1
|
Ren S, Wang J, Wang Y, Luo Q, Pu W, Meng X, Liu S. Oxidative stress and type 2 diabetes: a review of lactic acid bacteria as potential prophylactic and therapeutic interventions. Food Sci Biotechnol 2025; 34:2403-2416. [PMID: 40492034 PMCID: PMC12145373 DOI: 10.1007/s10068-024-01775-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 11/18/2024] [Accepted: 11/25/2024] [Indexed: 06/11/2025] Open
Abstract
Oxidative stress, which results from the overproduction of reactive oxygen species (ROS) that induce protein, lipid, and DNA oxidation, has emerged as a key factor in the pathogenesis of various diseases, including type 2 diabetes (T2D). Recently, the relationship between oxidative stress and T2D has gained considerable attention. Widely utilized as probiotics in fermented foods and beverages, lactic acid bacteria (LAB) exhibit potent antioxidant properties. However, the precise mechanisms enabling LAB to behave as antioxidants remain elusive. LAB play a pivotal role in promoting and maintaining host health while mitigating the development and progression of various disorders, including T2D. Against the backdrop of a large number of studies highlighting the beneficial role of LAB in mitigating oxidative stress-related diseases, this review explores potential biomarkers for the prevention of oxidative stress and examines the potential contribution of LAB to the fight against T2D.
Collapse
Affiliation(s)
- Siheng Ren
- Institute of Public Health, Chengdu University of Traditional Chinese Medicine, No.1166, Liutai Avenue, Wenjiang District, Chengdu, 611137 Sichuan China
| | - Juan Wang
- Institute of Public Health, Chengdu University of Traditional Chinese Medicine, No.1166, Liutai Avenue, Wenjiang District, Chengdu, 611137 Sichuan China
| | - Yuxi Wang
- Institute of Public Health, Chengdu University of Traditional Chinese Medicine, No.1166, Liutai Avenue, Wenjiang District, Chengdu, 611137 Sichuan China
| | - Qian Luo
- Institute of Public Health, Chengdu University of Traditional Chinese Medicine, No.1166, Liutai Avenue, Wenjiang District, Chengdu, 611137 Sichuan China
| | - Wei Pu
- Institute of Public Health, Chengdu University of Traditional Chinese Medicine, No.1166, Liutai Avenue, Wenjiang District, Chengdu, 611137 Sichuan China
| | - Xiao Meng
- Institute of Public Health, Chengdu University of Traditional Chinese Medicine, No.1166, Liutai Avenue, Wenjiang District, Chengdu, 611137 Sichuan China
| | - Shukun Liu
- Institute of Public Health, Chengdu University of Traditional Chinese Medicine, No.1166, Liutai Avenue, Wenjiang District, Chengdu, 611137 Sichuan China
| |
Collapse
|
2
|
Renesteen E, Boyajian JL, Islam P, Kassab A, Abosalha A, Makhlouf S, Santos M, Chen H, Shum-Tim C, Prakash S. Microbiome Engineering for Biotherapeutic in Alzheimer's Disease Through the Gut-Brain Axis: Potentials and Limitations. Int J Mol Sci 2025; 26:5351. [PMID: 40508160 PMCID: PMC12154405 DOI: 10.3390/ijms26115351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2025] [Revised: 05/21/2025] [Accepted: 05/27/2025] [Indexed: 06/16/2025] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative condition characterized by considerable cognitive decline and functional impairment, primarily due to the progressive alteration of neurons, microglia, and astrocytes. Pathological manifestations of AD include the loss of synaptic plasticity, reduction in synaptic strength by amyloid-beta, aggregation, and neurotoxicity from tau protein post-translational modifications, all contributing to the disruption of neural networks. Despite its current pharmacological treatment for AD, different approaches to treat such disease are being developed, from a microbiome perspective. The microbiome encompasses a diverse microorganism, including beneficial bacteria that create a positive impact to diminish AD pathogenesis. Growing evidence suggests that probiotic, prebiotic, synbiotic, and postbiotics can positively modulate the gut-brain axis, reducing systemic inflammation, restoring neurotransmitter balance, and improving gut health, thereby possibly mitigating AD pathogenesis. Moreover, there is paraprobiotics as the most recently developed biotherapeutic with beneficial effects. This review explores the correlation between AD and gut-brain axis as a novel biotherapeutic target. The underlying mechanism of the microbiota-gut-brain axis in AD is examined. Novel insights into the current applications as potential treatment and its limitations are highlighted.
Collapse
Affiliation(s)
- Editha Renesteen
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC H3A 2B4, Canada; (E.R.); (J.L.B.); (A.K.); (S.M.)
| | - Jacqueline L. Boyajian
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC H3A 2B4, Canada; (E.R.); (J.L.B.); (A.K.); (S.M.)
| | - Paromita Islam
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC H3A 2B4, Canada; (E.R.); (J.L.B.); (A.K.); (S.M.)
| | - Amal Kassab
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC H3A 2B4, Canada; (E.R.); (J.L.B.); (A.K.); (S.M.)
| | - Ahmed Abosalha
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC H3A 2B4, Canada; (E.R.); (J.L.B.); (A.K.); (S.M.)
- Pharmaceutical Technology Department, Faculty of Pharmacy, Tanta University, Tanta Al-Geish St., The Medical Campus, Tanta 31527, Egypt
| | - Stephanie Makhlouf
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC H3A 2B4, Canada; (E.R.); (J.L.B.); (A.K.); (S.M.)
| | - Madison Santos
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC H3A 2B4, Canada; (E.R.); (J.L.B.); (A.K.); (S.M.)
| | - Hongmei Chen
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC H3A 2B4, Canada; (E.R.); (J.L.B.); (A.K.); (S.M.)
| | - Cedrique Shum-Tim
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC H3A 2B4, Canada; (E.R.); (J.L.B.); (A.K.); (S.M.)
| | - Satya Prakash
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC H3A 2B4, Canada; (E.R.); (J.L.B.); (A.K.); (S.M.)
| |
Collapse
|
3
|
Shao X, Wu T, Li M, Zheng M, Lin H, Qi X. Enterococcus faecalis Exerts Neuroprotective Effects via the Vagus Nerve in a Mouse Model of Parkinson's Disease. Mol Neurobiol 2025; 62:7875-7891. [PMID: 39954164 DOI: 10.1007/s12035-025-04741-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 02/02/2025] [Indexed: 02/17/2025]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease worldwide. Current treatment methods for PD are unable to halt disease progression. The gut microbiota contributes to the neurodevelopment of PD; however, the gut-brain connections and underlying neural bases that regulate this complex behavior are not yet clear. Enterococcus faecalis (EF) is a common commensal bacterium of the gut and a common pathogen associated with hospital-acquired infections. Here, we demonstrated the significant therapeutic effects of a non-pathogenic strain of EF (EF ATCC19433) on PD. In this study, we established a mouse model of PD by intraperitoneal injection of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). We found that EF treatment alleviated behavioral impairment, dopaminergic neuronal loss, blood-brain barrier damage, and neuroinflammation induced by MPTP in the mice. Additionally, 16S rRNA sequencing revealed that dysbiosis of PD-related microbial communities induced by MPTP was reversed by EF treatment. Moreover, EF treatment relieved gastrointestinal dysfunction in the mice. The therapeutic efficacy of EF in MPTP-induced PD mice is markedly diminished when the activity of EF is lost. Further mechanistic studies indicated that the neuroprotective effects of EF in PD were associated with the vagus nerve pathway. Following the surgical severance of the vagus nerve through subdiaphragmatic vagotomy, the protective effects of EF on PD were markedly diminished. Our study suggests that EF can alleviate neurofunctional impairments and gastrointestinal disorders associated with PD, indicating that gut-derived microbes influence brain function through the vagus nerve pathway.
Collapse
Affiliation(s)
- Xian Shao
- Department of Medical Research Center, Shaoxing People's Hospital, Zhejiang University Shaoxing Hospital, Shaoxing, 312000, Zhejiang, China
| | - Tao Wu
- School of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang, China
| | - Mengyun Li
- Department of Medical Research Center, Shaoxing People's Hospital, Zhejiang University Shaoxing Hospital, Shaoxing, 312000, Zhejiang, China
| | - Matao Zheng
- Department of Neurosurgery, Shaoxing People's Hospital, Zhejiang University Shaoxing Hospital, Shaoxing, 312000, Zhejiang, China
| | - Hui Lin
- Healthy Science Center, The Affiliated Lihuili Hospital of Ningbo University, Ningbo University, Ningbo, 315211, Zhejiang, China.
| | - Xuchen Qi
- Department of Neurosurgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310020, Zhejiang, China.
| |
Collapse
|
4
|
Yang X, Yang Z, Wang Y, Zeng H, Wang B. Proteomics and metabolomics elucidate the biosynthetic pathway of acid stress-induced exopolysaccharides and its impact on growth phenotypes in Lactiplantibacillus plantarum HMX2. Food Chem 2025; 476:143431. [PMID: 39977986 DOI: 10.1016/j.foodchem.2025.143431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/07/2025] [Accepted: 02/13/2025] [Indexed: 02/22/2025]
Abstract
Lactiplantibacillus plantarum has been well acknowledged to produce exopolysaccharides (EPS) as a defense mechanism against acid stress. However, the complete biosynthetic pathway of EPS in L. plantarum and its impact on the cell growth and primary metabolism were still unclear. To fill these gaps, we carried out phenotypic, proteomic and metabolomics analysis of L. plantarum HMX2 cultured under different acidic conditions. Component and structure analysis showed that the repeating unit of EPS consisted of N-acetylmannosamine, N-acetylglucosamine, galactose, mannoses and glucoses. Multiomics analysis facilitated the curation and entablement of the complete EPS biosynthetic pathway ready for use in genome-scale metabolic models. Furthermore, proteomics and metabolomics data indicated that compared to the pH 6.5 condition, the acid stress at pH 4.5 significantly accelerated glycolysis and EPS biosynthesis processes while reduced the metabolic fluxes through the TCA cycle and the lactic acid fermentation, which suggested a trade-off between primary and secondary metabolism.
Collapse
Affiliation(s)
- Xinyu Yang
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Ministry of Education, Beijing 100048, China
| | - Zhijie Yang
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Ministry of Education, Beijing 100048, China
| | - Yanbo Wang
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Ministry of Education, Beijing 100048, China; Beijing Engineering and Technology Research Center of Food Additives, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Hong Zeng
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Ministry of Education, Beijing 100048, China; Beijing Engineering and Technology Research Center of Food Additives, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China.
| | - Bei Wang
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Ministry of Education, Beijing 100048, China; Beijing Engineering and Technology Research Center of Food Additives, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China.
| |
Collapse
|
5
|
Qi Y, Xie S, Chen J, Zhang C, Ma X, Yu Y, Yu X, Wang Y. Gut microbiota regulation by Lactiplantibacillus plantarum SG5 enhances mitochondrial function in Parkinson's disease mice via the GLP-1/PGC-1α pathway. J Nutr Biochem 2025:109954. [PMID: 40368220 DOI: 10.1016/j.jnutbio.2025.109954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 04/24/2025] [Accepted: 05/05/2025] [Indexed: 05/16/2025]
Abstract
Motor dysfunction constitutes a prominent characteristic of Parkinson's disease (PD), a neurodegenerative disorder associated with compromised mitochondrial activity, perturbed gut microbial composition, and neuronal loss. In this study, we examined the regulatory mechanisms of Lactiplantibacillus plantarum SG5 (SG5) on mitochondrial function in PD mouse models, with a particular emphasis on its interaction with the GLP-1/PGC-1α pathway. Findings revealed that MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, MPTP) induced (male 6-8 weeks C57BL/6 mice) motor impairments and damage to dopaminergic (DA) neurons in PD mice, resulting in mitochondrial dysfunction, decreased mitochondrial biogenesis, disrupted dynamics, and autophagy, while promoting fission and apoptosis. Additionally, MPTP modified gut microbial diversity and community structure. Nevertheless, supplementation with SG5 alleviated motor deficits and DA neurons damage in PD mice, enhancing mitochondrial quality by elevating PGC-1α expression and restoring biogenesis, dynamics, and autophagy levels. Mechanistic investigations demonstrated that SG5 increased colonic GLP-1 expression, suggesting that GLP-1 might regulate mitochondrial function via the GLP-1R-mediated PGC-1α. Furthermore, SG5 counteracted MPTP-induced gut dysbiosis. Notably, both GLP-1R antagonists and PGC-1α inhibitors attenuated the protective effects of SG5 in PD mice. In conclusion, L. plantarum SG5 may enhance mitochondrial function in the substantia nigra (SN) of PD mice through the GLP-1/PGC-1α pathway, potentially delaying neurodegeneration. Its mechanism is closely related to the regulation of the gut microenvironment and GLP-1 levels, presenting novel microbiota-based therapeutic targets for PD.
Collapse
Affiliation(s)
- Yueyan Qi
- Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Collaborative Innovation Center for Eco-Environment, Hebei Key Laboratory of Physiology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China
| | - Siyou Xie
- Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Collaborative Innovation Center for Eco-Environment, Hebei Key Laboratory of Physiology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China
| | - Jinhu Chen
- Department of endocrinology, Hebei General Hospital, Shijiazhuang, 050051, China
| | - Cancan Zhang
- Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Collaborative Innovation Center for Eco-Environment, Hebei Key Laboratory of Physiology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China
| | - Xin Ma
- Thankcome Biotechnology (Su Zhou) Co., Suzhou, China
| | - Yang Yu
- Thankcome Biotechnology (Su Zhou) Co., Suzhou, China
| | - Xueping Yu
- Thankcome Biotechnology (Su Zhou) Co., Suzhou, China
| | - Yanqin Wang
- Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Collaborative Innovation Center for Eco-Environment, Hebei Key Laboratory of Physiology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China.
| |
Collapse
|
6
|
Saleem ASA, Abdelnour S, Bassiony SM, Abdel-Monem UM, Elaref MY, Al-Marakby KM. Probiotic supplementation in sustainable sheep production: impacts on health, performance, and methane mitigation. Trop Anim Health Prod 2025; 57:206. [PMID: 40320471 PMCID: PMC12050230 DOI: 10.1007/s11250-025-04439-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 04/11/2025] [Indexed: 05/08/2025]
Abstract
Probiotics, defined as live microorganisms conferring health benefits, are increasingly recognized for their potential to enhance animal productivity, mitigate environmental impact, and improve overall animal health. Ruminants, including sheep, are significant contributors to greenhouse gas emissions, a key factor in climate change. Literature from 2003 to 2024 was retrieved from PubMed (Medline), Web of Science, and CAB Direct using the keywords: sheep, sustainability, probiotics, methane emission, and greenhouse gas emissions. The inclusion of probiotics in sheep diets demonstrates potential as a methane mitigation strategy through the stimulation of beneficial bacteria and the suppression of methanogenic microbial activity. Probiotics can improve rumen fermentation parameters by increasing volatile fatty acid production, decreasing protozoal numbers, and improving gas production. Additionally, probiotics can sustain intestinal health, boost nutrient digestibility, and strengthen the immune system. Although promising, the variable effectiveness of probiotics underscores the importance of refining formulations and delivery methods, taking into account strain, dose, and administration. Further studies are crucial to understand the underlying mechanisms and maximize their impact on sheep productivity. This review delves into the potential of probiotics to improve growth, health, and environmental sustainability in the sheep industry, drawing on insights from in vitro and in vivo studies.
Collapse
Affiliation(s)
- Ali S A Saleem
- Animal Production Department, Faculty of Agriculture, Sohag University, Sohag, Egypt.
