, Akiba Y, Furuyama T, Adelson DW, Iwamoto K, Watanabe M, Kuwahara A, Kaunitz JD. Free fatty acid receptor 3 activation suppresses neurogenic motility in rat proximal colon. Neurogastroenterol Motil. 2018 doi: 10.1111/nmo.13157. doi: 10.1111/nmo.13157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Kang T, Zhu L, Xue Y, Yang Q, Lei Q, Wang Q. Overexpression of olfactory receptor 78 ameliorates brain injury in cerebral ischaemia-reperfusion rats by activating Prkaca-mediated cAMP/PKA-MAPK pathway. J Cell Mol Med. 2024;28:e18366. doi: 10.1111/jcmm.18366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Katsurada K, Yada T. Neural effects of gut- and brain-derived glucagon-like peptide-1 and its receptor agonist. J Diabetes Investig 7 Suppl. 2016;1:64–69. doi: 10.1111/jdi.12464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Keating DJ, Spencer NJ. What is the role of endogenous gut serotonin in the control of gastrointestinal motility? Pharmacol Res. 2019;140:50–55. doi: 10.1016/j.phrs.2018.06.017. [DOI] [PubMed] [Google Scholar]
  111. Keshavarzian A, Green SJ, Engen PA, Voigt RM, Naqib A, Forsyth CB, Mutlu E, Shannon KM. Colonic bacterial composition in Parkinson’s disease. Mov Disord. 2015;30:1351–1360. doi: 10.1002/mds.26307. [DOI] [PubMed] [Google Scholar]
  112. Kidd SK, Schneider JS. Protection of dopaminergic cells from MPP+-mediated toxicity by histone deacetylase inhibition. Brain Res. 2010;1354:172–178. doi: 10.1016/j.brainres.2010.07.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Kim JS, Sung HY. Gastrointestinal autonomic dysfunction in patients with Parkinson’s disease. J Mov Disord. 2015;8:76–82. doi: 10.14802/jmd.15008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Kim M, Qie Y, Park J, Kim CH. Gut microbial metabolites fuel host antibody responses. Cell Host Microbe. 2016;20:202–214. doi: 10.1016/j.chom.2016.07.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Kim S, Kwon SH, Kam TI, Panicker N, Karuppagounder SS, Lee S, Lee JH, Kim WR, Kook M, Foss CA, Shen C, Lee H, Kulkarni S, Pasricha PJ, Lee G, Pomper MG, Dawson VL, Dawson TM, Ko HS. Transneuronal propagation of pathologic alpha-synuclein from the gut to the brain models Parkinson’s disease. Neuron. 2019;103:627–641e627. doi: 10.1016/j.neuron.2019.05.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Klose CSN, Mahlakõiv T, Moeller JB, Rankin LC, Flamar AL, Kabata H, Monticelli LA, Moriyama S, Putzel GG, Rakhilin N, Shen X, Kostenis E, König GM, Senda T, Carpenter D, Farber DL, Artis D. The neuropeptide neuromedin U stimulates innate lymphoid cells and type 2 inflammation. Nature. 2017;549:282–286. doi: 10.1038/nature23676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Kortekaas R, Leenders KL, van Oostrom JC, Vaalburg W, Bart J, Willemsen AT, Hendrikse NH. Blood-brain barrier dysfunction in parkinsonian midbrain in vivo. Ann Neurol. 2005;57:176–179. doi: 10.1002/ana.20369. [DOI] [PubMed] [Google Scholar]
  118. Kulkarni S, et al. Adult enteric nervous system in health is maintained by a dynamic balance between neuronal apoptosis and neurogenesis. Proc Natl Acad Sci U S A. 2017;114:e3709–3718. doi: 10.1073/pnas.1619406114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Kumari S, Goyal V, Kumaran SS, Dwivedi SN, Srivastava A, Jagannathan NR. Quantitative metabolomics of saliva using proton NMR spectroscopy in patients with Parkinson’s disease and healthy controls. Neurol Sci. 2020;41:1201–1210. doi: 10.1007/s10072-019-04143-4. [DOI] [PubMed] [Google Scholar]
  120. Kumari S, Kumaran SS, Goyal V, Sharma RK, Sinha N, Dwivedi SN, Srivastava AK, Jagannathan NR. Identification of potential urine biomarkers in idiopathic parkinson’s disease using NMR. Clin Chim Acta. 2020;510:442–449. doi: 10.1016/j.cca.2020.08.005. [DOI] [PubMed] [Google Scholar]
  121. Kuo YM, Li Z, Jiao Y, Gaborit N, Pani AK, Orrison BM, Bruneau BG, Giasson BI, Smeyne RJ, Gershon MD, Nussbaum RL. Extensive enteric nervous system abnormalities in mice transgenic for artificial chromosomes containing Parkinson disease-associated alpha-synuclein gene mutations precede central nervous system changes. Hum Mol Genet. 2010;19:1633–1650. doi: 10.1093/hmg/ddq038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Lal S, Kirkup AJ, Brunsden AM, Thompson DG, Grundy D. Vagal afferent responses to fatty acids of different chain length in the rat. Am J Physiol Gastrointest Liver Physiol. 2001;281:G907–915. doi: 10.1152/ajpgi.2001.281.4.G907. [DOI] [PubMed] [Google Scholar]
  123. Larraufie P, Martin-Gallausiaux C, Lapaque N, Dore J, Gribble FM, Reimann F, Blottiere HM. SCFAs strongly stimulate PYY production in human enteroendocrine cells. Sci Rep. 2018;8:74. doi: 10.1038/s41598-017-18259-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Lau WL, Chang Y, Vaziri ND. The consequences of altered microbiota in immune-related chronic kidney disease. Nephrol Dial Transplant. 2021;36:1791–1798. doi: 10.1093/ndt/gfaa087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Le Poul E, Loison C, Struyf S, Springael JY, Lannoy V, Decobecq ME, Brezillon S, Dupriez V, Vassart G, Van Damme J, Parmentier M, Detheux M. Functional characterization of human receptors for short chain fatty acids and their role in polymorphonuclear cell activation. J Biol Chem. 2003;278:25481–25489. doi: 10.1074/jbc.M301403200. [DOI] [PubMed] [Google Scholar]
  126. Li P, Killinger BA, Ensink E, Beddows I, Yilmaz A, Lubben N, Lamp J, Schilthuis M, Vega IE, Woltjer R, Pospisilik JA, Brundin P, Brundin L, Graham SF, Labrie V. Gut microbiota dysbiosis is associated with elevated bile acids in Parkinson’s disease. Metabolites. 2021;11:29. doi: 10.3390/metabo11010029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Li W, Wu X, Hu X, Wang T, Liang S, Duan Y, Jin F, Qin B. Structural changes of gut microbiota in Parkinson’s disease and its correlation with clinical features. Sci China Life Sci. 2017;60:1223–1233. doi: 10.1007/s11427-016-9001-4. [DOI] [PubMed] [Google Scholar]
