1
|
Zhang Y, Liang Y, Gu Y. The dopaminergic system and Alzheimer's disease. Neural Regen Res 2025; 20:2495-2512. [PMID: 39314145 PMCID: PMC11801300 DOI: 10.4103/nrr.nrr-d-24-00230] [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: 02/25/2024] [Revised: 06/21/2024] [Accepted: 07/31/2024] [Indexed: 09/25/2024] Open
Abstract
Alzheimer's disease is a common neurodegenerative disorder in older adults. Despite its prevalence, its pathogenesis remains unclear. In addition to the most widely accepted causes, which include excessive amyloid-beta aggregation, tau hyperphosphorylation, and deficiency of the neurotransmitter acetylcholine, numerous studies have shown that the dopaminergic system is also closely associated with the occurrence and development of this condition. Dopamine is a crucial catecholaminergic neurotransmitter in the human body. Dopamine-associated treatments, such as drugs that target dopamine receptor D and dopamine analogs, can improve cognitive function and alleviate psychiatric symptoms as well as ameliorate other clinical manifestations. However, therapeutics targeting the dopaminergic system are associated with various adverse reactions, such as addiction and exacerbation of cognitive impairment. This review summarizes the role of the dopaminergic system in the pathology of Alzheimer's disease, focusing on currently available dopamine-based therapies for this disorder and the common side effects associated with dopamine-related drugs. The aim of this review is to provide insights into the potential connections between the dopaminergic system and Alzheimer's disease, thus helping to clarify the mechanisms underlying the condition and exploring more effective therapeutic options.
Collapse
Affiliation(s)
- Yuhan Zhang
- International Medical College, Chongqing Medical University, Chongqing, China
| | - Yuan Liang
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Yixue Gu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| |
Collapse
|
2
|
Zhang L, Wei J, Liu X, Li D, Pang X, Chen F, Cao H, Lei P. Gut microbiota-astrocyte axis: new insights into age-related cognitive decline. Neural Regen Res 2025; 20:990-1008. [PMID: 38989933 PMCID: PMC11438350 DOI: 10.4103/nrr.nrr-d-23-01776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 03/04/2024] [Indexed: 07/12/2024] Open
Abstract
With the rapidly aging human population, age-related cognitive decline and dementia are becoming increasingly prevalent worldwide. Aging is considered the main risk factor for cognitive decline and acts through alterations in the composition of the gut microbiota, microbial metabolites, and the functions of astrocytes. The microbiota-gut-brain axis has been the focus of multiple studies and is closely associated with cognitive function. This article provides a comprehensive review of the specific changes that occur in the composition of the gut microbiota and microbial metabolites in older individuals and discusses how the aging of astrocytes and reactive astrocytosis are closely related to age-related cognitive decline and neurodegenerative diseases. This article also summarizes the gut microbiota components that affect astrocyte function, mainly through the vagus nerve, immune responses, circadian rhythms, and microbial metabolites. Finally, this article summarizes the mechanism by which the gut microbiota-astrocyte axis plays a role in Alzheimer's and Parkinson's diseases. Our findings have revealed the critical role of the microbiota-astrocyte axis in age-related cognitive decline, aiding in a deeper understanding of potential gut microbiome-based adjuvant therapy strategies for this condition.
Collapse
Affiliation(s)
- Lan Zhang
- Haihe Laboratory of Cell Ecosystem, Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Jingge Wei
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Xilei Liu
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Dai Li
- Haihe Laboratory of Cell Ecosystem, Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaoqi Pang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Fanglian Chen
- Tianjin Neurological Institution, Tianjin Medical University General Hospital, Tianjin, China
| | - Hailong Cao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Ping Lei
- Haihe Laboratory of Cell Ecosystem, Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
3
|
Xu CC, Zhao WX, Sheng Y, Yun YJ, Ma T, Fan N, Song JQ, Wang J, Zhang Q. Serum homocysteine showed potential association with cognition and abnormal gut microbiome in major depressive disorder. World J Psychiatry 2025; 15:102567. [DOI: 10.5498/wjp.v15.i3.102567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/18/2024] [Accepted: 01/06/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Cognitive impairment is one of the common clinical manifestations of depression, causing negative distress to patients. Elevated homocysteine (Hcy) concentrations and gut microbiome dysfunction may be observed in patients with depression.
AIM To investigate the relationship between Hcy, microbiome, and cognition in depressive patients.
METHODS We recruited 67 patients with major depressive disorder (MDD) (MDD group) and 94 healthy controls (HCs) individuals (HCs group). Serum Hcy levels were determined using the enzyme circulation method. 16s rRNA sequencing was used to classify and identify the fecal bacteria. 17 Hamilton depression rating scale and MATRICS consensus cognitive battery were used to evaluate mood states and cognition in patients with MDD. Correlation analysis was performed to explore the correlation between fecal flora, Hcy, and depressive cognitive function.
RESULTS Elevated serum levels of Hcy were seen in patients with MDD compared to healthy individuals. Patients with MDD indicated significant decreases in cognitive scores (P < 0.001) in six modules: Speed of processing, working memory, visual learning, reasoning and problem-solving, social cognition, and total scores. Hcy levels showed a negative correlation with processing speed, social cognition, and total MDD scores (P < 0.05). Hcy was also significantly negatively correlated with Alistipes, Ruminococcae, Tenericides, and Porphyromonas (P < 0.05).
CONCLUSION Our results highlight that Hcy was correlated with cognition and gut microbiome in MDD. This interaction may be related to the physiological and pathological mechanisms underlying cognitive deficits in depression.
Collapse
Affiliation(s)
- Chen-Chen Xu
- Department of Psychiatry, The Affiliated Wuxi Mental Health Center of Nanjing Medical University, Wuxi 214151, Jiangsu Province, China
| | - Wen-Xuan Zhao
- Department of Psychiatry, Beijing Huilongguan Hospital, Peking University Huilongguan Clinical Medical School, Beijing 100096, China
| | - Yu Sheng
- Department of Psychiatry, Chinese People’s Liberation Army Unit 94710, Wuxi 214141, Jiangsu Province, China
| | - Ya-Jun Yun
- Department of Psychiatry, Beijing Huilongguan Hospital, Peking University Huilongguan Clinical Medical School, Beijing 100096, China
| | - Ting Ma
- Department of Psychiatry, Beijing Huilongguan Hospital, Peking University Huilongguan Clinical Medical School, Beijing 100096, China
| | - Ning Fan
- Department of Psychiatry, Beijing Huilongguan Hospital, Peking University Huilongguan Clinical Medical School, Beijing 100096, China
| | - Jia-Qi Song
- Department of Psychiatry, Beijing Huilongguan Hospital, Peking University Huilongguan Clinical Medical School, Beijing 100096, China
| | - Jun Wang
- Department of Psychiatry, The Affiliated Wuxi Mental Health Center of Nanjing Medical University, Wuxi 214151, Jiangsu Province, China
- Department of Psychiatry, The Affiliated Mental Health Center of Jiangnan University, Wuxi 214151, Jiangsu Province, China
| | - Qi Zhang
- Department of Psychiatry, The Affiliated Wuxi Mental Health Center of Nanjing Medical University, Wuxi 214151, Jiangsu Province, China
- Department of Psychiatry, The Affiliated Mental Health Center of Jiangnan University, Wuxi 214151, Jiangsu Province, China
| |
Collapse
|
4
|
Zhang Q, Zhao W, Yun Y, Ma T, An H, Fan N, Wang J, Wang Z, Yang F. Multiomics analysis reveals aberrant tryptophan-kynurenine metabolism and immunity linked gut microbiota with cognitive impairment in major depressive disorder. J Affect Disord 2025; 373:273-283. [PMID: 39716675 DOI: 10.1016/j.jad.2024.12.070] [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: 06/07/2024] [Revised: 11/04/2024] [Accepted: 12/20/2024] [Indexed: 12/25/2024]
Abstract
OBJECTIVE Cognitive impairment occurs throughout the entire course of and affects the work and life of patients with major depressive disorder (MDD). The gut microbiota, kynurenine pathway (KP) and inflammatory response may have important roles in the mechanism of cognitive impairment in MDD patients. Consequently, our goal was to investigate the association among the gut microbiota, inflammation, KP, and cognition in MDD. METHOD We enrolled patients with MDD (N = 86) and healthy controls (HCs, N = 120) in this research. The study involved participant data regarding the levels of serum inflammatory factors (interleukin [IL]-1β, IL-4, IL-6, brain-derived neurotropic factor [BNDF], migration inhibitory factor [MIF], tumor necrosis factor [TNF]-α, vascular endothelial growth factor [VEGF]), gut microbiota and cognitive function (MCCB) were collected. RESULTS Patients demonstrated poorer cognitive function. Gut microbiota, such as Bacteroide, Prevotella, Faecalibacterium and Parabacteroides between MDDs and HCs were significantly different. Moreover, in patients with MDD, we found that different microbiomes were related to cognition and that Acidaminococcus was positively correlated with multiple domains of cognition. Allisonella and Acidaminococcus were significantly positively correlated with BDNF and negatively correlated with MIF. Alloprevotella, Blautia, and Megamonas were positively correlated with kynurenine/tryptophan (KYN/TRP). Acidaminococcus was negatively correlated with 3-hydroxykynurenine (3-HK). BDNF levels was significantly positive correlated with kynurenic acid (KA) and quinolinic acid (QA). CONCLUSION The results of the present study suggest that the gut microbiota is associated with cognitive function, cytokine levels and KP metabolism in patients with MDD; however, the mechanism of the interaction between cognition and gut microbiota in MDD patients require further investigation.
Collapse
Affiliation(s)
- Qi Zhang
- The Affiliated Mental Health Center of Jiangnan University, Wuxi Central Rehabilitaion Hospital, Wuxi, China
| | - Wenxuan Zhao
- Beijing HuiLongGuan Hospital, Peking University HuiLongGuan Clinical Medical School, Beijing, China
| | - Yajun Yun
- Beijing HuiLongGuan Hospital, Peking University HuiLongGuan Clinical Medical School, Beijing, China
| | - Ting Ma
- Beijing HuiLongGuan Hospital, Peking University HuiLongGuan Clinical Medical School, Beijing, China
| | - Huimei An
- Beijing HuiLongGuan Hospital, Peking University HuiLongGuan Clinical Medical School, Beijing, China
| | - Ning Fan
- Beijing HuiLongGuan Hospital, Peking University HuiLongGuan Clinical Medical School, Beijing, China
| | - Jun Wang
- The Affiliated Mental Health Center of Jiangnan University, Wuxi Central Rehabilitaion Hospital, Wuxi, China.
| | - Zhiren Wang
- Beijing HuiLongGuan Hospital, Peking University HuiLongGuan Clinical Medical School, Beijing, China.
| | - Fude Yang
- Beijing HuiLongGuan Hospital, Peking University HuiLongGuan Clinical Medical School, Beijing, China.
| |
Collapse
|
5
|
Pereira DE, de Cássia de Araújo Bidô R, da Costa Alves M, Frazão Tavares de Melo MF, Dos Santos Costa AC, Gomes Dutra LM, de Morais MM, Gomes da Câmara CA, Viera VB, Alves AF, de Araujo WJ, Leite EL, Bruno de Oliveira CJ, Rufino Freitas JC, Barbosa Soares JK. Maternal supplementation with Dipteryx alata Vog. modulates fecal microbiota diversity, accelerates reflex ontogeny, and improves non-associative and spatial memory in the offspring of rats. Brain Res 2025; 1850:149383. [PMID: 39647597 DOI: 10.1016/j.brainres.2024.149383] [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: 08/04/2024] [Revised: 11/09/2024] [Accepted: 12/03/2024] [Indexed: 12/10/2024]
Abstract
Maternal diet plays a crucial role in offspring development, directly affecting neural development and gut microbiota composition. This study aimed to assess if baru almond and oil (Dipteryx alata Vog.) could modulate intestinal microbiota, brain fatty acid profile, and enhance memory in offspring of rats treated during early life stages. Three groups were formed: Control- received distilled water by gavage; Oil- received 2000 mg/kg of baru oil, and Almond - received 2000 mg/kg of baru almond. Somatic development and reflex ontogenesis were evaluated in offspring during the first 21 days. In adolescence and adulthood, memory was tested using Open Field Habituation, Object Recognition, and Morris Water Maze. Brain histology and fatty acid were measured, and fecal microbiota analysis was performed. Both almond and oil groups showed increased PUFAs in breast milk and brains, accelerated reflex ontogeny, improved somatic development and better performance in the memory tests in both life stages (p < 0.05). Supplementation enhanced fecal microbiota abundance associated with neuroprotective effects. The almond group showed a 29 % increase in Eubacterium, Candidates-Arthromitus, Collinsella, and Christensenellaceae-R-7. Both oil and almond groups had higher Blautia and Clostridia-UCG-014 compared to controls. The oil group had about 10 % more Ruminococcus, UCG-005, Acetatifactor, Negativibacillus, and Lachnospiraceae-ND3007 than the others. With the present data, we can observe the safety of baru consumption by pregnant and lactating rats and verify its effects on modulating the microbiota, inducing adequate development of the offspring's nervous system, contributing to anticipated reflex maturation and improving memory.