- Animal Production Department, Faculty of Agriculture, Zagazig University, Zagazig, Egypt.
| | - Sameh Abdelnour
- Animal Production Department, Faculty of Agriculture, Zagazig University, Zagazig, Egypt.
| | - Sabry M Bassiony
- Animal Production Department, Faculty of Agriculture, Zagazig University, Zagazig, Egypt
| | - Usama M Abdel-Monem
- Animal Production Department, Faculty of Agriculture, Zagazig University, Zagazig, Egypt
| | - Mohamed Y Elaref
- Animal Production Department, Faculty of Agriculture, Sohag University, Sohag, Egypt
| | - Khaled M Al-Marakby
- Animal Production Department, Faculty of Agriculture, Zagazig University, Zagazig, Egypt
| |
Collapse
|
7
|
Yang X, Zhao Y, Liang L, Qu Y, Yu C, Zhang J, Lian W, Zhao Y. Protective effect of ginsenoside CK against MPTP-induced Parkinson' s disease mouse model by suppressing oxidative stress and inflammation, and modulating the gut microbiota. Microb Pathog 2025; 202:107409. [PMID: 40010656 DOI: 10.1016/j.micpath.2025.107409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 02/14/2025] [Accepted: 02/20/2025] [Indexed: 02/28/2025]
Abstract
Ginsenoside CK (CK) is a metabolite of natural diol ginsenoside in the intestine, which has a unique chemical structure and pharmacological activity. CK has great potential in the treatment of neurologic dysfunction diseases. However, the therapeutic effect and potential mechanism of CK on Parkinson's disease (PD) have not been studied. Accordingly, this study used microbiome analysis to correlate behavioral, physiological and biochemical indices, and combined with WB to elucidate the mechanism of CK's improvement on PD. CK showed significant therapeutic effects on PD mice, which improved behavioral abnormalities such as spatial memory ability and motor coordination in PD mice, increased the activities of T-AOC and GSH-Px, decreased the MDA content, thus alleviating oxidative stress injury, suppressed the levels of pro-inflammatory factors IL-1β, IL-6, and TNF-α, and activated the expression of anti-inflammatory factor IL-2, which then exerted against neuroinflammation, inhibited the apoptosis of dopaminergic neurons in the substantia nigra of PD mice, increased the expression of TH, and prevented the aggregation of α-Syn in the substantia nigra. Microbiomics analysis showed that CK treatment could reshape the gut microbiota of PD mice by increasing the abundance of probiotics (Bacteroides anomalies) and decreasing the number of pathogenic bacteria (Actinomycetes). Correlation analysis showed that gut microbiota had potential correlation with behavioral, physiological and biochemical indexes. Western blot results showed that CK inhibited the expression levels of apoptotic proteins Bax, caspase-3, and Bcl-2, which revealed that CK treatment could improve the dysfunction of MPTP-induced PD mice from the molecular level. Collectively, these findings will provide a basis for further development of CK as an anti-PD drug.
Collapse
Affiliation(s)
- Xu Yang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117, Jilin Province, China
| | - Yuting Zhao
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117, Jilin Province, China
| | - Lily Liang
- Gynecology of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, Jilin Province, China
| | - Yi Qu
- Jilin Province Product Quality Supervision and Inspection Institute, Changchun, 130117, Jilin Province, China
| | - Chunhui Yu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117, Jilin Province, China
| | - Jinnan Zhang
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, Jilin Province, China
| | - Wenhui Lian
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117, Jilin Province, China.
| | - Yu Zhao
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117, Jilin Province, China.
| |
Collapse
|
8
|
Guo X, Qiao X, Li X, Zhou W, Liu C, Yu F, Chen Q, Pan M, Niu X, Wang X, Li G, Wang L. Lactoferrin-modified organic-inorganic hybrid mesoporous silica for co-delivery of levodopa and curcumin in the synergistic treatment of Parkinson's disease. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156547. [PMID: 40054175 DOI: 10.1016/j.phymed.2025.156547] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 02/05/2025] [Accepted: 02/18/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Parkinson's disease (PD) is a chronic neurodegenerative disorder primarily characterized by oxidative stress and dopaminergic neuron damage. While levodopa remains the cornerstone of PD treatment, its efficacy is limited by poor bioavailability and neuroprotective effects. Curcumin, a potent antioxidant derived from turmeric, demonstrates neuroprotective promise but also suffers from low bioavailability, hindering its therapeutic application. The combined therapeutic use of levodopa and curcumin offers a potential synergistic approach, though its neuroprotection potential through brain-targeted delivery remains underexplored. PURPOSE To develop a lactoferrin-modified organic-inorganic hybrid mesoporous silica nanoparticle system (Lf-lip@LC-MSNs) for co-delivering levodopa and curcumin, aiming to enhance neuroprotective efficacy and achieve brain-targeted delivery in PD. METHODS Lf-lip@LC-MSNs were engineered to encapsulate levodopa within a curcumin-loaded lipid bilayer, modified with lactoferrin for optimized brain-targeted delivery. In vitro studies were conducted on rotenone-damaged neuronal models to evaluate oxidative stress, mitochondrial dysfunction, α-synuclein aggregation, and neuronal survival. In vivo experiments on MPTP-induced PD mouse models evaluated biodistribution, therapeutic efficacy, and safety in healthy mice, focusing on motor function recovery. RESULTS The combination of levodopa and curcumin significantly reduced oxidative stress and α-synuclein accumulation, enhancing neuronal survival compared to monotherapies. Lf-lip@LC-MSNs further amplified these effects, achieving superior brain-targeted delivery and improved motor function restoration with minimal systemic toxicity. CONCLUSIONS The combination of curcumin and levodopa provided synergistic neuroprotection in PD models. By employing a targeted delivery system, the Lf-lip@LC-MSNs not only facilitated efficient brain targeting but also potentiated therapeutic outcomes, providing a compelling strategy for treating PD and paving the way for advancements in managing other neurodegenerative diseases.
Collapse
Affiliation(s)
- Xiuping Guo
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050 PR China
| | - Xubai Qiao
- Beijing United Family Hospital, Beijing 100015, PR China
| | - Xinru Li
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100093 PR China
| | - Wenkai Zhou
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050 PR China
| | - Chang Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050 PR China
| | - Feifei Yu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050 PR China
| | - Qingbo Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050 PR China
| | - Mingyu Pan
- University of California-Riverside, Riverside 92521, USA
| | - Xia Niu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050 PR China
| | - Xiaomei Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050 PR China
| | - Guiling Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050 PR China.
| | - Lulu Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050 PR China.
| |
Collapse
|
9
|
Izquierdo-Altarejos P, Arenas YM, Montoliu C, Pérez-Martínez G, Llansola M, Felipo V. Extracellular vesicles from Lacticaseibacillus paracasei reduce neuroinflammation in hippocampus and restore some cognitive functions in hyperammonemic rats. Microbiol Res 2025; 294:128101. [PMID: 39978142 DOI: 10.1016/j.micres.2025.128101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 02/22/2025]
Abstract
Cirrhotic patients may show minimal hepatic encephalopathy (MHE) which impairs life quality and span. There is a need of new safe treatments for MHE. Hyperammonemia is a main contributor to MHE. Hyperammonemic rats reproduce the cognitive impairment present in patients with MHE, which is mediated by neuroinflammation and altered glutamatergic neurotransmission in hippocampus. Probiotics induce positive effects in MHE patients, which could be mediated by bacterial extracellular vesicles (EVs). The aims of this work were to evaluate in hyperammonemic rats: 1) if intravenous administration of EVs from L. paracasei improves memory and learning and 2) reduces neuroinflammation in hippocampus and 3) to study the mechanisms involved using an ex vivo approach. It is shown that intravenous injection of EVs from L. paracasei reverses glial activation in hippocampus and cognitive impairment in hyperammonemic rats. Ex vivo studies in hippocampal slices show that hyperammonemia increases TNFα and TNFR1 and S1PR2 membrane expression and activation, leading to increased IL-1β content and activation of IL-1 receptor and of Src. This increases CCL2 and BDNF and TrkB activation. This leads to increased membrane expression of the NR2B subunit of the NMDA receptor and of the GluA2 subunit of AMPA receptors and reduced membrane expression of the GluA1 subunit, leading to cognitive impairment. EVs from L. paracasei reduce neuroinflammation in hyperammonemic rats and restore the function of the TNFα-TNFR1-S1PR2-IL-1β-CCL2-BDNF-TrkB pathway, glutamatergic neurotransmission and cognitive function in rats with hyperammonemia and MHE. This suggests that these EVs could also improve cognitive function in cirrhotic patients with MHE.
Collapse
Affiliation(s)
| | - Yaiza M Arenas
- Laboratory of Lactic Acid Bacteria and Probiotics, Department of Biotechnology, Instituto de Agroquímica y Tecnología de Alimentos (C.S.I.C.), Valencia, Spain; Departamento de Patología, Facultad de Medicina, Universidad Valencia, Valencia, Spain; INCLIVA Instituto de Investigación Sanitaria, Valencia, Spain.
| | - Carmina Montoliu
- Departamento de Patología, Facultad de Medicina, Universidad Valencia, Valencia, Spain; INCLIVA Instituto de Investigación Sanitaria, Valencia, Spain.
| | - Gaspar Pérez-Martínez
- Laboratory of Lactic Acid Bacteria and Probiotics, Department of Biotechnology, Instituto de Agroquímica y Tecnología de Alimentos (C.S.I.C.), Valencia, Spain.
| | - Marta Llansola
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, Valencia, Spain.
| | - Vicente Felipo
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, Valencia, Spain.
| |
Collapse
|
10
|
Kumari N, Addepalli V, More A, Patil A, Suryawanshi M. Gut microbiota and Parkinson's Disease: a new frontier in understanding neurological health. Inflammopharmacology 2025:10.1007/s10787-025-01726-w. [PMID: 40244491 DOI: 10.1007/s10787-025-01726-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 01/31/2025] [Indexed: 04/18/2025]
Abstract
Increasingly recognized as a neurodegenerative disease with motor manifestations and progressive cognitive decline, PD has more frequently been linked to the gut microbiome. The gut-brain axis, a bidirectional communication system between the gut and brain, plays a crucial role in PD pathogenesis. Exploration of the intricacies in the interplay between PD and the gut microbiome, together with the important mechanisms involved, will form the basis of this review. Gut microbiome activities as contributors to PD actions include altered intestinal permeability, neuroinflammation, alpha-syn aggregation, oxidative stress, and neurotransmitter production. Gut-brain axis communication that is highly facilitated through immune, metabolic, and neural pathways enables communication between the gut and the brain. Recent evidence suggests that the disease may begin in the gut, with GI symptoms typically preceding loss of motor control. Research has shown a significant connection between Parkinson's disease and the gut microbiome, affecting disease onset, progression, and symptoms. Therapeutic strategies targeting the gut microbiome, such as probiotics, prebiotics, and FMT, may improve PD outcomes. Personalized medicine and neuroprotective therapies are promising for managing PD. Researchers are exploring the connection between the gut microbiome and PD to create new treatments for bettering the lives of those with the disease. By understanding the intricate relationship between the gut microbiome and PD, researchers can develop novel therapeutic approaches to improve the quality of life for individuals with this debilitating disease.
Collapse
Affiliation(s)
- Nishchhal Kumari
- Dr. D.Y. Patil Institute of Pharmaceutical Sciences and Research, Pune, Maharashtra, 411018, India
| | - Veeranjaneyulu Addepalli
- Dr. D.Y. Patil Institute of Pharmaceutical Sciences and Research, Pune, Maharashtra, 411018, India
| | - Aniket More
- Dr. D.Y. Patil Institute of Pharmaceutical Sciences and Research, Pune, Maharashtra, 411018, India
| | - Ashwani Patil
- Dr. D.Y. Patil Institute of Pharmaceutical Sciences and Research, Pune, Maharashtra, 411018, India.
| | - Meghraj Suryawanshi
- Department of Pharmaceutics, Sandip Institute of Pharmaceutical Sciences (SIPS), Affiliated To Savitribai Phule Pune University (SPPU, Pune), Nashik, Maharashtra, 422213, India.
| |
Collapse
|
11
|
Xu K, Wang G, Gong J, Yang X, Cheng Y, Li D, Sheng S, Zhang F. Akkermansia muciniphila protects against dopamine neurotoxicity by modulating butyrate to inhibit microglia-mediated neuroinflammation. Int Immunopharmacol 2025; 152:114374. [PMID: 40056512 DOI: 10.1016/j.intimp.2025.114374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/19/2025] [Accepted: 02/24/2025] [Indexed: 03/10/2025]
Abstract
Parkinson's disease (PD) is an age-related and second most common neurodegenerative disease. To date, safe and efficient therapeutic drugs are deficient. In recent years, the relationship between gut microbiota and CNS have received more attention. Homeostatic imbalance of gut microbiota was revealed to participate in the progression of PD. This study detected that Akkermansia muciniphila (A. muciniphila) was apparently decreased in the feces of PD rats via 16S rRNA amplicon sequencing. Furtherly, we found that exogenous supplementation of A. muciniphila could improve 6-OHDA-induced motor dysfunction and dopamine (DA) neuronal damage and neuroinflammatory factors release in PD rats. Moreover, the short-chain fatty acids (SCFAs) sequencing demonstrated that A. muciniphila addition increased butyrate content both in gut and brain. The subsequent functional experiments confirmed that the exogenous supplementation of butyrate conferred neuroprotection against DA neurotoxicity. Mechanically, butyrate targeted microglia to attenuate DA neuronal injury via inhibiting microglia activation and neuroinflammatory factors production. In conclusion, A. muciniphila protected DA neuronal damage by modulating butyrate to inhibit microglia-elicited neuroinflammation. These findings provided a potential application of A. muciniphila on PD treatment.
Collapse
Affiliation(s)
- Kaifei Xu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Centre, Zunyi Medical University, Zunyi, Guizhou, China
| | - Guoqing Wang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Centre, Zunyi Medical University, Zunyi, Guizhou, China
| | - Jiantao Gong
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Centre, Zunyi Medical University, Zunyi, Guizhou, China
| | - Xinxing Yang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Centre, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yufeng Cheng
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Centre, Zunyi Medical University, Zunyi, Guizhou, China
| | - Daidi Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Centre, Zunyi Medical University, Zunyi, Guizhou, China
| | - Shuo Sheng
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Centre, Zunyi Medical University, Zunyi, Guizhou, China
| | - Feng Zhang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Centre, Zunyi Medical University, Zunyi, Guizhou, China.
| |
Collapse
|
12
|
Ren M, Jin T, Tong J, Song D, Xie Q, Li X, Li Y, Liu K, Gao J, Liu M, Cheng J. Anti-Inflammatory Effects of Weissella cibaria SDS2.1 Against Klebsiella pneumoniae-Induced Mammary Gland Inflammation. Animals (Basel) 2025; 15:1139. [PMID: 40281973 PMCID: PMC12024108 DOI: 10.3390/ani15081139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/03/2025] [Accepted: 04/13/2025] [Indexed: 04/29/2025] Open
Abstract
Dairy cows are highly susceptible to mastitis caused by Klebsiella pneumoniae, and treating these infections poses a challenge due to the resistance of the bacterium to common antibiotics. This study aimed to evaluate the safety of W. cibaria SDS2.1 and investigate its protective effects against K. pneumoniae-induced mastitis. The safety of W. cibaria SDS2.1 was assessed through comprehensive analyses, including antibiotic resistance profiling, hemolysis assays, cell cytotoxicity tests, and whole-genome sequencing. Furthermore, its ability to protect against cellular and tissue damage caused by K. pneumoniae-induced mastitis was evaluated using both in vitro and in vivo models. Our results revealed that W. cibaria SDS2.1 was non-hemolytic, non-cytotoxic, and significantly inhibited the growth of K. pneumoniae (p < 0.05). Additionally, W. cibaria SDS2.1 effectively reduced the adhesion and invasion of K. pneumoniae. In the K. pneumoniae-induced mouse mastitis model, W. cibaria SDS2.1 significantly reduced myeloperoxidase (MPO) activity, mammary tissue damage, and the expression of inflammatory cytokines (IL-6, IL-1β, and TNF-α) (p < 0.05). In K. pneumoniae-infected bovine mammary epithelial cells (bMECs), W. cibaria SDS2.1 significantly decreased lactate dehydrogenase (LDH) release, indicating reduced cellular damage. These findings demonstrate that W. cibaria SDS2.1 exhibits anti-inflammatory properties in experimental models, suggesting its potential role in mitigating K. pneumoniae-induced mastitis.