  128. Li Y, Perry T, Kindy MS, Harvey BK, Tweedie D, Holloway HW, Powers K, Shen H, Egan JM, Sambamurti K, Brossi A, Lahiri DK, Mattson MP, Hoffer BJ, Wang Y, Greig NH. GLP-1 receptor stimulation preserves primary cortical and dopaminergic neurons in cellular and rodent models of stroke and Parkinsonism. Proc Natl Acad Sci U S A. 2009;106:1285–1290. doi: 10.1073/pnas.0806720106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Li Z, Chalazonitis A, Huang YY, Mann JJ, Margolis KG, Yang QM, Kim DO, Côté F, Mallet J, Gershon MD. Essential roles of enteric neuronal serotonin in gastrointestinal motility and the development/survival of enteric dopaminergic neurons. J Neurosci. 2011;31:8998–9009. doi: 10.1523/JNEUROSCI.6684-10.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Lin Y, Lv Y, Mao Z, Chen X, Chen Y, Zhu B, Yu Y, Ding Z, Zhou F. Polysaccharides from Tetrastigma Hemsleyanum Diels et Gilg ameliorated inflammatory bowel disease by rebuilding the intestinal mucosal barrier and inhibiting inflammation through the SCFA-GPR41/43 signaling pathway. Int J Biol Macromol. 2023;250:126167. doi: 10.1016/j.ijbiomac.2023.126167. [DOI] [PubMed] [Google Scholar]
  131. Liu J, Wang F, Liu S, Du J, Hu X, Xiong J, Fang R, Chen W, Sun J. Sodium butyrate exerts protective effect against Parkinson’s disease in mice via stimulation of glucagon like peptide-1. J Neurol Sci. 2017;381:176–181. doi: 10.1016/j.jns.2017.08.3235. [DOI] [PubMed] [Google Scholar]
  132. Liu J, Lv X, Ye T, Zhao M, Chen Z, Zhang Y, Yang W, Xie H, Zhan L, Chen L, Liu WC, Su KP, Sun J. Microbiota-microglia crosstalk between Blautia producta and neuroinflammation of Parkinson’s disease: a bench-to-bedside translational approach. Brain Behav Immun. 2024;117:270–282. doi: 10.1016/j.bbi.2024.01.010. [DOI] [PubMed] [Google Scholar]
  133. Lu VB, Gribble FM, Reimann F. Free fatty acid receptors in enteroendocrine cells. Endocrinology. 2018;159:2826–2835. doi: 10.1210/en.2018-00261. [DOI] [PubMed] [Google Scholar]
  134. Ma X, Wang Q, Yuan W, Wang Y, Zhou F, Kang K, Tong X, Liu Z. Electroacupuncture alleviates neuroinflammation and motor dysfunction by regulating intestinal barrier function in a mouse model of Parkinson disease. J Neuropathol Exp Neurol. 2021;80:844–855. doi: 10.1093/jnen/nlab046. [DOI] [PubMed] [Google Scholar]
  135. Mac M, Nalecz KA. Expression of monocarboxylic acid transporters (MCT) in brain cells. Implication for branched chain alpha-ketoacids transport in neurons. Neurochem Int. 2003;43:305–309. doi: 10.1016/s0197-0186(03)00016-0. [DOI] [PubMed] [Google Scholar]
  136. Mackie PM, Koshy J, Bhogade M, Hammoor T, Hachmeister W, Lloyd GM, Paterno G, Bolen M, Tansey MG, Giasson BI, Khoshbouei H. Complement C1q-dependent engulfment of alpha-synuclein induces ENS-resident macrophage exhaustion and accelerates Parkinson’s-like gut pathology. BioRxiv. 2023 doi: 10.1101/2023.10.24.563832. [Google Scholar]
  137. Maisterrena A, Matas E, Mirfendereski H, Balbous A, Marchand S, Jaber M. The state of the dopaminergic and glutamatergic systems in the valproic acid mouse model of autism spectrum disorder. Biomolecules. 2022;12:1691. doi: 10.3390/biom12111691. [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Mamedova LK, Krogstad KC, McDonald PO, Pokhrel L, Hua DH, Titgemeyer EC, Bradford BJ. Investigation of HCAR2 antagonists as a potential strategy to modulate bovine leukocytes. J Anim Sci Biotechnol. 2024;15:38. doi: 10.1186/s40104-024-00999-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Mann ER, Lam YK, Uhlig HH. Short-chain fatty acids: linking diet, the microbiome and immunity. Nat Rev Immunol. 2024;24:577–595. doi: 10.1038/s41577-024-01014-8. [DOI] [PubMed] [Google Scholar]
  140. Marques de Souza PR, Keenan CM, Wallace LE, Habibyan YB, Davoli-Ferreira M, Ohland C, Vicentini FA, McCoy KD, Sharkey KA. T cells regulate intestinal motility and shape enteric neuronal responses to intestinal microbiota. Gut Microbes. 2025;17:2442528. doi: 10.1080/19490976.2024.2442528. [DOI] [PubMed] [Google Scholar]
  141. Martin AM, Jones LA, Wei L, Spencer NJ, Sanders KM, Ro S, Keating DJ. Distinguishing the contributions of neuronal and mucosal serotonin in the regulation of colonic motility. Neurogastroenterol Motil. 2022;34:e14361. doi: 10.1111/nmo.14361. [DOI] [PubMed] [Google Scholar]
  142. Masse KE, Lu VB. Short-chain fatty acids, secondary bile acids and indoles: gut microbial metabolites with effects on enteroendocrine cell function and their potential as therapies for metabolic disease. Front Endocrinol (Lausanne) 2023;14:1169624. doi: 10.3389/fendo.2023.1169624. [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Matheis F, Muller PA, Graves CL, Gabanyi I, Kerner ZJ, Costa-Borges D, Ahrends T, Rosenstiel P, Mucida D. Adrenergic signaling in muscularis macrophages limits infection-induced neuronal loss. Cell. 2020;180:64–78.e16. doi: 10.1016/j.cell.2019.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Mazzetti S, De Leonardis M, Gagliardi G, Calogero AM, Basellini MJ, Madaschi L, Costa I, Cacciatore F, Spinello S, Bramerio M, Cilia R, Rolando C, Giaccone G, Pezzoli G, Cappelletti G. Phospho-HDAC6 gathers into protein aggregates in parkinson’s disease and atypical Parkinsonisms. Front Neurosci. 2020;14:624. doi: 10.3389/fnins.2020.00624. [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. McBride DA, Dorn NC, Yao M, Johnson WT, Wang W, Bottini N, Shah NJ. Short-chain fatty acid-mediated epigenetic modulation of inflammatory T cells in vitro. Drug Deliv Transl Res. 2023;13:1912–1924. doi: 10.1007/s13346-022-01284-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Meka ST, Bojja SL, Kumar G, Birangal SR, Rao CM. Novel HDAC inhibitors provide neuroprotection in MPTP-induced Parkinson’s disease model of rats. Eur J Pharmacol. 2023;959:176067. doi: 10.1016/j.ejphar.2023.176067. [DOI] [PubMed] [Google Scholar]
  147. Mi N, Ma L, Li X, Fu J, Bu X, Liu F, Yang F, Zhang Y, Yao L. Metabolomic analysis of serum short-chain fatty acid concentrations in a mouse of MPTP-induced Parkinson’s disease after dietary supplementation with branched-chain amino acids. Open Med (Wars) 2023;18:20230849. doi: 10.1515/med-2023-0849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  148. Miki Y, Mori F, Tanji K, Kakita A, Takahashi H, Wakabayashi K. Accumulation of histone deacetylase 6, an aggresome-related protein, is specific to Lewy bodies and glial cytoplasmic inclusions. Neuropathology. 2011;31:561–568. doi: 10.1111/j.1440-1789.2011.01200.x. [DOI] [PubMed] [Google Scholar]
