Your new experience awaits. Try the new design now and help us make it even better

REVIEW article

Front. Aging Neurosci., 09 October 2025

Sec. Parkinson’s Disease and Aging-related Movement Disorders

Volume 17 - 2025 | https://doi.org/10.3389/fnagi.2025.1600782

Parkinson’s disease beyond the brain: implications for treatments

Setareh Malekian Naeini&#x;Setareh Malekian Naeini1Marc Danzell Lopez&#x;Marc Danzell Lopez2Paul E. Fraser,Paul E. Fraser2,3Anurag Tandon,
Anurag Tandon2,4*
  • 1Department of Physiology, University of Toronto, Toronto, ON, Canada
  • 2Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
  • 3Department of Medicine and Medical Biophysics, Toronto, ON, Canada
  • 4Department of Medicine, University of Toronto, Toronto, ON, Canada

The presence of α-synuclein pathology in peripheral nervous system neurons is linked to early prodromal, non-motor symptoms in a segment of Parkinson’s disease (PD) patients referred to as “body-first.” These features of the disease suggest a convergence of environmental, genetic, immune and age-related factors within the enteric nervous system as initiating triggers of disease. This review explores the changes in the gut microbiome and intestinal permeability that may drive systemic inflammation and precede neurodegeneration in PD. The pathways leading to the formation of α-synuclein aggregates are explored as well as their role in transneuronal propagation and the spreading of pathology within the brain. Lastly, advances in systemic gene therapy that could be used to target multiple PD-affected organs following systemic injection are highlighted. By integrating insights from molecular biology and clinical research, it may be possible to shed light on the multifactorial etiology of PD and the interconnectedness of the gut and brain, which could lead to novel diagnostic and therapeutic strategies.

1 Introduction

Parkinson’s disease (PD) is a neurodegenerative disorder that is estimated to affect 12.0 million people by 2040 (Dorsey et al., 2018). The disease exerts a substantial economic burden; in the U. S. alone, the direct and indirect costs of PD summed up to $51.9 billion in 2017 (Yang et al., 2020). Therefore, understanding the underlying mechanisms of disease is essential in providing care to patients and reducing the economic burden on healthcare systems.

PD manifests clinically with motor disturbances including resting tremor, muscle rigidity, bradykinesia, and gait instability (Halliday et al., 2011). Some patients also experience non-motor symptoms such as rapid eye movement (REM) sleep behavior disorders (RBD), depression, and constipation prior to the appearance of motor deficits that elicit a PD diagnosis (Postuma and Berg, 2016). These prodromal symptoms implicate the involvement of multiple brain regions and the peripheral nervous system prior to substantia nigra degeneration. The triggers for PD remain unknown, though a combination of aging, genetics, and environmental factors are involved (Pang et al., 2019). These factors contribute to the toxic aggregation of proteins, most commonly the synaptic protein α-Synuclein (α-Syn), that disrupts neuronal homeostasis and leads to apoptosis. α-Syn aggregates found within the soma or along axodendritic projections are termed Lewy bodies (LB) and Lewy neurites (LN), respectively, and are associated with neuronal dysfunction. Thus, the principal pathological change in PD is the α-Syn aggregate-driven impairments and degeneration leading to the loss of dopaminergic neurons within the substantia nigra, a midbrain region involved in regulating movement via projections to the striatum (Halliday et al., 2011; Sonne et al., 2022).

The Braak gut-brain axis hypothesis of PD could explain the involvement of the peripheries, particularly the enteric nervous system (ENS), in the initiation and progression of disease (Beach et al., 2021; Braak et al., 2003). Entry of foreign pathogens through nasal or gut pathways could initiate or exacerbate pathology in enteric neuron and its passage via the vagus nerve to the brainstem (Braak et al., 2003; Braak et al., 2003). The presence of gastrointestinal (GI) symptoms such as constipation, dysphagia, gastroparesis, and excessive strain during defecation before the appearance of motor symptoms indicates early GI dysfunction and ENS involvement in PD (Sung et al., 2014; Gan et al., 2018; Yu et al., 2018). Lewy bodies (LBs) have been detected in the submucosal and myenteric layers of the GI system of PD patients mostly in the vasoactive intestinal peptide (VIP)-positive cholinergic neurons and minimally in tyrosine hydroxylase (TH)-positive neurons (Wakabayashi et al., 1988; Wakabayashi et al., 1989). Notably, one-third of age-matched non-parkinsonian patients also displayed enteric LBs and were suggested to have been in the premotor phase of PD (Wakabayashi et al., 1988; Wakabayashi et al., 1990). Enteric LBs have also been observed in the esophagus (Braak et al., 2006), as well as in nerve fibers of gastric, duodenal, and colonic biopsies of pre-clinical stage patients (Hilton et al., 2014).

It is important to note this staging for PD progression does not apply to all PD cases (Braak et al., 2003; Parkkinen et al., 2008). Conversely, enteric LBs are not detected in all PD patients nor do they serve as a sensitive or specific biomarker for PD (Chung et al., 2016; Ganguly et al., 2021; Visanji et al., 2015). A study examining 111 PD patients found minimal enteric LBs in only 5 patients. Since enteric LBs were absent in those without brain pathology, the researchers suggested that Parkinson’s disease may initiate solely in the brain (Beach et al., 2021). Such discrepancies between findings may be better accommodated by the hypothesis that postulates the existence of at least two subtypes of PD based on the origin of α-Syn pathology: body-first and brain-first. Patients of the body-first model are predicted to experience GI symptoms, cardiac denervation, and RBD prior to motor symptom onset while the opposite order is experienced by those of the brain-first phenotype (Borghammer and Van Den Berge, 2019; Borghammer, 2021). Although the numbers vary between studies, approximately 50% of premotor PD patients report constipation as compared to ~10% in age-matched controls with an even greater overrepresentation in the PD groups when broader GI symptoms were included (Cersosimo et al., 2013). Nevertheless, the periphery is still affected in both subtypes, whether in premotor or in the late stages of PD. This review aims to summarize how the disease may progress from the periphery to the brain, highlighting contributing factors such as gut dysbiosis and inflammation, and suggests the use of systemic AAV-mediated gene delivery as a potential therapeutic strategy that may concurrently treat the peripheral and central pathologies.

2 Gut dysbiosis

For patients of the body-first phenotype, disturbances to the gastrointestinal (GI) system are commonly observed in the premotor phase of PD. Constipation and inflammation are the most common gut symptoms affecting 50–60% of cases and about 25% of all PD cases develop these before motor symptoms (Gan et al., 2018; Yu et al., 2018). GI diseases involving inflammation like inflammatory bowel disease and Crohn’s disease also increase the risk of developing PD (Yu et al., 2018; Chen et al., 2019; Zhu et al., 2022; Fasano et al., 2015). A key player that may drive or counteract these symptoms is the gut microbiome.

The gut microbiome is composed of diverse microorganisms involved in metabolism, immunity, and regulation of the intestinal barrier (Tan et al., 2022). Microbial imbalance, termed dysbiosis, results in an alteration of microbial families present and the metabolites they produce, which can affect gastrointestinal and neurological processes (Tan et al., 2022). Thanks to technological advances such as next-generation sequencing, clinical studies have been able to identify the differences between the gut microbiota of PD and healthy patients using stool samples (Barichella et al., 2019; Cirstea et al., 2020; Tan et al., 2021). These studies have revealed deleterious taxonomic and metabolic shifts in PD patients that could be associated with constipation (Cirstea et al., 2020; Tan et al., 2021) and cognitive deficits. The importance of having a healthy microbiome has been highlighted by “gut reset” experiments wherein a healthy microbiome is introduced to subjects, commonly through a stool transplant (also known as fecal microbiota transplant). For example, in transgenic mice overexpressing α-Syn, transplantation of fecal microbiota from PD patients leads to worsened motor dysfunction and increased brain pathology compared to those given healthy fecal transplants, indicating the involvement of the microbiota in neurodegeneration (Sampson et al., 2016). Another study that employed a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) induced mouse model of PD found that stool transplants from wild-type mice alleviated motor symptoms, improved gut dysbiosis, and increased the number of striatal dopaminergic neurons (Sun et al., 2018). Lastly, two case studies that tested the gut reset in PD patients also found beneficial effects on motor symptoms, bowel movements, mood, and sleep (Huang et al., 2019; Xue et al., 2020).

Although decoding the changes in the microbiome in PD is complicated, there is an overall increase in gram-negative bacteria that produce the endotoxin lipopolysaccharide (LPS). LPS damages the intestinal barrier, rendering it more permeable (more commonly known as a “leaky gut”) (Tan et al., 2022). Injection of LPS to mice has been shown to increase intestinal permeability and elevate the expression and pathological phosphorylation of α-Syn in colonic neurons, which are also observed in PD patients (Kelly et al., 2014). In another study, oral administration of LPS to mice over-expressing α-Syn led to the emergence of motor impairments (Gorecki et al., 2019). Gram-negative bacteria could also lead to LPS-mediated damage, as demonstrated by the administration of a gram-negative bacterium, Proteus mirabilis, which induced motor deficits, neuroinflammation, dopaminergic neuron loss, and α-Syn aggregation in the intestine and brains of mice (Choi et al., 2018). These pathological changes were attributed to LPS-mediated damage to the gut barrier and inflammation. Indeed, LPS mouse models of PD have emerged with a variety of administration routes, including nasal, oral, systemic injection, and localized injection in the substantia nigra and the striatum (Deng and Bobrovskaya, 2022). Impaired gut permeability, as demonstrated by mucosal α-Syn expression and serum LPS binding protein detected in PD patients, is indicative of the translocation of microbial byproducts (Bhattacharyya et al., 2019; Forsyth et al., 2011; Kim et al., 2016).

LPS could drive pathology through multiple mechanisms. Indeed, increased plasma LPS binding protein, a promoter of innate immunity and biomarker of intestinal permeability, has been associated with an increased risk for PD (Zhao et al., 2023). Endotoxins like LPS cause pathology through interactions with intestinal receptors, particularly toll-like receptor 4 (TLR4). TLRs are pattern-recognition receptors activated by endogenous damage or pathogens and triggering inflammatory responses implicated, for example, in inflammatory bowel disease (Tam et al., 2021). LPS interactions with intestinal TLR4s have been shown to mediate defects in epithelial tight junctions through TLR4’s downstream mechanisms, promoting gut leakiness and inflammation (Guo et al., 2015). Further, activation of TLR4 via LPS initiates a cascade of mechanisms that leads to pro-inflammatory gene expression and cytokine release, which further recruit leukocytes and enhance gut inflammation (Morris et al., 2015). This is supported by colonic samples from PD patients which revealed increased pro-inflammatory gene markers and cytokine expression (Perez-Pardo et al., 2019). In addition, TLR4 knock-out mice treated with a PD-inducing pesticide exhibited reduced intestinal inflammation, motor dysfunction, neuroinflammation, and neurodegeneration compared to wild-type mice, highlighting the importance of TLR4-mediated mechanisms in PD (Perez-Pardo et al., 2019). Studies have also shown that LPS can modulate α-Syn aggregation and initiate synucleinopathy in vivo (Kim et al., 2016).

Microbial alterations could also affect the metabolic byproducts produced by the microbiota, such as short-chain-fatty-acid (SCFA) levels. SCFAs, such as propionic acid and butyric acid, are produced when anaerobic bacteria digest fiber (Metzdorf and Tönges, 2021). SCFAs protect the intestinal barrier and reduce inflammation (Chen et al., 2018; Zhang et al., 2023). Some reports have shown an increase in some SCFA-producing genera and a simultaneous decrease in related microbiota in the fecal samples of PD patients as compared to healthy controls (Li et al., 2023). While the changes in bacterial strains may be due to PD drug interactions or natural ageing processes, these findings point to the complicated interactions of the microbiota and their role in human health. In addition, multiple studies have reported reduced SCFA levels in the stool samples of PD patients, while some studies show increased serum levels (Wu et al., 2022; Aho et al., 2021; Baert et al., 2021; Unger et al., 2016). Despite conflicting reports on SCFA levels, numerous in vivo studies have underscored the beneficial properties of SCFA in PD. Administration of sodium butyrate to a neurotoxin-induced mouse model of PD alleviated motor disability, inhibited neuroinflammation, and increased tyrosine hydroxylase levels in the substantia nigra (Hou et al., 2021). Similarly, intragastric administration of sodium butyrate into rotenone-treated mice protected against gut dysfunction and motor deficits (Zhang et al., 2022). Another study found that injection of Blautia producta, a butyrate-producing bacterium shown to be reduced in PD patients, improved motor deficits and attenuated dopaminergic neuron loss in a PD mouse model (Liu et al., 2024). Conversely, feeding SCFAs to germ-free mice over-expressing α-Syn exacerbated inflammation and motor dysfunction (Sampson et al., 2016). These contradictory findings may be attributed to the heterogeneity of PD and SCFAs, which require further elucidation via standardization of experimental protocols regarding SCFA type, SCFA dosage, and animal models used (Zhang et al., 2023).