Collapse
Affiliation(s)
- Meiyi Ren
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China; (M.R.); (T.J.); (J.T.); (D.S.); (Q.X.); (X.L.); (Y.L.); (K.L.)
| | - Tianxiong Jin
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China; (M.R.); (T.J.); (J.T.); (D.S.); (Q.X.); (X.L.); (Y.L.); (K.L.)
| | - Jingdi Tong
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China; (M.R.); (T.J.); (J.T.); (D.S.); (Q.X.); (X.L.); (Y.L.); (K.L.)
| | - Deyuan Song
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China; (M.R.); (T.J.); (J.T.); (D.S.); (Q.X.); (X.L.); (Y.L.); (K.L.)
| | - Qinna Xie
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China; (M.R.); (T.J.); (J.T.); (D.S.); (Q.X.); (X.L.); (Y.L.); (K.L.)
| | - Xiaohan Li
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China; (M.R.); (T.J.); (J.T.); (D.S.); (Q.X.); (X.L.); (Y.L.); (K.L.)
| | - Yan Li
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China; (M.R.); (T.J.); (J.T.); (D.S.); (Q.X.); (X.L.); (Y.L.); (K.L.)
- Key Laboratory of Healthy Breeding in Dairy Cattle (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Hebei Agricultural University, Baoding 071001, China
| | - Kangping Liu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China; (M.R.); (T.J.); (J.T.); (D.S.); (Q.X.); (X.L.); (Y.L.); (K.L.)
| | - Jian Gao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China;
| | - Mingchao Liu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China; (M.R.); (T.J.); (J.T.); (D.S.); (Q.X.); (X.L.); (Y.L.); (K.L.)
- Key Laboratory of Healthy Breeding in Dairy Cattle (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Hebei Agricultural University, Baoding 071001, China
| | - Jia Cheng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China; (M.R.); (T.J.); (J.T.); (D.S.); (Q.X.); (X.L.); (Y.L.); (K.L.)
- Key Laboratory of Healthy Breeding in Dairy Cattle (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Hebei Agricultural University, Baoding 071001, China
| |
Collapse
|
13
|
Wu S, Luo Y, Wei F, Li Y, Fan J, Chen Y, Zhang W, Li X, Xu Y, Chen Z, Xia C, Hu M, Li P, Gu Q. Lactic acid bacteria target NF-κB signaling to alleviate gastric inflammation. Food Funct 2025; 16:3101-3119. [PMID: 40152095 DOI: 10.1039/d4fo06308b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Helicobacter pylori (H. pylori) infection and the resulting gastric inflammation are major contributors to gastric cancer development. Probiotics, particularly Lactobacillus, are promising for their anti-inflammatory potential, yet their exact mechanisms in inhibiting H. pylori-induced inflammation are unclear. In our previous study, Lactiplantibacillus plantarum ZJ316 (L. plantarum ZJ316) demonstrated strong anti-inflammatory effects against H. pylori infection in vivo, but its precise mechanisms were not fully understood. Here, we aimed to investigate how L. plantarum ZJ316 inhibits the inflammatory response to H. pylori infection. Our results demonstrated that L. plantarum ZJ316 effectively reduced the expression of pro-inflammatory cytokines in H. pylori-infected AGS cells. Mechanistically, L. plantarum ZJ316 inhibited the NF-κB signaling pathway by preventing the degradation of IκBα, suppressing p65 phosphorylation, and blocking the nuclear translocation of phosphorylated p65. Treatment with the NF-κB inhibitor BAY 11-7082 further decreased tumor necrosis factor-α (TNF-α), interleukin-8 (IL-8), and interleukin-1β (IL-1β) levels, confirming the inhibitory effect of L. plantarum ZJ316 on the NF-κB pathway. In H. pylori-infected mice, oral administration of L. plantarum ZJ316 significantly alleviated inflammatory cell infiltration, reduced TNF-α and pepsinogen II (PGII) levels, and increased interleukin-10 (IL-10) levels in serum. A comparative metagenomic analysis of the gastric microbiota revealed a decrease in Prevotella and Desulfovibrio, alongside an increase in Ligilactobacillus and Akkermansia, supporting the protective effects of L. plantarum ZJ316 and correlating with their decreased inflammatory response. In summary, administration of L. plantarum ZJ316 demonstrated robust anti-inflammatory effects against H. pylori infection by suppressing NF-κB signaling and promoting favorable changes in the gastric microbiota composition. Therefore, L. plantarum ZJ316 holds promise as a novel functional food for protecting the body against H. pylori infection.
Collapse
Affiliation(s)
- Shiying Wu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Yuenuo Luo
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Fangtong Wei
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Yanan Li
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, Jiangsu 210023, China
| | - Jiayi Fan
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Yongqiang Chen
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Wenjie Zhang
- Hangzhou Helixinjian Industry Co., Ltd, No. 48 Zijinghua Road, Gudang Street, Xihu District, Hangzhou, Zhejiang 310050, China
| | - Xuelong Li
- Hangzhou Helixinjian Industry Co., Ltd, No. 48 Zijinghua Road, Gudang Street, Xihu District, Hangzhou, Zhejiang 310050, China
| | - Yang Xu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Ziqi Chen
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Chenlan Xia
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Mingyang Hu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Ping Li
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Qing Gu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| |
Collapse
|
14
|
Flores-Soto ME, Nápoles-Medina AY, Tejeda-Martínez AR, Solís-Pacheco JR, Chaparro-Huerta V, Gutiérrez-Sevilla JE, Aguilar-Uscanga BR. Supplementation of the Probiotic LLH135 Reduces Oxidative Stress in a Model of Hemiparkinsonism. Behav Neurol 2025; 2025:8401392. [PMID: 40256256 PMCID: PMC12006709 DOI: 10.1155/bn/8401392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 03/24/2025] [Indexed: 04/22/2025] Open
Abstract
Oxidative stress and neuroinflammation are considered as the two main etiological reasons behind idiopathic Parkinson's disease (PD). Nevertheless, the actual treatments are focused on improving motor symptoms by restoring dopamine (DA) presence, leaving said causes unattended. Probiotics could be a promising strategy for the improvement of these physiological features behind the disease and therefore constitute a complementary treatment for those having PD. This study evaluated the effect of the oral administration of a probiotic bacteria mixture from 3 strains of Limosilactobacillus fermentum LH01, Limosilactobacillus reuteri LH03, and Lactiplantibacillus plantarum LH05 (LLH135), of human milk origin, for 4 weeks, on mice under the hemiparkinsonism model of intrastriatal administration of 6-hidroxidopamine (6-OHDA). We measured total antioxidant capacity (TAC), super oxide dismutase (SOD) activity, and 8-deoxyguanosine (8-OHdG) regarding oxidative stress. Concerning neuroinflammation, immunoreactivity for GFAP, IBA-1, and CD68 was measured by immunohistochemistry and the latter markers corroborated in colocalization with immunofluorescence to assess activated microglia. The probiotic mixture diminished the oxidative stress features of SOD activity as well as 8-OHdG generated by the model of hemiparkinsonism. These effects were accompanied as well by the dampening of the glial immunoreactivity and colocalization of IBA-1 and CD68 that were present under the model. Our findings suggest that the administration of the probiotic LLH135 exerts neuroprotective effects by promoting an antioxidant response which could be explained by the modulation of the response from glial cells to dopaminergic neuronal damage induced with 6-OHDA.
Collapse
Affiliation(s)
- Mario E. Flores-Soto
- Laboratorio de Neurobiología Celular y Molecular, División de Neurociencias, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social, Guadalajara, Jalisco, Mexico
| | - Angelica Y. Nápoles-Medina
- Laboratorio de Neurobiología Celular y Molecular, División de Neurociencias, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social, Guadalajara, Jalisco, Mexico
- Laboratorio de Investigación Leche humana, Departamento de Farmacobiología, Centro Universitario de Ciencias Exactas e Ingenierías, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Aldo R. Tejeda-Martínez
- Laboratorio de Neurobiología Celular y Molecular, División de Neurociencias, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social, Guadalajara, Jalisco, Mexico
| | - Josué R. Solís-Pacheco
- Laboratorio de Investigación Leche humana, Departamento de Farmacobiología, Centro Universitario de Ciencias Exactas e Ingenierías, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Verónica Chaparro-Huerta
- Laboratorio de Neurobiología Celular y Molecular, División de Neurociencias, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social, Guadalajara, Jalisco, Mexico
| | - Juan E. Gutiérrez-Sevilla
- Laboratorio de Neurobiología Celular y Molecular, División de Neurociencias, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social, Guadalajara, Jalisco, Mexico
| | - Blanca R. Aguilar-Uscanga
- Laboratorio de Investigación Leche humana, Departamento de Farmacobiología, Centro Universitario de Ciencias Exactas e Ingenierías, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| |
Collapse
|
15
|
Zhang X, Yan F, He XJ, Chen Y, Gu R, Dong X, Wei Y, Bai L, Bai J. Thioredoxin-1 Downregulation in the SNpc Exacerbates the Cognitive Impairment Induced by MPTP. Antioxid Redox Signal 2025. [PMID: 40135707 DOI: 10.1089/ars.2024.0630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
Aims: Parkinson's disease (PD) is characterized by dopaminergic (DAergic) neuron degeneration in the substantia nigra pars compacta (SNpc). Thioredoxin-1 (Trx-1) is a redox protein that protects neurons from various injuries. Our study revealed that Trx-1 overexpression improved the learning and memory impairments induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). However, the role of the specific transmission of signals from the SNpc to the hippocampus regulated by Trx-1 in cognition deficits associated with PD is still unknown. Results: We observed that Trx-1 downregulation in the SNpc aggravated cognitive dysfunction induced by MPTP. Importantly, we observed that the SNpc directly projects to the hippocampus. We found that the loss of DAergic neurons in the SNpc induced by MPTP resulted in a decrease in dopamine D1 receptor (D1R) expression in the hippocampus, which was promoted by Trx-1 downregulation in the SNpc. The levels of phosphorylated extracellular signal-regulated kinase (p-ERK1/2), phosphorylated cAMP-response element binding protein (p-CREB), brain-derived neurotrophic factor (BDNF), and postsynaptic density protein 95 (PSD95) in the hippocampus were decreased by MPTP and further decreased by Trx-1 downregulation in the SNpc. Finally, the number of synapses in the hippocampus was decreased by MPTP in the hippocampus and further reduced by Trx-1 downregulation in the SNpc. Innovation: Trx-1 downregulation accelerated the loss of DAergic neurons in the SNpc, leading to a decrease in the number dopaminergic projections to the hippocampus, subsequently inhibiting the D1R-ERK1/2-CREB-BDNF pathway in the hippocampus, and ultimately impairing hippocampus-dependent cognition. Conclusions: These results indicate that a decrease in Trx-1 level in the SNpc plays a critical regulatory role in cognitive dysfunction in individuals with PD by decreasing the hippocampal D1R signaling pathway. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Xianwen Zhang
- Medical Faculty, Laboratory of Molecular Neurobiology, Kunming University of Science and Technology, Kunming, China
| | - Fang Yan
- Medical Faculty, Laboratory of Molecular Neurobiology, Kunming University of Science and Technology, Kunming, China
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| | - Xiong Jie He
- Medical Faculty, Laboratory of Molecular Neurobiology, Kunming University of Science and Technology, Kunming, China
| | - Yali Chen
- Medical Faculty, Laboratory of Molecular Neurobiology, Kunming University of Science and Technology, Kunming, China
| | - Rou Gu
- Medical Faculty, Laboratory of Molecular Neurobiology, Kunming University of Science and Technology, Kunming, China
| | - Xianghuan Dong
- Medical Faculty, Laboratory of Molecular Neurobiology, Kunming University of Science and Technology, Kunming, China
| | - Yonghang Wei
- Medical Faculty, Laboratory of Molecular Neurobiology, Kunming University of Science and Technology, Kunming, China
| | - Liping Bai
- Medical Faculty, Laboratory of Molecular Neurobiology, Kunming University of Science and Technology, Kunming, China
| | - Jie Bai
- Medical Faculty, Laboratory of Molecular Neurobiology, Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
16
|
Yao SQ, Xu ZP, Guo WX, Zhang H, Zhang M, Jia JX, Yang ZJ, Wang H, Zhao ZY, Huo DS. Neuroprotective effect of Cistanche deserticola glycosides in MPTP-Induced Parkinson's disease mouse model involves Nrf2 activation. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2025; 88:576-588. [PMID: 40056093 DOI: 10.1080/15287394.2025.2470847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Parkinson's Disease (PD) a progressive neurodegenerative disorder is attributed to dopaminergic neuronal cell loss in the mid-brain substantia nigra pars compacta. A major risk factors associated with PD development is presence of excess oxidative stress. Previously, glycosides derived from Cistanche deserticola were reported to play a key role in counteracting PD; however, the underlying mechanisms remain to be determined. This study aimed to examine the neuroprotective effect attributed to glycosides derived from C. deserticola in PD model in mice. The model of PD was established by injecting intraperitoneally 1-methyl-4-penyl-1,2,3,6-tetrahydropyridine (MPTP). Rotarod and pole tests determined neurological behavior. The following immunohistochemistry, and metabolic biomarkers were measured mid-brain substantia nigra: (1) number of dopaminergic neuronal cell using immunohistochemistry (2) oxidative stress as evidenced by activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) as well levels of malondialdehyde (MDA), (3) inflammatory infiltration as measured by levels of IL-1β and TNF-α (4) by Western blot involvement of protein expression levels of Nrf2 signaling pathway. Data demonstrated that C. deserticola glycosides treatment improved behavioral performance, increased number of dopaminergic neurons, reduced cytokine levels of IL-1β and TNF-α accompanied by enhanced antioxidant activity in PD mice. These observations were associated with activation of Nrf2 signaling pathway. Data suggest that C. deserticola glycosides may thus be considered as an alternative compound for PD treatment.
Collapse
Affiliation(s)
- Shu-Qing Yao
- Department of Human Anatomy, Baotou Medical College, Inner Mongolia, China
- Key Laboratory of Human Anatomy, Education Department of Inner Mongolia Autonomous Region, Baotou, China
| | - Zhi-Peng Xu
- Department of Human Anatomy, Baotou Medical College, Inner Mongolia, China
- Key Laboratory of Human Anatomy, Education Department of Inner Mongolia Autonomous Region, Baotou, China
| | - Wu-Xia Guo
- Department of Pain, The Fourth Hospital of Baotou, Inner Mongolia, China
| | - He Zhang
- Department of Human Anatomy, Baotou Medical College, Inner Mongolia, China
- Key Laboratory of Human Anatomy, Education Department of Inner Mongolia Autonomous Region, Baotou, China
| | - Ming Zhang
- Department of Human Anatomy, Baotou Medical College, Inner Mongolia, China
- Key Laboratory of Human Anatomy, Education Department of Inner Mongolia Autonomous Region, Baotou, China
| | - Jian-Xin Jia
- Department of Human Anatomy, Baotou Medical College, Inner Mongolia, China
- Key Laboratory of Human Anatomy, Education Department of Inner Mongolia Autonomous Region, Baotou, China
| | - Zhan-Jun Yang
- Key Laboratory of Human Anatomy, Education Department of Inner Mongolia Autonomous Region, Baotou, China
- Department of Human Anatomy, Chifeng University, Inner Mongolia, China
| | - He Wang
- School of Health Sciences, University of Newcastle, Newcastle, Australia
| | - Zhi-Ying Zhao
- Department of Human Anatomy, Baotou Medical College, Inner Mongolia, China
- Key Laboratory of Human Anatomy, Education Department of Inner Mongolia Autonomous Region, Baotou, China
| | - Dong-Sheng Huo
- Department of Human Anatomy, Baotou Medical College, Inner Mongolia, China
- Key Laboratory of Human Anatomy, Education Department of Inner Mongolia Autonomous Region, Baotou, China
| |
Collapse
|
17
|
Wang M, Liu Y, Zhong L, Wu F, Wang J. Advancements in the investigation of gut microbiota-based strategies for stroke prevention and treatment. Front Immunol 2025; 16:1533343. [PMID: 40103814 PMCID: PMC11914130 DOI: 10.3389/fimmu.2025.1533343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 02/11/2025] [Indexed: 03/20/2025] Open
Abstract
Stroke represents a predominant cause of mortality and disability on a global scale, impacting millions annually and exerting a considerable strain on healthcare systems. The incidence of stroke exhibits regional variability, with ischemic stroke accounting for the majority of occurrences. Post-stroke complications, such as cognitive impairment, motor dysfunction, and recurrent stroke, profoundly affect patients' quality of life. Recent advancements have elucidated the microbiota-gut-brain axis (MGBA), underscoring the complex interplay between gut health and brain function. Dysbiosis, characterized by an imbalance in gut microbiota, is significantly linked to an elevated risk of stroke and unfavorable outcomes. The MGBA plays a crucial role in modulating immune function, neurotransmitter levels, and metabolic byproducts, which may intensify neuroinflammation and impair cerebral health. This review elucidates the role of MGBA in stroke pathophysiology and explores potential gut-targeted therapeutic strategies to reduce stroke risk and promote recovery, including probiotics, prebiotics, pharmacological interventions, and dietary modifications. However, the current prevention and treatment strategies based on intestinal flora still face many problems, such as the large difference of individual intestinal flora, the stability of efficacy, and the long-term safety need to be considered. Further research needs to be strengthened to promote its better application in clinical practice.