  149. Mishra SP, Jain S, Wang B, Wang S, Miller BC, Lee JY, Borlongan CV, Jiang L, Pollak J, Taraphder S, Layden BT, Rane SG, Yadav H. Abnormalities in microbiota/butyrate/FFAR3 signaling in aging gut impair brain function. JCI Insight. 2024;9:e168443. doi: 10.1172/jci.insight.168443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Moleón J, González-Correa C, Miñano S, Robles-Vera I, de la Visitación N, Barranco AM, Gómez-Guzmán M, Sánchez M, Riesco P, Guerra-Hernández E, Toral M, Romero M, Duarte J. Protective effect of microbiota-derived short chain fatty acids on vascular dysfunction in mice with systemic lupus erythematosus induced by toll like receptor 7 activation. Pharmacol Res. 2023;198:106997. doi: 10.1016/j.phrs.2023.106997. [DOI] [PubMed] [Google Scholar]
  151. Morimoto R, Satoh F, Murakami O, Totsune K, Saruta M, Suzuki T, Sasano H, Ito S, Takahashi K. Expression of peptide YY in human brain and pituitary tissues. Nutrition. 2008;24:878–884. doi: 10.1016/j.nut.2008.06.011. [DOI] [PubMed] [Google Scholar]
  152. Morris HR, Spillantini MG, Sue CM, Williams-Gray CH. The pathogenesis of Parkinson’s disease. Lancet. 2024;403:293–304. doi: 10.1016/S0140-6736(23)01478-2. [DOI] [PubMed] [Google Scholar]
  153. Muller PA, Koscsó B, Rajani GM, Stevanovic K, Berres ML, Hashimoto D, Mortha A, Leboeuf M, Li XM, Mucida D, Stanley ER, Dahan S, Margolis KG, Gershon MD, Merad M, Bogunovic M. Crosstalk between muscularis macrophages and enteric neurons regulates gastrointestinal motility. Cell. 2014;158:300–313. doi: 10.1016/j.cell.2014.04.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Nakamori H, Niimi A, Mitsui R, Hashitani H. Lipopolysaccharide accelerates peristalsis by stimulating glucagon-like peptide-1 release from L cells in the rat proximal colon. J Physiol. 2024;602:4803–4820. doi: 10.1113/JP286258. [DOI] [PubMed] [Google Scholar]
  155. Ness J, Hoth A, Barnett MJ, Shorr RI, Kaboli PJ. Anticholinergic medications in community-dwelling older veterans: prevalence of anticholinergic symptoms, symptom burden, and adverse drug events. Am J Geriatr Pharmacother. 2006;4:42–51. doi: 10.1016/j.amjopharm.2006.03.008. [DOI] [PubMed] [Google Scholar]
  156. Ni R. Imaging alpha-synuclein pathology in Parkinson’s disease. Neural Regen Res. 2025 doi: 10.4103/NRR.NRR-D-24-01348. doi: 10.4103/NRR.NRR-D-24-01348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  157. Nishida A, Miyamoto J, Shimizu H, Kimura I. Gut microbial short-chain fatty acids-mediated olfactory receptor 78 stimulation promotes anorexigenic gut hormone peptide YY secretion in mice. Biochem Biophys Res Commun. 2021;557:48–54. doi: 10.1016/j.bbrc.2021.03.167. [DOI] [PubMed] [Google Scholar]
  158. Nishiwaki H, Ito M, Hamaguchi T, Maeda T, Kashihara K, Tsuboi Y, Ueyama J, Yoshida T, Hanada H, Takeuchi I, Katsuno M, Hirayama M, Ohno K. Short chain fatty acids-producing and mucin-degrading intestinal bacteria predict the progression of early Parkinson’s disease. NPJ Parkinsons Dis. 2022;8:65. doi: 10.1038/s41531-022-00328-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Nishiwaki H, Ito M, Ishida T, Hamaguchi T, Maeda T, Kashihara K, Tsuboi Y, Ueyama J, Shimamura T, Mori H, Kurokawa K, Katsuno M, Hirayama M, Ohno K. Meta-analysis of gut dysbiosis in Parkinson’s disease. Mov Disord. 2020;35:1626–1635. doi: 10.1002/mds.28119. [DOI] [PubMed] [Google Scholar]
  160. Nishiwaki H, Hamaguchi T, Ito M, Ishida T, Maeda T, Kashihara K, Tsuboi Y, Ueyama J, Shimamura T, Mori H, Kurokawa K, Katsuno M, Hirayama M, Ohno K. Short-chain fatty acid-producing gut microbiota is decreased in Parkinson’s disease but not in rapid-eye-movement sleep behavior disorder. mSystems. 2020;5:e00797–00820. doi: 10.1128/mSystems.00797-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  161. Nissen SK, Shrivastava K, Schulte C, Otzen DE, Goldeck D, Berg D, Moller HJ, Maetzler W, Romero-Ramos M. Alterations in blood monocyte functions in Parkinson’s disease. Mov Disord. 2019;34:1711–1721. doi: 10.1002/mds.27815. [DOI] [PubMed] [Google Scholar]
  162. Nohr MK, Pedersen MH, Gille A, Egerod KL, Engelstoft MS, Husted AS, Sichlau RM, Grunddal KV, Poulsen SS, Han S, Jones RM, Offermanns S, Schwartz TW. GPR41/FFAR3 and GPR43/FFAR2 as cosensors for short-chain fatty acids in enteroendocrine cells vs FFAR3 in enteric neurons and FFAR2 in enteric leukocytes. Endocrinology. 2013;154:3552–3564. doi: 10.1210/en.2013-1142. [DOI] [PubMed] [Google Scholar]
  163. Nøhr MK, Pedersen MH, Gille A, Egerod KL, Engelstoft MS, Husted AS, Sichlau RM, Grunddal KV, Poulsen SS, Han S, Jones RM, Offermanns S, Schwartz TW. GPR41/FFAR3 and GPR43/FFAR2 as cosensors for short-chain fatty acids in enteroendocrine cells vs FFAR3 in enteric neurons and FFAR2 in enteric leukocytes. Endocrinology. 2013;154:3552–3564. doi: 10.1210/en.2013-1142. [DOI] [PubMed] [Google Scholar]
  164. O’Riordan KJ, Collins MK, Moloney GM, Knox EG, Aburto MR, Fulling C, Morley SJ, Clarke G, Schellekens H, Cryan JF. Short chain fatty acids: microbial metabolites for gut-brain axis signalling. Mol Cell Endocrinol. 2022;546:111572. doi: 10.1016/j.mce.2022.111572. [DOI] [PubMed] [Google Scholar]
  165. Ohlsson B, Englund E. Atrophic myenteric and submucosal neurons are observed in Parkinson’s disease. Parkinsons Dis. 2019;2019:7935820. doi: 10.1155/2019/7935820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  166. Oldendorf WH. Carrier-mediated blood-brain barrier transport of short-chain monocarboxylic organic acids. Am J Physiol. 1973;224:1450–1453. doi: 10.1152/ajplegacy.1973.224.6.1450. [DOI] [PubMed] [Google Scholar]
  167. Ostendorf F, Metzdorf J, Gold R, Haghikia A, Tonges L. Propionic acid and fasudil as treatment against rotenone toxicity in an in vitro model of Parkinson’s disease. Molecules. 2020;25:2502. doi: 10.3390/molecules25112502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  168. Ouchi Y, Yoshikawa E, Sekine Y, Futatsubashi M, Kanno T, Ogusu T, Torizuka T. Microglial activation and dopamine terminal loss in early Parkinson’s disease. Ann Neurol. 2005;57:168–175. doi: 10.1002/ana.20338. [DOI] [PubMed] [Google Scholar]