The composition of gut microbiota can change through the process of ageing. Some of the effects of ageing on the gut include increased gut permeability, loss of microbial diversity, and a shift toward more harmful and inflammatory microbes (Cryan et al., 2019). For instance, housing young germ-free mice with older mice resulted in an increase in intestinal permeability and inflammatory markers in the gut of the germ-free mice due to microbial colonization with the microbiota from older mice (Thevaranjan et al., 2017). Germ-free mice were also shown to be resistant to neurotoxin-induced PD, however, they demonstrated motor dysfunction, gut permeability and intestinal inflammation after colonization by fecal bacterial content from older mice. These findings indicate a potential involvement of age-related gut microbiota dysbiosis in PD (Lima et al., 2023).

Beyond microbiota composition, certain bacterial proteins, such as the amyloid curli, can also be pathogenic. Curli fibers, composed of amyloid CsgA monomers, are expressed by commensal E. coli and are found in about 40% of human fecal isolates in the United States (Ravva et al., 2016; Miller et al., 2021). Curli is crucial for biofilm formation, which may occur after ingesting pathogenic microbial strains like Salmonella typhi and curli-positive E. coli. Notably, CsgA has a similar amyloidogenic structure to α-Syn fibrils and other related pathogens, enabling it to cross-seed the misfolding of α-Syn aggregates (Cherny et al., 2005; Evans et al., 2015; Lundmark et al., 2005). Indeed, aged Fischer 344 rats, Caenorhabditis elegans, and transgenics overexpressing α-Syn exhibited increased aggregation when fed a diet containing curli-positive bacteria (Sampson et al., 2016; Chen et al., 2016).

Overall, gut dysbiosis with increased LPS-producing bacteria contributes to gut permeability, allowing the translocation of microbial byproducts and gut contents into the underlying enteric neurons and into the systemic blood flow. This may promote the aggregation and spread of α-Syn as well as inflammation in the body-first PD cases.

3 Inflammation and PD

Gut inflammation has been demonstrated in PD patients with a pro-inflammatory microbial makeup and elevated stool levels of inflammatory markers, such as calprotectin, a marker of gut immune system activity (Klingberg et al., 2019; Mulak et al., 2019) and cytokines that include interleukin-1α, interleukin-1β, and C-reactive protein (Houser et al., 2018). Biopsies from the ascending colon of PD patients also reveal elevated mRNA expression levels of pro-inflammatory cytokines as well as the enteric glial cell markers, glial fibrillary acidic protein and Sox-10, consistent with enteric inflammation (Devos et al., 2013). This is in line with the finding that individuals with inflammatory bowel diseases have a higher risk of future PD development (Zhu et al., 2022; Li et al., 2023). In addition, anti-tumor necrosis factor therapy, a therapy for inflammatory bowel disease has been associated with a reduced risk of PD development through limiting peripheral inflammation (Peter et al., 2018). Therefore, an inflammatory gut environment is one of the homeostatic disturbances implicated in PD. As mentioned above, inflammation leads to increased gut permeability which could allow translocation of pro-inflammatory molecules, bacteria, bacterial metabolites, and amyloidogenic proteins to the underlying mucosal tissue where ENS neurons are located and subsequently into the circulatory system.

Systemic endotoxin translocation has also been reported in PD patients with lower levels of plasma LPS binding protein, an indicator of gram-negative bacteria exposure (Forsyth et al., 2011; Perez-Pardo et al., 2019). LPS can induce a systemic inflammatory response through the activation of monocytes and macrophages that produce pro-inflammatory cytokines (Kim et al., 2022). Indeed, studies on early PD patients have revealed the elevation of certain cytokines in the blood, like interleukin-2, interleukin-6, and interleukin-1ß in the early stages of the disease (Kim et al., 2018; Umemura et al., 2014). In fact, interleukin-2 and 6 were associated with non-motor progression, particularly mood/apathy-related symptoms (Kim et al., 2022). Systemic inflammation, whether through the leakage of deleterious gut contents or induced by gut inflammation, could in turn cause and exacerbate central neuroinflammation. For instance, infectious diseases like pneumonia are known to worsen the outcomes of PD patients and accelerate motor deterioration, due to systemic inflammation exacerbating neurodegeneration (Umemura et al., 2014; Brugger et al., 2015). In line with this, suppressing inflammation with nonsteroidal anti-inflammatory drugs has been shown to reduce the risk of future PD development (Chen et al., 2005; Chen et al., 2003).

Communication between immune pathways and the central nervous system (CNS) can happen in several ways (Figure 1). Firstly, inflammatory molecules must penetrate the protective blood–brain-barrier (BBB) – the selectively permeable membrane of the cerebral blood vessels – to enter the brain. While some cytokines like tumor necrosis factor-alpha (TNFɑ) and interleukin-1ß can cross the BBB through active transport, entry of leukocytes and other blood contents are restricted (Gao and Hong, 2008). Systemic inflammatory molecules could disrupt the BBB. For example, injection of rats with interleukin-6 and interleukin-2 significantly increased the blood-to-brain transfer constant, indicative of BBB permeability (Saija et al., 1995). Similarly, exposure to TNFɑ, interleukin-6, and interleukin-1ß in a rat cerebral endothelial cell model increased permeability, as determined by trans-endothelial electrical resistance (TEER), which measures the integrity of the membrane as an electrical barrier (de Vries et al., 1996). In addition, numerous studies have demonstrated the ability of endotoxins like LPS to disrupt BBB integrity. One study modelled infection by injecting E. coli, an LPS-producing bacterium, into rats and found an increase in BBB permeability through the vasodilatory mechanisms of nitric oxide (Shukla et al., 1995). Another demonstrated enhanced permeability of cultured cerebral endothelial cells treated with LPS, as indicated with reduced TEER (De Vries et al., 1996). A similar reduction of TEER was also observed in cultured rat brain endothelial cells treated with LPS, accompanied by increased oxidative stress and nitric oxide levels (Veszelka et al., 2007). Effects on BBB function have been reported in the substantia nigra of PD patients using magnetic resonance imaging, where a higher transfer rate of contrast agent into the brain compared to healthy controls indicated increased permeability attributed to microvasculature dysfunction (Al-Bachari et al., 2020). A meta-analysis also found BBB disruption in PD patients using biofluid markers, such as elevated ratios of albumin levels in cerebrospinal fluid (Wong et al., 2022).

Figure 1
Illustration depicting the mechanism of neuronal damage involving reactive astrocytes, activated microglia, and dopaminergic neurons. Reactive astrocytes release pro-inflammatory cytokines, influencing activated microglia. Activated microglia respond to toxins and produce cytokines and superoxides, affecting dopaminergic neurons. The neurons undergo damage due to alpha-synuclein toxicity and inflammation, leading to apoptosis and other cellular releases, further interacting with the astrocytes and microglia in a damaging cycle.

Figure 1. The cycle of neuroinflammation and neuronal damage in PD. Microglia are activated via circulating toxins, α-Synuclein aggregates released from cells, or systemic inflammatory agents crossing the BBB. Upon activation, microglia release pro-inflammatory markers and reactive oxygen species that activate astrocytes and mediate toxicity in neurons. Meanwhile, inflammatory toxicity further damages neurons that contain toxic alpha-Synuclein aggregates. This damage ultimately leads to cell death and the release of cell contents, reactive oxygen species, and more aggregates into the extracellular space which further activates microglia. This feed-forward cycle leads to exacerbated inflammatory responses and chronic neuroinflammation. Created in BioRender. Fraser, P. (2025) https://BioRender.com/tk5fbnp

A compromised BBB could increase the transfer of blood contents, including circulating toxins and systemic inflammatory molecules, into the brain parenchyma and induce neuroinflammation. Chronic neuroinflammation is implicated in neurodegeneration in PD and post-mortem analyses of substantia nigra and striatum revealed increased microglial activation, leukocytes, and the presence of inflammatory cytokines (Gao and Hong, 2008; Wong et al., 2022; Mogi et al., 1994; Brochard et al., 2009). Central inflammation and microglial activation were also observed in de novo PD patients compared to healthy controls (Yacoubian et al., 2023). Systemic inflammation and its effectors could induce neuroinflammation as shown by a mouse study where the systemic injection of TNFɑ or LPS caused microglial activation and drove cytokine production in the brain. Neuroinflammation was attributed to a systemic increase in TNFɑ which infiltrated the BBB through interactions with the TNFɑ receptor, without which neuroinflammation was not observed (Qin et al., 2007). Adaptive immunity could also infiltrate the brain, as evidenced by the presence of helper and cytotoxic T cells in the post-mortem substantia nigra of PD patients (Brochard et al., 2009). This investigation demonstrated the presence of fluorescent-tagged T cells in the substantia nigra and striatum of mice after neurotoxin-induced PD. This was accompanied by the leakage of a serum protein, albumin, in the brain which suggests that a leaky BBB contributes to the migration of leukocytes into the brain (Brochard et al., 2009). Systemic inflammation induced by gut dysfunction in an LPS-injected rat model of ulcerative colitis also exacerbated dopaminergic cell loss, microglial activation, serum and central cytokine levels, and BBB dysfunction and dopaminergic cell loss (Villarán et al., 2010). These data suggest that peripheral inflammation could migrate into the brain and modulate neuroinflammation, although, this is not a one-way interaction as central inflammation could also influence and recruit peripheral inflammation (Ferrari and Tarelli, 2011).

Another signal that promotes neuroinflammation comes from the contents of dying neurons. Apoptotic neurons release their cytoplasmic contents, including α-Syn aggregates, into the extracellular space which can signal microglial activation. This release results in phagocytosis and the activation of microglial downstream mechanisms that produce reactive oxygen species and further exacerbate dopaminergic neurotoxicity (Zhang et al., 2005). Dying cells also release damage-associated molecular patterns that are recognized by microglia (Calabresi et al., 2023). Microglial activation also promotes the release of pro-inflammatory cytokines, like interleukin-1β, interleukin-6, interleukin-8, and TNFɑ (Grozdanov et al., 2019). Some of these cytokines, like TNFɑ and interleukin-1ɑ, promote reactive astrocytosis, a process which promotes genetic changes in astrocytes that induce a role shift from providing trophic support to neurotoxic activity (Liddelow et al., 2017). This leads to reduced survival of human dopaminergic neurons co-cultured with transformed astrocytes. The presence of these toxic astrocytes were also observed in the post-mortem tissue of PD patients, including in the substantia nigra, (Liddelow et al., 2017) suggesting that neurotoxic astrocytes activated by microglial cytokines contribute to dopaminergic cell death in PD.

While inflammation can mediate neurotoxicity and cell death, neuronal damage would further drive inflammation by releasing chemokines and markers that activate microglia and astrocytes. This creates a feed-forward loop that exacerbates inflammation and drives further neurodegeneration (Wu et al., 2005). Neurodegeneration could further compromise BBB through elevated levels of vascular endothelial growth factor (VEGF), which is responsible for angiogenesis during states of neuroinflammation. VEGF is a potent disruptor of BBB function and has been found to be elevated in the substantia nigra of PD patients (Rite et al., 2007; Wada et al., 2006). BBB damage could further allow systemic inflammation to infiltrate the brain and exacerbate neuroinflammation. This cycle would drive neurodegeneration in susceptible brain areas in PD, such as the substantia nigra (Figure 1). A higher presence of microglia in the substantia nigra compared to other regions in the brain could explain the increased susceptibility of dopaminergic cells in this region to microglial-mediated neurotoxicity (Qin et al., 2007). Dopaminergic neurons are also inherently more vulnerable to reactive oxygen species due to their low antioxidant capacity. Therefore, they are more readily damaged by the oxidative stress produced by neuroinflammation (Block and Hong, 2005).

Aging remains one of the strongest risk factors in developing PD which may be linked to elevated inflammatory pathways. It has been suggested that microglia become “primed” with advanced age, producing more pronounced inflammatory responses (Perry et al., 2007). As such, aged rodents and non-human primates show increased microglial expression of major histocompatibility complex II (MHCII), a marker of microglial activation (Perry et al., 1993; Sheffield and Berman, 1998). Inducing inflammation by peripheral injection of LPS in aged mice also resulted in more long-lasting pro-inflammatory cytokines and oxidative stress compared to younger adult mice, indicating an exaggerated inflammatory response (Godbout et al., 2005). Cellular senescence, an aging process that promotes cell cycle and genetic changes which alter cell function, is another aging phenomenon contributing to inflammation. Aging cells often exhibit mitochondrial dysfunction, elevated reactive oxygen species levels, and senescence-associated secretory phenotype (SASP) that make them more susceptible to PD-related pathology. SASP includes the release of cytokines and chemokines that activate an inflammatory response (Russo and Riessland, 2022). In addition, the senescence of endothelial cells in the vasculature has been shown to promote BBB dysfunction through reduced tight junction coverage in an accelerated aging mouse model (Yamazaki et al., 2016). Therefore, ageing could contribute to the development of PD by exacerbating inflammation, reducing anti-oxidative capacity, and disrupting BBB function. Therefore, systemic inflammation that originates from the gut or other sources may disrupt the BBB integrity, allow the entry of pro-inflammatory cytokines, and induce neuroinflammation. Inflammation also intersects with the aggregation of α-Syn, the hallmark protein implicated in PD.