Collapse
Affiliation(s)
| | | | | | | | - Jinjin Wang
- Department of Gastroenterology, The First People’s Hospital of Xiaoshan District, Hangzhou, Zhejiang, China
| |
Collapse
|
18
|
Kim D, Nguyen TTM, Moon Y, Kim J, Nam H, Cha DS, An YJ, de Guzman ACV, Park S. Time-Resolved Evaluation of L-Dopa Metabolism in Bacteria-Host Symbiotic System and the Effect on Parkinson's Molecular Pathology. SMALL METHODS 2025; 9:e2400469. [PMID: 39058017 PMCID: PMC11926514 DOI: 10.1002/smtd.202400469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Indexed: 07/28/2024]
Abstract
The gut microbiome influences drug metabolism and therapeutic efficacy. Still, the lack of a general label-free approach for monitoring bacterial or host metabolic contribution hampers deeper insights. Here, a 2D nuclear magnetic resonance (NMR) approach is introduced that enables real-time monitoring of the metabolism of Levodopa (L-dopa), an anti-Parkinson drug, in both live bacteria and bacteria-host (Caenorhabditis elegans) symbiotic systems. The quantitative method reveals that discrete Enterococcus faecalis substrains produce different amounts of dopamine in live hosts, even though they are a single species and all have the Tyrosine decarboxylase (TyrDC) gene involved in L-dopa metabolism. The differential bacterial metabolic activity correlates with differing Parkinson's molecular pathology concerning alpha-synuclein aggregation as well as behavioral phenotypes. The gene's existence or expression is not an indicator of metabolic activity is also shown, underscoring the significance of quantitative metabolic estimation in vivo. This simple approach is widely adaptable to any chemical drug to elucidate pharmacomicrobiomic relationships and may help rapidly screen bacterial metabolic effects in drug development.
Collapse
Affiliation(s)
- Doyeon Kim
- Natural Products Research InstituteCollege of PharmacySeoul National UniversitySeoul08826South Korea
| | - Tin Tin Manh Nguyen
- Natural Products Research InstituteCollege of PharmacySeoul National UniversitySeoul08826South Korea
| | - Yechan Moon
- Natural Products Research InstituteCollege of PharmacySeoul National UniversitySeoul08826South Korea
| | - Jin‐Mo Kim
- Natural Products Research InstituteCollege of PharmacySeoul National UniversitySeoul08826South Korea
| | - Hoonsik Nam
- Natural Products Research InstituteCollege of PharmacySeoul National UniversitySeoul08826South Korea
| | - Dong Seok Cha
- College of Pharmacy Woosuk UniversityJeonbuk55338South Korea
| | - Yong Jin An
- Natural Products Research InstituteCollege of PharmacySeoul National UniversitySeoul08826South Korea
| | | | - Sunghyouk Park
- Natural Products Research InstituteCollege of PharmacySeoul National UniversitySeoul08826South Korea
| |
Collapse
|
19
|
van der Maden MM, Verbeek MM, Beckers M. Lactobacillaceae and Parkinson's disease: An apparent paradox. JOURNAL OF PARKINSON'S DISEASE 2025; 15:269-281. [PMID: 39973489 DOI: 10.1177/1877718x241312401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder predominantly known for its motor symptoms such as bradykinesia, rigidity and tremor, but the disorder is also increasingly recognized for its association with impaired gastrointestinal function. The composition of the gut microbiome is known to be different in PD compared with healthy individuals. One of the bacterial families with increased abundance in people with PD is Lactobacillaceae. Interestingly, opposite effects have been ascribed to Lactobacillaceae in PD. A number of studies have linked Lactobacillaceae spp. in the gut to worse motor function, and to premature degradation of levodopa. However, other studies have linked administration of Lactobacillaceae-containing probiotics to improved motor function and reduced gastrointestinal problems. In this narrative review, we investigate this apparent paradox. The key to its understanding appears to lie in the specific species of Lactobacillaceae. The species L. plantarum in particular seemed to show a correlation with improved motor symptoms, as well as a reduction in intestinal inflammation, whereas L. brevis, L. curvatus and L. fermentum have properties that might be detrimental to people with PD.
Collapse
Affiliation(s)
| | - Marcel M Verbeek
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboudumc Centre of Expertise for Parkinson & Movement Disorders, Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Milan Beckers
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboudumc Centre of Expertise for Parkinson & Movement Disorders, Nijmegen, The Netherlands
| |
Collapse
|
20
|
Tang Y, Zhang Y, Chen C, Cao Y, Wang Q, Tang C. Gut microbiota: A new window for the prevention and treatment of neuropsychiatric disease. J Cent Nerv Syst Dis 2025; 17:11795735251322450. [PMID: 39989718 PMCID: PMC11846125 DOI: 10.1177/11795735251322450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/11/2024] [Accepted: 01/27/2025] [Indexed: 02/25/2025] Open
Abstract
Under normal physiological conditions, gut microbiota and host mutually coexist. They play key roles in maintaining intestinal barrier integrity, absorption, and metabolism, as well as promoting the development of the central nervous system (CNS) and emotional regulation. The dysregulation of gut microbiota homeostasis has attracted significant research interest, specifically in its impact on neurological and psychiatric disorders. Recent studies have highlighted the important role of the gut- brain axis in conditions including Alzheimer's Disease (AD), Parkinson's Disease (PD), and depression. This review aims to elucidate the regulatory mechanisms by which gut microbiota affect the progression of CNS disorders via the gut-brain axis. Additionally, we discuss the current research landscape, identify gaps, and propose future directions for microbial interventions against these diseases. Finally, we provide a theoretical reference for clinical treatment strategies and drug development for AD, PD, and depression.
Collapse
Affiliation(s)
- Yali Tang
- Department of Pharmacy, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Yizhu Zhang
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Chen Chen
- Department of Pharmacy, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Ying Cao
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, People’s Republic of China
| | - Qiaona Wang
- School of Ecology and Applied Meteorology, Nanjing University of Information Science & Technology, Nanjing, People’s Republic of China
| | - Chuanfeng Tang
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| |
Collapse
|
21
|
Tang MB, Liu YX, Hu ZW, Luo HY, Zhang S, Shi CH, Xu YM. Study insights in the role of PGC-1α in neurological diseases: mechanisms and therapeutic potential. Front Aging Neurosci 2025; 16:1454735. [PMID: 40012862 PMCID: PMC11861300 DOI: 10.3389/fnagi.2024.1454735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 12/30/2024] [Indexed: 02/28/2025] Open
Abstract
Peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α), which is highly expressed in the central nervous system, is known to be involved in the regulation of mitochondrial biosynthesis, metabolic regulation, neuroinflammation, autophagy, and oxidative stress. This knowledge indicates a potential role of PGC-1α in a wide range of functions associated with neurological diseases. There is emerging evidence indicating a protective role of PGC-1α in the pathogenesis of several neurological diseases. As such, a deeper and broader understanding of PGC-1α and its role in neurological diseases is urgently needed. The present review provides a relatively complete overview of the current knowledge on PGC-1α, including its functions in different types of neurons, basic structural characteristics, and its interacting transcription factors. Furthermore, we present the role of PGC-1α in the pathogenesis of various neurological diseases, such as intracerebral hemorrhage, ischemic stroke, Alzheimer's disease, Parkinson's disease, Amyotrophic lateral sclerosis, Huntington's disease, and other PolyQ diseases. Importantly, we discuss some compounds or drug-targeting strategies that have been studied to ameliorate the pathology of these neurological diseases and introduce the possible mechanistic pathways. Based on the available studies, we propose that targeting PGC-1α could serve as a promising novel therapeutic strategy for one or more neurological diseases.
Collapse
Affiliation(s)
- Mi-bo Tang
- Department of Geriatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Yi-xuan Liu
- Department of Geriatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Zheng-wei Hu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Hai-yang Luo
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Shuo Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Chang-he Shi
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Yu-ming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, Zhengzhou, Henan, China
- Henan Key Laboratory of Cerebrovascular Diseases, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
22
|
Zhu X, Lu H, Li W, Niu S, Xue J, Sun H, Zhang J, Zhang Z. Ferroptosis Induces gut microbiota and metabolic dysbiosis in Collagen-Induced arthritis mice via PAD4 enzyme. Gene 2025; 936:149106. [PMID: 39561902 DOI: 10.1016/j.gene.2024.149106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/17/2024] [Accepted: 11/15/2024] [Indexed: 11/21/2024]
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease characterized by chronic inflammation and joint destruction, with emerging evidence implicating gut microbiota dysbiosis in its pathogenesis. The current study explores the role of ferroptosis, a form of regulated cell death driven by iron-dependent lipid peroxidation, in modulating gut microbiota and metabolic dysregulation through the enzyme peptidyl arginine deiminase 4 (PAD4) in collagen-induced arthritis (CIA) mouse model. Our findings demonstrate that ferroptosis exacerbates RA-related inflammatory responses and joint damage by upregulating PAD4 expression, which, in turn, influences the gut microbial composition and associated metabolite profiles. Erastin, a known ferroptosis agonist, significantly increased the relative abundance of pro-inflammatory bacteria such as Proteobacteria while reducing beneficial taxa like Firmicutes and Bacteroidetes. This microbial shift was associated with heightened oxidative stress and an imbalance in key metabolites, such as lysophosphatidyl ethanolamine 14:0 (LysoPE 14:0), further exacerbated by ferroptosis. Co-treatment with GSK484, a PAD4 inhibitor, reversed these effects, restoring microbial homeostasis and reducing joint inflammation. This study suggests that ferroptosis-mediated PAD4 activity contributes to RA pathogenesis by disrupting the gut-joint axis, providing novel insights into potential therapeutic targets for RA. Our results highlight the intricate interplay between immune-mediated cell death, gut microbiota, and systemic inflammation, emphasizing the importance of ferroptosis as a therapeutic target in mitigating RA progression.
Collapse
Affiliation(s)
- Xiaoying Zhu
- Department of Rheumatology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Hanya Lu
- Department of Rheumatology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Wenjing Li
- Department of Rheumatology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Sijia Niu
- Department of Rheumatology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Jiawei Xue
- Department of Rheumatology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Haoyuan Sun
- Department of Orthopedics, Heilongjiang Provincial Hospital, Harbin 150001, China
| | - Juan Zhang
- Department of Rheumatology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China.
| | - Zhiyi Zhang
- Department of Rheumatology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China.
| |
Collapse
|
23
|
Wang L, Cui Y, Han B, Du Y, Salewala KS, Wang S, Zhao W, Zhang H, Wang S, Xu X, Ma J, Zhu Y, Tuo H. Gut microbiota and Parkinson's disease. Chin Med J (Engl) 2025; 138:289-297. [PMID: 39501822 PMCID: PMC11771718 DOI: 10.1097/cm9.0000000000003318] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Indexed: 01/29/2025] Open
Abstract
ABSTRACT Emerging evidence suggests that dysbiosis of the gut microbiota is associated with the pathogenesis of Parkinson's disease (PD), a prevalent neurodegenerative disorder. The microbiota-gut-brain axis plays a crucial role in the development and progression of PD, and numerous studies have demonstrated the potential therapeutic benefits of modulations in the intestinal microbiota. This review provides insights into the characterization of the gut microbiota in patients with PD and highlights associations with clinical symptoms and underlying mechanisms. The discussion underscores the increased influence of the gut microbiota in the pathogenesis of PD. While the relationship is not fully elucidated, existing research demonstrates a strong correlation between changes in the composition of gut microbiota and disease development, and further investigation is warranted to explain the specific underlying mechanisms.
Collapse
Affiliation(s)
- Lin Wang
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Ying Cui
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Bingyu Han
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Yitong Du
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | | | - Shiya Wang
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Wenlu Zhao
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Hongxin Zhang
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Sichen Wang
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Xinran Xu
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Jianpeng Ma
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Yan Zhu
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Houzhen Tuo
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| |
Collapse
|
24
|
Gao W, Wang Y, Wang F, Wu X, Lu F, Liu F. Ergothioneine exerts neuroprotective effects in Parkinson's disease: Targeting α-synuclein aggregation and oxidative stress. Food Res Int 2025; 201:115590. [PMID: 39849723 DOI: 10.1016/j.foodres.2024.115590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 11/16/2024] [Accepted: 12/28/2024] [Indexed: 01/25/2025]
Abstract
Ergothioneine (EGT) is a natural dietary antioxidant derived from certain edible mushrooms, commonly used as a food additive and supplement, but its effects on Parkinson's Disease (PD) are still unclear. The accumulation of α-synuclein (α-syn) plays a pivotal role in the pathogenesis and development of PD. Here, this study demonstrated that EGT effectively inhibits α-syn aggregation, disrupts mature fibers, and reduces associated cytotoxicity and oxidative stress. The beneficial effects of EGT were confirmed in Caenorhabditis elegans, where it protected dopaminergic neurons, prolonged lifespan and enhanced behavioral functions by reducing α-syn plaque accumulation and associated oxidative stress. Molecular dynamics simulation revealed that EGT interacts directly with α-syn pentamer through van der Waals and electrostatic forces, disrupting the structural stability of the preformed pentamer. Furthermore, animal studies validated that EGT alleviated neuronal damage and improved behavioral deficits by reducing α-syn aggregation, oxidative stress and inflammatory response. In conclusion, EGT presents promising potential as a dietary supplement for preventing and alleviating PD.
Collapse
Affiliation(s)
- Wen Gao
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin 300457, PR China; Tianjin Key Laboratory of Industrial Microbiology, Tianjin 300457, PR China; College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Yang Wang
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin 300457, PR China; Tianjin Key Laboratory of Industrial Microbiology, Tianjin 300457, PR China; College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Fuhao Wang
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin 300457, PR China; Tianjin Key Laboratory of Industrial Microbiology, Tianjin 300457, PR China; College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Xinni Wu
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin 300457, PR China; Tianjin Key Laboratory of Industrial Microbiology, Tianjin 300457, PR China; College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Fuping Lu
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin 300457, PR China; Tianjin Key Laboratory of Industrial Microbiology, Tianjin 300457, PR China; College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Fufeng Liu
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin 300457, PR China; Tianjin Key Laboratory of Industrial Microbiology, Tianjin 300457, PR China; College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, PR China.
| |
Collapse
|
25
|
Ma J, Liu Y, Hu J, Liu X, Xia Y, Xia W, Shen Z, Kong X, Wu X, Mao L, Li Q. Tirzepatide administration improves cognitive impairment in HFD mice by regulating the SIRT3-NLRP3 axis. Endocrine 2025; 87:486-497. [PMID: 39222203 DOI: 10.1007/s12020-024-04013-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
PURPOSE High-fat diet (HFD) currently is reported that in connection with cognitive impairment. Tirzepatide is a novel dual receptor agonist for glycemic control. But whether Tirzepatide exerts a protective effect in HFD-related cognitive impairment remains to be explore. METHODS During the study, the cognitive dysfunction mice model induced by HFD were established. The expressions synapse-associated protein and other target proteins were detected. The oxidative stress parameters, levels of inflammatory cytokine were also detected. RESULTS Our findings proved that Tirzepatide administration attenuates high fat diet-related cognitive impairment. Tirzepatide administration suppresses microglia activation, alleviates oxidative stress as well as suppressed the expression of NLRP3 in HFD mice by up-regulating SIRT3 expression. In conclusion, Tirzepatide attenuates HFD-induced cognitive impairment through reducing oxidative stress and neuroinflammation via SIRT3-NLRP3 signaling. CONCLUSION This study suggest that Tirzepatide has neuroprotective effects in HFD-related cognitive dysfunction mice model, which provides a promising treatment of HFD-related cognitive impairment.