  169. Ozturk O, Celebi G, Duman UG, Kupcuk E, Uyanik M, Sertoglu E. Short-chain fatty acid levels in stools of patients with inflammatory bowel disease are lower than those in healthy subjects. Eur J Gastroenterol Hepatol. 2024;36:890–896. doi: 10.1097/MEG.0000000000002789. [DOI] [PubMed] [Google Scholar]
  170. Pabois J, Durand T, Le Berre C, Gonzales J, Neunlist M, Bourreille A, Naveilhan P, Neveu I. T cells show preferential adhesion to enteric neural cells in culture and are close to neural cells in the myenteric ganglia of Crohn’s patients. J Neuroimmunol. 2020;349:577422. doi: 10.1016/j.jneuroim.2020.577422. [DOI] [PubMed] [Google Scholar]
  171. Pabois J, Durand T, Le Berre C, Filippone RT, Noël T, Durieu E, Bossard C, Bruneau S, Rolli-Derkinderen M, Nurgali K, Neunlist M, Bourreille A, Neveu I, Naveilhan P. Role of ICAM-1 in the adhesion of t cells to enteric glia: perspectives in the formation of plexitis in Crohn’s disease. Cell Mol Gastroenterol Hepatol. 2024;18:133–153. doi: 10.1016/j.jcmgh.2024.02.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  172. Paiva I, Pinho R, Pavlou MA, Hennion M, Wales P, Schutz AL, Rajput A, Szego EM, Kerimoglu C, Gerhardt E, Rego AC, Fischer A, Bonn S, Outeiro TF. Sodium butyrate rescues dopaminergic cells from alpha-synuclein-induced transcriptional deregulation and DNA damage. Hum Mol Genet. 2017;26:2231–2246. doi: 10.1093/hmg/ddx114. [DOI] [PubMed] [Google Scholar]
  173. Parada Venegas D, De la Fuente MK, Landskron G, González MJ, Quera R, Dijkstra G, Harmsen HJM, Faber KN, Hermoso MA. Short chain fatty acids (SCFAs)-mediated gut epithelial and immune regulation and its relevance for inflammatory Bowel diseases. Front Immunol. 2019;10:277. doi: 10.3389/fimmu.2019.00277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  174. Pellegrini C, Ippolito C, Segnani C, Dolfi A, Errede M, Virgintino D, Fornai M, Antonioli L, Garelli F, Nericcio A, Colucci R, Cerri S, Blandini F, Blandizzi C, Bernardini N. Pathological remodelling of colonic wall following dopaminergic nigrostriatal neurodegeneration. Neurobiol Dis. 2020;139:104821. doi: 10.1016/j.nbd.2020.104821. [DOI] [PubMed] [Google Scholar]
  175. Pendse M, De Selle H, Vo N, Quinn G, Dende C, Li Y, Salinas CN, Srinivasan T, Propheter DC, Crofts AA, Koo E, Hassell B, Ruhn KA, Raj P, Obata Y, Hooper LV. Macrophages regulate gastrointestinal motility through complement component 1q. Elife. 2023;12:e78558. doi: 10.7554/eLife.78558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  176. Peng J, Li X, Zheng L, Duan L, Gao Z, Hu D, Li J, Li X, Shen X, Xiao H. Ban-Lan-Gen granule alleviates dextran sulfate sodium-induced chronic relapsing colitis in mice via regulating gut microbiota and restoring gut SCFA Derived-GLP-1 production. J Inflamm Res. 2022;15:1457–1470. doi: 10.2147/JIR.S352863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  177. Perez-Pardo P, Dodiya HB, Engen PA, Forsyth CB, Huschens AM, Shaikh M, Voigt RM, Naqib A, Green SJ, Kordower JH, Shannon KM, Garssen J, Kraneveld AD, Keshavarzian A. Role of TLR4 in the gut-brain axis in Parkinson’s disease: a translational study from men to mice. Gut. 2019;68:829–843. doi: 10.1136/gutjnl-2018-316844. [DOI] [PubMed] [Google Scholar]
  178. Petrov VA, Saltykova IV, Zhukova IA, Alifirova VM, Zhukova NG, Dorofeeva YB, Tyakht AV, Kovarsky BA, Alekseev DG, Kostryukova ES, Mironova YS, Izhboldina OP, Nikitina MA, Perevozchikova TV, Fait EA, Babenko VV, Vakhitova MT, Govorun VM, Sazonov AE. Analysis of gut microbiota in patients with Parkinson’s disease. Bull Exp Biol Med. 2017;162:734–737. doi: 10.1007/s10517-017-3700-7. [DOI] [PubMed] [Google Scholar]
  179. Pietrucci D, Cerroni R, Unida V, Farcomeni A, Pierantozzi M, Mercuri NB, Biocca S, Stefani A, Desideri A. Dysbiosis of gut microbiota in a selected population of Parkinson’s patients. Parkinsonism Relat Disord. 2019;65:124–130. doi: 10.1016/j.parkreldis.2019.06.003. [DOI] [PubMed] [Google Scholar]
  180. Pisani V, Stefani A, Pierantozzi M, Natoli S, Stanzione P, Franciotta D, Pisani A. Increased blood-cerebrospinal fluid transfer of albumin in advanced Parkinson’s disease. J Neuroinflammation. 2012;9:188. doi: 10.1186/1742-2094-9-188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  181. Priyadarshini M, Kotlo KU, Dudeja PK, Layden BT. Role of short chain fatty acid receptors in intestinal physiology and pathophysiology. Compr Physiol. 2018;8:1091–1115. doi: 10.1002/cphy.c170050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  182. Psichas A, Sleeth ML, Murphy KG, Brooks L, Bewick GA, Hanyaloglu AC, Ghatei MA, Bloom SR, Frost G. The short chain fatty acid propionate stimulates GLP-1 and PYY secretion via free fatty acid receptor 2 in rodents. Int J Obes (Lond) 2015;39:424–429. doi: 10.1038/ijo.2014.153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  183. 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. doi: 10.1007/s12035-022-03157-y. [DOI] [PubMed] [Google Scholar]
  184. Qi J, Yang Q, Xia Q, Huang F, Guo H, Cui H, Xie Y, Ren Z, Gou L, Cai D, Kumbhar MA, Fang J, Zuo Z. Low glucose plus β-hydroxybutyrate induces an enhanced inflammatory response in Yak alveolar macrophages via activating the GPR109A/NF-κB signaling pathway. Int J Mol Sci. 2023;24:11331. doi: 10.3390/ijms241411331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  185. Qi Y, Dong Y, Chen J, Xie S, Ma X, Yu X, Yu Y, Wang Y. Lactiplantibacillus plantarum SG5 inhibits neuroinflammation in MPTP-induced PD mice through GLP-1/PGC-1α pathway. Exp Neurol. 2025;383:115001. doi: 10.1016/j.expneurol.2024.115001. [DOI] [PubMed] [Google Scholar]
  186. Qiao CM, Sun MF, Jia XB, Shi Y, Zhang BP, Zhou ZL, Zhao LP, Cui C, Shen YQ. Sodium butyrate causes alpha-synuclein degradation by an Atg5-dependent and PI3K/Akt/mTOR-related autophagy pathway. Exp Cell Res. 2020;387:111772. doi: 10.1016/j.yexcr.2019.111772. [DOI] [PubMed] [Google Scholar]
  187. Qiao CM, Sun MF, Jia XB, Li Y, Zhang BP, Zhao LP, Shi Y, Zhou ZL, Zhu YL, Cui C, Shen YQ. Sodium butyrate exacerbates parkinson’s disease by aggravating neuroinflammation and colonic inflammation in MPTP-induced mice model. Neurochem Res. 2020;45:2128–2142. doi: 10.1007/s11064-020-03074-3. [DOI] [PubMed] [Google Scholar]