4 α-Synuclein aggregation and transmission

α-Syn pathology spreads in a transneuronal prion-like fashion, where the aggregates are infectious protein “seeds” that can be transferred between donor and recipient cells (Figure 2). This process leads to the recruitment of endogenous monomeric α-Syn into the oligomerized form and amplifies aggregation (Lopes et al., 2022; Rahayel et al., 2022). It has been shown in vitro that α-Syn localizes to intracellular vesicles and can be secreted, even in aggregated form (Lee et al., 2008). Extracellular α-syn in increased by neuronal activity suggesting that α-syn release is regulated, at least in part, some form of physiological secretion (Yamada and Iwatsubo, 2018). Aggregates could be released through unconventional mechanisms that are independent of the Golgi and the endoplasmic reticulum like exosome-mediated vesicular exocytosis and tunneling nanotubule-mediated transfer through intercellular channels (Emmanouilidou et al., 2010; Abounit et al., 2016; Danzer et al., 2012). Exogenous α-Syn can also be internalized by neighboring cells through mechanisms like phagocytosis or receptor-mediated endocytosis (Choi et al., 2021; Lee et al., 2008; Sung et al., 2001). Once in the cell, aggregates enter the endocytic pathway and are found in endosomes and lysosomes to be degraded (Karpowicz et al., 2017; Konno et al., 2012). Investigations have also demonstrated that α-Syn aggregates in primary neuron cultures result in vesicular rupture in the endolysosomal pathway, coinciding with an increase in reactive oxygen species (Freeman et al., 2013; Chen et al., 2013; Samuel et al., 2016). Similar results were shown in other investigations which demonstrated that lysosomal rupture by α-Syn aggregates was subsequently followed by the aggregation of endogenous α-Syn (Kakuda et al., 2024). The exact mechanisms of lysosomal vesicle rupture remain unknown, however, studies have suggested aggregate-mediated membrane permeabilization, curvature induction, and oxidative stress-mediated membrane damage as possible causes (Senol et al., 2021; Varkey et al., 2010; Volles and Lansbury, 2002). Seeding aggregation disrupts normal neuronal physiology through impairing proteasome function which maintains proteomic homeostasis (Popova et al., 2021; Zondler et al., 2017), disturbing cellular trafficking mechanisms (Cooper et al., 2006; Wang and Hay, 2015), and impairing mitochondrial function (Geibl et al., 2023; Lurette et al., 2023; Ramezani et al., 2023), resulting in oxidative stress (Figure 2).

Figure 2
Diagram illustrating neuronal function and dysfunction. It shows vesicle release between a presynaptic and postsynaptic neuron, leading to endocytosis. Inside the postsynaptic neuron, processes like lysosomal evasion, mitochondrial and trafficking dysfunction, exocytosis, healthy alpha-synuclein recruitment, and apoptosis are depicted. A legend indicates symbols for healthy alpha-synuclein, alpha-synuclein aggregates, reactive oxygen species, and transport vesicles.

Figure 2. The donor-recipient transfer of aggregated α-Synuclein. Alpha-Synuclein aggregates can be transferred from infected donor cells to neurons in close vicinity, particularly those with synaptic connections. α-Synuclein aggregates leave the donor neuron through different mechanisms, like exocytosis, and are taken up by the recipient neuron via receptor-mediated endocytosis. Once inside the healthy recipient, the aggregates impair lysosomal function and escape degradation via lysosome rupture. In the cytosol, aggregates recruit the cell’s endogenous monomeric α-Synuclein and cause further aggregation. The accumulation of aggregates disrupts cell homeostasis, impairing Golgi-mediated trafficking, and mitochondrial function, leading to reactive oxygen species production and cell apoptosis. Created in BioRender. Fraser, P. (2025) https://BioRender.com/i0zlebg

α-Syn aggregates clearly play a critical role in the pathogenesis of PD and oligomers have been proposed to be more toxic than mature fibrillar aggregates (Mori et al., 2006; Sidhu et al., 2004; Tarutani et al., 2016). Additionally, α-Syn strains that create stable pS129 deposits are less toxic than those that are more readily destabilized (Lau et al., 2020). Furthermore, it has been found that α-Syn pathology precedes inflammation and glial activation (Izco et al., 2021). These data cumulatively suggest that α-Syn aggregation, mitochondrial dysfunction, and ROS generation all converge and cooperate toward neuronal death. As such, α-Syn aggregates and oligomers correlate with neuronal death and symptoms based on the affected areas (Kövari et al., 2003; Wakabayashi et al., 2006). This allows the aggregates to serve as a detectable and quantifiable measure of disease progression.

Neuron-to-neuron transfer of aggregates provide a mechanism for pathology to migrate from the gut to the brain via the vagus nerve (Figure 3). Indeed, α-Syn aggregates have been observed in the vagus nerve in pre-clinical studies. Mice injected in the intestine with PD brain lysates containing α-Syn aggregates showed a time-dependent transport along the vagus nerve, ultimately reaching the brainstem (Holmqvist et al., 2014). Subsequent investigation demonstrated that injecting pre-formed α-Syn fibrils into the intestine of mice resulted in neurotoxicity in the brain and motor deficits (Challis et al., 2020; Van Den Berge et al., 2021); importantly, these studies showed α-Syn pathology did not progress beyond the brainstem unless the animals were aged. The age-dependent gut-brain propagation can be circumvented by α-Syn overexpression (Van Den Berge et al., 2019). The gut-to-brain spread of pathology and the resulting motor and cognitive deficits were prevented by truncal (full) vagotomy and in α-Syn deficient mice, highlighting the importance of the vagus nerve for the gut-brain transfer of pathology and the necessary recruitment of endogenous α-Syn substrate for this process (Kim et al., 2019). Indeed, in humans, α-Syn aggregates were observed in post-mortem cervical vagus nerve samples of PD patients (Mu et al., 2013). Additionally, two large cohort studies of vagotomized humans also showed a lower incidence of PD development for individuals that underwent truncal, but not selective (partial) vagotomy at least 5 years prior to diagnosis (Liu et al., 2017; Svensson et al., 2015). Together, the prion-like spread of α-Syn along the vagal pathway is a key event in the propagation of pathology to the brain.

Figure 3
Diagram illustrating gut dysbiosis. Panel A shows microbial imbalance in the intestines. Panel B depicts microbial byproduct leakage. Panel C shows compromised enteric nervous system with bacterial products interacting with neurons. Panel D indicates alpha-synuclein aggregation and propagation to the brain. Includes a legend with icons for foreign pathogens, bacteria, bacterial products (LPS), intestinal epithelium, enteric neurons, and alpha-syn aggregates.

Figure 3. An overview of the gut etiology of Parkinson’s disease. A foreign pathogen (A) causes the intestine wall to become permeable (B), allowing microbial products and other intestine contents to cross over the intestine wall and interact with neurons that are situated in the underlying tissue (C). These microbial products interfere with the normal function of neurons and cause the formation of α-Synuclein aggregates (D). These aggregates can move from the digestive system to the brain via the vagus nerve, which connects the brain to internal organs. Created in BioRender. Fraser, P. (2025) https://BioRender.com/jqu4fxg

It is important to note that the gut-first model does not apply to all PD cases since some patients report GI symptoms without protein aggregation in the brain and others have protein aggregation without clinical GI symptoms (Gaig et al., 2007; Parkkinen et al., 2005). Clinically, not all patients experience premotor GI symptoms (Yu et al., 2018). The modern body-first and brain-first models of PD imply two subtypes of PD, each with unique patterns of α-Syn spread that ultimately reach and cause neuronal death in the SN (Borghammer and Van Den Berge, 2019; Borghammer, 2021). This is substantiated by the conformer strain hypothesis which states that different conformations of misfolded α-Syn lead to strains with unique aggregation kinetics, resistance to degradation, and patterns of spread in the brain. For example, aggregates generated from α-Syn monomers in low and high salt concentrations showed different resistance to proteinase K, depolymerization thresholds, and toxicity levels in vitro and in vivo (Lau et al., 2020; So et al., 2024; Peelaerts et al., 2015). The spread of pathology and motor disruptions in the animals inoculated with α-Syn aggregates also occurred in a strain-dependent manner wherein fibrils formed in high salt conditions caused greater detriment, leading to shorter lifespans (Lau et al., 2020; So et al., 2024). As such, it is possible that the body-first and brain-first subtypes of PD, having different sites of origins, are characterized by different strains of PD. Nevertheless, the GI tract is affected in about 50–70% of PD patients and remains a therapeutic target, even among patients of brain-first PD as GI dysfunction occurs in their later stages of disease (Gan et al., 2018; Borghammer and Van Den Berge, 2019).

Therefore, propagation patterns in the brain can be diverse, as expected with the heterogenous nature of PD, and can be influenced by factors like α-Syn strain, cell vulnerability, and anatomic connectivity. These findings highlight the need for PD therapeutics that can address the heterogenous nature of PD pathology and symptoms as well as target not only the brain but also peripheral organs. The difficulty of simultaneously targeting the central and peripheral neurons can be addressed by adeno-associated virus-mediated gene delivery.

5 Gene transfer to the nervous system by systemic AAV

As discussed in the previous sections, the early appearance in the periphery of α-Syn aggregation and inflammation in some PD cases suggests potential benefits of initiating therapeutics to target peripheral sites. Multiple anti-inflammatory drugs have been tested and shown to be beneficial in different animal models of PD. However, although benefits on motor symptoms and inflammation were observed, these are not permanent and the spread of brain pathology is not completely halted (Çınar et al., 2022). To halt the feed-forward loop between neuroinflammation and α-Syn aggregation, synucleinopathy must also be addressed. Delivering gene therapies that can reduce α-Syn pathology in neurons has also shown positive improvements in motor symptoms, cognition and clearing α-Syn aggregates. Among the beneficial gene products are nucleotides that reduce the amounts of SNCA mRNA like microRNAs or short hairpin-RNAs, as well as proteins that drive the clearance of native or fibrillar α-Syn like intrabodies (Menon et al., 2021). The mechanisms of action of these therapies are beyond the scope of this review. This review focuses rather on the delivery of gene therapy vectors to the gut and the brain, specifically by adeno-associated viruses (AAV).

AAV has been an attractive option to use for gene delivery for numerous reasons. First, AAV has innately low immunogenicity as it cannot replicate without adenovirus. Second, it enables efficient gene delivery of sequences up to approximately 4.5 kbp (Hermonat and Muzyczka, 1984). Importantly, in the case of PD, the delivered transgenes have long-term expression even in post-mitotic cells (Royo et al., 2008). This allows a single transduction event to be beneficial throughout the duration of the neuron’s lifespan. Lastly, AAVs delivered in the bloodstream can transduce PD-related organs. Together, this paints a possible future where multiple PD-related organs are treated with a single intravenous injection of AAV-anti-α-Syn gene therapy.

Many studies that test gene transfer to the brain usually use direct injection of the gene carried by the AAV1 or AAV2 serotype (Haery et al., 2019). In developing an AAV-mediated gene therapy for PD, preclinical tests have attempted AAV brain transduction through the bloodstream. Among all the naturally occurring AAV serotypes, AAV9 has the best ability to transduce the BBB from the circulation. However, AAV9 may only pass through the BBB with high doses (Gray et al., 2011), in neonates whose BBBs are still underdeveloped (Foust et al., 2009), or with the help of magnetic resonance imaging-guided focused ultrasound (FUS) with microbubbles which can transiently increase the permeability of the BBB (Thévenot et al., 2012). Indeed, mice that received an intravenous dose of AAV9-SNCA-shRNA and FUS showed transgene expression and reduced α-Syn in the FUS-targeted brain areas (Xhima et al., 2018).

To improve AAV9 BBB penetration, a library of AAV9 derivatives was engineered using the Cre-recombinase-dependent AAV targeted evolution (CREATE) method to evolve AAV9 selective for improved brain transduction following intravenous administration (Deverman et al., 2016). Briefly, the CREATE method involves making a library of AAV9 derivatives with evolved capsids. The edited AAV9 library is then tagged with a loxP sequence and injected into transgenic mice that express Cre-recombinase in predefined cell types. PCR is then performed to determine which AAV9 variant transduced the Cre-positive cells. By selecting for transduction in Cre-positive neurons, selection is biased toward those that bypass the BBB.

One of the first BBB-penetrant AAV9 derivatives is AAV-PHP. B (hereafter just PHP. B), known for its brain transduction capabilities (Deverman et al., 2016). An intravenous dose of PHP. B-GFP transduces the entire brain and spinal cord significantly more effectively than the parent AAV9, even at a tenth of AAV9’s dosage. Additionally, PHP. B exhibited significantly less off-target peripheral expression in the pancreas and adrenal gland, with a decreasing trend observed in the liver (Deverman et al., 2016). Further evolution of PHP. B by CREATE led to the development of AAV-PHP.eB (enhanced/evolved brain transduction). PHP.eB demonstrates even greater BBB penetrance and neuronal transduction while maintaining similar astrocyte tropism compared to its PHP. B predecessor (Chan et al., 2017). However, because PHP. B and PHP.eB can transduce the entire brain, they lack spatial resolution which is a critical consideration when designing treatments. If greater localization of gene therapy is required, AAV9 with FUS could be utilized.