Collapse
Affiliation(s)
- Jingjing Ma
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Qinhuai District, Nanjing, 21006, China
- Department of Endocrinology, The Affifiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, 223300, China
| | - Yuanyuan Liu
- Department of Endocrinology, The Affifiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, 223300, China
| | - Junya Hu
- Department of Pharmacy, Nanjing First Hospital, China Pharmaceutical University, Nanjing, 210006, China
| | - Xingjing Liu
- Department of Endocrinology, The Affifiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, 223300, China
| | - Yin Xia
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Qinhuai District, Nanjing, 21006, China
| | - Wenqing Xia
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Qinhuai District, Nanjing, 21006, China
| | - Ziyang Shen
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Qinhuai District, Nanjing, 21006, China
| | - Xiaocen Kong
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Qinhuai District, Nanjing, 21006, China
| | - Xia Wu
- Department of Endocrinology, Affiliated Jiangyin Hospital of Nantong University, Wuxi, 214400, China
| | - Li Mao
- Department of Endocrinology, The Affifiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, 223300, China.
| | - Qian Li
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Qinhuai District, Nanjing, 21006, China.
| |
Collapse
|
26
|
Jiang M, Chi J, Qiao Y, Wang J, Zhang Z, Liu J, Sheng X, Yuan L. Ginsenosides Rg1, Rb1 and rare ginsenosides: Promising candidate agents for Parkinson's disease and Alzheimer's disease and network pharmacology analysis. Pharmacol Res 2025; 212:107578. [PMID: 39756554 DOI: 10.1016/j.phrs.2025.107578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 12/12/2024] [Accepted: 01/01/2025] [Indexed: 01/07/2025]
Abstract
Ginseng has been commonly used as a traditional Chinese medicine in Asian countries for thousands of years. Ginsenosides are the main pharmacologically active ingredients isolated from ginseng and have neuroprotective effects in the treatment of neurodegenerative disorders, such as Parkinson's disease (PD) and Alzheimer's disease (AD). To summarise and investigate the protective roles of ginsenosides and their underlying mechanisms in PD and AD, we used ''Ginsenoside", ''Parkinson's disease", ''Alzheimer's disease", ''anti-inflammatory", ''antioxidant", and ''apoptosis" as keywords to search and extract relevant literature information from scientific databases such as Elsevier, PubMed, and Google Scholar databases. In particular, we used network pharmacology to identify the potential targets of ginsenosides Rg1 and Rb1 in PD and AD. By analysing the existing research advances and network pharmacology results, we found that the neuroprotective effects of ginsenosides, primarily mediated through anti-inflammation, anti-apoptosis and anti-oxidative stress, etc, may be associated with the PI3K/Akt, BDNF/TrkB, MAPKs, NF-κB, Nrf2 and Wnt/β-catenin signalling pathways. This review systematically summarises the different roles and mechanisms of ginsenosides Rg1, Rb1, and rare ginsenosides in PD and AD and provides new strategies for the treatment of neurodegenerative disorders. Network pharmacology provides a new research paradigm for the treatment of PD and AD using Rg1 and Rb1.
Collapse
Affiliation(s)
- Mingchun Jiang
- School of Clinical Medicine and Basic Medical Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China; The Second Affiliated Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China
| | - Jiaxin Chi
- School of Clinical Medicine and Basic Medical Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China
| | - Yifan Qiao
- School of Clinical Medicine and Basic Medical Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China
| | - Jinpeng Wang
- School of Clinical Medicine and Basic Medical Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China
| | - Zhixin Zhang
- School of pharmacy, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China
| | - Jia Liu
- School of Clinical Medicine and Basic Medical Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China
| | - Xinhao Sheng
- School of Clinical Medicine and Basic Medical Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China
| | - Liangjie Yuan
- School of Clinical Medicine and Basic Medical Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China; The Second Affiliated Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China.
| |
Collapse
|
27
|
Yin D, Zhao L, Deng S, Xie Y, Ro KS, Yang Z, Du L, Xie J, Wei D. Lactiplantibacillus plantarum X7022 Plays Roles on Aging Mice with Memory Impairment Induced by D-Galactose Through Restoring Neuronal Damage, Relieving Inflammation and Oxidative Stress. Probiotics Antimicrob Proteins 2025; 17:1-14. [PMID: 38183568 DOI: 10.1007/s12602-023-10208-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2023] [Indexed: 01/08/2024]
Abstract
In this study, Lactiplantibacillus plantarum X7022 was applied to ameliorate memory impairment of aging mice induced by D-galactose. The strain showed specific choloylglycine hydrolysis ability based on in vitro investigation. Morris water maze test showed L. plantarum X7022 administration improved learning ability and spatial memory of aging mice. The gavage of L. plantarum X7022 displayed a promising ability of relieving cerebral oxidative stress and hippocampal inflammatory condition according to the increased GSH level and SOD activity and decreased MDA level, as well as decreased TNF-α, IL-1β, and IL-6 levels. The intervention with the strain could protect neuron by regulating cell apoptosis and AChE overexpression and inhibiting amyloid-β deposition, as well as affect neuron functions by regulating CREB-BDNF signaling pathways and iNOS expression. Besides, the strain could improve fecal SCFA contents and increase the abundance of anti-inflammatory and antioxidant-related genera such as Lactobacillus, Akkermansia, and Adlercreutzia. These results suggest that L. plantarum X7022 could be a prospective therapeutic alternative for the improvement of memory impairment among the elderly.
Collapse
Affiliation(s)
- Deyi Yin
- State Key Laboratory of Bioreactor Engineering, Department of Food Science and Technology, School of Biotechnology, East China University of Science and Technology, 130 # Meilong Rd, Shanghai, 200237, People's Republic of China
| | - Li Zhao
- State Key Laboratory of Bioreactor Engineering, Department of Food Science and Technology, School of Biotechnology, East China University of Science and Technology, 130 # Meilong Rd, Shanghai, 200237, People's Republic of China
| | - Sijing Deng
- State Key Laboratory of Bioreactor Engineering, Department of Food Science and Technology, School of Biotechnology, East China University of Science and Technology, 130 # Meilong Rd, Shanghai, 200237, People's Republic of China
| | - Yaqi Xie
- State Key Laboratory of Bioreactor Engineering, Department of Food Science and Technology, School of Biotechnology, East China University of Science and Technology, 130 # Meilong Rd, Shanghai, 200237, People's Republic of China
| | - Kum-Song Ro
- State Key Laboratory of Bioreactor Engineering, Department of Food Science and Technology, School of Biotechnology, East China University of Science and Technology, 130 # Meilong Rd, Shanghai, 200237, People's Republic of China
- Department of Biotechnology, Faculty of Life Science, Kim Hyong Jik University of Education, Pyongyang, 999093, Democratic People's Republic of Korea
| | - Zeyong Yang
- Department of Anesthesiology, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Embryo Original Disease, Shanghai Municipal Key Clinical Specialty, Shanghai, 200030, People's Republic of China
| | - Lei Du
- State Key Laboratory of Bioreactor Engineering, Department of Food Science and Technology, School of Biotechnology, East China University of Science and Technology, 130 # Meilong Rd, Shanghai, 200237, People's Republic of China.
| | - Jingli Xie
- State Key Laboratory of Bioreactor Engineering, Department of Food Science and Technology, School of Biotechnology, East China University of Science and Technology, 130 # Meilong Rd, Shanghai, 200237, People's Republic of China.
- Shanghai Collaborative Innovation Center for Biomanufacturing (SCICB), Shanghai, 200237, People's Republic of China.
| | - Dongzhi Wei
- State Key Laboratory of Bioreactor Engineering, Department of Food Science and Technology, School of Biotechnology, East China University of Science and Technology, 130 # Meilong Rd, Shanghai, 200237, People's Republic of China
- Shanghai Collaborative Innovation Center for Biomanufacturing (SCICB), Shanghai, 200237, People's Republic of China
| |
Collapse
|
28
|
Qin Y, Zhou Y, Xiong J, Lu C, Zhou J, Su X, Han J. Limosilactobacillus reuteri RE225 alleviates gout by modulating the TLR4/MyD88/NF-κB inflammatory pathway and the Nrf2/HO-1 oxidative stress pathway, and by regulating gut microbiota. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2025; 105:1185-1193. [PMID: 39297558 DOI: 10.1002/jsfa.13908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/25/2024] [Accepted: 08/29/2024] [Indexed: 12/12/2024]
Abstract
BACKGROUND Gout poses a significant health threat. The use of Lactobacillus from the gut microbiota is one potential remedy. However, the intricate molecular mechanisms governing the impact of Lactobacillus on gout remain largely uncharted. In this study, a strain of Limosilactobacillus reuteri RE225 was separated from the gut of mice and colitis was treated with polypeptide intervention. RESULTS Limosilactobacillus reuteri RE225 reduced foot tumefaction markedly in mice with gout and extended the pain threshold time in their feet. It also improved the health of gut microbiota. Intervention with L. reuteri RE225 also suppressed the TLR4/MyD88/NF-κB and nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) pathways in the mice, reduced the levels of pro-inflammatory cytokines - interleukin 1β (IL-1β), interleukin 6 (IL-6), and tumor necrosis factor-α (TNF-α) - and increased the level of the anti-inflammatory cytokine interleukin 10 (IL-10), thereby mitigating inflammation. CONCLUSION This study provides a theoretical basis for the comprehensive development of Limosilactobacillus reuteri and new ideas for the non-pharmacological treatment of gout. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Yang Qin
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Science, Ningbo University, Ningbo, China
| | - Yucong Zhou
- College of Biological and Environmental Science, Zhejiang Wanli University, Ningbo, China
| | - Jiayi Xiong
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Science, Ningbo University, Ningbo, China
| | - Chenyang Lu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Science, Ningbo University, Ningbo, China
| | - Jun Zhou
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Science, Ningbo University, Ningbo, China
| | - Xiurong Su
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Science, Ningbo University, Ningbo, China
| | - Jiaojiao Han
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Science, Ningbo University, Ningbo, China
| |
Collapse
|
29
|
Kumar D, Bishnoi M, Kondepudi KK, Sharma SS. Gut Microbiota-Based Interventions for Parkinson's Disease: Neuroprotective Mechanisms and Current Perspective. Probiotics Antimicrob Proteins 2025:10.1007/s12602-024-10433-x. [PMID: 39809955 DOI: 10.1007/s12602-024-10433-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2024] [Indexed: 01/16/2025]
Abstract
Recent evidence links gut microbiota alterations to neurodegenerative disorders, including Parkinson's disease (PD). Replenishing the abnormal composition of gut microbiota through gut microbiota-based interventions "prebiotics, probiotics, synbiotics, postbiotics, and fecal microbiota transplantation (FMT)" has shown beneficial effects in PD. These interventions increase gut metabolites like short-chain fatty acids (SCFAs) and glucagon-like peptide-1 (GLP-1), which may protect dopaminergic neurons via the gut-brain axis. Neuroprotective effects of these interventions are mediated by several mechanisms, including the enhancement of neurotrophin and activation of the PI3K/AKT/mTOR signaling pathway, GLP-1-mediated gut-brain axis signaling, Nrf2/ARE pathway, and autophagy. Other pathways, such as free fatty acid receptor activation, synaptic plasticity improvement, and blood-brain and gut barrier integrity maintenance, also contribute to neuroprotection. Furthermore, the inhibition of the TLR4/NF-кB pathway, MAPK pathway, GSK-3β signaling pathway, miR-155-5p-mediated neuroinflammation, and ferroptosis could account for their protective effects. Clinical studies involving gut microbiota-based interventions have shown therapeutic benefits in PD patients, particularly in improving gastrointestinal dysfunction and some neurological symptoms. However, the effectiveness in alleviating motor symptoms remains mild. Large-scale clinical trials are still needed to confirm these findings. This review emphasizes the neuroprotective mechanisms of gut microbiota-based interventions in PD as supported by both preclinical and clinical studies.
Collapse
Affiliation(s)
- Deepak Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Mohali, Punjab, 160062, India
| | - Mahendra Bishnoi
- Centre for Excellence in Functional Foods, Division of Food and Nutritional Biotechnology, National Agri-Food Biomanufacturing Institute (NABI), Knowledge City-Sector 81, S.A.S. Nagar, Punjab, 140306, India
| | - Kanthi Kiran Kondepudi
- Centre for Excellence in Functional Foods, Division of Food and Nutritional Biotechnology, National Agri-Food Biomanufacturing Institute (NABI), Knowledge City-Sector 81, S.A.S. Nagar, Punjab, 140306, India
| | - Shyam Sunder Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Mohali, Punjab, 160062, India.
| |
Collapse
|
30
|
Ma Y, Tu Y, Chen Y, Chen X, Pan X, Sun M, Fu X, Zou J, Gao F. An Oral H 2S Responsive Cu 5.4O Nanozyme Platform with Strong ROS/H 2S Scavenging Capacity for the Treatment of Colitis. ACS APPLIED MATERIALS & INTERFACES 2025; 17:617-631. [PMID: 39722133 DOI: 10.1021/acsami.4c17782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2024]
Abstract
Inflammatory bowel disease involves excess reactive oxygen species (ROS) and hydrogen sulfide (H2S) at inflammatory sites. Nanozyme-mediated ROS and H2S scavenging therapy is promising for colitis treatment. Here, we synthesized a multiple ROS scavenging Cu5.4O nanoparticle and first explored its H2S scavenging capacity. Chitosan oligosaccharide modified with alpha-lipoic acid was coated on the nanoparticles to further enhance the H2S scavenging capacity. Furthermore, calcium alginate was coated on the surface to develop an oral nanoplatform (Cu5.4O@SAG) possessing dual-pH/H2S-responsive release characteristics. Importantly, Cu5.4O@SAG exhibited enrichment at the colonic inflammation site and relieved the inflammatory index, containing the recovery of colon length, spleen index, liver index, and body weight, as well as inflammatory cell infiltration. In vivo and in vitro experiments revealed the dual ROS and H2S scavenging capacities of the nanoplatform. Additionally, Cu5.4O@SAG regulated tight junctions, mucus layers, and gut microbiota, which was accompanied by the downregulation of inflammatory cytokines. Notably, Cu5.4O@SAG also had excellent biocompatibility. In conclusion, this oral multiple-scavenging nanozyme platform provides a new and safe paradigm for the development of nanozymes for colitis treatment.
Collapse
Affiliation(s)
- Ying Ma
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yixing Tu
- Department of Pharmacy, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yang Chen
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Xinyi Chen
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Xier Pan
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Mingyue Sun
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Xiuzhi Fu
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Jiafeng Zou
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China
- Pharmaceutical Engineering and Process of Chemical Engineering Research Center of Ministry of Education, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Feng Gao
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China
- Pharmaceutical Engineering and Process of Chemical Engineering Research Center of Ministry of Education, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
31
|
Li M, Chen M, Li H, Gao D, Zhao L, Zhu M. Glial cells improve Parkinson's disease by modulating neuronal function and regulating neuronal ferroptosis. Front Cell Dev Biol 2025; 12:1510897. [PMID: 39830208 PMCID: PMC11739109 DOI: 10.3389/fcell.2024.1510897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 12/11/2024] [Indexed: 01/22/2025] Open
Abstract
The main characteristics of Parkinson's disease (PD) are the loss of dopaminergic (DA) neurons and abnormal aggregation of cytosolic proteins. However, the exact pathogenesis of PD remains unclear, with ferroptosis emerging as one of the key factors driven by iron accumulation and lipid peroxidation. Glial cells, including microglia, astrocytes, and oligodendrocytes, serve as supportive cells in the central nervous system (CNS), but their abnormal activation can lead to DA neuron death and ferroptosis. This paper explores the interactions between glial cells and DA neurons, reviews the changes in glial cells during the pathological process of PD, and reports on how glial cells regulate ferroptosis in PD through iron homeostasis and lipid peroxidation. This opens up a new pathway for basic research and therapeutic strategies in Parkinson's disease.