  188. Rahman AA, Stavely R, Pan W, Ott L, Ohishi K, Ohkura T, Han C, Hotta R, Goldstein AM. Optogenetic activation of cholinergic enteric neurons reduces inflammation in experimental colitis. Cell Mol Gastroenterol Hepatol. 2024;17:907–921. doi: 10.1016/j.jcmgh.2024.01.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  189. Romano S, Savva GM, Bedarf JR, Charles IG, Hildebrand F, Narbad A. Meta-analysis of the Parkinson’s disease gut microbiome suggests alterations linked to intestinal inflammation. NPJ Parkinsons Dis. 2021;7:27. doi: 10.1038/s41531-021-00156-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  190. Rota L, Pellegrini C, Benvenuti L, Antonioli L, Fornai M, Blandizzi C, Cattaneo A, Colla E. Constipation, deficit in colon contractions and alpha-synuclein inclusions within the colon precede motor abnormalities and neurodegeneration in the central nervous system in a mouse model of alpha-synucleinopathy. Transl Neurodegener. 2019;8:5. doi: 10.1186/s40035-019-0146-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  191. Saikachain N, Sungkaworn T, Muanprasat C, Asavapanumas N. Neuroprotective effect of short-chain fatty acids against oxidative stress-induced SH-SY5Y injury via GPR43-dependent pathway. J Neurochem. 2023;166:201–214. doi: 10.1111/jnc.15827. [DOI] [PubMed] [Google Scholar]
  192. Sampson TR, Debelius JW, Thron T, Janssen S, Shastri GG, Ilhan ZE, Challis C, Schretter CE, Rocha S, Gradinaru V, Chesselet MF, Keshavarzian A, Shannon KM, Krajmalnik-Brown R, Wittung-Stafshede P, Knight R, Mazmanian SK. Gut microbiota regulate motor deficits and neuroinflammation in a model of Parkinson’s disease. Cell. 2016;167:1469–1480.e1412. doi: 10.1016/j.cell.2016.11.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  193. Sanchez-Ruiz M, Iorgu AM, Küster F, Hellmich M, Brunn A, Deckert M. CD8 T cell-derived perforin and TNF-α are crucial mediators of neuronal destruction in experimental autoimmune enteric ganglionitis. Am J Pathol. 2021;191:1064–1076. doi: 10.1016/j.ajpath.2021.02.021. [DOI] [PubMed] [Google Scholar]
  194. Schertzer JD, Lam TKT. Peripheral and central regulation of insulin by the intestine and microbiome. Am J Physiol Endocrinol Metab. 2021;320:e234–239. doi: 10.1152/ajpendo.00547.2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  195. Schulthess J, Pandey S, Capitani M, Rue-Albrecht KC, Arnold I, Franchini F, Chomka A, Ilott NE, Johnston DGW, Pires E, McCullagh J, Sansom SN, Arancibia-Carcamo CV, Uhlig HH, Powrie F. The short chain fatty acid butyrate imprints an antimicrobial program in macrophages. Immunity. 2019;50:432–445.e437. doi: 10.1016/j.immuni.2018.12.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  196. Schwiertz A, Spiegel J, Dillmann U, Grundmann D, Burmann J, Fassbender K, Schafer KH, Unger MM. Fecal markers of intestinal inflammation and intestinal permeability are elevated in Parkinson’s disease. Parkinsonism Relat Disord. 2018;50:104–107. doi: 10.1016/j.parkreldis.2018.02.022. [DOI] [PubMed] [Google Scholar]
  197. Scott NA, et al. Antibiotics induce sustained dysregulation of intestinal T cell immunity by perturbing macrophage homeostasis. Sci Transl Med. 2018;10:eaao4755. doi: 10.1126/scitranslmed.aao4755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  198. Seguella L, Sarnelli G, Esposito G. Leaky gut, dysbiosis, and enteric glia activation: the trilogy behind the intestinal origin of Parkinson’s disease. Neural Regen Res. 2020;15:1037–1038. doi: 10.4103/1673-5374.270308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  199. Sepahi A, Liu Q, Friesen L, Kim CH. Dietary fiber metabolites regulate innate lymphoid cell responses. Mucosal Immunol. 2021;14:317–330. doi: 10.1038/s41385-020-0312-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  200. Sharkey KA, Mawe GM. The enteric nervous system. Physiol Rev. 2023;103:1487–1564. doi: 10.1152/physrev.00018.2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  201. Sharma S, Taliyan R. Targeting histone deacetylases: a novel approach in Parkinson’s disease. Parkinsons Dis. 2015;2015:303294. doi: 10.1155/2015/303294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  202. Sharma S, Taliyan R, Singh S. Beneficial effects of sodium butyrate in 6-OHDA induced neurotoxicity and behavioral abnormalities: modulation of histone deacetylase activity. Behav Brain Res. 2015;291:306–314. doi: 10.1016/j.bbr.2015.05.052. [DOI] [PubMed] [Google Scholar]
  203. Shen L, Wang Z, Huang R, Wu L, Huang Y, Qi Q, Zhong R, Chen Y, Li L, Wu H. Electroacupuncture modulates 5-HT(4R)-mediated cAMP/PKA signaling to improve intestinal motility disorders in a Thy1-αSyn Parkinson’s mouse model. Evid Based Complement Alternat Med. 2022;2022:8659462. doi: 10.1155/2022/8659462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  204. Shi Y, Li S, Zhang H, Zhu J, Che T, Yan B, Li J, Liu C. The effect of macrophage polarization on the expression of the oxytocin signalling system in enteric neurons. J Neuroinflammation. 2021;18:261. doi: 10.1186/s12974-021-02313-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  205. Shih LC, Lin RJ, Chen YL, Fu SC. Unravelling the mechanisms of underweight in Parkinson’s disease by investigating into the role of gut microbiome. NPJ Parkinsons Dis. 2024;10:28. doi: 10.1038/s41531-023-00587-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  206. Shin C, Lim Y, Lim H, Ahn TB. Plasma short-chain fatty acids in patients with Parkinson’s disease. Mov Disord. 2020;35:1021–1027. doi: 10.1002/mds.28016. [DOI] [PubMed] [Google Scholar]
  207. Singaram C, Ashraf W, Gaumnitz EA, Torbey C, Sengupta A, Pfeiffer R, Quigley EM. Dopaminergic defect of enteric nervous system in Parkinson’s disease patients with chronic constipation. Lancet. 1995;346:861–864. doi: 10.1016/s0140-6736(95)92707-7. [DOI] [PubMed] [Google Scholar]