Collapse
Affiliation(s)
- Diego Elias Pereira
- Program of Food Science and Technology, Federal University of Paraíba, João Pessoa, PB, Brazil; Laboratory of Experimental Nutrition, Department of Nutrition, Federal University of Campina Grande, Cuité, CG, Brazil
| | - Rita de Cássia de Araújo Bidô
- Program of Food Science and Technology, Federal University of Paraíba, João Pessoa, PB, Brazil; Laboratory of Experimental Nutrition, Department of Nutrition, Federal University of Campina Grande, Cuité, CG, Brazil
| | - Maciel da Costa Alves
- Department of Biofísica and Pharmacology, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | | | - Ana Carolina Dos Santos Costa
- Laboratory of Experimental Nutrition, Department of Nutrition, Federal University of Campina Grande, Cuité, CG, Brazil; Department of Rural Technology, Federal Rural University of Pernambuco, Recife, Brazil
| | - Larissa Maria Gomes Dutra
- Program of Food Science and Technology, Federal University of Paraíba, João Pessoa, PB, Brazil; Laboratory of Experimental Nutrition, Department of Nutrition, Federal University of Campina Grande, Cuité, CG, Brazil.
| | | | | | - Vanessa Bordin Viera
- Laboratory of Bromatology, Department of Nutrition, Federal University of Campina Grande, Cuité, CG, Brazil
| | - Adriano Francisco Alves
- Laboratory of General Pathology, Department of Physiology and General Pathology, Federal University of Paraíba, João Pessoa, PB, Brazil
| | - Wydemberg José de Araujo
- Laboratory for the Evaluation of Products of Animal Origin, Department of Animal Science, Federal University of Paraíba - Areia, PB, Brazil
| | - Elma Lima Leite
- Laboratory for the Evaluation of Products of Animal Origin, Department of Animal Science, Federal University of Paraíba - Areia, PB, Brazil
| | - Celso José Bruno de Oliveira
- Laboratory for the Evaluation of Products of Animal Origin, Department of Animal Science, Federal University of Paraíba - Areia, PB, Brazil
| | | | - Juliana Késsia Barbosa Soares
- Program of Food Science and Technology, Federal University of Paraíba, João Pessoa, PB, Brazil; Laboratory of Experimental Nutrition, Department of Nutrition, Federal University of Campina Grande, Cuité, CG, Brazil
| |
Collapse
|
6
|
Peng R, Wang W, Liang L, Han R, Li Y, Wang H, Wang Y, Li W, Feng S, Zhou J, Huang Y, Wu F, Wu K. The brain-gut microbiota network (BGMN) is correlated with symptom severity and neurocognition in patients with schizophrenia. Neuroimage 2025; 308:121052. [PMID: 39875038 DOI: 10.1016/j.neuroimage.2025.121052] [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: 12/14/2023] [Revised: 01/19/2025] [Accepted: 01/23/2025] [Indexed: 01/30/2025] Open
Abstract
The association between the human brain and gut microbiota, known as the "brain-gut-microbiota axis", is involved in the neuropathological mechanisms of schizophrenia (SZ); however, its association patterns and correlations with symptom severity and neurocognition are still largely unknown. In this study, 43 SZ patients and 55 normal controls (NCs) were included, and resting-state functional magnetic resonance imaging (rs-fMRI) and gut microbiota data were acquired for each participant. First, the brain features of brain images and functional brain networks were computed from rs-fMRI data; the gut features of gut microbiota abundance and the gut microbiota network were computed from gut microbiota data. Second, we propose a novel methodology to construct an individual brain-gut microbiota network (BGMN) for each participant by combining the brain and gut features via multiple strategies. Third, discriminative models between SZ patients and NCs were built using the connectivity matrices of the BGMN as input features. Moreover, the correlations between the most discriminative features and the scores of symptom severity and neurocognition were analyzed in SZ patients. The results showed that the best discriminative model between SZ patients and NCs was achieved using the connectivity matrices of the BGMN when all the brain and gut features were integrated, with an accuracy of 0.90 and an area under the curve value of 0.97. The most discriminative features were related primarily to the genera Faecalibacterium and Collinsella, in which the genus Faecalibacterium was linked to the visual system and subcortical cortices and the genus Collinsella was linked to the default network and subcortical cortices. Furthermore, parts of the most discriminative features were significantly correlated with the scores of neurocognition in the SZ patients. The methodology for constructing individual BGMNs proposed in this study can help us reveal the associations between the brain and gut microbiota and understand the neuropathology of SZ.
Collapse
Affiliation(s)
- Runlin Peng
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
| | - Wei Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
| | - Liqin Liang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
| | - Rui Han
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
| | - Yi Li
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
| | - Haiyuan Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
| | - Yuran Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
| | - Wenhao Li
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
| | - Shixuan Feng
- Department of Psychiatry, The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou 510370, China
| | - Jing Zhou
- School of Material Science and Engineering, South China University of Technology, Guangzhou 510006, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
| | - Yuanyuan Huang
- Department of Psychiatry, The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou 510370, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou 510370, China
| | - Fengchun Wu
- Department of Psychiatry, The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou 510370, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou 510370, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou 510370, China.
| | - Kai Wu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China; Department of Aging Research and Geriatric Medicine, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan.
| |
Collapse
|
7
|
Junyi L, Yueyang W, Bin L, Xiaohong D, Wenhui C, Ning Z, Hong Z. Gut Microbiota Mediates Neuroinflammation in Alzheimer's Disease: Unraveling Key Factors and Mechanistic Insights. Mol Neurobiol 2025; 62:3746-3763. [PMID: 39317889 DOI: 10.1007/s12035-024-04513-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] [Received: 03/24/2024] [Accepted: 09/18/2024] [Indexed: 09/26/2024]
Abstract
The gut microbiota, the complex community of microorganisms that inhabit the gastrointestinal tract, has emerged as a key player in the pathogenesis of neurodegenerative disorders, including Alzheimer's disease (AD). AD is characterized by progressive cognitive decline and neuronal loss, associated with the accumulation of amyloid-β plaques, neurofibrillary tangles, and neuroinflammation in the brain. Increasing evidence suggests that alterations in the composition and function of the gut microbiota, known as dysbiosis, may contribute to the development and progression of AD by modulating neuroinflammation, a chronic and maladaptive immune response in the central nervous system. This review aims to comprehensively analyze the current role of the gut microbiota in regulating neuroinflammation and glial cell function in AD. Its objective is to deepen our understanding of the pathogenesis of AD and to discuss the potential advantages and challenges of using gut microbiota modulation as a novel approach for the diagnosis, treatment, and prevention of AD.
Collapse
Affiliation(s)
- Liang Junyi
- Heilongjiang University of Traditional Chinese Medicine, Harbin, 150040, Heilongjiang Province, China
| | - Wang Yueyang
- Heilongjiang University of Traditional Chinese Medicine, Harbin, 150040, Heilongjiang Province, China
| | - Liu Bin
- Heilongjiang University of Traditional Chinese Medicine, Harbin, 150040, Heilongjiang Province, China.
| | - Dong Xiaohong
- Jiamusi College, Heilongjiang University of Traditional Chinese Medicine, Jiamusi, Heilongjiang Province, China
| | - Cai Wenhui
- Heilongjiang University of Traditional Chinese Medicine, Harbin, 150040, Heilongjiang Province, China
| | - Zhang Ning
- Heilongjiang University of Traditional Chinese Medicine, Harbin, 150040, Heilongjiang Province, China
| | - Zhang Hong
- Heilongjiang Jiamusi Central Hospital, Jiamusi, Heilongjiang Province, China
| |
Collapse
|
8
|
Manfredi JN, Gupta SK, Vyavahare S, Deak F, Lu X, Buddha L, Wankhade U, Lohakare J, Isales C, Fulzele S. Gut microbiota dysbiosis in Alzheimer's disease (AD): Insights from human clinical studies and the mouse AD models. Physiol Behav 2025; 290:114778. [PMID: 39672482 DOI: 10.1016/j.physbeh.2024.114778] [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: 08/20/2024] [Revised: 11/19/2024] [Accepted: 12/07/2024] [Indexed: 12/15/2024]
Abstract
Alzheimer's Disease (AD) is a debilitating neurocognitive disorder with an unclear underlying mechanism. Recent studies have implicated gut microbiota dysbiosis with the onset and progression of AD. The connection between gut microbiota and AD can significantly affect the prevention and treatment of AD patients. This systematic review summarizes primary outcomes of human and mouse AD models concerning gut microbiota alterations. A systematic literature search in February through March 2023 was conducted on PubMed, Embase, and Web of Science. We identified 711 as potential manuscripts of which 672 were excluded because of irrelevance to the identified search criteria. Primary outcomes include microbiota compositions of control and AD models in humans and mice. In total, 39 studies were included (19 mouse and 20 human studies), published between 2017 and 2023. We included studies involving well-established mice models of AD (5xFAD, 3xTg-AD, APP/PS1, Tg2576, and APPPS2) which harbor mutations and genes that drive the formation of Aß plaques. All human studies were included on those with AD or mild cognitive impairment. Among alterations in gut microbiota, most studies found a decreased abundance of the phyla Firmicutes and Bifidobacteria, a genus of the phylum Actinomycetota. An increased abundance of the phyla Bacteroidetes and Proteobacteria were identified in animal and human studies. Studies indicated that gut microbiota alter the pathogenesis of AD through its impact on neuroinflammation and permeability of the gastrointestinal tract. The ensuing increase in blood-brain barrier permeability may accelerate Aβ penetrance and formation of neuritic plaques that align with the amyloid hypothesis of AD pathogenesis. Further studies should assess the relationship between gut microbiota and AD progression and therapy preserving beneficial gut microbiota.
Collapse
Affiliation(s)
- John N Manfredi
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Sonu Kumar Gupta
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Sagar Vyavahare
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Ferenc Deak
- Deptment of Neuroscience & Regenerative Medicine, Augusta, GA 30912, USA
| | - Xinyun Lu
- Deptment of Neuroscience & Regenerative Medicine, Augusta, GA 30912, USA
| | - Lasya Buddha
- Arkansas Children's Nutrition Center, Department of Pediatrics, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Umesh Wankhade
- Arkansas Children's Nutrition Center, Department of Pediatrics, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Jayant Lohakare
- College of Agriculture, Food, and Natural Resources, Prairie View A&M University, Prairie View, TX 77446, USA
| | - Carlos Isales
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA; Deptment of Neuroscience & Regenerative Medicine, Augusta, GA 30912, USA; Centre for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Sadanand Fulzele
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA; Deptment of Neuroscience & Regenerative Medicine, Augusta, GA 30912, USA; College of Agriculture, Food, and Natural Resources, Prairie View A&M University, Prairie View, TX 77446, USA; Centre for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA, USA; Department of Cell Biology and Anatomy, Medical College of Georgia, Augusta University, GA, USA; Department of Orthopedic Surgery, Medical College of Georgia, Augusta University, Augusta, GA, USA.
| |
Collapse
|
9
|
Ren R, Zhang G, Ma J, Zheng Y, Zhao Y, Zhang Y, Zhao L. Nebulized seabuckthorn seed oil inhalation attenuates Alzheimer's disease progression in APP/PS1 mice. Sci Rep 2025; 15:6368. [PMID: 39984555 PMCID: PMC11845625 DOI: 10.1038/s41598-025-89747-x] [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: 08/16/2024] [Accepted: 02/07/2025] [Indexed: 02/23/2025] Open
Abstract
Seabuckthorn (Hippophae rhamnoides L.) is known for its medicinal properties in treating various diseases, including neurological conditions. However, the therapeutic effect of inhaled seabuckthorn seed oil (SSO) on Alzheimer's disease (AD) remains not fully understood. This study explores the effects of nebulized inhalation of SSO in 9-month-old APP/PS1 mice over 21 days. The results showed that nebulized SSO improved memory and cognition. Using 7.0T MRI to monitor blood oxygenation level dependent (BOLD) signals revealed that SSO altered the Amplitude of Low Frequency Fluctuations (ALFF) and Regional Homogeneity (ReHo) signaling such as in the amygdala and substantia innominate, and hippocampus. Enzyme-linked immuno sorbent assay (ELISA) and pathological analyses indicated reduced neuroinflammation in plasma and brain, decreased neuronal necrosis, lower β-amyloid (Aβ) protein levels, reduced amyloid deposition, and increased tyrosine hydroxylase activity. Additionally, SSO promoted gut microbiota remodeling by increasing alpha diversity and boosting levels of probiotics such as Verrucomicrobia, Bifidobacterium, Prevotella, and Akkermansia, without adverse effects on lung tissue. Nebulized inhalation of SSO may slow AD progression by modulating inflammation and amyloid deposition. Nebulized inhalation offered a potential method for enhancing drug delivery across the blood-brain barrier with reduced systemic side effects.