Gene delivery to the BBB is also important to rescue its integrity in PD patients who have increased BBB permeability (Al-Bachari et al., 2020). Among the first PHP variants that were discovered is PHP. V1 which showed greater endothelial cell transduction than AAV9 (Ravindra Kumar et al., 2020). Building on this, an improved multiplexed-CREATE (M-CREATE) method was used on AAV9 to create AAV-X1 and -X1.1 which display enhanced selectivity for BBB endothelial cells and avoidance of liver transduction (Chen et al., 2023). Additionally, these two AAV vectors allow for multiple AAV gene delivery and serotype switching from the blood to the BBB which avoid triggering immune responses from the subject. A potential therapeutic strategy for PD was tested by the creators of AAV-X1 and -X1.1. The endothelial cells were turned into a “biofactory” by the transduction of AAV-X1 carrying genes for the matricellular protein Hevin which is helpful for neuronal and astrocytic health (Chen et al., 2023). This biofactory strategy can be used to create BBB resistance against systemic inflammation in PD or other neurodegenerative diseases that also damage the BBB.

In the context of the body-first model of PD, gene delivery to the enteric neurons must also be considered. Once again, AAV9 displays the highest transduction levels in enteric neurons and glia over the other naturally occurring AAVs (Gombash et al., 2014). The development of AAV9 derivative, AAV-PHP. S, which, although not highly efficient at crossing the BBB, exhibits robust expression in the spinal cord and sensory afferents leading to the brainstem (Chan et al., 2017). PHP. S demonstrates enhanced neuronal transduction compared to AAV9 in the dorsal root ganglia (DRG) of the spinal cord, cardiac ganglia, and both the myenteric and submucosal plexuses of the ENS, all relevant to the body-first subtype of PD. In the ENS, PHP. S shows superior transduction efficiency and uniformity along the length of the small intestine compared to AAV9 (Chan et al., 2017). Systemic injection (iv) of PHP. S-GBA1 transduced enteric neurons and increased duodenal GCase expression (Challis et al., 2020). This treatment mitigated the pathology induced by α-syn pre-formed fibrils and restored gut function to healthy wild-type levels.

Since PD leads to dysfunction of both the CNS and ENS, optimizing gene delivery to both the brain and gut could be beneficial. This can be done through MaCPNS1 and MaCPNS2, two derivatives of PHP. S that target the brain and small intestine with reduced liver expression (Chen et al., 2022). Additionally, both vectors exhibit approximately a two-fold increase in tropism for the dorsal root ganglia of the spine and nodose ganglion of the vagus nerve. The ability to target multiple peripheral nerves without significant liver tropism is beneficial as AAV’s natural high affinity for the liver reduces the bioavailability for the target cells and may risk liver damage (Flotte, 2021; Zincarelli et al., 2008). Lower liver tropism may also decrease the required AAV dose and help avoid undesired immune responses that could exacerbate inflammation (Mingozzi and High, 2013).

Together, the current understanding and engineering of AAVs sets the scene for a gene delivery therapeutic for PD that could be administered by a single or few intravenous injections. The treatment serotype used could also be personalized to the patient based on the location of the disease (Table 1). For example, PHP. S-like serotypes may be used for patients in the early stage of disease that follows the gut-to-brain model while those who follow a brain-centric disease progression may benefit more from PHP.eB-like serotypes or the combination AAV9 and FUS. MaCPNS-like serotypes may be beneficial for patients in whom central and peripheral symptoms are observed. If the BBB is the target of gene delivery, then serotypes with the properties of AAV-X1 and -X1.1 may be used. Importantly, all the possible options mentioned avoid highly invasive brain or intestinal injections.

Table 1
www.frontiersin.org

Table 1. Summary table of AAV serotypes with tropism to PD-related organs via the bloodstream.

6 Conclusion

In many cases, Parkinson’s disease is associated with the presence of α-Syn pathology and pre-motor gastrointestinal symptoms, the neurotoxic effects of pesticides, herbicides, and toxins injected in the gastrointestinal system, and post-mortem patterns of aggregates in brain structures. The observation of pro-inflammatory shifts in the gut microbiome makeup of PD patients suggests a potential source for neurotoxin production and initiation of systemic and neuroinflammatory responses that may drive α-Syn aggregation. Disruptions to the homeostatic state of neurons, their inherent physical connectivity, and chronic inflammatory responses collectively drive neurodegeneration in susceptible dopaminergic cells, primarily through mitochondrial dysfunction. However, key questions remain, including the mechanisms through which dysbiosis and toxins contribute to α-Syn aggregation, whether α-Syn aggregation acts as a defensive mechanism or a pathology driver, how α-Syn bypasses endolysosomal degradation, and the reasons behind the vulnerability of dopaminergic cells. Also, further studies on pre-clinical stage PD patients are required to help elucidate early changes in the microbiota and the extent of the subsequent damage in the enteric nervous system.

Since the body-first model of PD with early peripheral nervous system involvement holds true for a subset of PD patients, a brain-first manner of propagation has also been hypothesized. This is consistent with clinical manifestation where some patients do not exhibit autonomic symptoms prior to motor deficits caused by dopaminergic loss in the striatum and substantia nigra. Deciphering whether different α-Syn strains are involved with the two subtypes could help elucidate the etiology of PD. α-Syn-related pathology is implicated not only in PD, but also in other neurodegenerative diseases, such as dementia with Lewy bodies, multiple system atrophy, and pure autonomic failure (Calabresi et al., 2023). These synucleinopathies have different aggregation types but also overlapping clinical and pathological features, meriting further research into the applicability of the gut-first hypothesis, or parts of it, to other disorders.

The implication of multiple sites of inflammation and α-Syn pathology posits the need of delivering beneficial gene products to the affected sites. Multiple gene therapy strategies that target aggregated α-Syn can reduce inflammation in the targeted sites. However, more research on therapies that avoid invasive brain injections or surgeries is required. To this end, current advances in AAV gene delivery may simultaneously deliver these gene products to multiple PD-related organs, like the gut and brain, through a single intravenous injection. The AAV toolkit discussed in this review also points to the possibility of personalizing the gene delivery treatment, thus addressing the multi-faceted characteristic of PD or other synucleinopathies. Future preclinical and clinical experiments may test whether and how systemic gene therapy may rescue, reduce, delay, or prevent synucleinopathies.

Author contributions

SN: Writing – original draft. ML: Writing – original draft. PF: Writing – review & editing, Funding acquisition. AT: Writing – review & editing, Funding acquisition.

Funding

The author(s) declare that financial support was received for the research and/or publication of this article. This work was supported by the Canadian Institutes of Health Research (PJT-173497 to PF; PJT-180582 to AT).

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Generative AI statement

The authors declare that no Gen AI was used in the creation of this manuscript.

Any alternative text (alt text) provided alongside figures in this article has been generated by Frontiers with the support of artificial intelligence and reasonable efforts have been made to ensure accuracy, including review by the authors wherever possible. If you identify any issues, please contact us.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Abbreviations

PD, Parkinson’s Disease; α-Syn, α-Synuclein; AAV, Adeno-Associated Virus; CNS, Central Nervous System; ENS, Enteric Nervous System; LPS, Lipopolysaccharide; TLR4, Toll-Like Receptor 4; SCFA, Short-Chain Fatty Acid; TEER, Trans-Endothelial Electrical Resistance; BBB, Blood Brain Barrier; TNFɑ, Tumor Necrosis Factor Alpha; VEGF, Vascular Endothelial Growth Factor; MHCII, Major Histocompatibility Complex II; SASP, Senescence-Associated Secretory Phenotype; LPT, Long-Term Potentiation; NADPH, Nicotinamide Adenine Dinucleotide Phosphate; DMV, Dorsal Motor Nucleus of the Vagus.

References

Abounit, S., Bousset, L., Loria, F., Zhu, S., Chaumont, F.d., Pieri, L., et al. (2016). Tunneling nanotubes spread fibrillar α-synuclein by intercellular trafficking of lysosomes 35:2120. doi: 10.15252/embj.201593411

Crossref Full Text | Google Scholar

Aho, V. T. E., Houser, M. C., Pereira, P. A. B., Chang, J., Rudi, K., Paulin, L., et al. (2021). Relationships of gut microbiota, short-chain fatty acids, inflammation, and the gut barrier in Parkinson’s disease. Mol. Neurodegener. 16:427. doi: 10.1186/s13024-021-00427-6

PubMed Abstract | Crossref Full Text | Google Scholar

Al-Bachari, S., Naish, J. H., Parker, G. J. M., Emsley, H. C. A., and Parkes, L. M. (2020). Blood–brain barrier leakage is increased in Parkinson’s disease. Front. Physiol. 11:593026. doi: 10.3389/fphys.2020.593026

PubMed Abstract | Crossref Full Text | Google Scholar

Baert, F., Matthys, C., Maselyne, J., Poucke, C., Coillie, E., Bergmans, B., et al. (2021). Parkinson's disease patients' short chain fatty acids production capacity after in vitro fecal fiber fermentation. NPJ Parkinsons Dis. 7:72. doi: 10.1038/s41531-021-00215-5

PubMed Abstract | Crossref Full Text | Google Scholar

Barichella, M., Severgnini, M., Cilia, R., Cassani, E., Bolliri, C., Caronni, S., et al. (2019). Unraveling gut microbiota in Parkinson's disease and atypical parkinsonism 34, 396–405. doi: 10.1002/mds.27581

Crossref Full Text | Google Scholar

Beach, T. G., Adler, C. H., Sue, L. I., Shill, H. A., Driver-Dunckley, E., Mehta, S. H., et al. (2021). Vagus nerve and stomach Synucleinopathy in Parkinson's disease, incidental Lewy body disease, and Normal elderly subjects: evidence against the 'body-first' hypothesis. J. Parkinsons Dis. 11, 1833–1843. doi: 10.3233/JPD-212733

PubMed Abstract | Crossref Full Text | Google Scholar

Bhattacharyya, D., Mohite, G. M., Krishnamoorthy, J., Gayen, N., Mehra, S., Navalkar, A., et al. (2019). Lipopolysaccharide from gut microbiota modulates α-synuclein aggregation and alters its biological function. ACS Chem. Neurosci. 10, 2229–2236. doi: 10.1021/acschemneuro.8b00733

PubMed Abstract | Crossref Full Text | Google Scholar

Block, M. L., and Hong, J. S. (2005). Microglia and inflammation-mediated neurodegeneration: multiple triggers with a common mechanism. Prog. Neurobiol. 76, 77–98. doi: 10.1016/j.pneurobio.2005.06.004

PubMed Abstract | Crossref Full Text | Google Scholar

Borghammer, P. (2021). The α-Synuclein origin and connectome model (SOC model) of Parkinson's disease: explaining motor asymmetry, non-motor phenotypes, and cognitive decline. J. Parkinsons Dis. 11, 455–474. doi: 10.3233/JPD-202481

PubMed Abstract | Crossref Full Text | Google Scholar

Borghammer, P., and Van Den Berge, N. (2019). Brain-first versus gut-first Parkinson’s disease: a hypothesis. J. Parkinson’s Dis. 9:S281. doi: 10.3233/JPD-191721

Crossref Full Text | Google Scholar

Braak, H., De Vos, R. A. I., Bohl, J., and Del Tredici, K. (2006). Gastric alpha-synuclein immunoreactive inclusions in Meissner's and Auerbach's plexuses in cases staged for Parkinson's disease-related brain pathology 396, 67–72. doi: 10.1016/j.neulet.2005.11.012

Crossref Full Text | Google Scholar

Braak, H., Del Tredici, K., Rüb, U., De Vos, R. A. I., Jansen Steur, E. N. H., and Braak, E. (2003). Staging of brain pathology related to sporadic Parkinson’s disease 24, 197–211. doi: 10.1016/s0197-4580(02)00065-9

Crossref Full Text | Google Scholar

Braak, H., Rüb, U., Gai, W. P., and Del Tredici, K. (2003). Idiopathic Parkinson's disease: possible routes by which vulnerable neuronal types may be subject to neuroinvasion by an unknown pathogen. J. Neural Transm. 110, 517–536. doi: 10.1007/s00702-002-0808-2

PubMed Abstract | Crossref Full Text | Google Scholar

Brochard, V., Combadière, B., Prigent, A., Laouar, Y., Perrin, A., Beray-Berthat, V., et al. (2009). Infiltration of CD4+ lymphocytes into the brain contributes to neurodegeneration in a mouse model of Parkinson disease. J. Clin. Inv. 119, 182–192. doi: 10.1172/JCI36470

Crossref Full Text | Google Scholar

Brugger, F., Erro, R., Balint, B., Kägi, G., Barone, P., and Bhatia, K. P. (2015). Why is there motor deterioration in Parkinson’s disease during systemic infections-a hypothetical view. NPJ Parkinsons Dis. 1:15014. doi: 10.1038/npjparkd.2015.14

Crossref Full Text | Google Scholar

Calabresi, P., Mechelli, A., Natale, G., Volpicelli-Daley, L., Di Lazzaro, G., and Ghiglieri, V. (2023). Alpha-synuclein in Parkinson’s disease and other synucleinopathies: from overt neurodegeneration back to early synaptic dysfunction. Cell Death Dis. 14, 1–16. doi: 10.1038/s41419-023-05672-9

Crossref Full Text | Google Scholar

Cersosimo, M. G., Raina, G. B., Pecci, C., Pellene, A., Calandra, C. R., Gutiérrez, C., et al. (2013). Gastrointestinal manifestations in Parkinson’s disease: prevalence and occurrence before motor symptoms. J. Neurol. 260, 1332–1338. doi: 10.1007/s00415-012-6801-2