  208. Singh N, Gurav A, Sivaprakasam S, Brady E, Padia R, Shi H, Thangaraju M, Prasad PD, Manicassamy S, Munn DH, Lee JR, Offermanns S, Ganapathy V. Activation of Gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis. Immunity. 2014;40:128–139. doi: 10.1016/j.immuni.2013.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  209. Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly YM, Glickman JN, Garrett WS. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science. 2013;341:569–573. doi: 10.1126/science.1241165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  210. Soret R, Chevalier J, De Coppet P, Poupeau G, Derkinderen P, Segain JP, Neunlist M. Short-chain fatty acids regulate the enteric neurons and control gastrointestinal motility in rats. Gastroenterology. 2010;138:1772–1782. doi: 10.1053/j.gastro.2010.01.053. [DOI] [PubMed] [Google Scholar]
  211. St Laurent R, O’Brien LM, Ahmad ST. Sodium butyrate improves locomotor impairment and early mortality in a rotenone-induced Drosophila model of Parkinson’s disease. Neuroscience. 2013;246:382–390. doi: 10.1016/j.neuroscience.2013.04.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  212. Stakenborg M, et al. Enteric glial cells favor accumulation of anti-inflammatory macrophages during the resolution of muscularis inflammation. Mucosal Immunol. 2022;15:1296–1308. doi: 10.1038/s41385-022-00563-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  213. Stakenborg N, et al. Preoperative administration of the 5-HT4 receptor agonist prucalopride reduces intestinal inflammation and shortens postoperative ileus via cholinergic enteric neurons. Gut. 2019;68:1406–1416. doi: 10.1136/gutjnl-2018-317263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  214. Stamatovic SM, Phillips CM, Martinez-Revollar G, Keep RF, Andjelkovic AV. Involvement of epigenetic mechanisms and non-coding RNAs in blood-brain barrier and neurovascular unit injury and recovery after stroke. Front Neurosci. 2019;13:864. doi: 10.3389/fnins.2019.00864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  215. Su Y, Liu N, Zhang Z, Li H, Ma J, Yuan Y, Shi M, Liu J, Zhao Z, Zhang Z, Holscher C. Cholecystokinin and glucagon-like peptide-1 analogues regulate intestinal tight junction, inflammation, dopaminergic neurons and α-synuclein accumulation in the colon of two Parkinson’s disease mouse models. Eur J Pharmacol. 2022;926:175029. doi: 10.1016/j.ejphar.2022.175029. [DOI] [PubMed] [Google Scholar]
  216. Sun H, Zhao F, Liu Y, Ma T, Jin H, Quan K, Leng B, Zhao J, Yuan X, Li Z, Li F, Kwok LY, Zhang S, Sun Z, Zhang J, Zhang H. Probiotics synergized with conventional regimen in managing Parkinson’s disease. NPJ Parkinsons Dis. 2022;8:62. doi: 10.1038/s41531-022-00327-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  217. Sun J, Wang F, Hu X, Yang C, Xu H, Yao Y, Liu J. Clostridium butyricum attenuates chronic unpredictable mild stress-induced depressive-like behavior in mice via the gut-brain axis. Journal of agricultural and food chemistry. 2018;66:8415–8421. doi: 10.1021/acs.jafc.8b02462. [DOI] [PubMed] [Google Scholar]
  218. Sun J, Li H, Jin Y, Yu J, Mao S, Su KP, Ling Z, Liu J. Probiotic Clostridium butyricum ameliorated motor deficits in a mouse model of Parkinson’s disease via gut microbiota-GLP-1 pathway. Brain Behav Immun. 2021;91:703–715. doi: 10.1016/j.bbi.2020.10.014. [DOI] [PubMed] [Google Scholar]
  219. Svensson E, Horvath-Puho E, Thomsen RW, Djurhuus JC, Pedersen L, Borghammer P, Sorensen HT. Vagotomy and subsequent risk of Parkinson’s disease. Ann Neurol. 2015;78:522–529. doi: 10.1002/ana.24448. [DOI] [PubMed] [Google Scholar]
  220. Talbot J, Hahn P, Kroehling L, Nguyen H, Li D, Littman DR. Feeding-dependent VIP neuron-ILC3 circuit regulates the intestinal barrier. Nature. 2020;579:575–580. doi: 10.1038/s41586-020-2039-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  221. Tan AH, Chong CW, Lim SY, Yap IKS, Teh CSJ, Loke MF, Song SL, Tan JY, Ang BH, Tan YQ, Kho MT, Bowman J, Mahadeva S, Yong HS, Lang AE. Gut microbial ecosystem in parkinson disease: new clinicobiological insights from multi-omics. Ann Neurol. 2021;89:546–559. doi: 10.1002/ana.25982. [DOI] [PubMed] [Google Scholar]
  222. Tanei ZI, Saito Y, Ito S, Matsubara T, Motoda A, Yamazaki M, Sakashita Y, Kawakami I, Ikemura M, Tanaka S, Sengoku R, Arai T, Murayama S. Lewy pathology of the esophagus correlates with the progression of Lewy body disease: a Japanese cohort study of autopsy cases. Acta Neuropathol. 2021;141:25–37. doi: 10.1007/s00401-020-02233-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  223. Tao E, Zhu Z, Hu C, Long G, Chen B, Guo R, Fang M, Jiang M. Potential roles of enterochromaffin cells in early life stress-induced irritable Bowel syndrome. Front Cell Neurosci. 2022;16:837166. doi: 10.3389/fncel.2022.837166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  224. Thomasi BBM, Valdetaro L, Ricciardi MCG, Hayashide L, Fernandes A, Mussauer A, da Silva ML, da Cunha Faria-Melibeu A, Ribeiro MGL, de Mattos Coelho-Aguiar J, Campello-Costa P, Moura-Neto V, Tavares-Gomes AL. Enteric glial cell reactivity in colonic layers and mucosal modulation in a mouse model of Parkinson’s disease induced by 6-hydroxydopamine. Brain Res Bull. 2022;187:111–121. doi: 10.1016/j.brainresbull.2022.06.013. [DOI] [PubMed] [Google Scholar]
  225. Tian YM, Chen X, Luo DZ, Zhang XH, Xue H, Zheng LF, Yang N, Wang XM, Zhu JX. Alteration of dopaminergic markers in gastrointestinal tract of different rodent models of Parkinson’s disease. Neuroscience. 2008;153:634–644. doi: 10.1016/j.neuroscience.2008.02.033. [DOI] [PubMed] [Google Scholar]
  226. Toczylowska B, Zieminska E, Michalowska M, Chalimoniuk M, Fiszer U. Changes in the metabolic profiles of the serum and putamen in Parkinson’s disease patients - In vitro and in vivo NMR spectroscopy studies. Brain Res. 2020;1748:147118. doi: 10.1016/j.brainres.2020.147118. [DOI] [PubMed] [Google Scholar]
  227. Toledano-Pinedo M, et al. Contilisant+tubastatin a hybrids: polyfunctionalized indole derivatives as new HDAC inhibitor-based multitarget small molecules with in vitro and in vivo activity in neurodegenerative diseases. J Med Chem. 2024;67:16533–16555. doi: 10.1021/acs.jmedchem.4c01367. [DOI] [PubMed] [Google Scholar]
  228. Tolhurst G, Heffron H, Lam YS, Parker HE, Habib AM, Diakogiannaki E, Cameron J, Grosse J, Reimann F, Gribble FM. Short-chain fatty acids stimulate glucagon-like peptide-1 secretion via the G-protein-coupled receptor FFAR2. Diabetes. 2012;61:364–371. doi: 10.2337/db11-1019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  229. Uhernik AL, Tucker C, Smith JP. Control of MCT1 function in cerebrovascular endothelial cells by intracellular pH. Brain Res. 2011;1376:10–22. doi: 10.1016/j.brainres.2010.12.060. [DOI] [PubMed] [Google Scholar]