Collapse
Affiliation(s)
- Ruichen Ren
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Gaorui Zhang
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Junqing Ma
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Yongze Zheng
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Yuxuan Zhao
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Yang Zhang
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, 250012, China.
| | - Lin Zhao
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan, 250353, China.
| |
Collapse
|
10
|
Man Y, Li X, Cui L, Song J, Cheng C, Zhang X, Niu F. Dydrogesterone alleviates periodontitis in perimenopausal women undergoing periodontal therapy by decreasing inflammation and mediating oral microbiota. Microb Pathog 2025; 201:107380. [PMID: 39956343 DOI: 10.1016/j.micpath.2025.107380] [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: 09/25/2024] [Revised: 02/13/2025] [Accepted: 02/13/2025] [Indexed: 02/18/2025]
Abstract
OBJECTIVE Dydrogesterone (DG), a synthetic isomer of progesterone, plays a potential regulatory role in the periodontal environment. The aim of this study was to investigate the potential effects of DG on periodontitis under periodontal therapy (PT) and the underlying mechanisms related to oral microbiota. METHODS As a cohort study, perimenopausal women with periodontitis and abnormal uterine bleeding associated with ovulatory dysfunction were screened. A total of 30 women received PT (PT group) and 30 women received PT and oral DG 10 mg twice/day for 10 days/month (PT + DG group). At baseline and 3 months after treatment, pocket probing depth (PPD), bleeding index (BI), bleeding on probing (BOP), plaque index, CRP, IL-6, and TNF-α were measured. Additionally, 16S rDNA sequencing was performed to determine the characteristics of oral microbiota, mainly in terms of abundance, diversity, composition, and community structure. RESULTS Three months after treatment, the levels of PPD, BI, and BOP, as well as the levels of CRP, IL-6, and TNF-α in gingival crevicular fluid were significantly lower in the PT + DG group than those in the PT group. After treatment, a relatively lower microbial abundance, and some differences in microbial composition were revealed between the PT and PT + DG groups. At the genus level, significantly fewer Escherichia-Shigella, Porphyromonas, and Absconditabacteriales (SR1), and more Lactobacillus, Gordonia, Bifidobacterium, and Oribacterium were found in the PT group than in the PT + DG group. CONCLUSIONS DG enhances the effect of PT on inhibiting inflammatory response in women with periodontitis by mediating oral microbiota.
Collapse
Affiliation(s)
- Ying Man
- Department of Stomatology, Shengli Oilfield Central Hospital, China
| | - Xiaofei Li
- Department of Stomatology, Shengli Oilfield Central Hospital, China
| | - Liyun Cui
- Department of Stomatology, Shengli Oilfield Central Hospital, China
| | - Jiajia Song
- Department of Stomatology, Shengli Oilfield Central Hospital, China
| | - Cheng Cheng
- Dongying District Hospital of Traditional Chinese Medicine, China
| | - Xinyue Zhang
- Department of Stomatology, Shengli Oilfield Central Hospital, China.
| | - Feifei Niu
- Department of Gynaecology, Shengli Oilfield Central Hospital, China.
| |
Collapse
|
11
|
Bashir B, Gulati M, Vishwas S, Gupta G, Dhanasekaran M, Paudel KR, Chellappan DK, Anand K, Negi P, Singh PK, Rajput A, Dua K, Singh SK. Bridging gap in the treatment of Alzheimer's disease via postbiotics: Current practices and future prospects. Ageing Res Rev 2025; 105:102689. [PMID: 39952328 DOI: 10.1016/j.arr.2025.102689] [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: 12/20/2024] [Accepted: 02/08/2025] [Indexed: 02/17/2025]
Abstract
Aging is an extremely significant risk associated with neurodegeneration. The most prevalent neurodegenerative disorders (NDs), such as Alzheimer's disease (AD) are distinguished by the prevalence of proteinopathy, aberrant glial cell activation, oxidative stress, neuroinflammation, defective autophagy, cellular senescence, mitochondrial dysfunction, epigenetic changes, neurogenesis suppression, increased blood-brain barrier permeability, and intestinal dysbiosis that is excessive for the patient's age. Substantial body studies have documented a close relationship between gut microbiota and AD, and restoring a healthy gut microbiota may reduce or even ameliorate AD symptoms and progression. Thus, control of the microbiota in the gut has become an innovative model for clinical management of AD, and rising emphasis is focused on finding new techniques for preventing and/or managing the disease. The etiopathogenesis of gut microbiota in driving AD progression and supplementing postbiotics as a preventive and therapeutic treatment for AD is discussed. The review additionally discusses the use of postbiotics in AD prophylaxis and therapy, portraying them as substances that address senescence-triggered dysfunctions and are worthy of translating from bench to biopharmaceutical market in response to "silver consumers" needs. The current review examines and evaluates the impact of postbiotics as whole and specific metabolites, such as short-chain fatty acids (SCFAs), lactate, polyamines, polyphenols, tryptophan metabolites, exopolysaccharides, and bacterial extracellular vesicles, on the aging-associated processes that reinforce AD. Moreover, it provides an overview of the most recent data from both clinical and preclinical research involving the use of postbiotics in AD.
Collapse
Affiliation(s)
- Bushra Bashir
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India
| | - Sukriti Vishwas
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India
| | - Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India; Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Muralikrishnan Dhanasekaran
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL 36849, USA
| | - Keshav Raj Paudel
- Centre of Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW 2007, Australia
| | | | - Krishnan Anand
- Precision Medicine and Integrated Nano-Diagnostics (P-MIND) Research Group, Office of the Dean, Faculty of Health Sciences, University of the Free State, Bloemfontein, South Africa
| | - Poonam Negi
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India
| | - Pankaj Kumar Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, India
| | - Amarjitsing Rajput
- Department of Pharmaceutics, Bharti Vidyapeeth Deemed to be University, Poona College of Pharmacy, Erandwane, Pune 411038, Maharashtra, India
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India; Sunway Biofunctional Molecules Discovery Centre (SBMDC), School of Medical and Life Sciences, Sunway University, Sunway, Malaysia.
| |
Collapse
|
12
|
Rout S, Dash R, Satish V, Venugopal G, Rao BN, Bandhyopadhyay D, Bhoi SK, Ramadass B. Exploring the role of acylated ghrelin and gut microbiome in delineating cognitive health in the elderly. Aging (Albany NY) 2025; null:206200. [PMID: 39927883 DOI: 10.18632/aging.206200] [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: 09/10/2024] [Accepted: 01/30/2025] [Indexed: 02/11/2025]
Abstract
INTRODUCTION With increased life expectancy, there is an increase in aging population and prevalence of dementia. Ghrelin is a key regulator of spatial memory and cognition. The gut microbiome may affect the circulating levels of unacylated ghrelin (UAG) and acylated ghrelin (AG). Thus, we explore the potential association of the gut microbiome, AG, and cognitive health in the aging dementia patient. METHODS 40 dementia patients and 40 controls were recruited. Fecal Microbiome analysis using 16S rRNA sequencing was performed on 18 samples. A mixed-method approach was employed for robust interpretation. RESULTS Dementia patients had an increased serum AG and AG/UAG ratio. With the increase in AG among dementia subjects, a significant decrease in species richness was observed. Bifidobacterium longum, Eubacterium biforme, Fecalibacterium prausnitzii, Lactobacillus ruminis, and Prevotella copri contributed to substantial differences in beta-diversity. Blautia obeum was associated with Mini-Mental State Examination (MMSE), and Fecalibacterium prausnitzii was associated with Montreal Cognitive Assessment (MoCA) Scale. DISCUSSION This pilot study indicates a complex interaction between AG, gut microbiome, and cognitive scores. Increased AG corresponds to both dementia and gut dysbiosis, intricately interconnecting the gut-brain axis. The circulating AG and associated gut microbiome might be a putative biomarker for dementia.
Collapse
Affiliation(s)
- Sudeshna Rout
- Department of Biochemistry, All India Institute of Medical Sciences Bhubaneswar, Odisha 751019, India
| | - Rishikesh Dash
- Centre of Excellence for Clinical Microbiome and Research (CCMR), All India Institute of Medical Sciences Bhubaneswar, Odisha 751019, India
| | - Varsha Satish
- Centre of Excellence for Clinical Microbiome and Research (CCMR), All India Institute of Medical Sciences Bhubaneswar, Odisha 751019, India
| | - Giriprasad Venugopal
- Centre of Excellence for Clinical Microbiome and Research (CCMR), All India Institute of Medical Sciences Bhubaneswar, Odisha 751019, India
| | - Bodepudi Narasimha Rao
- Department of Physiology, All India Institute of Medical Sciences Bhubaneswar, Odisha 751019, India
| | - Debapriya Bandhyopadhyay
- Department of Biochemistry, All India Institute of Medical Sciences Bhubaneswar, Odisha 751019, India
| | - Sanjeev Kumar Bhoi
- Department of Neurology, All India Institute of Medical Sciences Bhubaneswar, Odisha 751019, India
| | - Balamurugan Ramadass
- Department of Biochemistry, All India Institute of Medical Sciences Bhubaneswar, Odisha 751019, India
- Centre of Excellence for Clinical Microbiome and Research (CCMR), All India Institute of Medical Sciences Bhubaneswar, Odisha 751019, India
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA 5000, Australia
| |
Collapse
|
13
|
Jiang H, Zeng W, Zhu F, Zhang X, Cao D, Peng A, Wang H. Exploring the associations of gut microbiota with inflammatory and the early hematoma expansion in intracerebral hemorrhage: from change to potential therapeutic objectives. Front Cell Infect Microbiol 2025; 15:1462562. [PMID: 39963412 PMCID: PMC11830820 DOI: 10.3389/fcimb.2025.1462562] [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: 07/10/2024] [Accepted: 01/07/2025] [Indexed: 02/20/2025] Open
Abstract
Background Although a great deal of research has explored the possibility of a systemic inflammatory response and dysbiosis of the gut microbiota after an intracerebral hemorrhage (ICH), the relationships between gut microbiota and blood inflammatory indicators as well as their role in the hematoma expansion following an early-stage mild-to-moderate ICH (emICH) remain unknown. This study analyzes these changes and associations in order to predict and prevent hematoma expansion after emICH. Methods The study included 100 participants, with 70 individuals diagnosed with emICH (30 with hematoma expansion and 40 without hematoma expansion, referred to as the HE and NE groups) and 30 healthy controls matched in terms of age and gender (HC). We used 16S rRNA gene sequencing to explore the gut microbial structure and its underlying associations with blood inflammatory parameters in the HE group. Results Our findings showed a significant decrease in the diversity and even distribution of microorganisms in the HE group when compared to the HC and NE groups. The composition of the gut microbiota experienced notable alterations in the emICH group, especially in HE. These changes included a rise in the number of gram-negative pro-inflammatory bacteria and a decline in the level of probiotics. Furthermore, we observed strong positive connections between bacteria enriched in the HE group and levels of systemic inflammation. Several microbial biomarkers (e.g. Escherichia_Shigella, Enterobacter, and Porphyromonas) were revealed in disparateiating HE from HC and NE. Analysis of the Kyoto Encyclopedia of Genes and Genomes (KEGG) exposed disturbances in essential physiological pathways, especially those related to inflammation (such as the Toll-like receptor signaling pathway), in the HE group. Conclusions Our exploration indicated that individuals with emICH, especially those with HE, demonstrate notably different host-microbe interactions when compared to healthy individuals. We deduced that emICH could rapidly trigger the dysbiosis of intestinal flora, and the disturbed microbiota could, in turn, exacerbate inflammatory response and increase the risk of hematoma expansion. Our comprehensive research revealed the potential of intestinal flora as a potent diagnostic tool, emphasizing its significance as a preventive target for HE.
Collapse
Affiliation(s)
- Haixiao Jiang
- Department of Neurosurgery, The Affiliated Hospital of Yangzhou University, Yangzhou, China
| | - Wei Zeng
- Department of Neurosurgery, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, Yancheng, China
| | - Fei Zhu
- Department of Neurosurgery, The Affiliated Hospital of Yangzhou University, Yangzhou, China
| | - Xiaoli Zhang
- Department of Medical Imaging, The Affiliated Hospital of Yangzhou University, Yangzhou, China
| | - Demao Cao
- Department of Neurosurgery, The Affiliated Hospital of Yangzhou University, Yangzhou, China
| | - Aijun Peng
- Department of Neurosurgery, The Affiliated Hospital of Yangzhou University, Yangzhou, China
| | - Hongsheng Wang
- Department of Neurosurgery, The Affiliated Hospital of Yangzhou University, Yangzhou, China
| |
Collapse
|
14
|
Lista S, Munafò A, Caraci F, Imbimbo C, Emanuele E, Minoretti P, Pinto-Fraga J, Merino-País M, Crespo-Escobar P, López-Ortiz S, Monteleone G, Imbimbo BP, Santos-Lozano A. Gut microbiota in Alzheimer's disease: Understanding molecular pathways and potential therapeutic perspectives. Ageing Res Rev 2025; 104:102659. [PMID: 39800223 DOI: 10.1016/j.arr.2025.102659] [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: 07/19/2024] [Revised: 11/29/2024] [Accepted: 01/09/2025] [Indexed: 01/15/2025]
Abstract
Accumulating evidence suggests that gut microbiota (GM) plays a crucial role in Alzheimer's disease (AD) pathogenesis and progression. This narrative review explores the complex interplay between GM, the immune system, and the central nervous system in AD. We discuss mechanisms through which GM dysbiosis can compromise intestinal barrier integrity, enabling pro-inflammatory molecules and metabolites to enter systemic circulation and the brain, potentially contributing to AD hallmarks. Additionally, we examine other pathophysiological mechanisms by which GM may influence AD risk, including the production of short-chain fatty acids, secondary bile acids, and tryptophan metabolites. The role of the vagus nerve in gut-brain communication is also addressed. We highlight potential therapeutic implications of targeting GM in AD, focusing on antibiotics, probiotics, prebiotics, postbiotics, phytochemicals, and fecal microbiota transplantation. While preclinical studies showed promise, clinical evidence remains limited and inconsistent. We critically assess clinical trials, emphasizing challenges in translating GM-based therapies to AD patients. The reviewed evidence underscores the need for further research to elucidate precise molecular mechanisms linking GM to AD and determine whether GM dysbiosis is a contributing factor or consequence of AD pathology. Future studies should focus on large-scale clinical trials to validate GM-based interventions' efficacy and safety in AD.