PubMed Abstract | Crossref Full Text | Google Scholar

Challis, C., Hori, A., Sampson, T. R., Yoo, B. B., Challis, R. C., Hamilton, A. M., et al. (2020). Gut-seeded α-synuclein fibrils promote gut dysfunction and brain pathology specifically in aged mice. Nat. Neurosci. 23, 327–336. doi: 10.1038/s41593-020-0589-7

PubMed Abstract | Crossref Full Text | Google Scholar

Chan, K. Y., Jang, M. J., Yoo, B. B., Greenbaum, A., Ravi, N., Wu, W. L., et al. (2017). Engineered AAVs for efficient noninvasive gene delivery to the central and peripheral nervous systems. Nat. Neurosci. 20, 1172–1179. doi: 10.1038/nn.4593

PubMed Abstract | Crossref Full Text | Google Scholar

Chen, Q. Q., Haikal, C., Li, W., and Li, J. Y. (2019). Gut inflammation in association with pathogenesis of Parkinson’s disease. Front. Mol. Neurosci. 12:218. doi: 10.3389/fnmol.2019.00218

PubMed Abstract | Crossref Full Text | Google Scholar

Chen, H., Jacobs, E., Schwarzschild, M. A., McCullough, M. L., Calle, E. E., Thun, M. J., et al. (2005). Nonsteroidal antiinflammatory drug use and the risk for Parkinson's disease. Ann. Neurol. 58, 963–967. doi: 10.1002/ana.20682

PubMed Abstract | Crossref Full Text | Google Scholar

Chen, G., Ran, X., Li, B., Li, Y., He, D., Huang, B., et al. (2018). Sodium butyrate inhibits inflammation and maintains epithelium barrier integrity in a TNBS-induced inflammatory bowel disease mice model. EBioMedicine 30, 317–325. doi: 10.1016/j.ebiom.2018.03.030

PubMed Abstract | Crossref Full Text | Google Scholar

Chen, X., Ravindra Kumar, S., Adams, C. D., Yang, D., Wang, T., Wolfe, D. A., et al. (2022). Engineered AAVs for non-invasive gene delivery to rodent and non-human primate nervous systems. Neuron 110, 2242–22576. doi: 10.1016/j.neuron.2022.05.003

PubMed Abstract | Crossref Full Text | Google Scholar

Chen, Z. F., Shi, S. M., Hu, R. X., Zhang, M., Liang, H., and Zhou, Z. Y. (2003). Nonsteroidal anti-inflammatory drugs and the risk of Parkinson disease 60, 1059–1065. doi: 10.1001/archneur.60.8.1059

Crossref Full Text | Google Scholar

Chen, S. G., Stribinskis, V., Rane, M. J., Demuth, D. R., Gozal, E., Roberts, A. M., et al. (2016). Exposure to the functional bacterial amyloid protein curli enhances alpha-synuclein aggregation in aged Fischer 344 rats and Caenorhabditis elegans. Sci. Rep. 6:477. doi: 10.1038/srep34477

PubMed Abstract | Crossref Full Text | Google Scholar

Chen, R. H. C., Wislet-Gendebien, S., Samuel, F., Visanji, N. P., Zhang, G., Marsilio, D., et al. (2013). α-synuclein membrane association is regulated by the Rab3a recycling machinery and presynaptic activity. J. Biol. Chem. 288, 7438–7449. doi: 10.1074/jbc.M112.439497

Crossref Full Text | Google Scholar

Chen, X., Wolfe, D. A., Bindu, D. S., Zhang, M., Taskin, N., Goertsen, D., et al. (2023). Functional gene delivery to and across brain vasculature of systemic AAVs with endothelial-specific tropism in rodents and broad tropism in primates. Nat. Commun. 14:38582. doi: 10.1038/s41467-023-38582-7

PubMed Abstract | Crossref Full Text | Google Scholar

Cherny, I., Rockah, L., Levy-Nissenbaum, O., Gophna, U., Ron, E. Z., and Gazit, E. (2005). The formation of Escherichia coli curli amyloid fibrils is mediated by prion-like peptide repeats. J. Mol. Biol. 352, 245–252. doi: 10.1016/j.jmb.2005.07.028

PubMed Abstract | Crossref Full Text | Google Scholar

Choi, J. G., Kim, N., Ju, I. G., Eo, H., Lim, S. M., Jang, S. E., et al. (2018). Oral administration of Proteus mirabilis damages dopaminergic neurons and motor functions in mice. Sci. Rep. 8:1275. doi: 10.1038/s41598-018-19646-x

PubMed Abstract | Crossref Full Text | Google Scholar

Choi, Y. R., Park, S. J., and Park, S. M. (2021). Molecular events underlying the cell-to-cell transmission of α-synuclein. FASEB J 288, 6593–6602. doi: 10.1111/febs.15674

Crossref Full Text | Google Scholar

Chung, S. J., Kim, J., Lee, H. J., Ryu, H. S., Kim, K., Lee, J. H., et al. (2016). Alpha-synuclein in gastric and colonic mucosa in Parkinson's disease: limited role as a biomarker. Mov. Disord. 31, 241–249. doi: 10.1002/mds.26473

PubMed Abstract | Crossref Full Text | Google Scholar

Çınar, E., Tel, B. C., and Şahin, G. (2022). Neuroinflammation in Parkinson’s disease and its treatment opportunities. Balkan Med. J. 39, 318–333. doi: 10.4274/balkanmedj.galenos.2022.2022-7-100

PubMed Abstract | Crossref Full Text | Google Scholar

Cirstea, M. S., Yu, A. C., Golz, E., Sundvick, K., Kliger, D., Radisavljevic, N., et al. (2020). Microbiota composition and metabolism are associated with gut function in Parkinson's disease. Mov. Disord. 35, 1208–1217. doi: 10.1002/mds.28052

PubMed Abstract | Crossref Full Text | Google Scholar

Cooper, A. A., Gitler, A. D., Cashikar, A., Haynes, C. M., Hill, K. J., Bhullar, B., et al. (2006). α-Synuclein blocks ER-Golgi traffic and Rab1 rescues neuron loss in Parkinson's models. Science 313, 324–328. doi: 10.1126/science.1129462

PubMed Abstract | Crossref Full Text | Google Scholar

Cryan, J. F., O, K. J., Cowan, C. S. M., Sandhu, K. V., Bastiaanssen, T. F. S., Boehme, M., et al. (2019). The microbiota-gut-brain Axis. Physiol. Rev. 99, 1877–2013. doi: 10.1152/physrev.00018.2018

Crossref Full Text | Google Scholar

Danzer, K. M., Kranich, L. R., Ruf, W. P., Cagsal-Getkin, O., Winslow, A. R., Zhu, L., et al. (2012). Exosomal cell-to-cell transmission of alpha synuclein oligomers. Mol. Neurodegener. 7:42. doi: 10.1186/1750-1326-7-42

PubMed Abstract | Crossref Full Text | Google Scholar

de Vries, H. E., Blom-Roosemalen, M. C., de Boer, A. G., van Berkel, T. J., Breimer, D. D., and Kuiper, J. (1996). Effect of endotoxin on permeability of bovine cerebral endothelial cell layers in vitro. J. Pharmacol. Exp. Ther. 277, 1418–1423. doi: 10.1016/S0022-3565(25)13090-5

Crossref Full Text | Google Scholar

De Vries, H. E., Blom-Roosemalen, M. C. M., Van Oosten, M., De Boer, A. G., Van Berkel, T. J. C., Breimer, D. D., et al. (1996). The influence of cytokines on the integrity of the blood-brain barrier in vitro. J. Neuroimmunol. 64, 37–43.

Google Scholar

Deng, I., and Bobrovskaya, L. (2022). Lipopolysaccharide mouse models for Parkinson’s disease research: a critical appraisal 17:2413. doi: 10.4103/1673-5374.331866

Crossref Full Text | Google Scholar

Deverman, B. E., Pravdo, P. L., Simpson, B. P., Kumar, S. R., Chan, K. Y., Banerjee, A., et al. (2016). Cre-dependent selection yields AAV variants for widespread gene transfer to the adult brain. Nat. Biotechnol. 34, 204–209. doi: 10.1038/nbt.3440

PubMed Abstract | Crossref Full Text | Google Scholar

Devos, D., Lebouvier, T., Lardeux, B., Biraud, M., Rouaud, T., Pouclet, H., et al. (2013). Colonic inflammation in Parkinson's disease. Neurobiol. Dis. 50, 42–48. doi: 10.1016/j.nbd.2012.09.007

PubMed Abstract | Crossref Full Text | Google Scholar

Dorsey, E. R., and Bloem, B. R.The Parkinson Pandemic—A Call to Action (2018). The Parkinson pandemic—a call to action. JAMA Neurol. 75, 9–10. doi: 10.1001/jamaneurol.2017.3299

PubMed Abstract | Crossref Full Text | Google Scholar

Emmanouilidou, E., Melachroinou, K., Roumeliotis, T., Garbis, S. D., Ntzouni, M., Margaritis, L. H., et al. (2010). Cell-produced alpha-synuclein is secreted in a calcium-dependent manner by exosomes and impacts neuronal survival. J. Neurosci. 30, 6838–6851. doi: 10.1523/JNEUROSCI.5699-09.2010

PubMed Abstract | Crossref Full Text | Google Scholar

Evans, M. L., Chorell, E., Taylor, J. D., Åden, J., Götheson, A., Li, F., et al. (2015). The bacterial curli system possesses a potent and selective inhibitor of amyloid formation. Mol. Cell 57, 445–455. doi: 10.1016/j.molcel.2014.12.025

PubMed Abstract | Crossref Full Text | Google Scholar

Fasano, A., Visanji, N. P., Liu, L. W. C., Lang, A. E., and Pfeiffer, R. F. (2015). Gastrointestinal dysfunction in Parkinson's disease. Lancet Neurol. 14, 625–639. doi: 10.1016/S1474-4422(15)00007-1

PubMed Abstract | Crossref Full Text | Google Scholar

Ferrari, C. C., and Tarelli, R. (2011). Parkinson's disease and systemic. Inflammation 2011:436813. doi: 10.4061/2011/436813

Crossref Full Text | Google Scholar

Flotte, T. R. (2021). Liver targeting with rAAV7: balancing tropism with immune profiles. Gene Ther. 28, 115–116. doi: 10.1038/s41434-021-00230-4

PubMed Abstract | Crossref Full Text | Google Scholar

Forsyth, C. B., Shannon, K. M., Kordower, J. H., Voigt, R. M., Shaikh, M., Jaglin, J. A., et al. (2011). Increased intestinal permeability correlates with sigmoid mucosa alpha-synuclein staining and endotoxin exposure markers in early Parkinson's disease. PLoS One 6:28032. doi: 10.1371/journal.pone.0028032

PubMed Abstract | Crossref Full Text | Google Scholar

Foust, K. D., Nurre, E., Montgomery, C. L., Hernandez, A., Chan, C. M., and Kaspar, B. K. (2009). Intravascular AAV9 preferentially targets neonatal neurons and adult astrocytes. Nat. Biotechnol. 27, 59–65. doi: 10.1038/nbt.1515

PubMed Abstract | Crossref Full Text | Google Scholar

Freeman, D., Cedillos, R., Choyke, S., Lukic, Z., McGuire, K., Marvin, S., et al. (2013). Alpha-synuclein induces lysosomal rupture and cathepsin dependent reactive oxygen species following endocytosis. PLoS ONE 8:62143. doi: 10.1371/journal.pone.0062143

Crossref Full Text | Google Scholar

Gaig, C., Martí, M. J., Ezquerra, M., Rey, M. J., Cardozo, A., and Tolosa, E. (2007). G2019s LRRK2 mutation causing Parkinson's disease without Lewy bodies. J. Neurol. Neurosurg. Psychiatry 78, 626–628. doi: 10.1136/jnnp.2006.107904

PubMed Abstract | Crossref Full Text | Google Scholar

Gan, J., Wan, Y., Shi, J., Zhou, M., Lou, Z., and Liu, Z. (2018). A survey of subjective constipation in Parkinson's disease patients in shanghai and literature review. BMC Neurol. 18:29. doi: 10.1186/s12883-018-1034-3

PubMed Abstract | Crossref Full Text | Google Scholar

Ganguly, U., Singh, S., Pal, S., Prasad, S., Agrawal, B. K., Saini, R. V., et al. (2021). Alpha-Synuclein as a biomarker of Parkinson’s disease: good, but not good enough. Front. Aging Neurosci. 13:702639. doi: 10.3389/fnagi.2021.702639

PubMed Abstract | Crossref Full Text | Google Scholar

Gao, H. M., and Hong, J. S. (2008). Why neurodegenerative diseases are progressive: uncontrolled inflammation drives disease progression. Trends Immunol. 29, 357–365. doi: 10.1016/j.it.2008.05.002

PubMed Abstract | Crossref Full Text | Google Scholar

Geibl, F. F., Henrich, M. T., Xie, Z., Zampese, E., Tkatch, T., Wokosin, D. L., et al. (2023). α-Synuclein pathology disrupts mitochondrial function in dopaminergic and cholinergic neurons at-risk in Parkinson's disease. Mol. Neurodegen. 19:69.