Collapse
Affiliation(s)
- Mengzhu Li
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Mengxuan Chen
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Haiyan Li
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Da Gao
- Shenzhen Clinical College of Integrated Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Lijun Zhao
- Shenzhen Clinical College of Integrated Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Meiling Zhu
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| |
Collapse
|
32
|
Kim JH, Choi Y, Lee S, Oh MS. Probiotics as Potential Treatments for Neurodegenerative Diseases: a Review of the Evidence from in vivo to Clinical Trial. Biomol Ther (Seoul) 2025; 33:54-74. [PMID: 39676295 PMCID: PMC11704393 DOI: 10.4062/biomolther.2024.215] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 12/08/2024] [Accepted: 12/09/2024] [Indexed: 12/17/2024] Open
Abstract
Neurodegenerative diseases (NDDs), characterized by the progressive deterioration of the structure and function of the nervous system, represent a significant global health challenge. Emerging research suggests that the gut microbiota plays a critical role in regulating neurodegeneration via modulation of the gut-brain axis. Probiotics, defined as live microorganisms that confer health benefits to the host, have garnered significant attention owing to their therapeutic potential in NDDs. This review examines the current research trends related to the microbiome-gut-brain axis across various NDDs, highlighting key findings and their implications. Additionally, the effects of specific probiotic strains, including Lactobacillus plantarum, Bifidobacterium breve, and Lactobacillus rhamnosus, on neurodegenerative processes were assessed, focusing on their potential therapeutic benefits. Overall, this review emphasizes the potential of probiotics as promising therapeutic agents for NDDs, underscoring the importance of further investigation into this emerging field.
Collapse
Affiliation(s)
- Jin Hee Kim
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Yujin Choi
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Seungmin Lee
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Myung Sook Oh
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Oriental Pharmaceutical Science and Kyung Hee East-West Pharmaceutical Research Institute, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
33
|
Wang R, Deng L, Wang Y, Liu N, Yang M, Qiu J, Chen C. Synergistic effects of combined lead and iprodione exposure on P53 signaling-mediated hepatotoxicity, enterotoxicity and transgenerational toxicity in zebrafish. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 958:178127. [PMID: 39708747 DOI: 10.1016/j.scitotenv.2024.178127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 11/04/2024] [Accepted: 12/12/2024] [Indexed: 12/23/2024]
Abstract
Environmental heavy metal contamination, combined with inappropriate use of fungicides, has led to the co-existence of lead (Pb) and iprodione (IPR), presenting signification risks to ecosystems and human health. The toxic effects resulting from concurrent exposure to Pb and IPR, however, remain poorly understood. In the study, we conducted a comprehensive 60-day subchronic study to investigate the toxic effects on the liver and gut in parental male zebrafish through employing multi-omics analyses. We also explored the potential transgenerational toxicity to unexposed offspring embryos. The results demonstrated that exposure to both Pb and IPR exacerbated intestinal pathological damage, decreased the expression of intestinal tight junction molecules, and activated the expression of intestinal inflammatory molecules in the gut. Metabolic and microbial analyses, utilizing 16S rRNA sequencing and non-targeted metabolic profiling, revealed alterations in the intestinal flora structure and disruptions in metabolite synthesis. Notably, we observed a significant negative correlation between the abundance of the Lactobacillus genus and uracil synthesis. Furthermore, liver RNA-seq analysis identified a marked enrichment of the P53 signaling pathway, confirmed by the activation of P53-mediated apoptotic markers, which was consistent with the observed increase in inflammatory infiltration and pathological damage within the liver. Importantly, P53-mediated apoptosis and inflammatory responses were activated in offspring embryos, suggesting that long-term parental exposure to Pb and IPR may induce transgenerational toxicity, potentially impacting offspring health. Despite the identification of these molecular changes, the phenotypic effects remain to be elucidated. Future studies are necessary to evaluate the potential phenotypic changes in offspring to fully understand the long-term effects of Pb and IPR exposure. Overall, these findings enhance the understanding of the molecular mechanisms underlying the toxic effects of Pb and IPR and emphasize the importance of a comprehensive risk assessment of environmental pollutants.
Collapse
Affiliation(s)
- Ruike Wang
- School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong, China
| | - Ligang Deng
- Institute of Agricultural Quality Standards and Testing Technology Research, Shandong Academy of Agricultural Sciences, Jinan, China; Shandong Provincial Key Laboratory of Test Technology on Food Quality and Safety, Jinan, China
| | - Yanhua Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Quality and Standard for Agro-products, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, Zhejiang, China
| | - Na Liu
- School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong, China
| | - Menglian Yang
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong, China
| | - Jing Qiu
- Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing, People's Republic of China
| | - Chen Chen
- School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong, China.
| |
Collapse
|
34
|
Li S, Liu Y, Lu S, Xu J, Liu X, Yang D, Yang Y, Hou L, Li N. A crazy trio in Parkinson's disease: metabolism alteration, α-synuclein aggregation, and oxidative stress. Mol Cell Biochem 2025; 480:139-157. [PMID: 38625515 DOI: 10.1007/s11010-024-04985-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 03/06/2024] [Indexed: 04/17/2024]
Abstract
Parkinson's disease (PD) is an aging-associated neurodegenerative disorder, characterized by the progressive loss of dopaminergic neurons in the pars compacta of the substantia nigra and the presence of Lewy bodies containing α-synuclein within these neurons. Oligomeric α-synuclein exerts neurotoxic effects through mitochondrial dysfunction, glial cell inflammatory response, lysosomal dysfunction and so on. α-synuclein aggregation, often accompanied by oxidative stress, is generally considered to be a key factor in PD pathology. At present, emerging evidences suggest that metabolism alteration is closely associated with α-synuclein aggregation and PD progression, and improvement of key molecules in metabolism might be potentially beneficial in PD treatment. In this review, we highlight the tripartite relationship among metabolic changes, α-synuclein aggregation, and oxidative stress in PD, and offer updated insights into the treatments of PD, aiming to deepen our understanding of PD pathogenesis and explore new therapeutic strategies for the disease.
Collapse
Affiliation(s)
- Sheng Li
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Yanbing Liu
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Sen Lu
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Jiayi Xu
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Xiaokun Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Di Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Yuxuan Yang
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Lin Hou
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Ning Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
35
|
Chen K, Wang J, Hu T, Zhao Y, Wu Y, Wang X, Li W, Yang G, Zhang L, Wang J, Zhu Y. Salmonella enterica serovar typhimurium effectors spiA and spiC promote replication by modulating iron metabolism and oxidative stress. Vet Microbiol 2025; 300:110328. [PMID: 39674031 DOI: 10.1016/j.vetmic.2024.110328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 10/22/2024] [Accepted: 11/30/2024] [Indexed: 12/16/2024]
Abstract
Salmonella enterica serovar Typhimurium (S. Typhimurium) poses a major threat to the health and safety of animal-derived foods worldwide. Recently, we have reported that S. Typhimurium uses iron to promote its own proliferation, leading to iron metabolism disorders. However, the mechanism by which S. Typhimurium induces iron metabolism disturbances remains unclear. In this study, we found that the S. Typhimurium effectors spiA and spiC promote the expression of iron regulatory protein 2 (IRP2), transferrin receptor 1 (TfR1) and divalent metal transporter protein 1 (DMT1) and inhibit the expression of ferroportin after transfection with the recombinant plasmids pEGFP-C1-spiA and pEGFP-C1-spiC, which in turn contributes to the accumulation of iron and oxidative stress. Furthermore, we aimed to verify the role of these two effector proteins in S. Typhimurium-induced disorders of iron metabolism. We constructed spiA or spiC mutant strains and their corresponding complementation strains. Our data showed that when spiA or spiC was knocked out, the upregulation of iron metabolism proteins (IRP2, TfR1 and DMT1), the accumulation of iron and oxidative stress caused by the wild-type strain were clearly alleviated in vitro and in vivo, which plays a key role in reducing the intracellular replication of S. Typhimurium and attenuating pathological damage to the liver and ileum of mice. Our findings highlighted that S. Typhimurium induces the disruption of iron metabolism via the virulence factors spiA and spiC, thereby facilitating S. Typhimurium proliferation and causing oxidative damage to the liver and ileum, which provides prospective insights into the search for effective antimicrobial targets for the defense against salmonellosis.
Collapse
Affiliation(s)
- Keyuan Chen
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China; College of Veterinary Medicine, Sanya Institute of China Agricultural University, Sanya 572000, China.
| | - Jing Wang
- Key Laboratory of Applied Biotechnology on Animal Science & Veterinary Medicine of Zhejiang Province, Zhejiang Engineering Research Center for Veterinary Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, Belt and Road International Joint Laboratory for One Health and Food Safety, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Veterinary Medicine of Zhejiang A&F University, 666 Wusu Street, Lin'an District, Hangzhou, Zhejiang Province 311300, China.
| | - Ting Hu
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China; College of Veterinary Medicine, Sanya Institute of China Agricultural University, Sanya 572000, China.
| | - Yiqing Zhao
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China; College of Veterinary Medicine, Sanya Institute of China Agricultural University, Sanya 572000, China.
| | - Yi Wu
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China; College of Veterinary Medicine, Sanya Institute of China Agricultural University, Sanya 572000, China.
| | - Xue Wang
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China; College of Veterinary Medicine, Sanya Institute of China Agricultural University, Sanya 572000, China.
| | - Wei Li
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China; College of Veterinary Medicine, Sanya Institute of China Agricultural University, Sanya 572000, China.
| | - Guiyan Yang
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China; College of Veterinary Medicine, Sanya Institute of China Agricultural University, Sanya 572000, China.
| | - Linlin Zhang
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China; College of Veterinary Medicine, Sanya Institute of China Agricultural University, Sanya 572000, China.
| | - Jiufeng Wang
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China; College of Veterinary Medicine, Sanya Institute of China Agricultural University, Sanya 572000, China.
| | - Yaohong Zhu
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China; College of Veterinary Medicine, Sanya Institute of China Agricultural University, Sanya 572000, China.
| |
Collapse
|
36
|
Liu B, Zhang Z, Zhao J, Li X, Wang Y, Liu L, Qiao W, Chen L. Lactiplantibacillus plantarum HM-P2 influences gestational gut microbiome and microbial metabolism. Front Nutr 2024; 11:1489359. [PMID: 39758313 PMCID: PMC11695228 DOI: 10.3389/fnut.2024.1489359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 12/02/2024] [Indexed: 01/07/2025] Open
Abstract
Introduction Human milk-derived probiotics are beneficial bacteria that provide gestational health benefits, for both pregnant women and their offspring. The study aims to investigate whether the administration of human milk-derived probiotic L. plantarum HM-P2 could effectively influence gestational health. Methods The gestational humanized microbiome model was built by fecal microbiome transplant from gestational women into germ-free (GF) mice. Results HM-P2 was successfully planted and increased the top crypt depth of the colon, and microbes such as L. reuteri, Anaerofilum sp. An201, and Gemmiger were up-regulated in the HM-P2 group throughout gestation. HM-P2 significantly promoted the contents of intestinal caproic acid, bile acids, and tryptophan catabolites such as serotonin. Gut microbes were associated with these bile acids and tryptophans. Discussion HM-P2 could modulate the microbial community and microbial metabolites in gestational humanized GF mice. This probiotic strain could be a potential gestational dietary supplement with health benefits.
Collapse
Affiliation(s)
- Bin Liu
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing, China
| | - Zhenzhen Zhang
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing, China
| | - Junying Zhao
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing, China
| | - Xianping Li
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing, China
| | - Yaru Wang
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing, China
| | - Lu Liu
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing, China
| | - Weicang Qiao
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing, China
| | - Lijun Chen
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing, China
| |
Collapse
|
37
|
Wang X, Hu R, Lin F, Yang T, Lu Y, Sun Z, Li T, Chen J. Lactobacillus reuteri or Lactobacillus rhamnosus GG intervention facilitates gut barrier function, decreases corticosterone and ameliorates social behavior in LPS-exposed offspring. Food Res Int 2024; 197:115212. [PMID: 39593298 DOI: 10.1016/j.foodres.2024.115212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/24/2024] [Accepted: 10/17/2024] [Indexed: 11/28/2024]
Abstract
Probiotic therapy with Lactobacillus reuteri and Lactobacillus rhamnosus (LGG) demonstrates potential as an adjunctive treatment for autism spectrum disorder (ASD). In a rat model of ASD induced by lipopolysaccharide (LPS) injection during pregnancy, we evaluated the effects of these probiotics on offspring. Administration of L. reuteri or LGG for three weeks post-birth improved social deficits and reduced anxiety in LPS-exposed rats. Additionally, probiotics significantly modified short-chain fatty acid profiles, increasing butyric acid levels and decreasing propionic acid levels. They also enhanced colonic barrier integrity by upregulating tight junction proteins, including ZO-1, Occludin, and Claudin4. RNA sequencing identified differential gene expression in pathways related to inflammation, the HPA axis, and reactive oxygen species metabolism, with NADPH oxidase 1 (NOX1) emerging as a crucial gene. Validation studies confirmed that Lactobacillus strains reduced inflammatory cytokines, inhibited corticosterone secretion, increased antioxidant levels, and suppressed the NF-κB/NOX1 pathway. In an H2O2-induced oxidative stress model using Caco-2 cells, pre-treatment with L. reuteri, LGG, or NF-κB inhibitors enhanced cellular antioxidants, inhibited NF-κB/NOX1 activation, and improved barrier function. Overall, L. reuteri and LGG administration improved social behavior, bolstered colonic barrier function, and mitigated HPA axis overactivation in LPS-exposed rats, while also alleviating oxidative stress in the colon and Caco-2 cells. These findings suggest that L. reuteri and LGG have substantial clinical potential for ASD treatment by targeting multiple pathophysiological mechanisms, including inflammation, HPA axis dysregulation, and oxidative stress, thereby presenting a promising adjunctive therapeutic strategy for enhancing social behavior and gut health in ASD.
Collapse
Affiliation(s)
- Xinyuan Wang
- Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing, China
| | - Riqiang Hu
- Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing, China
| | - Fang Lin
- Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing, China
| | - Ting Yang
- Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing, China
| | - Yiwen Lu
- School of Medicine, Jianghan University, Wuhan, China
| | - Zhujun Sun
- Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing, China
| | - Tingyu Li
- Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing, China
| | - Jie Chen
- Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing, China.
| |
Collapse
|
38
|
Li H, Shen N, Ren J, Yang S, Chen Y, Gao Z. Biotransformation characteristics of urate-lowering probiotic fermented apple juice and potential regulatory mechanisms for ameliorating hyperuricemia via mediating gut microbiota and metabolic pathways. Food Chem 2024; 460:140462. [PMID: 39032298 DOI: 10.1016/j.foodchem.2024.140462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 06/16/2024] [Accepted: 07/12/2024] [Indexed: 07/23/2024]
Abstract
Hyperuricemia has evolved into a global public health concern, and applying probiotics fermented apple juice holds promise for alleviating this condition. This study aimed to investigate the biotransformation and metabolic features of urate-lowering probiotics sequentially fermented dealcoholized apple juice (PSFA), and assess its ameliorative effects and potential mechanisms on hyperuricemia mice. Results showed that CICC 6074 and 20,292 possessed excellent purine, nucleotide and nucleoside degradation and acid and bile salt resistance; sequential fermentation decreased the fructose in apple juice, and viable counts reached 3.76 × 108 CFU/mL. Histopathological analysis showed that PSFA ameliorated kidney damage in hyperuricemia mice. Furthermore, PSFA significantly reduced Urea, Creatinine and Uric acid levels in hyperuricemia mice; and inhibited xanthine oxidase activity and the expression of pro-inflammatory factors. Importantly, PSFA reversed gut microbiota dysbiosis and raised the abundance of beneficial bacteria (Lactobacillush, Faecalibaculum and Lachnospiraceae_NK4A136_group). KEGG and COG functional prediction results revealed that the potential mechanism of PSFA to ameliorate hyperuricemia may be lipid metabolism and glycolysis pathways.