Collapse
Affiliation(s)
- Simone Lista
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid 47012, Spain.
| | - Antonio Munafò
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence 50139, Italy.
| | - Filippo Caraci
- Department of Drug and Health Sciences, University of Catania, Catania 95125, Italy; Oasi Research Institute-IRCCS, Troina 94018, Italy.
| | - Camillo Imbimbo
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia 27100, Italy.
| | | | | | - José Pinto-Fraga
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid 47012, Spain.
| | - María Merino-País
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid 47012, Spain.
| | - Paula Crespo-Escobar
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid 47012, Spain.
| | - Susana López-Ortiz
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid 47012, Spain.
| | - Giovanni Monteleone
- Department of Systems Medicine, University of Rome Tor Vergata, Rome 00133, Italy; Unit of Gastroenterology, Policlinico Tor Vergata University Hospital, Rome 00133, Italy.
| | - Bruno P Imbimbo
- Department of Research and Development, Chiesi Farmaceutici, Parma 43122, Italy.
| | - Alejandro Santos-Lozano
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid 47012, Spain; Physical Activity and Health Research Group (PaHerg), Research Institute of the Hospital 12 de Octubre ('imas12'), Madrid 28041, Spain.
| |
Collapse
|
15
|
Gasmi M, Silvia Hardiany N, van der Merwe M, Martins IJ, Sharma A, Williams-Hooker R. The influence of time-restricted eating/feeding on Alzheimer's biomarkers and gut microbiota. Nutr Neurosci 2025; 28:156-170. [PMID: 38953237 DOI: 10.1080/1028415x.2024.2359868] [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: 07/03/2024]
Abstract
OBJECTIVES Alzheimer's disease (AD) is a progressive neurodegenerative disorder affecting approximately 55 million individuals globally. Diagnosis typically occurs in advanced stages, and there are limited options for reversing symptoms. Preventive strategies are, therefore, crucial. Time Restricted Eating (TRE) or Time Restricted Feeding (TRF) is one such strategy. Here we review recent research on AD and TRE/TRF in addition to AD biomarkers and gut microbiota. METHODS A comprehensive review of recent studies was conducted to assess the impact of TRE/TRF on AD-related outcomes. This includes the analysis of how TRE/TRF influences circadian rhythms, beta-amyloid 42 (Aß42), pro-inflammatory cytokines levels, and gut microbiota composition. RESULTS TRE/TRF impacts circadian rhythms and can influence cognitive performance as observed in AD. It lowers beta-amyloid 42 deposition in the brain, a key AD biomarker, and reduces pro-ininflammatory cytokines. The gut microbiome has emerged as a modifiable factor in AD treatment. TRE/TRF changes the structure and composition of the gut microbiota, leading to increased diversity and a decrease in harmful bacteria. DISCUSSION These findings underscore the potential of TRE/TRF as a preventive strategy for AD. By reducing Aß42 plaques, modulating pro-inflammatory cytokines, and altering gut microbiota composition, TRE/TRF may slow the progression of AD. Further research is needed to confirm these effects and to understand the mechanisms involved. This review highlights TRE/TRF as a promising non-pharmacological intervention in the fight against AD.
Collapse
Affiliation(s)
- Maha Gasmi
- Higher Institute of Sport and Physical Education of Ksar said, Tunis, Tunisia
| | - Novi Silvia Hardiany
- Department of Biochemistry & Molecular Biology, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia
- Molecular Biology and Proteomic Core Facilities, Indonesia Medical Education and Research Institute, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia
| | - Marie van der Merwe
- Center for Nutraceuticals and Dietary Supplement Research, College of Health Sciences, University of Memphis, Memphis, TN, USA
| | - Ian J Martins
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia
| | - Aastha Sharma
- Department of Basic and Applied Science. School of Engineering and Science, University - GD Goenka University Gurugram, India
| | | |
Collapse
|
16
|
Chevalier C, Tournier BB, Marizzoni M, Park R, Paquis A, Ceyzériat K, Badina AM, Lathuiliere A, Saleri S, Cillis FD, Cattaneo A, Millet P, Frisoni GB. Fecal Microbiota Transplantation (FMT) From a Human at Low Risk for Alzheimer's Disease Improves Short-Term Recognition Memory and Increases Neuroinflammation in a 3xTg AD Mouse Model. GENES, BRAIN, AND BEHAVIOR 2025; 24:e70012. [PMID: 39801363 PMCID: PMC11725982 DOI: 10.1111/gbb.70012] [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: 09/20/2024] [Revised: 11/15/2024] [Accepted: 12/18/2024] [Indexed: 01/16/2025]
Abstract
Human microbiota-associated murine models, using fecal microbiota transplantation (FMT) from human donors, help explore the microbiome's role in diseases like Alzheimer's disease (AD). This study examines how gut bacteria from donors with protective factors against AD influence behavior and brain pathology in an AD mouse model. Female 3xTgAD mice received weekly FMT for 2 months from (i) an 80-year-old AD patient (AD-FMT), (ii) a cognitively healthy 73-year-old with the protective APOEe2 allele (APOEe2-FMT), (iii) a 22-year-old healthy donor (Young-FMT), and (iv) untreated mice (Mice-FMT). Behavioral assessments included novel object recognition (NOR), Y-maze, open-field, and elevated plus maze tests; brain pathology (amyloid and tau), neuroinflammation (in situ autoradiography of the 18 kDa translocator protein in the hippocampus); and gut microbiota were analyzed. APOEe2-FMT improved short-term memory in the NOR test compared to AD-FMT, without significant changes in other behavioral tests. This was associated with increased neuroinflammation in the hippocampus, but no effect was detected on brain amyloidosis and tauopathy. Specific genera, such as Parabacteroides and Prevotellaceae_UGC001, were enriched in the APOEe2-FMT group and associated with neuroinflammation, while genera like Desulfovibrio were reduced and linked to decreased neuroinflammation. Gut microbiota from a donor with a protective factor against AD improved short-term memory and induced neuroinflammation in regions strategic to AD. The association of several genera with neuroinflammation in the APOEe2-FMT group suggests a collegial effect of the transplanted microbiome rather than a single-microbe driver effect. These data support an association between gut bacteria, glial cell activation, and cognitive function in AD.
Collapse
Affiliation(s)
- Claire Chevalier
- Département de Readaptation et gériatrieUniversity of GenevaGenevaSwitzerland
| | | | - Moira Marizzoni
- Biological Psychiatry UnitIRCCS Centro San Giovanni di Dio FatebenefratelliBresciaItaly
| | - Rahel Park
- Département de Readaptation et gériatrieUniversity of GenevaGenevaSwitzerland
| | - Arthur Paquis
- Département de Readaptation et gériatrieUniversity of GenevaGenevaSwitzerland
| | - Kelly Ceyzériat
- Département de PsychiatrieUniversity of GenevaGenevaSwitzerland
| | | | | | - Samantha Saleri
- Biological Psychiatry UnitIRCCS Centro San Giovanni di Dio FatebenefratelliBresciaItaly
| | - Floriana De Cillis
- Biological Psychiatry UnitIRCCS Centro San Giovanni di Dio FatebenefratelliBresciaItaly
- Department of Pharmacological and Biomolecular SciencesUniversity of MilanMilanItaly
| | - Annamaria Cattaneo
- Biological Psychiatry UnitIRCCS Centro San Giovanni di Dio FatebenefratelliBresciaItaly
- Department of Pharmacological and Biomolecular SciencesUniversity of MilanMilanItaly
| | - Philippe Millet
- Département de PsychiatrieUniversity of GenevaGenevaSwitzerland
| | - Giovanni B. Frisoni
- Département de Readaptation et gériatrieUniversity of GenevaGenevaSwitzerland
| |
Collapse
|
17
|
Almutary AG, Begum MY, Kyada AK, Gupta S, Jyothi SR, Chaudhary K, Sharma S, Sinha A, Abomughaid MM, Imran M, Lakhanpal S, Babalghith AO, Abu-Seer EA, Avinash D, Alzahrani HA, Alhindi AA, Iqbal D, Kumar S, Jha NK, Alghamdi S. Inflammatory signaling pathways in Alzheimer's disease: Mechanistic insights and possible therapeutic interventions. Ageing Res Rev 2025; 104:102548. [PMID: 39419399 DOI: 10.1016/j.arr.2024.102548] [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: 01/26/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/19/2024]
Abstract
The complex pathophysiology of Alzheimer's disease (AD) poses challenges for the development of therapies. Recently, neuroinflammation has been identified as a key pathogenic mechanism underlying AD, while inflammation has emerged as a possible target for the management and prevention of AD. Several prior studies have demonstrated that medications modulating neuroinflammation might lessen AD symptoms, mostly by controlling neuroinflammatory signaling pathways such as the NF-κB, MAPK, NLRP3, etc, and their respective signaling cascade. Moreover, targeting these inflammatory modalities with inhibitors, natural products, and metabolites has been the subject of intensive research because of their anti-inflammatory characteristics, with many studies demonstrating noteworthy pharmacological capabilities and potential clinical applications. Therefore, targeting inflammation is considered a promising strategy for treating AD. This review comprehensively elucidates the neuroinflammatory mechanisms underlying AD progression and the beneficial effects of inhibitors, natural products, and metabolites in AD treatment.
Collapse
Affiliation(s)
- Abdulmajeed G Almutary
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, P.O. Box 59911, Abu Dhabi, United Arab Emirates
| | - M Yasmin Begum
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Ashish Kumar Kyada
- Marwadi University Research Center, Department of Pharmaceutical Sciences, Faculty of Health Sciences, Marwadi University, Rajkot, Gujarat 360003, India
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura, Uttar Pradesh, India
| | - S Renuka Jyothi
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Kamlesh Chaudhary
- Department of Neurology, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - Swati Sharma
- Chandigarh Pharmacy College, Chandigarh Group of Colleges, Jhanjeri, Mohali, Punjab 140307, India
| | - Aashna Sinha
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, Uttarakhand
| | - Mosleh Mohammad Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha 61922, Saudi Arabia
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia; Center for Health Research, Northern Border University, Arar, Saudi Arabia
| | - Sorabh Lakhanpal
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Ahmad O Babalghith
- Medical Genetics Department, College of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Eman Adnan Abu-Seer
- Department of Epidemiology and Medical Statistic, Faculty of Public Health and Health Informatics, Umm Al-Qura University, Makkah, Saudi Arabia
| | - D Avinash
- Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, India
| | - Hassan A Alzahrani
- Department of Respiratory Care, Medical Cities at the Minister of Interior, MCMOl, Riyadh, Saudi Arabia
| | | | - Danish Iqbal
- Department of Health Information Management, College of Applied Medical Sciences, Buraydah Private Colleges, Buraydah 51418, Saudi Arabia
| | - Sandeep Kumar
- School of Pharmacy, Sharda University, Greater Noida, India; DST-FIST Laboratory, Sharda University, Greater Noida, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Biosciences and Technology (SBT), Galgotias University, Greater Noida, India; Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura 140401, Punjab, India.
| | - Saad Alghamdi
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| |
Collapse
|
18
|
Zhou XP, Sun LB, Liu WH, Zhu WM, Li LC, Song XY, Xing JP, Gao SH. The complex relationship between gut microbiota and Alzheimer's disease: A systematic review. Ageing Res Rev 2025; 104:102637. [PMID: 39662839 DOI: 10.1016/j.arr.2024.102637] [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: 10/20/2024] [Revised: 12/06/2024] [Accepted: 12/06/2024] [Indexed: 12/13/2024]
Abstract
Alzheimer's disease (AD) is a progressive, degenerative disorder of the central nervous system. Despite extensive research conducted on this disorder, its precise pathogenesis remains unclear. In recent years, the microbiota-gut-brain axis has attracted considerable attention within the field of AD. The gut microbiota communicates bidirectionally with the central nervous system through the gut-brain axis, and alterations in its structure and function can influence the progression of AD. Consequently, regulating the gut microbiota to mitigate the progression of AD has emerged as a novel therapeutic approach. Currently, numerous studies concentrate on the intrinsic relationship between the microbiota-gut-brain axis and AD. In this paper, we summarize the multifaceted role of the gut microbiota in AD and present detailed therapeutic strategies targeting the gut microbiota, including the treatment of AD with Traditional Chinese Medicine (TCM), which has garnered increasing attention in recent years. Finally, we discuss potential therapeutic strategies for modulating the gut microbiota to alleviate the progression of AD, the current challenges in this area of research, and provide an outlook on future research directions in this field.