Google Scholar

Godbout, J. P., Chen, J., Abraham, J., Richwine, A. F., Berg, B. M., Kelley, K. W., et al. (2005). Exaggerated neuroinflammation and sickness behavior in aged mice after activation of the peripheral innate immune system. FASEB J 19, 1329–1331. doi: 10.1096/fj.05-3776fje

Crossref Full Text | Google Scholar

Gombash, S. E., Cowley, C. J., Fitzgerald, J. A., Hall, J. C. E., Mueller, C., Christofi, F. L., et al. (2014). Intravenous AAV9 efficiently transduces myenteric neurons in neonate and juvenile mice. Front. Mol. Neurosci. 7:81. doi: 10.3389/fnmol.2014.00081

PubMed Abstract | Crossref Full Text | Google Scholar

Gorecki, A. M., Preskey, L., Bakeberg, M. C., Kenna, J. E., Gildenhuys, C., MacDougall, G., et al. (2019). Altered gut microbiome in Parkinson's disease and the influence of lipopolysaccharide in a human α-synuclein over-expressing mouse model 13:473589. doi: 10.3389/fnins.2019.00839

Crossref Full Text | Google Scholar

Gray, S. J., Matagne, V., Bachaboina, L., Yadav, S., Ojeda, S. R., and Samulski, R. J. (2011). Preclinical differences of intravascular aav9 delivery to neurons and glia: a comparative study of adult mice and nonhuman primates. Mol. Ther. 19, 1058–1069. doi: 10.1038/mt.2011.72

PubMed Abstract | Crossref Full Text | Google Scholar

Grozdanov, V., Bousset, L., Hoffmeister, M., Bliederhaeuser, C., Meier, C., Madiona, K., et al. (2019). Increased immune activation by pathologic α-synuclein in Parkinson's disease. Ann. Neurol. 86, 593–606. doi: 10.1002/ana.25557

PubMed Abstract | Crossref Full Text | Google Scholar

Guo, S., Nighot, M., Al-Sadi, R., Alhmoud, T., Nighot, P., and Ma, T. Y. (2015). Lipopolysaccharide regulation of intestinal tight junction permeability is mediated by TLR-4 signal transduction pathway activation of FAK and MyD88. J. Immunol. 195, 4999–5010. doi: 10.4049/jimmunol.1402598

PubMed Abstract | Crossref Full Text | Google Scholar

Haery, L., Deverman, B. E., Matho, K. S., Cetin, A., Woodard, K., Cepko, C., et al. (2019). Adeno-associated virus technologies and methods for targeted neuronal manipulation. Front. Neuroanat. 13, 93. doi: 10.3389/fnana.2019.00093

PubMed Abstract | Crossref Full Text | Google Scholar

Halliday, G. M., Holton, J. L., Revesz, T., and Dickson, D. W. (2011). Neuropathology underlying clinical variability in patients with synucleinopathies. Acta Neuropathol. 122, 187–204. doi: 10.1007/s00401-011-0852-9

PubMed Abstract | Crossref Full Text | Google Scholar

Hermonat, P. L., and Muzyczka, N. (1984). Use of adeno-associated virus as a mammalian DNA cloning vector: transduction of neomycin resistance into mammalian tissue culture cells. Proc. Natl. Acad. Sci. USA 81, 6466–6470. doi: 10.1073/pnas.81.20.6466

PubMed Abstract | Crossref Full Text | Google Scholar

Hilton, D., Stephens, M., Kirk, L., Edwards, P., Potter, R., Zajicek, J., et al. (2014). Accumulation of α-synuclein in the bowel of patients in the pre-clinical phase of Parkinson's disease. Acta Neuropathol. 127, 235–241. doi: 10.1007/s00401-013-1214-6

PubMed Abstract | Crossref Full Text | Google Scholar

Holmqvist, S., Chutna, O., Bousset, L., Aldrin-Kirk, P., Li, W., Björklund, T., et al. (2014). Direct evidence of Parkinson pathology spread from the gastrointestinal tract to the brain in rats. Acta Neuropathol. 128, 805–820. doi: 10.1007/s00401-014-1343-6

PubMed Abstract | Crossref Full Text | Google Scholar

Hou, Y., Li, X., Liu, C., Zhang, M., Zhang, X., Ge, S., et al. (2021). Neuroprotective effects of short-chain fatty acids in MPTP induced mice model of Parkinson's disease. Exp. Gerontol. 150:111376. doi: 10.1016/j.exger.2021.111376

PubMed Abstract | Crossref Full Text | Google Scholar

Houser, M. C., Chang, J., Factor, S. A., Molho, E. S., Zabetian, C. P., Hill-Burns, E. M., et al. (2018). Stool immune profiles evince gastrointestinal inflammation in Parkinson's disease. Mov. Disord. 33, 793–804. doi: 10.1002/mds.27326

PubMed Abstract | Crossref Full Text | Google Scholar

Huang, H., Xu, H., Luo, Q., He, J., Li, M., Chen, H., et al. (2019). Fecal microbiota transplantation to treat Parkinson’s disease with constipation: a case report. Medicine 98, 1–5. doi: 10.1097/MD.0000000000016163

Crossref Full Text | Google Scholar

Izco, M., Blesa, J., Verona, G., Cooper, J. M., and Alvarez-Erviti, L. (2021). Glial activation precedes alpha-synuclein pathology in a mouse model of Parkinson's disease. Neurosci. Res. 170, 330–340. doi: 10.1016/j.neures.2020.11.004

PubMed Abstract | Crossref Full Text | Google Scholar

Kakuda, K., Ikenaka, K., Kuma, A., Doi, J., Aguirre, C., Wang, N., et al. (2024). Lysophagy protects against propagation of α-synuclein aggregation through ruptured lysosomal vesicles. Proc. Natl. Acad. Sci. USA 121:e2312306120. doi: 10.1073/pnas.2312306120

PubMed Abstract | Crossref Full Text | Google Scholar

Karpowicz, R. J., Haney, C. M., Mihaila, T. S., Sandler, R. M., Petersson, E. J., and Lee, V. M. Y. (2017). Selective imaging of internalized proteopathic α-synuclein seeds in primary neurons reveals mechanistic insight into transmission of synucleinopathies. J Biol. Chem. 292, 13482–13497. doi: 10.1074/jbc.M117.780296

Crossref Full Text | Google Scholar

Kelly, L. P., Carvey, P. M., Keshavarzian, A., Shannon, K. M., Shaikh, M., Bakay, R. A. E., et al. (2014). Progression of intestinal permeability changes and alpha-synuclein expression in a mouse model of Parkinson’s disease. Mov. Disord. 29, 999–1009. doi: 10.1002/mds.25736

PubMed Abstract | Crossref Full Text | Google Scholar

Kim, R., Kim, H. J., Kim, A., Jang, M., Kim, A., Kim, Y., et al. (2018). Peripheral blood inflammatory markers in early Parkinson's disease. J. Clin. Neurosci. 58, 30–33. doi: 10.1016/j.jocn.2018.10.079

PubMed Abstract | Crossref Full Text | Google Scholar

Kim, R., Kim, H. J., Shin, J. H., Lee, C. Y., Jeon, S. H., and Jeon, B. (2022). Serum inflammatory markers and progression of nonmotor symptoms in early Parkinson's disease. Mov. Disord. 37, 1535–1541. doi: 10.1002/mds.29056

PubMed Abstract | Crossref Full Text | Google Scholar

Kim, S., Kwon, S. H., Kam, T. I., Panicker, N., Karuppagounder, S. S., Lee, S., et al. (2019). Transneuronal propagation of pathologic α-synuclein from the gut to the brain models Parkinson’s disease. Neuron 103, 627–641.e7. doi: 10.1016/j.neuron.2019.05.035

PubMed Abstract | Crossref Full Text | Google Scholar

Kim, C., Lv, G., Lee, J. S., Jung, B. C., Masuda-Suzukake, M., Hong, C. S., et al. (2016). Exposure to bacterial endotoxin generates a distinct strain of α-synuclein fibril. Sci. Rep. 6:30891. doi: 10.1038/srep30891

PubMed Abstract | Crossref Full Text | Google Scholar

Klingberg, E., Magnusson, M. K., Strid, H., Deminger, A., Ståhl, A., Sundin, J., et al. (2019). A distinct gut microbiota composition in patients with ankylosing spondylitis is associated with increased levels of fecal calprotectin. Arthritis Res. Ther. 21:48. doi: 10.1186/s13075-019-2018-4

Crossref Full Text | Google Scholar

Konno, M., Hasegawa, T., Baba, T., Miura, E., Sugeno, N., Kikuchi, A., et al. (2012). Suppression of dynamin GTPase decreases α-synuclein uptake by neuronal and oligodendroglial cells: a potent therapeutic target for synucleinopathy. Mol. Neurodegener. 7:38. doi: 10.1186/1750-1326-7-38

PubMed Abstract | Crossref Full Text | Google Scholar

Kövari, E., Gold, G., Herrmann, F. R., Canuto, A., Hof, P. R., Bouras, C., et al. (2003). Lewy body densities in the entorhinal and anterior cingulate cortex predict cognitive deficits in Parkinson's disease. Acta Neuropathol. 106, 83–88. doi: 10.1007/s00401-003-0705-2

PubMed Abstract | Crossref Full Text | Google Scholar

Lau, A., So, R. W. L., Lau, H. H. C., Sang, J. C., Ruiz-Riquelme, A., Fleck, S. C., et al. (2020). α-Synuclein strains target distinct brain regions and cell types. Nat. Neurosci. 23, 21–31. doi: 10.1038/s41593-019-0541-x

PubMed Abstract | Crossref Full Text | Google Scholar

Lee, H. J., Suk, J. E., Bae, E. J., Lee, J. H., Paik, S. R., and Lee, S. J. (2008). Assembly-dependent endocytosis and clearance of extracellular alpha-synuclein. Int. J. Biochem. Cell Biol. 40, 1835–1849. doi: 10.1016/j.biocel.2008.01.017

PubMed Abstract | Crossref Full Text | Google Scholar

Li, Z., Liang, H., Hu, Y., Lu, L., Zheng, C., Fan, Y., et al. (2023). Gut bacterial profiles in Parkinson's disease: a systematic review. CNS Neurosci. Ther. 29, 140–157. doi: 10.1111/cns.13990

PubMed Abstract | Crossref Full Text | Google Scholar

Li, H. X., Zhang, C., Zhang, K., Liu, Y. Z., Peng, X. X., and Zong, Q. (2023). Inflammatory bowel disease and risk of Parkinson’s disease: evidence from a meta-analysis of 14 studies involving more than 13.4 million individuals. Front. Med. 10:1137366. doi: 10.3389/fmed.2023.1137366

PubMed Abstract | Crossref Full Text | Google Scholar

Liddelow, S. A., Guttenplan, K. A., Clarke, L. E., Bennett, F. C., Bohlen, C. J., Schirmer, L., et al. (2017). Neurotoxic reactive astrocytes are induced by activated microglia. Nature 541, 481–487. doi: 10.1038/nature21029

PubMed Abstract | Crossref Full Text | Google Scholar

Lima, I. S., Pêgo, A. C., Martins, A. C., Prada, A. R., Barros, J. T., Martins, G., et al. (2023). Gut dysbiosis: a target for protective interventions against Parkinson’s disease. Microorganisms 11:880. doi: 10.3390/microorganisms11040880

PubMed Abstract | Crossref Full Text | Google Scholar

Liu, B., Fang, F., Pedersen, N. L., Tillander, A., Ludvigsson, J. F., Ekbom, A., et al. (2017). Vagotomy and Parkinson disease: a Swedish register-based matched-cohort study. Neurology 88, 1996–2002. doi: 10.1212/WNL.0000000000003961

Crossref Full Text | Google Scholar

Liu, J., Lv, X., Ye, T., Zhao, M., Chen, Z., Zhang, Y., et al. (2024). Microbiota-microglia crosstalk between Blautia producta and neuroinflammation of Parkinson's disease: a bench-to-bedside translational approach. Brain Behav. Immun. 117, 270–282. doi: 10.1016/j.bbi.2024.01.010

PubMed Abstract | Crossref Full Text | Google Scholar

Lopes, D. M., Llewellyn, S. K., and Harrison, I. F. (2022). Propagation of tau and α-synuclein in the brain: therapeutic potential of the glymphatic system. Transl. Neurodegeneration 11, 1–23. doi: 10.1186/s40035-022-00293-2

Crossref Full Text | Google Scholar

Lundmark, K., Westermark, G. T., Olsén, A., and Westermark, P. (2005). Protein fibrils in nature can enhance amyloid protein a amyloidosis in mice: cross-seeding as a disease mechanism. Proc. Natl. Acad. Sci. USA 102, 6098–6102. doi: 10.1073/pnas.0501814102

PubMed Abstract | Crossref Full Text | Google Scholar

Lurette, O., Martín-Jiménez, R., Khan, M., Sheta, R., Jean, S., Schofield, M., et al. (2023). Aggregation of alpha-synuclein disrupts mitochondrial metabolism and induce mitophagy via cardiolipin externalization. Cell Death Disease 14, 1–14. doi: 10.1038/s41419-023-06251-8

Crossref Full Text | Google Scholar

Menon, S., Kofoed, R. H., Nabbouh, F., Xhima, K., Al-Fahoum, Y., Langman, T., et al. (2021). Viral alpha-synuclein knockdown prevents spreading synucleinopathy. Brain Communications 3:247. doi: 10.1093/braincomms/fcab247

PubMed Abstract | Crossref Full Text | Google Scholar

Metzdorf, J., and Tönges, L. (2021). Short-chain fatty acids in the context of Parkinson’s disease. Neural Reg. Res. 16:2015.