Collapse
Affiliation(s)
- Hongcai Li
- College of Food Science and Engineering, Northwest A&F University, 712100 Yangling, Shaanxi, People's Republic of China
| | - Ning Shen
- College of Food Science and Engineering, Northwest A&F University, 712100 Yangling, Shaanxi, People's Republic of China
| | - Jiani Ren
- College of Food Science and Engineering, Northwest A&F University, 712100 Yangling, Shaanxi, People's Republic of China
| | - Shuang Yang
- College of Food Science and Engineering, Northwest A&F University, 712100 Yangling, Shaanxi, People's Republic of China
| | - Yue Chen
- College of Food Science and Engineering, Northwest A&F University, 712100 Yangling, Shaanxi, People's Republic of China
| | - Zhenpeng Gao
- College of Food Science and Engineering, Northwest A&F University, 712100 Yangling, Shaanxi, People's Republic of China.
| |
Collapse
|
39
|
He G, Long H, He J, Zhu C. The Immunomodulatory Effects and Applications of Probiotic Lactiplantibacillus plantarum in Vaccine Development. Probiotics Antimicrob Proteins 2024; 16:2229-2250. [PMID: 39101975 DOI: 10.1007/s12602-024-10338-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2024] [Indexed: 08/06/2024]
Abstract
Lactiplantibacillus plantarum (previously known as Lactobacillus plantarum) is a lactic acid bacterium that exists in various niches. L. plantarum is a food-grade microorganism that is commonly considered a safe and beneficial microorganism. It is widely used in food fermentation, agricultural enhancement, and environmental protection. L. plantarum is also part of the normal flora that can regulate the intestinal microflora and promote intestinal health. Some strains of L. plantarum are powerful probiotics that induce and modulate the innate and adaptive immune responses. Due to its outstanding immunoregulatory capacities, an increasing number of studies have examined the use of probiotic L. plantarum strains as natural immune adjuvants or alternative live vaccine carriers. The present review summarizes the main immunomodulatory characteristics of L. plantarum and discusses the preliminary immunological effects of L. plantarum as a vaccine adjuvant and delivery carrier. Different methods for improving the immune capacities of recombinant vector vaccines are also discussed.
Collapse
Affiliation(s)
- Guiting He
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang, 421001, Hunan, China
| | - Huanbing Long
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang, 421001, Hunan, China
| | - Jiarong He
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang, 421001, Hunan, China
| | - Cuiming Zhu
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang, 421001, Hunan, China.
| |
Collapse
|
40
|
Liang W, Gao Y, Zhao Y, Gao L, Zhao Z, He Z, Li S. Lactiplantibacillus plantarum ELF051 Alleviates Antibiotic-Associated Diarrhea by Regulating Intestinal Inflammation and Gut Microbiota. Probiotics Antimicrob Proteins 2024; 16:1996-2006. [PMID: 37639209 PMCID: PMC11573863 DOI: 10.1007/s12602-023-10150-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/22/2023] [Indexed: 08/29/2023]
Abstract
Probiotics are widely recognized for their ability to prevent and therapy antibiotic-associated diarrhea (AAD). This study was designed to evaluate Lactiplantibacillus plantarum ELF051 ability to prevent colon inflammation and its effect on gut microbial composition in a mouse model of AAD. The mice were intragastrically administered triple antibiotics for 7 days and then subjected to L. plantarum ELF051 for 14 days. The administration of L. plantarum ELF051 ameliorated the pathological changes in the colon tissue, downregulated interleukin (IL)-1β and tumor necrosis factor (TNF)-α, and upregulated IL-10, and increased the intestinal short-chain fatty acids (SCFAs) level. Lactiplantibacillus plantarum ELF051 also regulated the Toll-like receptor/myeloid differentiation primary response 88/nuclear factor kappa light chain enhancer of activated B cells (TLR4/MyD88/NF-κB) and the phosphatidylinositol 3-kinase/protein kinase B/ NF-κB (PI3K/AKT/ NF-κB) inflammatory signaling pathways. 16S rRNA analyses showed that L. plantarum ELF051 increased the abundance and diversity of gut bacteria, restoring gut microbiota imbalance. A Spearman's rank correlation analysis showed that lactobacilli are closely associated with inflammatory markers and SCFAs. This work demonstrated that L. plantarum ELF051 can attenuate antibiotic-induced intestinal inflammation in a mouse AAD model by suppressing the pro-inflammatory response and modulating the gut microbiota.
Collapse
Affiliation(s)
- Wei Liang
- College of Chinese Medicinal Material, Jilin Agricultural University, Changchun, 130118, China
- Institute of Agro-Food Technology, Jilin Academy of Agricultural Sciences, No. 1363 Sheng-Tai Street, Changchun, 130033, China
| | - Yansong Gao
- Institute of Agro-Food Technology, Jilin Academy of Agricultural Sciences, No. 1363 Sheng-Tai Street, Changchun, 130033, China
| | - Yujuan Zhao
- Institute of Agro-Food Technology, Jilin Academy of Agricultural Sciences, No. 1363 Sheng-Tai Street, Changchun, 130033, China.
| | - Lei Gao
- Institute of Agro-Food Technology, Jilin Academy of Agricultural Sciences, No. 1363 Sheng-Tai Street, Changchun, 130033, China
| | - Zijian Zhao
- Institute of Agro-Food Technology, Jilin Academy of Agricultural Sciences, No. 1363 Sheng-Tai Street, Changchun, 130033, China
| | - Zhongmei He
- College of Chinese Medicinal Material, Jilin Agricultural University, Changchun, 130118, China
| | - Shengyu Li
- Institute of Agro-Food Technology, Jilin Academy of Agricultural Sciences, No. 1363 Sheng-Tai Street, Changchun, 130033, China.
| |
Collapse
|
41
|
Zhai S, Gao Y, Jiang Y, Li Y, Fan Q, Tie S, Wu Y, Gu S. Weizmannia coagulans BC99 affects valeric acid production via regulating gut microbiota to ameliorate inflammation and oxidative stress responses in Helicobacter pylori mice. J Food Sci 2024; 89:9985-10002. [PMID: 39556495 DOI: 10.1111/1750-3841.17514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/11/2024] [Accepted: 10/17/2024] [Indexed: 11/20/2024]
Abstract
Helicobacter pylori is a highly prevalent pathogen in human gastric mucosa epithelial cells with strong colonization ability. Weizmannia coagulans is a kind of active microorganism that is beneficial to the improvement of host gut microbiota balance and can prevent and treat intestinal diseases. We investigated the beneficial effects of W. coagulans BC99 in H. pylori infected mice and measured inflammation response, oxidative stress, and gut microbiota. Results showed that BC99 could alleviate the gastric inflammation, inhibit the increasing of inflammation parameters endotoxin, interleukin-10, transforming growth factor-β, and interferon-γ and oxidative stress myeloperoxidase and malondialdehyde, promote the levels of superoxide dismutase and catalase. Furthermore, 16S rRNA gene sequencing analysis revealed that BC99 reversed the change of gut microbiota by reducing the abundance of Olsenella, Candidatus_Saccharimonas, Monoglobus, and increasing the abundance of Tyzzerella. Meanwhile, BC99 caused elevated levels of Ligilactobacillus and Lactobacillus. In view of the beneficial effect of BC99 on the content of short-chain fatty acid, valeric acid with sodium valerate interfered with H. pylori infection in mice found that valeric acid had a good restorative effect of H. pylori infection relating inflammation and oxidative stress responses. These results suggest that W. coagulans BC99 can be used as a potential probiotic to prevent and treat H. pylori infection by regulating the inflammation, oxidative stress, and gut microbiota.
Collapse
Affiliation(s)
- Shirui Zhai
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, China
| | - Yinyin Gao
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, China
| | - Yiru Jiang
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, China
| | - Yuwan Li
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, China
- National Demonstration Center for Experimental Food Processing and Safety Education, Luoyang, China
| | - Qiuxia Fan
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, China
| | - Shanshan Tie
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, China
- National Demonstration Center for Experimental Food Processing and Safety Education, Luoyang, China
| | - Ying Wu
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, China
- Henan Engineering Research Center of Food Microbiology, Luoyang, China
| | - Shaobin Gu
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, China
- Henan Engineering Research Center of Food Microbiology, Luoyang, China
- National Demonstration Center for Experimental Food Processing and Safety Education, Luoyang, China
| |
Collapse
|
42
|
Li Z, Li M, Fang X, Yu D, Hu X. Dietary Lactobacillus johnsonii-derived extracellular vesicles ameliorate acute colitis by regulating gut microbiota and maintaining intestinal barrier homeostasis. Food Funct 2024; 15:11757-11779. [PMID: 39545264 DOI: 10.1039/d4fo04194a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
Inflammatory bowel disease (IBD) is a chronic gastrointestinal disease with intricate pathogenesis, and clinical treatment is still not ideal. The imbalance of gut microbiota is associated with IBD progression. Various probiotics have been used as functional foods for the prevention and treatment of IBD, but the specific mechanism is still not fully understood. Lactobacillus johnsonii (L. johnsonii) is a potential anti-inflammatory bacterium, and compared to other probiotic Lactobacillus species, its colonization in the gut of colitis patients is significantly reduced. In this study, we first found that dietary L. johnsonii exerts strong anti-inflammatory and antioxidant effects in colitis mice, and this beneficial effect is directly related to its derived extracellular vesicles (LJ-EVs). Further experimental results indicate that LJ-EVs effectively prevented colitis symptoms and modulated gut microbiota and metabolic pathways. Meanwhile, we have studied for the first time the protective effect of LJ-EVs on the intestinal barrier from the perspective of reducing oxidative stress. We found that LJ-EVs can be directly taken up by intestinal epithelial cells and activate the Nrf2/HO-1 antioxidant signaling pathway, reducing endotoxin damage to cells and maintaining intestinal barrier homeostasis, which cascades to alleviate intestinal inflammation response. This study reveals the mechanism of L. johnsonii in treating colitis and provides a new approach for the development of oral LJ-EVs for the treatment of colitis.
Collapse
Affiliation(s)
- Zhiguo Li
- China-Japan Union Hospital of Jilin University, Jilin University, Changchun, 130033, P. R. China.
| | - Mengyu Li
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, P. R. China
| | - Xuexun Fang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, P. R. China
| | - Dahai Yu
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, P. R. China
| | - Xin Hu
- China-Japan Union Hospital of Jilin University, Jilin University, Changchun, 130033, P. R. China.
| |
Collapse
|
43
|
Corazza E, Pizzi A, Parolin C, Giordani B, Abruzzo A, Bigucci F, Cerchiara T, Luppi B, Vitali B. Orange Peel Lactiplantibacillus plantarum: Development of A Mucoadhesive Nasal Spray with Antimicrobial and Anti-inflammatory Activity. Pharmaceutics 2024; 16:1470. [PMID: 39598593 PMCID: PMC11597421 DOI: 10.3390/pharmaceutics16111470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/14/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives: Due to the high frequency and severity of upper respiratory bacterial infections, probiotics could offer a new medical approach. We explored the antibacterial and anti-inflammatory properties of the new strain Lactiplantibacillus plantarum BIA and formulated a nasal spray. Methods:L. plantarum BIA was isolated from orange peel and taxonomically identified through 16S rRNA gene sequencing. Its antibacterial activity was tested against Pseudomonas aeruginosa, Streptococcus pyogenes, Bacillus subtilis, Escherichia coli, and Staphylococcus aureus, while anti-inflammatory potential was evaluated by Griess assay. BIA genome was fully sequenced and analyzed to assess its safety. BIA was formulated in a freeze-dried matrix, containing prebiotics and cryoprotectants, to be reconstituted with a polymer solution. Solutions containing two types of hydroxypropyl methylcellulose (HPMC) and hyaluronic acid were evaluated as resuspending media and compared in terms of pH, viscosity, and mucoadhesion ability. The biological activity of BIA formulated as nasal spray was verified together with the stability of the selected formulations. Results:L. plantarum BIA inhibited human pathogens' growth and showed anti-inflammatory activity and a safe profile. In the best-performing formulation, the probiotic is lyophilized in 10% fructooligosaccharides, 0.1% ascorbic acid, and 0.5% lactose and reconstituted with HPMC high viscosity 1% w/v. This composition ensured the probiotic's viability for up to six months in its dried form and one week after reconstitution. It also allowed interaction with the nasal mucosa, preserving its antimicrobial and anti-inflammatory activities. Conclusion: The developed nasal spray could become a promising formulation in the field of nasal infectious and inflammatory diseases.
Collapse
Affiliation(s)
- Elisa Corazza
- Drug Delivery Research Laboratory, Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via San Donato 19/2, 40127 Bologna, Italy; (E.C.); (A.A.); (F.B.); (T.C.)
| | - Asia Pizzi
- Beneficial Microbes Laboratory, Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via San Donato 19/2, 40127 Bologna, Italy; (A.P.); (C.P.); (B.G.); (B.V.)
| | - Carola Parolin
- Beneficial Microbes Laboratory, Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via San Donato 19/2, 40127 Bologna, Italy; (A.P.); (C.P.); (B.G.); (B.V.)
| | - Barbara Giordani
- Beneficial Microbes Laboratory, Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via San Donato 19/2, 40127 Bologna, Italy; (A.P.); (C.P.); (B.G.); (B.V.)
| | - Angela Abruzzo
- Drug Delivery Research Laboratory, Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via San Donato 19/2, 40127 Bologna, Italy; (E.C.); (A.A.); (F.B.); (T.C.)
| | - Federica Bigucci
- Drug Delivery Research Laboratory, Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via San Donato 19/2, 40127 Bologna, Italy; (E.C.); (A.A.); (F.B.); (T.C.)
| | - Teresa Cerchiara
- Drug Delivery Research Laboratory, Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via San Donato 19/2, 40127 Bologna, Italy; (E.C.); (A.A.); (F.B.); (T.C.)
| | - Barbara Luppi
- Drug Delivery Research Laboratory, Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via San Donato 19/2, 40127 Bologna, Italy; (E.C.); (A.A.); (F.B.); (T.C.)
| | - Beatrice Vitali
- Beneficial Microbes Laboratory, Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via San Donato 19/2, 40127 Bologna, Italy; (A.P.); (C.P.); (B.G.); (B.V.)
| |
Collapse
|
44
|
Zhou L, Zhou H, Wang P, Xu H, Wu J, Zhou Y, Feng J, Zheng W. Construction of engineered probiotic that adhere and display nanobody to neutralize porcine reproductive and respiratory syndrome virus. Arch Microbiol 2024; 206:466. [PMID: 39540965 DOI: 10.1007/s00203-024-04198-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/23/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
Pathogenic blue ear disease caused by porcine reproductive and respiratory syndrome virus (PRRSV) bring severe loss to breeding industry due to high infectivity and mortality. L. plantarum serves as the probiotic host strain, known for its beneficial properties in the gut microbiota. E. coli is used as a cloning host for the initial genetic engineering steps, facilitating the construction and amplification of the desired genetic constructs. In this study, using synthetic biology technology, we constructed engineered probiotics which could adhere and display nanobody on the surface to neutralize virus. Firstly, we screen an optimal nanobody to effectively bind with PRRSV by building library, expression and purification. Then, the integration of adhesion protein and nanobody into the genome of probiotics significantly improved its adhesion to IPEC-J2 cells. In addition, this engineered probiotic is almost non-toxic to cells with good safety, which can be used as a daily probiotics to prevent virus fecal transmission. Our study proposed this novel construction strategy of engineering probiotics with both adhesion and neutralization effects, which provided a new therapeutic view for intestinal virus clearance.