Collapse
Affiliation(s)
- Xuan-Peng Zhou
- China-Japan Union Hospital of Jilin University, Changchun, Jilin 130000, PR China
| | - Luan-Biao Sun
- China-Japan Union Hospital of Jilin University, Changchun, Jilin 130000, PR China
| | - Wen-Hao Liu
- China-Japan Union Hospital of Jilin University, Changchun, Jilin 130000, PR China
| | - Wu-Ming Zhu
- China-Japan Union Hospital of Jilin University, Changchun, Jilin 130000, PR China
| | - Lin-Chun Li
- China-Japan Union Hospital of Jilin University, Changchun, Jilin 130000, PR China
| | - Xin-Yuan Song
- The Chinese University of Hong Kong, New Territories 999077, Hong Kong
| | - Jian-Peng Xing
- China-Japan Union Hospital of Jilin University, Changchun, Jilin 130000, PR China.
| | - Shuo-Hui Gao
- China-Japan Union Hospital of Jilin University, Changchun, Jilin 130000, PR China.
| |
Collapse
|
19
|
Wei C, Wu X, Li C, Zhang Y, Yuan Q, Huang R. Aerobic exercise regulates gut microbiota profiles and metabolite in the early stage of Alzheimer's disease. FASEB J 2025; 39:e70327. [PMID: 39831888 PMCID: PMC11745210 DOI: 10.1096/fj.202402572r] [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: 10/24/2024] [Revised: 12/30/2024] [Accepted: 01/06/2025] [Indexed: 01/22/2025]
Abstract
Aerobic exercise (AE) has been shown to offer significant benefits for Alzheimer's disease (AD), potentially influencing the gut microbiota. However, the impact of changes in intestinal flora in early Alzheimer's disease induced by aerobic exercise on metabolic pathways and metabolites is not well understood. In this study, 3-month-old APP/PS1 and C57BL/6 mice were divided into two groups each: a control group (ADC for APP/PS1 and WTC for C57BL/6) and an aerobic exercise group (ADE for APP/PS1 and WTE for C57BL/6). The exercise groups underwent a 20-week aerobic training program on a motorized treadmill before the behavioral test (both the Morris water maze experiment (MWM) and the eight-arm maze test). Fecal samples were collected to analyze gut microbiota profiles via 16S rRNA gene sequencing. At the same time, the metabolic pathway analysis and the detection of metabolites were carried out. At the phylum level, the ADE group exhibited a significant reduced in the relative abundance of Bacteroidetes compared to the ADC group. At the genus level, both Ileibacterium and Faecalibaculum were found to be more abundant in the ADE group than in the ADC group. Additionally, PICRUSt analysis revealed that lipid metabolism and bile acid metabolism pathways were significantly enriched in the cecal microbiota of mice in the ADE group. The metabolites detected further confirmed the changes in the metabolic pathways mentioned above. Aerobic exercise may modify gut microbiota profiles and metabolites in APP/PS1 mice, thereby potentially playing a beneficial role in delaying cognitive impairment associated with early-stage Alzheimer's disease.
Collapse
Affiliation(s)
- Cuilan Wei
- School of Sports Medicine and HealthChengdu Sport UniversityChengduChina
- Sports Institute of Chengdu University of TechnologyChengduChina
| | - Xiaojing Wu
- Department of Orthopaedics, Sichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Chuikun Li
- School of Physical EducationChengdu UniversityChengduChina
| | - Yeting Zhang
- Civil Aviation Flight University of ChinaGuanghanChina
| | - Qiongjia Yuan
- School of Sports Medicine and HealthChengdu Sport UniversityChengduChina
| | - Rui Huang
- Department of Neurology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduChina
| |
Collapse
|
20
|
Palermo F, Marrocco N, Dacomo L, Grisafi E, Moresi V, Sanna A, Massimi L, Musella M, Maugeri L, Bukreeva I, Fiordaliso F, Corbelli A, Junemann O, Eckermann M, Cloetens P, Weitkamp T, Gigli G, de Rosbo NK, Balducci C, Cedola A. Investigating gut alterations in Alzheimer's disease: In-depth analysis with micro- and nano-3D X-ray phase contrast tomography. SCIENCE ADVANCES 2025; 11:eadr8511. [PMID: 39889000 PMCID: PMC11784835 DOI: 10.1126/sciadv.adr8511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 01/03/2025] [Indexed: 02/02/2025]
Abstract
Alzheimer's disease (AD), a debilitating neurodegenerative disorder, remains one of the foremost public health challenges affecting more than 30 million people worldwide with the etiology still largely enigmatic. The intricate gut-brain axis, serving as a vital communication network between the gut and the brain, appears to wield influence in the progression of AD. Our study showcases the remarkable precision of x-ray phase-contrast tomography (XPCT) in conducting an advanced three-dimensional examination of gut cellular composition and structure. The exploitation of micro- and nano-XPCT on various AD mouse models unveiled relevant alterations in villi and crypts, cellular transformations in Paneth and goblet cells, along with the detection of telocytes, neurons, erythrocytes, and mucus secretion by goblet cells within the gut cavity. The observed gut structural variations may elucidate the transition from dysbiosis to neurodegeneration and cognitive decline. Leveraging XPCT could prove pivotal in early detection and prognosis of the disease.
Collapse
Affiliation(s)
| | | | - Letizia Dacomo
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Elena Grisafi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | | | | | | | | | | | | | - Fabio Fiordaliso
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | | | | | | | | | | | - Giuseppe Gigli
- Institute of Nanotechnology – CNR, Lecce, Italy
- University of Salento, Lecce, Italy
| | | | - Claudia Balducci
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | | |
Collapse
|
21
|
Zheng Y, Yu Y, Chen M, Zhang H, Wang W, Fan X, Sun L, Tang L, Ta D. Abdominal LIPUS Stimulation Prevents Cognitive Decline in Hind Limb Unloaded Mice by Regulating Gut Microbiota. Mol Neurobiol 2025:10.1007/s12035-025-04709-8. [PMID: 39878866 DOI: 10.1007/s12035-025-04709-8] [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: 05/13/2024] [Accepted: 01/14/2025] [Indexed: 01/31/2025]
Abstract
Weightlessness usually causes disruption of the gut microbiota and impairs cognitive function. There is a close connection between gut microbiota and neurological diseases. Low-intensity pulsed ultrasound (LIPUS) has a beneficial effect on reducing intestinal inflammation. So we wondered if abdominal LIPUS stimulation can have a positive impact on weightlessness induced cognitive decline by reducing intestinal dysfunction. The findings revealed that the hind limb unloaded mice exhibited evident disruption in intestinal structure and gut microbial homeostasis, along with impairment in their learning and memory capabilities. However, 4-week abdominal LIPUS treatment improved intestinal function in hind limb unloaded mice, characterized by upregulation of tight junction proteins ZO-1 and Occludin expression in the colon, increased diversity and abundance of intestinal microbiota, decreased serum lipopolysaccharide (LPS), and increased short chain fatty acids in colon contents. The hind limb unloaded mice treated with LIPUS exhibited heightened activity levels, improved exploratory tendencies, and significantly enhanced learning and memory faculties, and elevated expression of neuroadaptation-related proteins such as PSD95, GAP43, P-CREB, BDNF, and its receptor TRKB in the hippocampus. Furthermore, the hind limb unloaded mice receiving fecal transplants from the mice whose abdomens were irradiated with LIPUS displayed enhanced cognitive abilities and improved intestinal structure, akin to the outcomes observed in hind limb unloaded mice who received LIPUS abdominal treatment directly. The above results indicate that LIPUS enhances intestinal structure and microbiota, which helps alleviate cognitive impairment caused by weightlessness. LIPUS could be a potential strategy to simultaneously improve gut dysfunction and cognitive decline in astronauts or bedridden patients.
Collapse
Affiliation(s)
- Yumei Zheng
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China
| | - Yanan Yu
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China
| | - Mengyao Chen
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China
| | - Huiyuan Zhang
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China
| | - Wanzhao Wang
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China
| | - Xiushan Fan
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China
| | - Lijun Sun
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China.
| | - Liang Tang
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China.
| | - Dean Ta
- Center for Biomedical Engineering, School of Information Science and Technology, Fudan University, Shanghai, 200433, China.
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
22
|
Brown JA, Bashir H, Zeng MY. Lifelong partners: Gut microbiota-immune cell interactions from infancy to old age. Mucosal Immunol 2025:S1933-0219(25)00006-6. [PMID: 39862964 DOI: 10.1016/j.mucimm.2025.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/10/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025]
Abstract
Our immune system and gut microbiota are intricately coupled from birth, both going through maturation during early life and senescence during aging almost in a synchronized fashion. The symbiotic relationship between the human host and microbiota is critically dependent on a healthy immune system to keep our microbiota in check, while the microbiota provides essential functions to promote the development and fitness of our immune system. The partnership between our immune system and microbiota is particularly important during early life, when microbial ligands and metabolites shape the development of the immune cells and immune tolerance; during aging, having sufficient beneficial gut bacteria is critical for the maintenance of intact mucosal barriers, immune metabolic fitness, and strong immunity against pathogens. The immune system during childhood is programmed, with the support of the microbiota, to develop robust immune tolerance, and limit autoimmunity and metabolic dysregulation, which are prevalent during aging. This review comprehensively explores the mechanistic underpinnings of gut microbiota-immune cell interactions during infancy and old age, with the goal to gain a better understanding of potential strategies to leverage the gut microbiota to combat age-related immune decline.
Collapse
Affiliation(s)
- Julia A Brown
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, United States; Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, United States
| | - Hilal Bashir
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, United States; Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, United States
| | - Melody Y Zeng
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, United States; Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, United States; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School, New York, NY 10065, United States.
| |
Collapse
|
23
|
Wang G, Li Y, Liu H, Yu X. Gut microbiota in patients with sarcopenia: a systematic review and meta-analysis. Front Microbiol 2025; 16:1513253. [PMID: 39911254 PMCID: PMC11794218 DOI: 10.3389/fmicb.2025.1513253] [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: 10/30/2024] [Accepted: 01/10/2025] [Indexed: 02/07/2025] Open
Abstract
Background Intestinal dysbiosis was considered a pivotal pathological mechanism underlying sarcopenia. Despite the fervor surrounding research in this domain, substantial controversy persists regarding the obtained outcomes. Objective To systematically summarized the disparities in gut microbiota composition between the group afflicted by sarcopenia and non-sarcopenia controls. Methods PubMed, Medline, CINAHL, EMBASE, Scopus, Web of Science and Google Scholer, Cochrane Library and gray literature sources were systematically searched for in randomized controlled trials. Meta-analysis and random-effects meta-regression were conducted using Rev. Man 5.3. Overall effect was measured using Hedges's g and determined using Z-statistics. Cochran's Q test and I 2 were used to investigate heterogeneity. The Newcastle-Ottawa Scale was used to assess overall quality of evidence. Results Ten studies, including 421 cases of sarcopenia and 1,642 cases of controls, were included in the meta-analysis. Patients with sarcopenia showed significantly reduced gut microbiota in α diversity, and β diversity was significantly different in 8/9 of included studies. We also found more abundance of phylum Proteobacteria and genus Escherichia-Shigella, and less abundance of phylum Firmicutes and genus Faecalibacterium, Prevotella 9, Blautia in the sarcopenia group. Conclusion The gut microbiota composition in patients with sarcopenia has undergone alterations, serving as a fundamental reference for further investigation into the potential pathogenic mechanisms and treatment strategies for sarcopenia.
Collapse
Affiliation(s)
- Guangning Wang
- Department of Critical Care Medicine, Qingdao Hospital, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Yujie Li
- Reproductive Medicine Center, Women and Children’s Hospital, Qingdao University, Qingdao, China
| | - Huisong Liu
- Department of Nursing, Qingdao Hospital, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Xinjuan Yu
- Department of Clinical Research Center, Qingdao Hospital, University of Health and Rehabilitation Sciences, Qingdao, China
| |
Collapse
|
24
|
Ghannadzadeh Kermani Pour R, Kamali Zounouzi S, Farshbafnadi M, Rezaei N. The interplay between gut microbiota composition and dementia. Rev Neurosci 2025:revneuro-2024-0113. [PMID: 39829047 DOI: 10.1515/revneuro-2024-0113] [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: 08/20/2024] [Accepted: 01/03/2025] [Indexed: 01/22/2025]
Abstract
Recently, researchers have been interested in the potential connection between gut microbiota composition and various neuropsychological disorders. Dementia significantly affects the socioeconomics of families. Gut microbiota is considered as a probable factor in its pathogenesis. Multiple bacterial metabolites such as short-chain fatty acids, lipopolysaccharides, and various neurotransmitters that are responsible for the incidence and progression of dementia can be produced by gut microbiota. Various bacterial species such as Bifidobacterium breve, Akkermansia muciniphila, Streptococcus thermophilus, Escherichia coli, Blautia hydrogenotrophica, etc. are implicated in the pathogenesis of dementia. Gut microbiota can be a great target for imitating or inhibiting their metabolites as an adjunctive therapy based on their role in its pathogenesis. Therefore, some diets can prevent or decelerate dementia by altering the gut microbiota composition. Moreover, probiotics can modulate gut microbiota composition by increasing beneficial bacteria and reducing detrimental species. These therapeutic modalities are considered novel methods that are probably safe and effective. They can enhance the efficacy of traditional medications and improve cognitive function.