Google Scholar

Miller, A. L., Bessho, S., Grando, K., and Tükel, Ç. (2021). Microbiome or infections: amyloid-containing biofilms as a trigger for complex human diseases. Front. Immunol. 12:638867. doi: 10.3389/fimmu.2021.638867

PubMed Abstract | Crossref Full Text | Google Scholar

Mingozzi, F., and High, K. A. (2013). Immune responses to AAV vectors: overcoming barriers to successful gene therapy. Blood 122, 23–36. doi: 10.1182/blood-2013-01-306647

PubMed Abstract | Crossref Full Text | Google Scholar

Mogi, M., Harada, M., Kondo, T., Riederer, P., Inagaki, H., Minami, M., et al. (1994). Interleukin-1β, interleukin-6, epidermal growth factor and transforming growth factor-α are elevated in the brain from parkinsonian patients. Neuro Lett 180, 147–150.

Google Scholar

Mori, F., Nishie, M., Kakita, A., Yoshimoto, M., Takahashi, H., and Wakabayashi, K. (2006). Relationship among α-synuclein accumulation, dopamine synthesis, and neurodegeneration in Parkinson disease substantia nigra. J. Neuropathol. Exp. Neurol. 65, 808–815. doi: 10.1097/01.jnen.0000230520.47768.1a

PubMed Abstract | Crossref Full Text | Google Scholar

Morris, M. C., Gilliam, E. A., and Li, L. (2015). Innate immune programing by endotoxin and its pathological consequences. Front. Immunol. 6:680. doi: 10.3389/fimmu.2014.00680

Crossref Full Text | Google Scholar

Mu, L., Sobotka, S., Chen, J., Su, H., Sanders, I., Adler, C. H., et al. (2013). Alpha-synuclein pathology and axonal degeneration of the peripheral motor nerves innervating pharyngeal muscles in Parkinson disease. J. Neurol. Exp. 72, 119–129. doi: 10.1097/NEN.0b013e3182801cde

Crossref Full Text | Google Scholar

Mulak, A., Koszewicz, M., Panek-Jeziorna, M., Koziorowska-Gawron, E., and Budrewicz, S. (2019). Fecal calprotectin as a marker of the gut immune system activation is elevated in Parkinson's disease. Front. Neurosci. 13:992. doi: 10.3389/fnins.2019.00992

PubMed Abstract | Crossref Full Text | Google Scholar

Pang, S. Y. Y., Ho, P. W. L., Liu, H. F., Leung, C. T., Li, L., Chang, E. E. S., et al. (2019). The interplay of aging, genetics and environmental factors in the pathogenesis of Parkinson's disease. Transl. Neurodegener. 8:23. doi: 10.1186/s40035-019-0165-9

PubMed Abstract | Crossref Full Text | Google Scholar

Parkkinen, L., Pirttilä, T., and Alafuzoff, I. (2008). Applicability of current staging/categorization of α-synuclein pathology and their clinical relevance. Acta Neuropathol. 115, 399–407. doi: 10.1007/s00401-008-0346-6

PubMed Abstract | Crossref Full Text | Google Scholar

Parkkinen, L., Pirttilä, T., Tervahauta, M., and Alafuzoff, I. (2005). Widespread and abundant alpha-synuclein pathology in a neurologically unimpaired subject 25, 304–314. doi: 10.1111/j.1440-1789.2005.00644.x

Crossref Full Text | Google Scholar

Peelaerts, W., Bousset, L., Van Der Perren, A., Moskalyuk, A., Pulizzi, R., Giugliano, M., et al. (2015). α-Synuclein strains cause distinct synucleinopathies after local and systemic administration. Nature 522, 340–344. doi: 10.1038/nature14547

PubMed Abstract | Crossref Full Text | Google Scholar

Perez-Pardo, P., Dodiya, H. B., Engen, P. A., Forsyth, C. B., Huschens, A. M., Shaikh, M., et al. (2019). Role of TLR4 in the gut-brain axis in Parkinson’s disease: a translational study from men to mice 68, 829–843. doi: 10.1136/gutjnl-2018-316844

Crossref Full Text | Google Scholar

Perry, V. H., Cunningham, C., and Holmes, C. (2007). Systemic infections and inflammation affect chronic neurodegeneration. Nat. Rev. Immunol. 7, 161–167. doi: 10.1038/nri2015

PubMed Abstract | Crossref Full Text | Google Scholar

Perry, V. H., Matyszak, M. K., and Fearn, S. (1993). Altered antigen expression of microglia in the aged rodent CNS. Glia 7, 60–67. doi: 10.1002/glia.440070111

PubMed Abstract | Crossref Full Text | Google Scholar

Peter, I., Dubinsky, M., Bressman, S., Park, A., Lu, C., Chen, N., et al. (2018). Anti-tumor necrosis factor therapy and incidence of Parkinson disease among patients with inflammatory bowel disease. JAMA Neurol. 75, 939–946. doi: 10.1001/jamaneurol.2018.0605

PubMed Abstract | Crossref Full Text | Google Scholar

Popova, B., Galka, D., Häffner, N., Wang, D., Schmitt, K., Valerius, O., et al. (2021). Α-synuclein decreases the abundance of proteasome subunits and alters ubiquitin conjugates in yeast. Cells 10:229. doi: 10.3390/cells10092229

PubMed Abstract | Crossref Full Text | Google Scholar

Postuma, R. B., and Berg, D. (2016). Advances in markers of prodromal Parkinson disease. Nat. Rev. Neurol. 12, 622–634. doi: 10.1038/nrneurol.2016.152

PubMed Abstract | Crossref Full Text | Google Scholar

Qin, L., Wu, X., Block, M. L., Liu, Y., Breese, G. R., Hong, J. S., et al. (2007). Systemic LPS causes chronic neuroinflammation and progressive neurodegeneration. Glia 55:453. doi: 10.1002/glia.20467

Crossref Full Text | Google Scholar

Rahayel, S., Mišić, B., Zheng, Y. Q., Liu, Z. Q., Abdelgawad, A., Abbasi, N., et al. (2022). Differentially targeted seeding reveals unique pathological alpha-synuclein propagation patterns. Brain 145, 1743–1756. doi: 10.1093/brain/awab440

PubMed Abstract | Crossref Full Text | Google Scholar

Ramezani, M., Wagenknecht-Wiesner, A., Wang, T., Holowka, D. A., Eliezer, D., and Baird, B. A. (2023). Alpha synuclein modulates mitochondrial Ca2+ uptake from ER during cell stimulation and under stress conditions. NPJ Parkinson's Disease 9, 1–18. doi: 10.1038/s41531-023-00578-x

Crossref Full Text | Google Scholar

Ravindra Kumar, S., Miles, T. F., Chen, X., Brown, D., Dobreva, T., Huang, Q., et al. (2020). Multiplexed Cre-dependent selection yields systemic AAVs for targeting distinct brain cell types. Nat. Methods 17, 541–550. doi: 10.1038/s41592-020-0799-7

PubMed Abstract | Crossref Full Text | Google Scholar

Ravva, S. V., Sarreal, C. Z., and Cooley, M. B. (2016). Expression of curli by Escherichia coli O157: H7 strains isolated from patients during outbreaks is different from similar strains isolated from leafy green production environments. Front. Cell. Infect. Microbiol. 6:189. doi: 10.3389/fcimb.2016.00189

PubMed Abstract | Crossref Full Text | Google Scholar

Rite, I., Machado, A., Cano, J., and Venero, J. L. (2007). Blood–brain barrier disruption induces in vivo degeneration of nigral dopaminergic neurons. J. Neurochem. 101, 1567–1582.

Google Scholar

Royo, N. C., Vandenberghe, L. H., Ma, J. Y., Hauspurg, A., Yu, L., Maronski, M., et al. (2008). Specific AAV serotypes stably transduce primary hippocampal and cortical cultures with high efficiency and low toxicity. Brain Res. 1190, 15–22. doi: 10.1016/j.brainres.2007.11.015

PubMed Abstract | Crossref Full Text | Google Scholar

Russo, T., and Riessland, M. (2022). Age-related midbrain inflammation and senescence in Parkinson’s disease. Front. Aging Neurosci. 14:917797. doi: 10.3389/fnagi.2022.917797

PubMed Abstract | Crossref Full Text | Google Scholar

Saija, A., Princi, P., Lanza, M., Scalese, M., Aramnejad, E., and Sarro, A. D. (1995). Systemic cytokine administration can affect blood-brain barrier permeability in the rat. Life Sci. 56, 775–784. doi: 10.1016/0024-3205(95)00008-t

PubMed Abstract | Crossref Full Text | Google Scholar

Sampson, T. R., Debelius, J. W., Thron, T., Janssen, S., Shastri, G. G., Ilhan, Z. E., et al. (2016). Gut microbiota regulate motor deficits and neuroinflammation in a model of Parkinson's disease 167, 1469–1480. doi: 10.1016/j.cell.2016.11.018

Crossref Full Text | Google Scholar

Samuel, F., Flavin, W. P., Iqbal, S., Pacelli, C., Sri Renganathan, S. D., Trudeau, L.-E., et al. (2016). Effects of serine 129 phosphorylation on α-Synuclein aggregation, membrane association, and internalization. J. Biol. Chem. 291, 4374–4385. doi: 10.1074/jbc.M115.705095

PubMed Abstract | Crossref Full Text | Google Scholar

Senol, A. D., Samarani, M., Syan, S., Guardia, C. M., Nonaka, T., Liv, N., et al. (2021). α-Synuclein fibrils subvert lysosome structure and function for the propagation of protein misfolding between cells through tunneling nanotubes. PLoS Biol. 19:e3001287. doi: 10.1371/journal.pbio.3001287

PubMed Abstract | Crossref Full Text | Google Scholar

Sheffield, L. G., and Berman, N. E. J. (1998). Microglial expression of MHC class II increases in normal aging of nonhuman primates. Neuro Biol. Aging 19, 47–55.

Google Scholar

Shukla, A., Dikshit, M., and Srimal, R. C. (1995). Nitric oxide modulates blood-brain barrier permeability during infections with an inactivated bacterium. Neuroreport 6, 1629–1632. doi: 10.1097/00001756-199508000-00010

PubMed Abstract | Crossref Full Text | Google Scholar

Sidhu, A., Wersinger, C., and Vernier, P. (2004). Does α-synuclein modulate dopaminergic synaptic content and tone at the synapse? FASEB J. 18, 637–647. doi: 10.1096/fj.03-1112rev

PubMed Abstract | Crossref Full Text | Google Scholar

So, R. W. L., Amano, G., Stuart, E., Amini, A. E., Aguzzi, A., Collingridge, G. L., et al. (2024). α-Synuclein strain propagation is independent of cellular prion protein expression in a transgenic synucleinopathy mouse model. PLoS Pathog. 20:1012517. doi: 10.1371/journal.ppat.1012517

PubMed Abstract | Crossref Full Text | Google Scholar

Sonne, J., Reddy, V., and Beato, M. R. (2022). Neuroanatomy, substantia Nigra. Tampa/St. Petersburg, FL: StatPearls Publishing.

Google Scholar

Sun, M. F., Zhu, Y. L., Zhou, Z. L., Jia, X. B., Xu, Y. D., Yang, Q., et al. (2018). Neuroprotective effects of fecal microbiota transplantation on MPTP-induced Parkinson's disease mice: gut microbiota, glial reaction and TLR4/TNF-α signaling pathway. Brain Behav. Immun. 70, 48–60. doi: 10.1016/j.bbi.2018.02.005

PubMed Abstract | Crossref Full Text | Google Scholar

Sung, J. Y., Kim, J., Paik, S. R., Park, J. H., Ahn, Y. S., and Chung, K. C. (2001). Induction of neuronal cell death by Rab5A-dependent endocytosis of alpha-synuclein. J Biol. Chem. 276, 27441–27448. doi: 10.1074/jbc.M101318200

Crossref Full Text | Google Scholar

Sung, H.-Y., Park, J.-W., and Kim, J.-S. (2014). The frequency and severity of gastrointestinal symptoms in patients with early Parkinson’s disease. J. Movem. Disorders 7:7. doi: 10.14802/jmd.14002

PubMed Abstract | Crossref Full Text | Google Scholar

Svensson, E., Horváth-Puhó, E., Thomsen, R. W., Djurhuus, J. C., Pedersen, L., Borghammer, P., et al. (2015). Vagotomy and subsequent risk of Parkinson's disease. Ann. Neurol. 78, 522–529. doi: 10.1002/ana.24448

PubMed Abstract | Crossref Full Text | Google Scholar

Tam, J. S. Y., Coller, J. K., Hughes, P. A., Prestidge, C. A., and Bowen, J. M. (2021). Toll-like receptor 4 (TLR4) antagonists as potential therapeutics for intestinal inflammation 40:5. doi: 10.1007/s12664-020-01114-y

Crossref Full Text | Google Scholar

Tan, A. H., Chong, C. W., Lim, S. Y., Yap, I. K. S., Teh, C. S. J., Loke, M. F., et al. (2021). Gut microbial ecosystem in Parkinson disease: New Clinicobiological insights from multi-omics. New York, NY: John Wiley & Sons, Ltd, 546–559.