  230. Unger MM, Spiegel J, Dillmann KU, Grundmann D, Philippeit H, Burmann J, Fassbender K, Schwiertz A, Schafer KH. Short chain fatty acids and gut microbiota differ between patients with Parkinson’s disease and age-matched controls. Parkinsonism Relat Disord. 2016;32:66–72. doi: 10.1016/j.parkreldis.2016.08.019. [DOI] [PubMed] [Google Scholar]
  231. van der Hee B, Wells JM. Microbial regulation of host physiology by short-chain fatty acids. Trends Microbiol. 2021;29:700–712. doi: 10.1016/j.tim.2021.02.001. [DOI] [PubMed] [Google Scholar]
  232. Vascellari S, Palmas V, Melis M, Pisanu S, Cusano R, Uva P, Perra D, Madau V, Sarchioto M, Oppo V, Simola N, Morelli M, Santoru ML, Atzori L, Melis M, Cossu G, Manzin A. Gut microbiota and metabolome alterations associated with Parkinson’s disease. mSystems. 2020;5:e00561. doi: 10.1128/mSystems.00561-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  233. Vendrik KE, Chernova VO, Kuijper EJ, Terveer EM, van Hilten JJ, Contarino MF, group FPs Safety and feasibility of faecal microbiota transplantation for patients with Parkinson’s disease: a protocol for a self-controlled interventional donor-FMT pilot study. BMJ Open. 2023;13:e071766. doi: 10.1136/bmjopen-2023-071766. [DOI] [PMC free article] [PubMed] [Google Scholar]
  234. Vicentini FA, Keenan CM, Wallace LE, Woods C, Cavin JB, Flockton AR, Macklin WB, Belkind-Gerson J, Hirota SA, Sharkey KA. Intestinal microbiota shapes gut physiology and regulates enteric neurons and glia. Microbiome. 2021;9:210. doi: 10.1186/s40168-021-01165-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  235. Viola MF, et al. Dedicated macrophages organize and maintain the enteric nervous system. Nature. 2023;618:818–826. doi: 10.1038/s41586-023-06200-7. [DOI] [PubMed] [Google Scholar]
  236. Voss U, Sand E, Hellström PM, Ekblad E. Glucagon-like peptides 1 and 2 and vasoactive intestinal peptide are neuroprotective on cultured and mast cell co-cultured rat myenteric neurons. BMC Gastroenterol. 2012;12:30. doi: 10.1186/1471-230X-12-30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  237. Wallen ZD, Appah M, Dean MN, Sesler CL, Factor SA, Molho E, Zabetian CP, Standaert DG, Payami H. Characterizing dysbiosis of gut microbiome in PD: evidence for overabundance of opportunistic pathogens. NPJ Parkinsons Dis. 2020;6:11. doi: 10.1038/s41531-020-0112-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  238. Wang L, Fleming SM, Chesselet MF, Tache Y. Abnormal colonic motility in mice overexpressing human wild-type alpha-synuclein. Neuroreport. 2008;19:873–876. doi: 10.1097/WNR.0b013e3282ffda5e. [DOI] [PMC free article] [PubMed] [Google Scholar]
  239. Wang L, Gourcerol G, Yuan PQ, Wu SV, Million M, Larauche M, Taché Y. Peripheral peptide YY inhibits propulsive colonic motor function through Y2 receptor in conscious mice. Am J Physiol Gastrointest Liver Physiol. 2010;298:G45–56. doi: 10.1152/ajpgi.00349.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  240. Wang L, Lv WQ, Yang JT, Lin X, Liu HM, Tan HJ, Quan RP, Long PP, Shen H, Shen J, Deng HW, Xiao HM. Enteric nervous system damage caused by abnormal intestinal butyrate metabolism may lead to functional constipation. Front Microbiol. 2023;14:1117905. doi: 10.3389/fmicb.2023.1117905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  241. Wang Y, Zhang J, Xu L, Ma J, Lu M, Ma J, Liu Z, Wang F, Tang X. Modified gegen qinlian decoction regulates Treg/Th17 balance to ameliorate DSS-induced acute experimental colitis in mice by altering the gut microbiota. Front Pharmacol. 2021;12:756978. doi: 10.3389/fphar.2021.756978. [DOI] [PMC free article] [PubMed] [Google Scholar]
  242. Wang Y, Chen R, Shi G, Huang X, Li K, Wang R, Cao X, Yang Z, Zhao N, Yan J. Chitosan alleviates symptoms of Parkinson’s disease by reducing acetate levels, which decreases inflammation and promotes repair of the intestinal barrier and blood-brain barrier. Neural Regen Res. 2024 doi: 10.4103/NRR.NRR-D-23-01511. doi: 10.4103/NRR.NRR-D-23-01511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  243. Wang Z, Cheng S. Effects of pramipexole combined with nerve growth factor on cognitive impairment and urinary AD7c-NTP expression in patients with Parkinson’s disease. Comput Math Methods Med. 2022;2022:3398732. doi: 10.1155/2022/3398732. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  244. Wang Z, Leng Y, Tsai LK, Leeds P, Chuang DM. Valproic acid attenuates blood-brain barrier disruption in a rat model of transient focal cerebral ischemia: the roles of HDAC and MMP-9 inhibition. J Cereb Blood Flow Metab. 2011;31:52–57. doi: 10.1038/jcbfm.2010.195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  245. Wang ZL, Yuan L, Li W, Li JY. Ferroptosis in Parkinson’s disease: glia-neuron crosstalk. Trends Mol Med. 2022;28:258–269. doi: 10.1016/j.molmed.2022.02.003. [DOI] [PubMed] [Google Scholar]
  246. Wenzel TJ, Gates EJ, Ranger AL, Klegeris A. Short-chain fatty acids (SCFAs) alone or in combination regulate select immune functions of microglia-like cells. Mol Cell Neurosci. 2020;105:103493. doi: 10.1016/j.mcn.2020.103493. [DOI] [PubMed] [Google Scholar]