Collapse
Affiliation(s)
| | - Sara Kamali Zounouzi
- School of Medicine, 48439 Tehran University of Medical Sciences , Tehran, 1416634793, Iran
| | - Melina Farshbafnadi
- School of Medicine, 48439 Tehran University of Medical Sciences , Tehran, 1416634793, Iran
- Universal Scientific Education and Research Network (USERN), Tehran, 1416634793, Iran
| | - Nima Rezaei
- Universal Scientific Education and Research Network (USERN), Tehran, 1416634793, Iran
- Research Center for Immunodeficiencies, Children's Medical Center, 48439 Tehran University of Medical Sciences , Tehran, 1416634793, Iran
- Department of Immunology, School of Medicine, 48439 Tehran University of Medical Sciences , Tehran, 1416634793, Iran
| |
Collapse
|
25
|
Kern L, Mastandrea I, Melekhova A, Elinav E. Mechanisms by which microbiome-derived metabolites exert their impacts on neurodegeneration. Cell Chem Biol 2025; 32:25-45. [PMID: 39326420 DOI: 10.1016/j.chembiol.2024.08.014] [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: 05/29/2024] [Revised: 07/18/2024] [Accepted: 08/27/2024] [Indexed: 09/28/2024]
Abstract
Recent developments in microbiome research suggest that the gut microbiome may remotely modulate central and peripheral neuronal processes, ranging from early brain development to age-related changes. Dysbiotic microbiome configurations have been increasingly associated with neurological disorders, such as neurodegeneration, but causal understanding of these associations remains limited. Most mechanisms explaining how the microbiome may induce such remote neuronal effects involve microbially modulated metabolites that influx into the 'sterile' host. Some metabolites are able to cross the blood-brain barrier (BBB) to reach the central nervous system, where they can impact a variety of cells and processes. Alternatively, metabolites may directly signal to peripheral nerves to act as neurotransmitters or exert modulatory functions, or impact immune responses, which, in turn, modulate neuronal function and associated disease propensity. Herein, we review the current knowledge highlighting microbiome-modulated metabolite impacts on neuronal disease, while discussing unknowns, controversies and prospects impacting this rapidly evolving research field.
Collapse
Affiliation(s)
- Lara Kern
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Ignacio Mastandrea
- Microbiome & Cancer Division, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anna Melekhova
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Eran Elinav
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel; Microbiome & Cancer Division, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
26
|
Sobral J, Empadinhas N, Esteves AR, Cardoso SM. Impact of Nutrition on the Gut Microbiota: Implications for Parkinson's Disease. Nutr Rev 2025:nuae208. [PMID: 39812804 DOI: 10.1093/nutrit/nuae208] [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] [Indexed: 01/16/2025] Open
Abstract
Parkinson's disease (PD) is a multifactorial neurodegenerative disease that is characterized by the degeneration of dopaminergic neurons in the substantia nigra pars compacta and by the anomalous accumulation of α-synuclein aggregates into Lewy bodies and Lewy neurites. Research suggests 2 distinct subtypes of PD: the brain-first subtype if the pathology arises from the brain and then spreads to the peripheral nervous system (PNS) and the body-first subtype, where the pathological process begins in the PNS and then spreads to the central nervous system. This review primarily focuses on the body-first subtype. The influence of the gut microbiota on the development of PD has been the subject of growing interest among researchers. It has been suggested that gut inflammation may be closely associated with pathogenesis in PD, therefore leading to the hypothesis that gut microbiota modulation could play a significant role in this process. Nutrition can influence gut health and alter the risk and progression of PD by altering inflammatory markers. This review provides an overview of recent research that correlates variations in gut microbiota composition between patients with PD and healthy individuals with the impact of certain nutrients and dietary patterns, including the Mediterranean diet, the Western diet, and the ketogenic diet. It explores how these diets influence gut microbiota composition and, consequently, the risk of PD. Last, it examines fecal transplantation and the use of prebiotics, probiotics, or synbiotics as potential therapeutic strategies to balance the gut microbiome, aiming to reduce the risk or delay the progression of PD.
Collapse
Affiliation(s)
- Joana Sobral
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra 3004-504, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra 3004-504, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra 3000-548, Portugal
| | - Nuno Empadinhas
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra 3004-504, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra 3004-504, Portugal
| | - Ana Raquel Esteves
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra 3004-504, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra 3004-504, Portugal
| | - Sandra Morais Cardoso
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra 3004-504, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra 3004-504, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra 3000-548, Portugal
| |
Collapse
|
27
|
Wang Y, Zhou Z, Broder JC, Woods RL, Orchard SG, Wolfe R, Ernst EJ, Ryan J, Ernst ME, Chan AT. Antibiotic Use and Subsequent Cognitive Decline and Dementia Risk in Healthy Older Adults. Neurology 2025; 104:e210129. [PMID: 39693592 DOI: 10.1212/wnl.0000000000210129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 09/25/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Antibiotics rapidly reduce intestinal bacterial diversity, leading to dysbiosis that persists for months to years. Although emerging evidence from retrospective and claims-based studies has linked dysbiosis to cognitive function, prospective data are lacking. We aim to examine the prospective association of antibiotics with cognitive aging among initially healthy older adults. METHODS We leveraged data from prospective follow-up and observational extension of ASPirin in Reducing Events in the Elderly, a completed randomized trial of community-based Australian older adults. Among participants whose prescription records were available and without dementia during the first 2 years of follow-up, we identified any or repeated antibiotic use based on the Anatomical Therapeutic Chemical code (J01). We assessed the associations of antibiotic use during the first 2 years with longitudinal changes in standardized composite and domain-specific cognitive scores (global cognition, episodic memory, language and executive function, and psychomotor speed) using linear mixed models, and with incident, clinically adjudicated dementia (Diagnostic and Statistical Manual for Mental Disorders, Fourth Edition criteria) and incident cognitive impairment, no dementia (CIND, without a dementia trigger but with significant, nontransient decline), using Cox proportional hazard models. RESULTS Over a median of 4.7 years after the second follow-up visit, we documented 461 dementia and 2,576 CIND cases among 13,571 participants (mean age ± SD 75.0 ± 4.1 years, 54.3% female). Compared with nonuse, antibiotic use was not associated with increased risks for dementia (hazard ratio [HR] 1.03; 95% CI 0.84-1.25), CIND (HR 1.02; 95% CI 0.94-1.11), or subsequent declines in cognitive scores, after adjusting for sociodemographic, lifestyle factors, family history of dementia, baseline cognitive function, and medications known to affect cognition. There were also no associations according to the cumulative frequency of antibiotic use, long-term use, specific antibiotic classes (e.g., beta-lactams, tetracyclines, and sulfonamides), and subgroups defined by risk factors. DISCUSSION Among initially healthy older adults, any or repeated antibiotic use was not associated with incident dementia, CIND, or accelerated cognitive decline. Although prescription data may not reflect the actual use, we examined the frequency of antibiotics within a defined period to capture the extent and duration of antibiotic exposure. Our results do not support an association between antibiotic-associated gut microbiome disruption and dementia risk.
Collapse
Affiliation(s)
- Yiqing Wang
- From the Clinical and Translational Epidemiology Unit (Y.W., A.T.C.), and Division of Gastroenterology (Y.W., A.T.C.), Massachusetts General Hospital and Harvard Medical School, Boston; School of Public Health and Preventive Medicine (Z.Z., J.C.B., R.L.W., S.G.O., R.W., J.R.), Monash University, Melbourne; Menzies Institute for Medical Research (Z.Z.), University of Tasmania, Australia; Department of Pharmacy Practice and Science (E.J.E.. M.E.E.), College of Pharmacy, and Department of Family Medicine (M.E.E.), Carver College of Medicine, University of Iowa, Iowa City; Department of Immunology and Infectious Diseases (A.T.C.), Harvard T.H. Chan School of Public Health, Boston; and Cancer Center (A.T.C.), Massachusetts General Hospital, Boston, MA
| | - Zhen Zhou
- From the Clinical and Translational Epidemiology Unit (Y.W., A.T.C.), and Division of Gastroenterology (Y.W., A.T.C.), Massachusetts General Hospital and Harvard Medical School, Boston; School of Public Health and Preventive Medicine (Z.Z., J.C.B., R.L.W., S.G.O., R.W., J.R.), Monash University, Melbourne; Menzies Institute for Medical Research (Z.Z.), University of Tasmania, Australia; Department of Pharmacy Practice and Science (E.J.E.. M.E.E.), College of Pharmacy, and Department of Family Medicine (M.E.E.), Carver College of Medicine, University of Iowa, Iowa City; Department of Immunology and Infectious Diseases (A.T.C.), Harvard T.H. Chan School of Public Health, Boston; and Cancer Center (A.T.C.), Massachusetts General Hospital, Boston, MA
| | - Jonathan C Broder
- From the Clinical and Translational Epidemiology Unit (Y.W., A.T.C.), and Division of Gastroenterology (Y.W., A.T.C.), Massachusetts General Hospital and Harvard Medical School, Boston; School of Public Health and Preventive Medicine (Z.Z., J.C.B., R.L.W., S.G.O., R.W., J.R.), Monash University, Melbourne; Menzies Institute for Medical Research (Z.Z.), University of Tasmania, Australia; Department of Pharmacy Practice and Science (E.J.E.. M.E.E.), College of Pharmacy, and Department of Family Medicine (M.E.E.), Carver College of Medicine, University of Iowa, Iowa City; Department of Immunology and Infectious Diseases (A.T.C.), Harvard T.H. Chan School of Public Health, Boston; and Cancer Center (A.T.C.), Massachusetts General Hospital, Boston, MA
| | - Robyn L Woods
- From the Clinical and Translational Epidemiology Unit (Y.W., A.T.C.), and Division of Gastroenterology (Y.W., A.T.C.), Massachusetts General Hospital and Harvard Medical School, Boston; School of Public Health and Preventive Medicine (Z.Z., J.C.B., R.L.W., S.G.O., R.W., J.R.), Monash University, Melbourne; Menzies Institute for Medical Research (Z.Z.), University of Tasmania, Australia; Department of Pharmacy Practice and Science (E.J.E.. M.E.E.), College of Pharmacy, and Department of Family Medicine (M.E.E.), Carver College of Medicine, University of Iowa, Iowa City; Department of Immunology and Infectious Diseases (A.T.C.), Harvard T.H. Chan School of Public Health, Boston; and Cancer Center (A.T.C.), Massachusetts General Hospital, Boston, MA
| | - Suzanne Gaye Orchard
- From the Clinical and Translational Epidemiology Unit (Y.W., A.T.C.), and Division of Gastroenterology (Y.W., A.T.C.), Massachusetts General Hospital and Harvard Medical School, Boston; School of Public Health and Preventive Medicine (Z.Z., J.C.B., R.L.W., S.G.O., R.W., J.R.), Monash University, Melbourne; Menzies Institute for Medical Research (Z.Z.), University of Tasmania, Australia; Department of Pharmacy Practice and Science (E.J.E.. M.E.E.), College of Pharmacy, and Department of Family Medicine (M.E.E.), Carver College of Medicine, University of Iowa, Iowa City; Department of Immunology and Infectious Diseases (A.T.C.), Harvard T.H. Chan School of Public Health, Boston; and Cancer Center (A.T.C.), Massachusetts General Hospital, Boston, MA
| | - Rory Wolfe
- From the Clinical and Translational Epidemiology Unit (Y.W., A.T.C.), and Division of Gastroenterology (Y.W., A.T.C.), Massachusetts General Hospital and Harvard Medical School, Boston; School of Public Health and Preventive Medicine (Z.Z., J.C.B., R.L.W., S.G.O., R.W., J.R.), Monash University, Melbourne; Menzies Institute for Medical Research (Z.Z.), University of Tasmania, Australia; Department of Pharmacy Practice and Science (E.J.E.. M.E.E.), College of Pharmacy, and Department of Family Medicine (M.E.E.), Carver College of Medicine, University of Iowa, Iowa City; Department of Immunology and Infectious Diseases (A.T.C.), Harvard T.H. Chan School of Public Health, Boston; and Cancer Center (A.T.C.), Massachusetts General Hospital, Boston, MA
| | - Erika J Ernst
- From the Clinical and Translational Epidemiology Unit (Y.W., A.T.C.), and Division of Gastroenterology (Y.W., A.T.C.), Massachusetts General Hospital and Harvard Medical School, Boston; School of Public Health and Preventive Medicine (Z.Z., J.C.B., R.L.W., S.G.O., R.W., J.R.), Monash University, Melbourne; Menzies Institute for Medical Research (Z.Z.), University of Tasmania, Australia; Department of Pharmacy Practice and Science (E.J.E.. M.E.E.), College of Pharmacy, and Department of Family Medicine (M.E.E.), Carver College of Medicine, University of Iowa, Iowa City; Department of Immunology and Infectious Diseases (A.T.C.), Harvard T.H. Chan School of Public Health, Boston; and Cancer Center (A.T.C.), Massachusetts General Hospital, Boston, MA
| | - Joanne Ryan
- From the Clinical and Translational Epidemiology Unit (Y.W., A.T.C.), and Division of Gastroenterology (Y.W., A.T.C.), Massachusetts General Hospital and Harvard Medical School, Boston; School of Public Health and Preventive Medicine (Z.Z., J.C.B., R.L.W., S.G.O., R.W., J.R.), Monash University, Melbourne; Menzies Institute for Medical Research (Z.Z.), University of Tasmania, Australia; Department of Pharmacy Practice and Science (E.J.E.. M.E.E.), College of Pharmacy, and Department of Family Medicine (M.E.E.), Carver College of Medicine, University of Iowa, Iowa City; Department of Immunology and Infectious Diseases (A.T.C.), Harvard T.H. Chan School of Public Health, Boston; and Cancer Center (A.T.C.), Massachusetts General Hospital, Boston, MA
| | - Michael E Ernst
- From the Clinical and Translational Epidemiology Unit (Y.W., A.T.C.), and Division of Gastroenterology (Y.W., A.T.C.), Massachusetts General Hospital and Harvard Medical School, Boston; School of Public Health and Preventive Medicine (Z.Z., J.C.B., R.L.W., S.G.O., R.W., J.R.), Monash University, Melbourne; Menzies Institute for Medical Research (Z.Z.), University of Tasmania, Australia; Department of Pharmacy Practice and Science (E.J.E.. M.E.E.), College of Pharmacy, and Department of Family Medicine (M.E.E.), Carver College of Medicine, University of Iowa, Iowa City; Department of Immunology and Infectious Diseases (A.T.C.), Harvard T.H. Chan School of Public Health, Boston; and Cancer Center (A.T.C.), Massachusetts General Hospital, Boston, MA
| | - Andrew T Chan
- From the Clinical and Translational Epidemiology Unit (Y.W., A.T.C.), and Division of Gastroenterology (Y.W., A.T.C.), Massachusetts General Hospital and Harvard Medical School, Boston; School of Public Health and Preventive Medicine (Z.Z., J.C.B., R.L.W., S.G.O., R.W., J.R.), Monash University, Melbourne; Menzies Institute for Medical Research (Z.Z.), University of Tasmania, Australia; Department of Pharmacy Practice and Science (E.J.E.. M.E.E.), College of Pharmacy, and Department of Family Medicine (M.E.E.), Carver College of Medicine, University of Iowa, Iowa City; Department of Immunology and Infectious Diseases (A.T.C.), Harvard T.H. Chan School of Public Health, Boston; and Cancer Center (A.T.C.), Massachusetts General Hospital, Boston, MA
| |
Collapse
|
28
|
Koppula S, Wankhede N, Kyada A, Ballal S, Arya R, Singh AK, Gulati M, Sute A, Sarode S, Polshettiwar S, Marde V, Taksande B, Upaganlawar A, Fareed M, Umekar M, Kopalli SR, Kale M. The gut-brain axis: Unveiling the impact of xenobiotics on neurological health and disorders. Prog Neuropsychopharmacol Biol Psychiatry 2025; 136:111237. [PMID: 39732317 DOI: 10.1016/j.pnpbp.2024.111237] [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: 05/25/2024] [Revised: 11/12/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024]
Abstract
The Gut-Brain Axis (GBA) is a crucial link between the gut microbiota and the central nervous system. Xenobiotics, originating from diverse sources, play a significant role in shaping this interaction. This review examines how these compounds influence neurotransmitter dynamics within the GBA. Environmental pollutants can disrupt microbial populations, impacting neurotransmitter synthesis-especially serotonin, gamma-aminobutyric acid (GABA), and dopamine pathways. Such disruptions affect mood regulation, cognition, and overall neurological function. Xenobiotics also contribute to the pathophysiology of neurological disorders, with changes in serotonin levels linked to mood disorders and imbalances in GABA and dopamine associated with anxiety, stress, and reward pathway disorders. These alterations extend beyond the GBA, leading to complications in neurological health, including increased risk of neurodegenerative diseases due to neuroinflammation triggered by neurotransmitter imbalances. This review provides a comprehensive overview of how xenobiotics influence the GBA and their implications for neurological well-being.