Google Scholar

Tan, A. H., Lim, S. Y., and Lang, A. E. (2022). The microbiome-gut-brain axis in Parkinson disease - from basic research to the clinic. Nat. Rev. Neurol. 18, 476–495. doi: 10.1038/s41582-022-00681-2

PubMed Abstract | Crossref Full Text | Google Scholar

Tarutani, A., Suzuki, G., Shimozawa, A., Nonaka, T., Akiyama, H., Hisanaga, S. I., et al. (2016). The effect of fragmented pathogenic α-synuclein seeds on prion-like propagation. J. Biol. Chem. 291, 18675–18688. doi: 10.1074/jbc.M116.734707

PubMed Abstract | Crossref Full Text | Google Scholar

Thevaranjan, N., Puchta, A., Schulz, C., Naidoo, A., Szamosi, J. C., Verschoor, C. P., et al. (2017). Age-associated microbial dysbiosis promotes intestinal permeability, systemic inflammation, and macrophage dysfunction 21, 455–466. doi: 10.1016/j.chom.2017.03.002

Crossref Full Text | Google Scholar

Thévenot, E., Jordão, J. F., O'Reilly, M. A., Markham, K., Weng, Y. Q., Foust, K. D., et al. (2012). Targeted delivery of self-complementary adeno-associated virus serotype 9 to the brain, using magnetic resonance imaging-guided focused ultrasound. Hum. Gene Ther. 23, 1144–1155. doi: 10.1089/hum.2012.013

PubMed Abstract | Crossref Full Text | Google Scholar

Umemura, A., Oeda, T., Tomita, S., Hayashi, R., Kohsaka, M., Park, K., et al. (2014). Delirium and high fever are associated with subacute motor deterioration in Parkinson disease: a nested case-control study. PLoS One 9:944. doi: 10.1371/journal.pone.0094944

PubMed Abstract | Crossref Full Text | Google Scholar

Unger, M. M., Spiegel, J., Dillmann, K. U., Grundmann, D., Philippeit, H., Bürmann, J., et al. (2016). Short chain fatty acids and gut microbiota differ between patients with Parkinson's disease and age-matched controls. Parkinsonism Relat. Disord. 32, 66–72. doi: 10.1016/j.parkreldis.2016.08.019

PubMed Abstract | Crossref Full Text | Google Scholar

Van Den Berge, N., Ferreira, N., Gram, H., Mikkelsen, T. W., Alstrup, A. K. O., Casadei, N., et al. (2019). Evidence for bidirectional and trans-synaptic parasympathetic and sympathetic propagation of alpha-synuclein in rats. Acta Neuropathol. 138, 535–550. doi: 10.1007/s00401-019-02040-w

PubMed Abstract | Crossref Full Text | Google Scholar

Van Den Berge, N., Ferreira, N., Mikkelsen, T. W., Olsen Alstrup, A. K., Tamgüney, G., Karlsson, P., et al. (2021). Ageing promotes pathological alpha-synuclein propagation and autonomic dysfunction in wild-type rats. Brain 144, 1853–1868. doi: 10.1093/brain/awab061

PubMed Abstract | Crossref Full Text | Google Scholar

Varkey, J., Isas, J. M., Mizuno, N., Jensen, M. B., Bhatia, V. K., Jao, C. C., et al. (2010). Membrane curvature induction and tubulation are common features of synucleins and apolipoproteins. J. Biol. Chem. 285, 32486–32493. doi: 10.1074/jbc.M110.139576

PubMed Abstract | Crossref Full Text | Google Scholar

Veszelka, S., Pásztói, M., Farkas, A. E., Krizbai, I., Dung, N. T. K., Niwa, M., et al. (2007). Pentosan polysulfate protects brain endothelial cells against bacterial lipopolysaccharide-induced damages 50, 219–228. doi: 10.1016/j.neuint.2006.08.006

Crossref Full Text | Google Scholar

Villarán, R. F., Espinosa-Oliva, A. M., Sarmiento, M., De Pablos, R. M., Argüelles, S., Delgado-Cortés, M. J., et al. (2010). Ulcerative colitis exacerbates lipopolysaccharide-induced damage to the nigral dopaminergic system: potential risk factor in Parkinson’s disease. J. Neurochem. 114, 1687–1700. doi: 10.1111/j.1471-4159.2010.06879.x

PubMed Abstract | Crossref Full Text | Google Scholar

Visanji, N. P., Marras, C., Kern, D. S., Al Dakheel, A., Gao, A., Liu, L. W. C., et al. (2015). Colonic mucosal a-synuclein lacks specificity as a biomarker for Parkinson disease. Neurology 84, 609–616. doi: 10.1212/WNL.0000000000001240

PubMed Abstract | Crossref Full Text | Google Scholar

Volles, M. J., and Lansbury, P. T. (2002). Vesicle permeabilization by protofibrillar α-synuclein is sensitive to Parkinson's disease-linked mutations and occurs by a pore-like mechanism 41, 4595–4602. doi: 10.1021/bi0121353

Crossref Full Text | Google Scholar

Wada, K., Arai, H., Takanashi, M., Fukae, J., Oizumi, H., Yasuda, T., et al. (2006). Expression levels of vascular endothelial growth factor and its receptors in Parkinson's disease. Neuroreport 17, 705–709. doi: 10.1097/01.wnr.0000215769.71657.65

PubMed Abstract | Crossref Full Text | Google Scholar

Wakabayashi, K., Mori, F., and Takahashi, H. (2006). Progression patterns of neuronal loss and Lewy body pathology in the substantia nigra in Parkinson's disease. Parkinsonism Relat. Disord. 12, S92–S98. doi: 10.1016/j.parkreldis.2006.05.028

Crossref Full Text | Google Scholar

Wakabayashi, K., Takahashi, H., Ohama, E., and Ikuta, F. (1990). Parkinson's disease: an immunohistochemical study of Lewy body-containing neurons in the enteric nervous system. Acta Neuropathol. 79, 581–583. doi: 10.1007/BF00294234

PubMed Abstract | Crossref Full Text | Google Scholar

Wakabayashi, K., Takahashi, H., Takeda, S., Ohama, E., and Ikuta, F. (1988). Parkinson's disease: the presence of Lewy bodies in Auerbach's and Meissner's plexuses. Acta Neuropathol. 76, 217–221. doi: 10.1007/BF00687767

PubMed Abstract | Crossref Full Text | Google Scholar

Wakabayashi, K., Takahashi, H., Takeda, S., Ohama, E., and Ikuta, F. (1989). Lewy bodies in the enteric nervous system in Parkinson's disease. Arch. Histol. Cytol. 52, 191–194.

Google Scholar

Wang, T., and Hay, J. C. (2015). Alpha-synuclein toxicity in the early secretory pathway: how it drives neurodegeneration in parkinsons disease. Front. Neurosci. 9:167161. doi: 10.3389/fnins.2015.00433

Crossref Full Text | Google Scholar

Wong, Y. Y., Wu, C. Y., Yu, D., Kim, E., Wong, M., Elez, R., et al. (2022). Biofluid markers of blood-brain barrier disruption and neurodegeneration in Lewy body spectrum diseases: a systematic review and meta-analysis. Parkinsonism Relat. Disord. 101, 119–128. doi: 10.1016/j.parkreldis.2022.06.004

PubMed Abstract | Crossref Full Text | Google Scholar

Wu, X. F., Block, M. L., Zhang, W., Qin, L., Wilson, B., Zhang, W. Q., et al. (2005). The role of microglia in paraquat-induced dopaminergic neurotoxicity. Antioxid. Redox Signal. 7, 654–661. doi: 10.1089/ars.2005.7.654

PubMed Abstract | Crossref Full Text | Google Scholar

Wu, G., Jiang, Z., Pu, Y., Chen, S., Wang, T., Wang, Y., et al. (2022). Serum short-chain fatty acids and its correlation with motor and non-motor symptoms in Parkinson’s disease patients. BioMed Central Ltd 22, 1–9.

Google Scholar

Xhima, K., Nabbouh, F., Hynynen, K., Aubert, I., and Tandon, A. (2018). Noninvasive delivery of an α-synuclein gene silencing vector with magnetic resonance–guided focused ultrasound. Mov. Disord. 33, 1567–1579. doi: 10.1002/mds.101

PubMed Abstract | Crossref Full Text | Google Scholar

Xue, L. J., Yang, X. Z., Tong, Q., Shen, P., Ma, S. J., Wu, S. N., et al. (2020). Fecal microbiota transplantation therapy for Parkinson's disease: a preliminary study. Medicine 99:e22035. doi: 10.1097/MD.0000000000022035

Crossref Full Text | Google Scholar

Yacoubian, T. A., Fang, Y. H. D., Gerstenecker, A., Amara, A., Stover, N., Ruffrage, L., et al. (2023). Brain and systemic inflammation in de novo Parkinson's disease 38, 743–754. doi: 10.1002/mds.29363

Crossref Full Text | Google Scholar

Yamada, K., and Iwatsubo, T. (2018). Extracellular α-synuclein levels are regulated by neuronal activity. Mol. Neurodegener. 13:9. doi: 10.1186/s13024-018-0241-0

PubMed Abstract | Crossref Full Text | Google Scholar

Yamazaki, Y., Baker, D. J., Tachibana, M., Liu, C. C., Van Deursen, J. M., Brott, T. G., et al. (2016). Vascular cell senescence contributes to blood-brain barrier breakdown. Stroke 47, 1068–1077. doi: 10.1161/STROKEAHA.115.010835

PubMed Abstract | Crossref Full Text | Google Scholar

Yang, W., Hamilton, J. L., Kopil, C., Beck, J. C., Tanner, C. M., Albin, R. L., et al. (2020). Current and projected future economic burden of Parkinson’s disease in the U.S. NPJ Parkinsons Dis. 6:117. doi: 10.1038/s41531-020-0117-1

PubMed Abstract | Crossref Full Text | Google Scholar

Yu, Q. J., Yu, S. Y., Zuo, L. J., Lian, T. H., Hu, Y., Wang, R. D., et al. (2018). Parkinson disease with constipation: clinical features and relevant factors. Sci. Rep. 8:16790. doi: 10.1038/s41598-017-16790-8

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, X., Tang, B., and Guo, J. (2023). Parkinson’s disease and gut microbiota: from clinical to mechanistic and therapeutic studies. Transl. Neurodegener. 12, 1–30. doi: 10.1186/s40035-023-00392-8

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, W., Wang, T., Pei, Z., Miller, D. S., Wu, X., Block, M. L., et al. (2005). Aggregated α-synuclein activates microglia: a process leading to disease progression in Parkinson's disease. New York, NY: John Wiley & Sons, Ltd, 533–542. doi: 10.1096/fj.04-2751com

Crossref Full Text | Google Scholar

Zhang, Y., Xu, S., Qian, Y., He, X., Mo, C., Yang, X., et al. (2022). Sodium butyrate attenuates rotenone-induced toxicity by activation of autophagy through epigenetically regulating PGC-1α expression in PC12 cells. Brain Res. 1776:147749. doi: 10.1016/j.brainres.2021.147749

PubMed Abstract | Crossref Full Text | Google Scholar

Zhao, Y., Walker, D. I., Lill, C. M., Bloem, B. R., Darweesh, S. K. L., Pinto-Pacheco, B., et al. (2023). Lipopolysaccharide-binding protein and future Parkinson’s disease risk: a European prospective cohort. BioMed Central Ltd 22, 1–8. doi: 10.1186/s12974-023-02846-2

Crossref Full Text | Google Scholar

Zhu, Y., Yuan, M., Liu, Y., Yang, F., Chen, W. Z., Xu, Z. Z., et al. (2022). Association between inflammatory bowel diseases and Parkinson's disease: systematic review and meta-analysis. Neural Regen. Res. 17, 344–353. doi: 10.4103/1673-5374.317981

PubMed Abstract | Crossref Full Text | Google Scholar

Zincarelli, C., Soltys, S., Rengo, G., and Rabinowitz, J. E. (2008). Analysis of AAV serotypes 1-9 mediated gene expression and tropism in mice after systemic injection. Mol. Ther. 16, 1073–1080. doi: 10.1038/mt.2008.76

PubMed Abstract | Crossref Full Text | Google Scholar

Zondler, L., Kostka, M., Garidel, P., Heinzelmann, U., Hengerer, B., Mayer, B., et al. (2017). Proteasome impairment by α-synuclein. PLoS One 12:184040. doi: 10.1371/journal.pone.0184040

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: synucleinopathy, enteric neurons, aggregation, lewy bodies, gastrointestinal tract

Citation: Malekian Naeini S, Lopez MD, Fraser PE and Tandon A (2025) Parkinson’s disease beyond the brain: implications for treatments. Front. Aging Neurosci. 17:1600782. doi: 10.3389/fnagi.2025.1600782

Received: 26 March 2025; Accepted: 28 August 2025;
Published: 09 October 2025.

Edited by:

Jeng-Ren Duann, National Yang Ming Chiao Tung University, Taiwan

Reviewed by:

Carsten Theiss, Ruhr University Bochum, Germany
Georgia Minakaki, Northwestern University, United States

Copyright © 2025 Malekian Naeini, Lopez, Fraser and Tandon. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Anurag Tandon, YS50YW5kb25AdXRvcm9udG8uY2E=

These authors have contributed equally to this work

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.