4.8 Article

Fecal microbiota transplantation protects rotenone-induced Parkinson's disease mice via suppressing inflammation mediated by the lipopolysaccharide-TLR4 signaling pathway through the microbiota-gut-brain axis

Journal

MICROBIOME
Volume 9, Issue 1, Pages -

Publisher

BMC
DOI: 10.1186/s40168-021-01107-9

Keywords

Fecal microbiota transplantation; Parkinson's disease; Rotenone-induced mouse model; Microbiota-gut-brain axis; 16S RNA sequencing

Categories

Funding

  1. National Sciences Foundation of China [81773718, 81630097, 81773589]
  2. National Key Research and Development Program of China [SQ2018YFA090025-04]
  3. CAMS Innovation Fund for Medical Sciences (China) [2016-I2M-3-011]
  4. Drug Innovation Major Project [2018ZX09711001-003-020, 2018ZX09711001-003005, 2018ZX09711001-008-005]
  5. CAMS The Fundamental Research Funds for the Central Universities (China) [2018RC350002]
  6. CAMS & PUMC Innovation Fund for Graduate (China) [2019-1007-23]

Ask authors/readers for more resources

This study demonstrates that FMT treatment can correct gut microbiota dysbiosis and ameliorate the rotenone-induced PD mouse model, possibly playing a significant role in suppressing inflammation via the LPS-TLR4 signaling pathway in both the gut and the brain. Furthermore, the study proves the involvement of rotenone-induced microbiota dysbiosis in the genesis of PD through the microbiota-gut-brain axis.
Background: Parkinson's disease (PD) is a prevalent neurodegenerative disorder, displaying not only well-known motor deficits but also gastrointestinal dysfunctions. Consistently, it has been increasingly evident that gut microbiota affects the communication between the gut and the brain in PD pathogenesis, known as the microbiota-gut-brain axis. As an approach to re-establishing a normal microbiota community, fecal microbiota transplantation (FMT) has exerted beneficial effects on PD in recent studies. Here, in this study, we established a chronic rotenone-induced PD mouse model to evaluate the protective effects of FMT treatment on PD and to explore the underlying mechanisms, which also proves the involvement of gut microbiota dysbiosis in PD pathogenesis via the microbiota-gut-brain axis. Results: We demonstrated that gut microbiota dysbiosis induced by rotenone administration caused gastrointestinal function impairment and poor behavioral performances in the PD mice. Moreover, 16S RNA sequencing identified the increase of bacterial genera Akkermansia and Desulfovibrio in fecal samples of rotenone-induced mice. By contrast, FMT treatment remarkably restored the gut microbial community, thus ameliorating the gastrointestinal dysfunctions and the motor deficits of the PD mice. Further experiments revealed that FMT administration alleviated intestinal inflammation and barrier destruction, thus reducing the levels of systemic inflammation. Subsequently, FMT treatment attenuated blood-brain barrier (BBB) impairment and suppressed neuroinflammation in the substantia nigra (SN), which further decreased the damage of dopaminergic neurons. Additional mechanistic investigation discovered that FMT treatment reduced lipopolysaccharide (LPS) levels in the colon, the serum, and the SN, thereafter suppressing the TLR4/MyD88/NF-kappa B signaling pathway and its downstream pro-inflammatory products both in the SN and the colon. Conclusions: Our current study demonstrates that FMT treatment can correct the gut microbiota dysbiosis and ameliorate the rotenone-induced PD mouse model, in which suppression of the inflammation mediated by the LPS-TLR4 signaling pathway both in the gut and the brain possibly plays a significant role. Further, we prove that rotenone-induced microbiota dysbiosis is involved in the genesis of PD via the microbiota-gut-brain axis.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available