Collapse
Affiliation(s)
- Sushruta Koppula
- College of Biomedical and Health Sciences, Konkuk University, Chungju-Si, Chungcheongbuk Do 27478, Republic of Korea
| | - Nitu Wankhede
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Ashishkumar Kyada
- Marwadi University Research Center, Department of Pharmaceutical Sciences, Faculty of Health Sciences, Marwadi University, Rajkot, -360003, Gujarat, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Renu Arya
- Department of Pharmacy, Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307, Punjab, India
| | | | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 1444411, India; ARCCIM, Faculty of Health, University of Technology Sydney, Ultimo, NSW 20227, Australia
| | - Astha Sute
- National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Sanskruti Sarode
- National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Shruti Polshettiwar
- National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Vaibhav Marde
- Indian Institute of Technology (IIT), Hyderabad, Telangana 502284, India
| | - Brijesh Taksande
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Aman Upaganlawar
- SNJB's Shriman Sureshdada Jain College of Pharmacy, Neminagar, Chandwad, Nashik, Maharashtra, India
| | - Mohammad Fareed
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Riyadh 11597, Saudi Arabia
| | - Milind Umekar
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Spandana Rajendra Kopalli
- Department of Bioscience and Biotechnology, Sejong University, Gwangjin-gu, Seoul 05006, Republic of Korea
| | - Mayur Kale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India.
| |
Collapse
|
29
|
Prajapati SK, Wang S, Mishra SP, Jain S, Yadav H. Protection of Alzheimer's disease progression by a human-origin probiotics cocktail. Sci Rep 2025; 15:1589. [PMID: 39794404 PMCID: PMC11724051 DOI: 10.1038/s41598-024-84780-8] [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: 01/27/2024] [Accepted: 12/26/2024] [Indexed: 01/13/2025] Open
Abstract
Microbiome abnormalities (dysbiosis) significantly contribute to the progression of Alzheimer's disease (AD). However, the therapeutic efficacy of microbiome modulators in protecting against these ailments remains poorly studied. Herein, we tested a cocktail of unique probiotics, including 5 Lactobacillus and 5 Enterococcus strains isolated from infant gut with proven microbiome modulating capabilities. We aimed to determine the probiotics cocktail's efficacy in ameliorating AD pathology in a humanized AD mouse model of APP/PS1 strains. Remarkably, feeding mice with 1 × 1011 CFU per day in drinking water for 16 weeks significantly reduced cognitive decline (measured by the Morris Water Maze test) and AD pathology markers, such as Aβ aggregation, microglia activation, neuroinflammation, and preserved blood-brain barrier (BBB) tight junctions. The beneficial effects were linked to a reduced inflammatory microbiome, leading to decreased gut permeability and inflammation in both systemic circulation and the brain. Although both male and female mice showed overall improvements in cognition and biological markers, females did not exhibit improvements in specific markers related to inflammation and barrier permeability, suggesting that the underlying mechanisms may differ depending on sex. In conclusion, our results suggest that this unique probiotics cocktail could serve as a prophylactic agent to reduce the progression of cognitive decline and AD pathology. This is achieved by beneficially modulating the microbiome, improving intestinal tight junction proteins, reducing permeability in both gut and BBB, and decreasing inflammation in the gut, blood circulation, and brain, ultimately mitigating AD pathology and cognitive decline.
Collapse
Affiliation(s)
- Santosh Kumar Prajapati
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA
- Department of Neurosurgery and Brain Repair, Center of Excellence in Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Shaohua Wang
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA
- Department of Neurosurgery and Brain Repair, Center of Excellence in Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Department of Biomedical Sciences, Infectious and Tropical Disease Institute, Ohio University Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Sidharth P Mishra
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA
- Department of Neurosurgery and Brain Repair, Center of Excellence in Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Shalini Jain
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA
- Department of Neurosurgery and Brain Repair, Center of Excellence in Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Hariom Yadav
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA.
- Department of Neurosurgery and Brain Repair, Center of Excellence in Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA.
- Department of Internal Medicine-Digestive Diseases and Nutrition, University of South Florida Morsani College of Medicine, Tampa, FL, USA.
| |
Collapse
|
30
|
Lin H, Ma Z, Li J, Zhu H, Huang X, Chen H, Tu L, Lian Y, Su Y. Community characteristics and relationship between gut microbiota and intratumoral microbiota in hepatocellular carcinoma. Front Immunol 2025; 15:1500863. [PMID: 39867901 PMCID: PMC11757874 DOI: 10.3389/fimmu.2024.1500863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 12/18/2024] [Indexed: 01/28/2025] Open
Abstract
Background The combination of local therapy with lenvatinib and programmed cell death protein-1 (PD-1) inhibitors represents an emerging treatment paradigm for unresectable hepatocellular carcinoma (uHCC). Our study sought to investigate the interrelationship between gut microbiota and intratumoral microbiota in the context of triple therapy, with a view to identifying potential biological markers. Methods The gut microbial community profiles of patients with primary untreated hepatocellular carcinoma (HCC) and those treated with local therapy combined with lenvatinib and PD-1 inhibitors were analyzed by 16S rRNA gene amplicon sequencing. Additionally, microbial community profiles of tumor tissues of patients with HCC and normal liver tissues were analyzed. Results In our investigation, we observed that patients with HCC who received triple therapy exhibited a notable enhancement in the abundance of Actinobacteriota and a considerable decrease in Escherichia Shigella. Patients who received hepatic artery infusion chemotherapy (HAIC) in combination with levatinib and PD-1 inhibitors exhibited significantly elevated levels of Faecalibacterium prausnitzii and Bacteroides stercoris in comparison to those who received transarterial chemoembolization (TACE) in combination with levatinib and PD-1 inhibitors. Furthermore, a notable decline in microbial diversity was observed within HCC tumors in comparison to normal liver tissues. The gut and intratumoral microbiota in HCC patients exhibited a high degree of similarity to the microbes present at the phylum level. Conclusions Gut microbiota is connected to triple therapy with local therapy combined with lenvatinib and PD-1 inhibitors for HCC. These discoveries underscore the potential of utilizing gut microbiota and intratumoral microbiota as biomarkers, as well as the possibility of triple therapy in the management of HCC.
Collapse
Affiliation(s)
- Huangpeng Lin
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, China
| | - Zexian Ma
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, China
| | - Jin Li
- Department of Health Management Center, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Heping Zhu
- Department of Hepatobiliary and Pancreatic Surgery, Xiamen Traditional Chinese Medicine Hospital, Xiamen, China
| | - Xuefeng Huang
- Department of Hepatobiliary and Pancreatic Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Huimin Chen
- Department of Hepatobiliary and Pancreatic Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Liang Tu
- Department of Hepatobiliary and Pancreatic Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yifan Lian
- Department of Gastroenterology, The National Key Clinical Specialty, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yongjie Su
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, China
| |
Collapse
|
31
|
Li X, Ding Q, Wan X, Wu Q, Ye S, Lou Y. Fecal microbiota transplantation attenuates Alzheimer's disease symptoms in APP/PS1 transgenic mice via inhibition of the TLR4-MyD88-NF-κB signaling pathway-mediated inflammation. BEHAVIORAL AND BRAIN FUNCTIONS : BBF 2025; 21:2. [PMID: 39780269 PMCID: PMC11715513 DOI: 10.1186/s12993-024-00265-8] [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: 08/15/2024] [Accepted: 12/30/2024] [Indexed: 01/11/2025]
Abstract
Alzheimer's disease (AD) is a prevalent and progressive neurodegenerative disorder that is the leading cause of dementia. The underlying mechanisms of AD have not yet been completely explored. Neuroinflammation, an inflammatory response mediated by certain mediators, has been exhibited to play a crucial role in the pathogenesis of AD. Additionally, disruption of the gut microbiota has been found to be associated with AD, and fecal microbiota transplantation (FMT) has emerged as a potential therapeutic approach. However, the precise mechanism of FMT in the treatment of AD remains elusive. In this study, FMT was performed by transplanting fecal microbiota from healthy wild-type mice into APP/PS1 mice (APPswe, PSEN1dE9) to assess the effectiveness of FMT in mitigating AD-associated inflammation and to reveal its precise mechanism of action. The results demonstrated that FMT treatment improved cognitive function and reduced the expression levels of inflammatory factors by regulating the TLR4/MyD88/NF-κB signaling pathway in mice, which was accompanied by the restoration of gut microbial dysbiosis. These findings suggest that FMT has the potential to ameliorate AD symptoms and delay the disease progression in APP/PS1 mice.