Collapse
Affiliation(s)
- Li Zhou
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, P. R. China
| | - Hanlin Zhou
- College of Life Science and Technology, Mudanjiang Normal University, 191 Wenhua Street, Mudanjiang, 157011, China
- College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luo Yu Road, Wuhan, 430074, China
- Liangzhun (Wuhan) Life Science & Technology Co. Ltd., 666 Gaoxin Avenue, Wuhan, 430073, China
| | - Panying Wang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, P. R. China
| | - Hang Xu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Affiliated to Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, P. R. China
| | - Jiayi Wu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, P. R. China
| | - Yuanzhuo Zhou
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, P. R. China
| | - Jiaying Feng
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, P. R. China
| | - Weiyi Zheng
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, P. R. China.
| |
Collapse
|
45
|
Alam M, Abbas K, Mustafa M, Usmani N, Habib S. Microbiome-based therapies for Parkinson's disease. Front Nutr 2024; 11:1496616. [PMID: 39568727 PMCID: PMC11576319 DOI: 10.3389/fnut.2024.1496616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 10/21/2024] [Indexed: 11/22/2024] Open
Abstract
The human gut microbiome dysbiosis plays an important role in the pathogenesis of Parkinson's disease (PD). The bidirectional relationship between the enteric nervous system (ENS) and central nervous system (CNS) under the mediation of the gut-brain axis control the gastrointestinal functioning. This review article discusses key mechanisms by which modifications in the composition and function of the gut microbiota (GM) influence PD progression and motor control loss. Increased intestinal permeability, chronic inflammation, oxidative stress, α-synuclein aggregation, and neurotransmitter imbalances are some key factors that govern gastrointestinal pathology and PD progression. The bacterial taxa of the gut associated with PD development are discussed with emphasis on the enteric nervous system (ENS), as well as the impact of gut bacteria on dopamine production and levodopa metabolism. The pathophysiology and course of the disease are associated with several inflammatory markers, including TNF-α, IL-1β, and IL-6. Emerging therapeutic strategies targeting the gut microbiome include probiotics, prebiotics, synbiotics, postbiotics, and fecal microbiota transplantation (FMT). The article explored how dietary changes may affect the gut microbiota (GM) and the ways that can affect Parkinson's disease (PD), with a focus on nutrition-based, Mediterranean, and ketogenic diets. This comprehensive review synthesizes current evidence on the role of the gut microbiome in PD pathogenesis and explores its potential as a therapeutic target. Understanding these complex interactions may assist in the development of novel diagnostic tools and treatment options for this neurodegenerative disorder.
Collapse
Affiliation(s)
- Mudassir Alam
- Indian Biological Sciences and Research Institute (IBRI), Noida, India
| | - Kashif Abbas
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Mohd Mustafa
- Department of Biochemistry, J.N. Medical College, Aligarh Muslim University, Aligarh, India
| | - Nazura Usmani
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Safia Habib
- Department of Biochemistry, J.N. Medical College, Aligarh Muslim University, Aligarh, India
| |
Collapse
|
46
|
Yao L, Yang Y, Yang X, Rezaei MJ. The Interaction Between Nutraceuticals and Gut Microbiota: a Novel Therapeutic Approach to Prevent and Treatment Parkinson's Disease. Mol Neurobiol 2024; 61:9078-9109. [PMID: 38587699 DOI: 10.1007/s12035-024-04151-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 03/25/2024] [Indexed: 04/09/2024]
Abstract
Parkinson's disease (PD) is a complex neurodegenerative disorder characterized by the progressive loss of dopaminergic neurons, leading to motor and non-motor symptoms. Emerging research has shed light on the role of gut microbiota in the pathogenesis and progression of PD. Nutraceuticals such as curcumin, berberine, phytoestrogens, polyphenols (e.g., resveratrol, EGCG, and fisetin), dietary fibers have been shown to influence gut microbiota composition and function, restoring microbial balance and enhancing the gut-brain axis. The mechanisms underlying these benefits involve microbial metabolite production, restoration of gut barrier integrity, and modulation of neuroinflammatory pathways. Additionally, probiotics and prebiotics have shown potential in promoting gut health, influencing the gut microbiome, and alleviating PD symptoms. They can enhance the gut's antioxidant capacity of the gut, reduce inflammation, and maintain immune homeostasis, contributing to a neuroprotective environment. This paper provides an overview of the current state of knowledge regarding the potential of nutraceuticals and gut microbiota modulation in the prevention and management of Parkinson's disease, emphasizing the need for further research and clinical trials to validate their effectiveness and safety. The findings suggest that a multifaceted approach involving nutraceuticals and gut microbiota may open new avenues for addressing the challenges of PD and improving the quality of life for affected individuals.
Collapse
Affiliation(s)
- Liyan Yao
- School of Public Health, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Yong Yang
- School of Public Health, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Xiaowei Yang
- School of Public Health, Mudanjiang Medical University, Mudanjiang, 157011, China.
| | - Mohammad J Rezaei
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
47
|
Domínguez Rojo N, Blanco Benítez M, Cava R, Fuentes JM, Canales Cortés S, González Polo RA. Convergence of Neuroinflammation, Microbiota, and Parkinson's Disease: Therapeutic Insights and Prospects. Int J Mol Sci 2024; 25:11629. [PMID: 39519181 PMCID: PMC11545862 DOI: 10.3390/ijms252111629] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 10/23/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024] Open
Abstract
Parkinson's disease (PD) is a complex neurodegenerative disorder. Recent evidence reveals connections between neuroinflammatory processes and intestinal microbiota alterations in the progression of this pathology. This comprehensive review explores the intricate relationships between them, highlighting their combined impact on PD. Neuroinflammation, characterized by immune activation in the central nervous system, is increasingly acknowledged as a critical factor in the development of PD. Concurrently, alterations in the gut microbiota composition have been linked to PD, suggesting a potential modulatory role in disease progression. Thus, bidirectional communication along the gut-brain axis has become pivotal in comprehending the pathogenesis of PD. Furthermore, we explore emerging therapeutic strategies that target these interconnected pathways, providing insights into potential avenues for PD treatment. The elucidation of these intricate relationships establishes a promising foundation for innovative therapeutic strategies aimed at altering disease progression and improving the quality of life for individuals affected by PD.
Collapse
Affiliation(s)
- Nerea Domínguez Rojo
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, 10003 Cáceres, Spain; (N.D.R.); (M.B.B.); (J.M.F.)
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 10003 Cáceres, Spain
| | - Mercedes Blanco Benítez
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, 10003 Cáceres, Spain; (N.D.R.); (M.B.B.); (J.M.F.)
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 10003 Cáceres, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas-Instituto de Salud Carlos III (CIBER-CIBERNED-ISCIII), 28029 Madrid, Spain
| | - Ramón Cava
- Tradinnoval Research Group, INBIO G+C, Departamento de Producción Animal y Ciencia de los Alimentos, Universidad de Extremadura, 10003 Cáceres, Spain;
| | - José Manuel Fuentes
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, 10003 Cáceres, Spain; (N.D.R.); (M.B.B.); (J.M.F.)
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 10003 Cáceres, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas-Instituto de Salud Carlos III (CIBER-CIBERNED-ISCIII), 28029 Madrid, Spain
| | - Saray Canales Cortés
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, 10003 Cáceres, Spain; (N.D.R.); (M.B.B.); (J.M.F.)
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 10003 Cáceres, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas-Instituto de Salud Carlos III (CIBER-CIBERNED-ISCIII), 28029 Madrid, Spain
| | - Rosa Ana González Polo
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, 10003 Cáceres, Spain; (N.D.R.); (M.B.B.); (J.M.F.)
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 10003 Cáceres, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas-Instituto de Salud Carlos III (CIBER-CIBERNED-ISCIII), 28029 Madrid, Spain
| |
Collapse
|
48
|
Huang X, Bao J, Yang M, Li Y, Liu Y, Zhai Y. The role of Lactobacillus plantarum in oral health: a review of current studies. J Oral Microbiol 2024; 16:2411815. [PMID: 39444695 PMCID: PMC11497578 DOI: 10.1080/20002297.2024.2411815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 07/09/2024] [Accepted: 08/27/2024] [Indexed: 10/25/2024] Open
Abstract
Background Oral non-communicable diseases, particularly dental caries and periodontal disease, impose a significant global health burden. The underlying microbial dysbiosis is a prominent factor, driving interest in strategies that promote a balanced oral microbiome. Lactobacillus plantarum, a gram-positive lactic acid bacterium known for its adaptability, has gained attention for its potential to enhance oral health. Recent studies have explored the use of probiotic L. plantarum in managing dental caries, periodontal disease, and apical periodontitis. However, a comprehensive review on its effects in this context is still lacking. Aims This narrative review evaluates current literature on L. plantarum's role in promoting oral health and highlights areas for future research. Content In general, the utilization of L. plantarum in managing non-communicable biofilm-dependent oral diseases is promising, but additional investigations are warranted. Key areas for future study include: exploring its mechanisms of action, identifying optimal strains or strain combinations of L. plantarum, determining effective delivery methods and dosages, developing commercial antibacterial agents from L. plantarum, and addressing safety considerations related to its use in oral care.
Collapse
Affiliation(s)
- Xinyan Huang
- School of Stomatology, Henan University, Kaifeng, China
- Kaifeng Key Laboratory of Periodontal Tissue Engineering, Kaifeng, China
- Eastman Institute for Oral Health, University of Rochester Medical Center, Rochester, NY, USA
| | - Jianhang Bao
- School of Stomatology, Henan University, Kaifeng, China
- Kaifeng Key Laboratory of Periodontal Tissue Engineering, Kaifeng, China
| | - Mingzhen Yang
- School of Stomatology, Henan University, Kaifeng, China
- Kaifeng Key Laboratory of Periodontal Tissue Engineering, Kaifeng, China
| | - Yingying Li
- Orthopedic Department, Luoyang Orthopedic Hospital of Henan Province (Orthopedic Hospital of Henan Province), Zhengzhou, China
| | - Youwen Liu
- Orthopedic Department, Luoyang Orthopedic Hospital of Henan Province (Orthopedic Hospital of Henan Province), Zhengzhou, China
| | - Yuankun Zhai
- School of Stomatology, Henan University, Kaifeng, China
- Kaifeng Key Laboratory of Periodontal Tissue Engineering, Kaifeng, China
| |
Collapse
|
49
|
Di Salvo C, D'Antongiovanni V, Benvenuti L, d'Amati A, Ippolito C, Segnani C, Pierucci C, Bellini G, Annese T, Virgintino D, Colucci R, Antonioli L, Fornai M, Errede M, Bernardini N, Pellegrini C. Lactiplantibacillus plantarum HEAL9 attenuates cognitive impairment and progression of Alzheimer's disease and related bowel symptoms in SAMP8 mice by modulating microbiota-gut-inflammasome-brain axis. Food Funct 2024; 15:10323-10338. [PMID: 39302233 DOI: 10.1039/d4fo02075h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
Background: Growing evidence highlights the relevance of the microbiota-gut-brain axis in Alzheimer's disease (AD). AD patients display gut dysbiosis, altered intestinal barrier and enteric inflammation that, besides bowel symptoms, can contribute to brain pathology. In this context, the modulation of gut microbiota is emerging as a therapeutical option to halt or slow down central pathology. Herein, we examined the effects of Lactiplantibacillus plantarum HEAL9 in a spontaneous mouse model of AD. Methods: Senescence-accelerated mouse prone 8 (SAMP8) mice and control SAMR1 mice were treated orally with HEAL9 1 × 109 CFU per mouse per day or placebo for two months to evaluate the effects of the probiotic during the earliest stages of AD, before the development of brain pathology. Cognitive impairment, in vivo and in vitro colonic motility, astrocyte and microglia reactive response, brain and colonic amyloid-β1-42 (Aβ1-42) levels, and inflammasome components activation (NLRP3, ASC, caspase-1 and interleukin-1β) were assessed. In addition, gut barrier alterations [circulating lipopolysaccharide-binding protein (LBP) levels] and acidic mucus were evaluated. Results: HEAL9 administration significantly attenuated cognitive impairment and counteracted colonic dysmotility in SAMP8 mice. Moreover, HEAL9 decreased astrogliosis and microgliosis, Aβ1-42 accumulation and inflammasome activation in colon and brain and normalized plasma LBP levels and colonic acidic mucus content. Conclusion: HEAL9 intake alleviated cognitive decline and normalized colonic motility in the prodromal phases of AD via the modulation of microbiota-gut-inflammasome-brain signalling. Thus, dietary supplementation with HEAL9 could be considered as a suitable therapeutical option for the treatment of AD and related intestinal symptoms in the early stages of the disease.
Collapse
Affiliation(s)
- C Di Salvo
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - V D'Antongiovanni
- Unit of Histology and Medical Embryology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.
| | - L Benvenuti
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - A d'Amati
- Human Anatomy and Histology Unit, Department of Basic Medical Sciences, Neuroscience, and Sensory Organs, University of Bari School of Medicine, Bari, Italy.
| | - C Ippolito
- Unit of Histology and Medical Embryology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.
| | - C Segnani
- Unit of Histology and Medical Embryology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.
| | - C Pierucci
- Unit of Histology and Medical Embryology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.
| | - G Bellini
- Unit of Neurology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - T Annese
- Human Anatomy and Histology Unit, Department of Basic Medical Sciences, Neuroscience, and Sensory Organs, University of Bari School of Medicine, Bari, Italy.
- Department of Medicine and Surgery, University LUM Giuseppe Degennaro, Casamassima, Bari, Italy
| | - D Virgintino
- Human Anatomy and Histology Unit, Department of Basic Medical Sciences, Neuroscience, and Sensory Organs, University of Bari School of Medicine, Bari, Italy.
| | - R Colucci
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - L Antonioli
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - M Fornai
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - M Errede
- Human Anatomy and Histology Unit, Department of Basic Medical Sciences, Neuroscience, and Sensory Organs, University of Bari School of Medicine, Bari, Italy.
| | - N Bernardini
- Unit of Histology and Medical Embryology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.
| | - C Pellegrini
- Unit of Histology and Medical Embryology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.
| |
Collapse
|
50
|
Gao W, Dong Q, Wu X, Wang Y, Li J, Zhang Q, Lu F, Liu F. Bifunctional Inhibitor Lentinan Inhibits Fibrillogenesis of Amyloid-β Protein and α-Synuclein and Alleviates Their Cytotoxicity: In Vitro and In Vivo Studies. ACS Chem Neurosci 2024; 15:3437-3448. [PMID: 39264814 DOI: 10.1021/acschemneuro.4c00164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/14/2024] Open
Abstract
Alzheimer's disease (AD) and Parkinson's disease (PD) are the two most common neurodegenerative diseases in the world. Misfolding of β-amyloid (Aβ) and α-synuclein (α-syn) and subsequent fibril formation are closely associated with the pathogenesis of AD and PD, respectively. Lentinan is a natural product commonly used in medicine and dietary supplements. It has potential antitumor, anti-inflammatory, and antiviral effects, but the underlying mechanism of its action on AD and PD remains unclear. In this study, lentinan inhibited the formation of Aβ and α-syn fibers in a dose-dependent manner and disrupted their mature fibers. Lentinan inhibited the conversion of Aβ and α-syn conformations to β-sheet-rich conformations. Additionally, lentinan protected Caenorhabditis elegans against damage caused by the accumulation of Aβ and α-syn aggregation and prolonged their lifespan. Notably, the beneficial effects of lentinan in AD and PD mice were also demonstrated, including ameliorating the cognitive and memory impairments in AD mice and behavioral deficits in PD mice. Finally, molecular interactions between lentinan and Aβ/α-syn pentamers were also explored using molecular docking.
Collapse
Affiliation(s)
- Wen Gao
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin 300457, P. R. China
- Tianjin Key Laboratory of Industrial Microbiology, Tianjin 300457, P. R. China
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P. R. China
| | - Qinchen Dong
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin 300457, P. R. China
- Tianjin Key Laboratory of Industrial Microbiology, Tianjin 300457, P. R. China
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P. R. China
| | - Xinni Wu
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin 300457, P. R. China
- Tianjin Key Laboratory of Industrial Microbiology, Tianjin 300457, P. R. China
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P. R. China
| | - Yang Wang
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin 300457, P. R. China
- Tianjin Key Laboratory of Industrial Microbiology, Tianjin 300457, P. R. China
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P. R. China
| | - Jinbi Li
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin 300457, P. R. China
- Tianjin Key Laboratory of Industrial Microbiology, Tianjin 300457, P. R. China
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P. R. China
| | - Qingfu Zhang
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin 300457, P. R. China
- Tianjin Key Laboratory of Industrial Microbiology, Tianjin 300457, P. R. China
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P. R. China
| | - Fuping Lu
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin 300457, P. R. China
- Tianjin Key Laboratory of Industrial Microbiology, Tianjin 300457, P. R. China
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P. R. China
| | - Fufeng Liu
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin 300457, P. R. China
- Tianjin Key Laboratory of Industrial Microbiology, Tianjin 300457, P. R. China
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P. R. China
| |
Collapse
|