  247. Wu G, Jiang Z, Pu Y, Chen S, Wang T, Wang Y, Xu X, Wang S, Jin M, Yao Y, Liu Y, Ke S, Liu S. Serum short-chain fatty acids and its correlation with motor and non-motor symptoms in Parkinson’s disease patients. BMC Neurol. 2022;22:13. doi: 10.1186/s12883-021-02544-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  248. Wu YL, Zhang CH, Teng Y, Pan Y, Liu NC, Liu PX, Zhu X, Su XL, Lin J. Propionate and butyrate attenuate macrophage pyroptosis and osteoclastogenesis induced by CoCrMo alloy particles. Mil Med Res. 2022;9:46. doi: 10.1186/s40779-022-00404-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  249. Xu H, et al. Transcriptional atlas of intestinal immune cells reveals that neuropeptide α-CGRP modulates group 2 innate lymphoid cell responses. Immunity. 2019;51:696–708.e699. doi: 10.1016/j.immuni.2019.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  250. Xu J, Moore BN, Pluznick JL. Short-chain fatty acid receptors and blood pressure regulation: council on hypertension mid-career award for research excellence 2021. Hypertension. 2022;79:2127–2137. doi: 10.1161/HYPERTENSIONAHA.122.18558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  251. Xu RC, Miao WT, Xu JY, Xu WX, Liu MR, Ding ST, Jian YX, Lei YH, Yan N, Liu HD. Neuroprotective effects of sodium butyrate and monomethyl fumarate treatment through GPR109A modulation and intestinal barrier restoration on PD mice. Nutrients. 2022;14:4163. doi: 10.3390/nu14194163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  252. 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. doi: 10.1002/iid3.1194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  253. Yan Q, Jia S, Li D, Yang J. The role and mechanism of action of microbiota-derived short-chain fatty acids in neutrophils: from the activation to becoming potential biomarkers. Biomed Pharmacother. 2023;169:115821. doi: 10.1016/j.biopha.2023.115821. [DOI] [PubMed] [Google Scholar]
  254. Yan Y, Ramanan D, Rozenberg M, McGovern K, Rastelli D, Vijaykumar B, Yaghi O, Voisin T, Mosaheb M, Chiu I, Itzkovitz S, Rao M, Mathis D, Benoist C. Interleukin-6 produced by enteric neurons regulates the number and phenotype of microbe-responsive regulatory T cells in the gut. Immunity. 2021;54:499–513.e495. doi: 10.1016/j.immuni.2021.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  255. Yang LL, Millischer V, Rodin S, MacFabe DF, Villaescusa JC, Lavebratt C. Enteric short-chain fatty acids promote proliferation of human neural progenitor cells. J Neurochem. 2020;154:635–646. doi: 10.1111/jnc.14928. [DOI] [PubMed] [Google Scholar]
  256. Yang W, Yu T, Huang X, Bilotta AJ, Xu L, Lu Y, Sun J, Pan F, Zhou J, Zhang W, Yao S, Maynard CL, Singh N, Dann SM, Liu Z, Cong Y. Intestinal microbiota-derived short-chain fatty acids regulation of immune cell IL-22 production and gut immunity. Nat Commun. 2020;11:4457. doi: 10.1038/s41467-020-18262-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  257. Yang X, Ai P, He X, Mo C, Zhang Y, Xu S, Lai Y, Qian Y, Xiao Q. Parkinson’s disease is associated with impaired gut-blood barrier for short-chain fatty acids. Mov Disord. 2022;37:1634–1643. doi: 10.1002/mds.29063. [DOI] [PubMed] [Google Scholar]
  258. Yang X, He X, Xu S, Zhang Y, Mo C, Lai Y, Song Y, Yan Z, Ai P, Qian Y, Xiao Q. Effect of Lacticaseibacillus paracasei strain Shirota supplementation on clinical responses and gut microbiome in Parkinson’s disease. Food Funct. 2023;14:6828–6839. doi: 10.1039/d3fo00728f. [DOI] [PubMed] [Google Scholar]
  259. Ye L, Bae M, Cassilly CD, Jabba SV, Thorpe DW, Martin AM, Lu HY, Wang J, Thompson JD, Lickwar CR, Poss KD, Keating DJ, Jordt SE, Clardy J, Liddle RA, Rawls JF. Enteroendocrine cells sense bacterial tryptophan catabolites to activate enteric and vagal neuronal pathways. Cell Host Microbe. 2021;29:179–196.e179. doi: 10.1016/j.chom.2020.11.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  260. Yu F, Wang Z, Tanaka M, Chiu CT, Leeds P, Zhang Y, Chuang DM. Posttrauma cotreatment with lithium and valproate: reduction of lesion volume, attenuation of blood-brain barrier disruption, and improvement in motor coordination in mice with traumatic brain injury. J Neurosurg. 2013;119:766–773. doi: 10.3171/2013.6.JNS13135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  261. Zaiatz-Bittencourt V, Jones F, Tosetto M, Scaife C, Cagney G, Jones E, Doherty GA, Ryan EJ. Butyrate limits human natural killer cell effector function. Sci Rep. 2023;13:2715. doi: 10.1038/s41598-023-29731-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  262. Zhai L, et al. Ruminococcus gnavus plays a pathogenic role in diarrhea-predominant irritable bowel syndrome by increasing serotonin biosynthesis. Cell Host Microbe. 2023;31:33–44.e35. doi: 10.1016/j.chom.2022.11.006. [DOI] [PubMed] [Google Scholar]
  263. Zhan K, Gong X, Chen Y, Jiang M, Yang T, Zhao G. Short-chain fatty acids regulate the immune responses via G protein-coupled receptor 41 in bovine rumen epithelial cells. Front Immunol. 2019;10:2042. doi: 10.3389/fimmu.2019.02042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  264. Zhang LY, Zhang SY, Wen R, Zhang TN, Yang N. Role of histone deacetylases and their inhibitors in neurological diseases. Pharmacol Res. 2024;208:107410. doi: 10.1016/j.phrs.2024.107410. [DOI] [PubMed] [Google Scholar]
  265. Zhang X, Tang B, Guo J. Parkinson’s disease and gut microbiota: from clinical to mechanistic and therapeutic studies. Transl Neurodegener. 2023;12:59. doi: 10.1186/s40035-023-00392-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  266. Zhang X, Li Y, Liu C, Fan R, Wang P, Zheng L, Hong F, Feng X, Zhang Y, Li L, Zhu J. Alteration of enteric monoamines with monoamine receptors and colonic dysmotility in 6-hydroxydopamine-induced Parkinson’s disease rats. Transl Res. 2015;166:152–162. doi: 10.1016/j.trsl.2015.02.003. [DOI] [PubMed] [Google Scholar]
  267. Zhang Y, Xu S, Qian Y, Mo C, Ai P, Yang X, Xiao Q. Sodium butyrate ameliorates gut dysfunction and motor deficits in a mouse model of Parkinson’s disease by regulating gut microbiota. Front Aging Neurosci. 2023;15:1099018. doi: 10.3389/fnagi.2023.1099018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  268. Zhao H, Zhou Y, Xu J, Zhang Y, Wang H, Zhao C, Huang H, Yang J, Huang C, Li Y, Wang L, Nie Y. Short-chain fatty acid-producing bacterial strains attenuate experimental ulcerative colitis by promoting M2 macrophage polarization via JAK/STAT3/FOXO3 axis inactivation. J Transl Med. 2024;22:369. doi: 10.1186/s12967-024-05122-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  269. Zhu P, Lu T, Chen Z, Liu B, Fan D, Li C, Wu J, He L, Zhu X, Du Y, Tian Y, Fan Z. 5-hydroxytryptamine produced by enteric serotonergic neurons initiates colorectal cancer stem cell self-renewal and tumorigenesis. Neuron. 2022;110:2268–2282.e2264. doi: 10.1016/j.neuron.2022.04.024. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Not applicable.


Articles from Neural Regeneration Research are provided here courtesy of Wolters Kluwer -- Medknow Publications

RESOURCES