Collapse
Affiliation(s)
- Xiang Li
- Wenzhou Key Laboratory of Sanitary Microbiology; School of Laboratory Medicine and Life Sciences; Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
- Colorectal Cancer Research Center, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
- One Health Research Institute, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
- Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
| | - Qingyong Ding
- Wenzhou Key Laboratory of Sanitary Microbiology; School of Laboratory Medicine and Life Sciences; Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- Colorectal Cancer Research Center, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- One Health Research Institute, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- Testing Center of the First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui Province, China
| | - Xinxin Wan
- Wenzhou Key Laboratory of Sanitary Microbiology; School of Laboratory Medicine and Life Sciences; Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- Colorectal Cancer Research Center, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- One Health Research Institute, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Qilong Wu
- Wenzhou Key Laboratory of Sanitary Microbiology; School of Laboratory Medicine and Life Sciences; Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- Colorectal Cancer Research Center, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- One Health Research Institute, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Shiqing Ye
- Wenzhou Key Laboratory of Sanitary Microbiology; School of Laboratory Medicine and Life Sciences; Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- Colorectal Cancer Research Center, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- One Health Research Institute, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yongliang Lou
- Wenzhou Key Laboratory of Sanitary Microbiology; School of Laboratory Medicine and Life Sciences; Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
- Colorectal Cancer Research Center, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
- One Health Research Institute, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
- Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
| |
Collapse
|
32
|
Kaur S, Kumari D, Dandekar MP. Importance of Gut Microbiota Dysbiosis and Circadian Disruption-Associated Biomarkers in Emergence of Alzheimer's Disease. Mol Neurobiol 2025:10.1007/s12035-024-04685-5. [PMID: 39775480 DOI: 10.1007/s12035-024-04685-5] [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: 07/19/2024] [Accepted: 12/26/2024] [Indexed: 01/11/2025]
Abstract
Alzheimer's disease (AD) is a major devastating neurodegenerative disorder afflicting majorly the geriatric population. Emerging studies augur the connection of gut dysbiosis and circadian disruption with the early onset of AD. Gut dysbiosis is characterized by dysregulated gut microbiota signature and compromised intestinal integrity, which provokes the translocation of bacterial metabolites into the systemic circulation. Noteworthy, gut-derived metabolites like calprotectin, trimethylamine-N-oxide, kynurenine, isoamylamine, and short-chain fatty acids play a key role in AD pathogenesis. Circadian dysregulation also corresponds with the exacerbated AD pathogenesis by accumulating Aβ and tau proteins. Moreover, circadian dysregulation is one of the causative factors for gut dysbiosis. This review discusses the complex interplay between the microbiota-gut-brain axis, circadian rhythmicity, and the emergence of AD. We reviewed preclinical and clinical studies on AD describing potential biomarkers of gut dysbiosis and circadian dysregulation. The identification of new biomarkers associated with the microbiota-gut-brain axis and circadian rhythmicity may help in early diagnosis and development of targeted therapies for mitigating neurodegenerative AD.
Collapse
Affiliation(s)
- Simranjit Kaur
- Department of Biological Sciences (Pharmacology and Toxicology), National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India, 500037
| | - Deepali Kumari
- Department of Biological Sciences (Pharmacology and Toxicology), National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India, 500037
| | - Manoj P Dandekar
- Department of Biological Sciences (Pharmacology and Toxicology), National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India, 500037.
| |
Collapse
|
33
|
Lana D, Ugolini F, Iovino L, Attorre S, Giovannini MG. Astrocytes phenomics as new druggable targets in healthy aging and Alzheimer's disease progression. Front Cell Neurosci 2025; 18:1512985. [PMID: 39835288 PMCID: PMC11743640 DOI: 10.3389/fncel.2024.1512985] [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: 10/17/2024] [Accepted: 12/13/2024] [Indexed: 01/22/2025] Open
Abstract
For over a century after their discovery astrocytes were regarded merely as cells located among other brain cells to hold and give support to neurons. Astrocytes activation, "astrocytosis" or A1 functional state, was considered a detrimental mechanism against neuronal survival. Recently, the scientific view on astrocytes has changed. Accumulating evidence indicate that astrocytes are not homogeneous, but rather encompass heterogeneous subpopulations of cells that differ from each other in terms of transcriptomics, molecular signature, function and response in physiological and pathological conditions. In this review, we report and discuss the recent literature on the phenomic differences of astrocytes in health and their modifications in disease conditions, focusing mainly on the hippocampus, a region involved in learning and memory encoding, in the age-related memory impairments, and in Alzheimer's disease (AD) dementia. The morphological and functional heterogeneity of astrocytes in different brain regions may be related to their different housekeeping functions. Astrocytes that express diverse transcriptomics and phenomics are present in strictly correlated brain regions and they are likely responsible for interactions essential for the formation of the specialized neural circuits that drive complex behaviors. In the contiguous and interconnected hippocampal areas CA1 and CA3, astrocytes show different, finely regulated, and region-specific heterogeneity. Heterogeneous astrocytes have specific activities in the healthy brain, and respond differently to physiological or pathological stimuli, such as inflammaging present in normal brain aging or beta-amyloid-dependent neuroinflammation typical of AD. To become reactive, astrocytes undergo transcriptional, functional, and morphological changes that transform them into cells with different properties and functions. Alterations of astrocytes affect the neurovascular unit, the blood-brain barrier and reverberate to other brain cell populations, favoring or dysregulating their activities. It will be of great interest to understand whether the differential phenomics of astrocytes in health and disease can explain the diverse vulnerability of the hippocampal areas to aging or to different damaging insults, in order to find new astrocyte-targeted therapies that might prevent or treat neurodegenerative disorders.
Collapse
Affiliation(s)
- Daniele Lana
- Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy
| | - Filippo Ugolini
- Section of Pathological Anatomy, Department of Health Sciences, University of Florence, Florence, Italy
| | - Ludovica Iovino
- Institute of Neuroscience, National Research Council (CNR), Pisa, Italy
| | - Selene Attorre
- Section of Pathological Anatomy, Department of Health Sciences, University of Florence, Florence, Italy
| | - Maria Grazia Giovannini
- Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy
| |
Collapse
|
34
|
Onisiforou A, Charalambous EG, Zanos P. Shattering the Amyloid Illusion: The Microbial Enigma of Alzheimer's Disease Pathogenesis-From Gut Microbiota and Viruses to Brain Biofilms. Microorganisms 2025; 13:90. [PMID: 39858858 PMCID: PMC11767882 DOI: 10.3390/microorganisms13010090] [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: 12/11/2024] [Revised: 12/18/2024] [Accepted: 01/03/2025] [Indexed: 01/27/2025] Open
Abstract
For decades, Alzheimer's Disease (AD) research has focused on the amyloid cascade hypothesis, which identifies amyloid-beta (Aβ) as the primary driver of the disease. However, the consistent failure of Aβ-targeted therapies to demonstrate efficacy, coupled with significant safety concerns, underscores the need to rethink our approach to AD treatment. Emerging evidence points to microbial infections as environmental factors in AD pathoetiology. Although a definitive causal link remains unestablished, the collective evidence is compelling. This review explores unconventional perspectives and emerging paradigms regarding microbial involvement in AD pathogenesis, emphasizing the gut-brain axis, brain biofilms, the oral microbiome, and viral infections. Transgenic mouse models show that gut microbiota dysregulation precedes brain Aβ accumulation, emphasizing gut-brain signaling pathways. Viral infections like Herpes Simplex Virus Type 1 (HSV-1) and Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) may lead to AD by modulating host processes like the immune system. Aβ peptide's antimicrobial function as a response to microbial infection might inadvertently promote AD. We discuss potential microbiome-based therapies as promising strategies for managing and potentially preventing AD progression. Fecal microbiota transplantation (FMT) restores gut microbial balance, reduces Aβ accumulation, and improves cognition in preclinical models. Probiotics and prebiotics reduce neuroinflammation and Aβ plaques, while antiviral therapies targeting HSV-1 and vaccines like the shingles vaccine show potential to mitigate AD pathology. Developing effective treatments requires standardized methods to identify and measure microbial infections in AD patients, enabling personalized therapies that address individual microbial contributions to AD pathogenesis. Further research is needed to clarify the interactions between microbes and Aβ, explore bacterial and viral interplay, and understand their broader effects on host processes to translate these insights into clinical interventions.
Collapse
Affiliation(s)
- Anna Onisiforou
- Translational Neuropharmacology Laboratory, Department of Psychology, University of Cyprus, 75 Kallipoleos Avenue, 1678 Nicosia, Cyprus;
- Center of Applied Neuroscience, 75 Kallipoleos Avenue, 1678 Nicosia, Cyprus
| | - Eleftheria G. Charalambous
- Translational Neuropharmacology Laboratory, Department of Psychology, University of Cyprus, 75 Kallipoleos Avenue, 1678 Nicosia, Cyprus;
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, 1–2, Ellernholzstr., 17489 Greifswald, Germany
| | - Panos Zanos
- Translational Neuropharmacology Laboratory, Department of Psychology, University of Cyprus, 75 Kallipoleos Avenue, 1678 Nicosia, Cyprus;
- Center of Applied Neuroscience, 75 Kallipoleos Avenue, 1678 Nicosia, Cyprus
| |
Collapse
|
35
|
Malan-Müller S, Martín-Hernández D, Caso JR, Matthijnssens J, Rodríguez-Urrutia A, Lowry CA, Leza JC. Metagenomic symphony of the intestinal ecosystem: How the composition affects the mind. Brain Behav Immun 2025; 123:510-523. [PMID: 39368785 DOI: 10.1016/j.bbi.2024.09.033] [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: 06/14/2024] [Revised: 09/04/2024] [Accepted: 09/27/2024] [Indexed: 10/07/2024] Open
Abstract
Mental health disorders and neurodegenerative diseases place a heavy burden on patients and societies, and, although great strides have been made to understand the pathophysiology of these conditions, advancement in drug development is lagging. The importance of gastrointestinal health in maintaining overall health and preventing disease is not a new concept. Hundreds of years ago, healers from various cultures and civilizations recognized the crucial role of the gut in sustaining health. More than a century ago, scientists began exploring the restorative effects of probiotics, marking the early recognition of the importance of gut microbes. The omics era brought more enlightenment and enabled researchers to identify the complexity of the microbial ecosystems we harbour, encompassing bacteria, eukaryotes (including fungi), archaea, viruses, and other microorganisms. The extensive genetic capacity of the microbiota is dynamic and influenced by the environment. The microbiota therefore serves as a significant entity within us, with evolutionarily preserved functions in host metabolism, immunity, development, and behavior. The significant role of the bacterial gut microbiome in mental health and neurodegenerative disorders has been realized and described within the framework of the microbiota-gut-brain axis. However, the bacterial members do not function unaccompanied, but rather in concert, and there is a substantial knowledge gap regarding the involvement of non-bacterial microbiome members in these disorders. In this review, we will explore the current literature that implicates a role for the entire metagenomic ensemble, and how their complex interkingdom relationships could influence CNS functioning in mental health disorders and neurodegenerative diseases.
Collapse
Affiliation(s)
- Stefanie Malan-Müller
- Department of Pharmacology and Toxicology, Faculty of Medicine, University Complutense of Madrid (UCM), Research Institute of Hospital 12 de Octubre (Imas12), Instituto Universitario de Investigación Neuroquímica (IUIN-UCM), Madrid, Spain; Biomedical Research Network Centre in Mental Health, Institute of Health Carlos III (CIBERSAM, ISCIII), Madrid, Spain.
| | - David Martín-Hernández
- Department of Pharmacology and Toxicology, Faculty of Medicine, University Complutense of Madrid (UCM), Research Institute of Hospital 12 de Octubre (Imas12), Instituto Universitario de Investigación Neuroquímica (IUIN-UCM), Madrid, Spain; Biomedical Research Network Centre in Mental Health, Institute of Health Carlos III (CIBERSAM, ISCIII), Madrid, Spain
| | - Javier R Caso
- Department of Pharmacology and Toxicology, Faculty of Medicine, University Complutense of Madrid (UCM), Research Institute of Hospital 12 de Octubre (Imas12), Instituto Universitario de Investigación Neuroquímica (IUIN-UCM), Madrid, Spain; Biomedical Research Network Centre in Mental Health, Institute of Health Carlos III (CIBERSAM, ISCIII), Madrid, Spain
| | - Jelle Matthijnssens
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute, Division of Clinical and Epidemiological Virology, Laboratory of Viral Metagenomics, Leuven, Belgium
| | - Amanda Rodríguez-Urrutia
- Biomedical Research Network Centre in Mental Health, Institute of Health Carlos III (CIBERSAM, ISCIII), Madrid, Spain; Department of Mental Health, Hospital Universitari Vall d'Hebron, Barcelona, Catalonia, Spain; Group of Psychiatry, Mental Health and Addictions, Vall d'Hebron Research Institute (VHIR), Barcelona, Catalonia, Spain; Department of Psychiatry and Forensic Medicine, Universitat Autònoma de Barcelona, Barcelona, Catalonia, Spain
| | - Christopher A Lowry
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Juan C Leza
- Department of Pharmacology and Toxicology, Faculty of Medicine, University Complutense of Madrid (UCM), Research Institute of Hospital 12 de Octubre (Imas12), Instituto Universitario de Investigación Neuroquímica (IUIN-UCM), Madrid, Spain; Biomedical Research Network Centre in Mental Health, Institute of Health Carlos III (CIBERSAM, ISCIII), Madrid, Spain
| |
Collapse
|
36
|
Leisgang Osse AM, Kinney JW, Cummings JL. The Common Alzheimer's Disease Research Ontology (CADRO) for biomarker categorization. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2025; 11:e70050. [PMID: 39935614 PMCID: PMC11812129 DOI: 10.1002/trc2.70050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/03/2025] [Accepted: 01/06/2025] [Indexed: 02/13/2025]
Abstract
Biomarkers are vital to Alzheimer's disease (AD) drug development and clinical trials, and will have an increasing role in clinical care. In this narrative review, we demonstrate the use of the National Institutes on Aging/Alzheimer's Association (NIA/AA) Common Alzheimer's Disease Research Ontology (CADRO) system for the categorization of biomarkers based on the primary mechanism on which they report. We show that biomarkers are available (in various levels of validation) for all CADRO processes. Application of the CADRO system demonstrates gaps in the field where novel biomarkers are needed for specific aspects of the disease, and assays to detect and measure biological changes, in individuals with symptomatic or preclinical AD. We demonstrate the CADRO system as a means of categorizing established and candidate AD biomarkers, showing the feasibility and practicality of the system. CADRO can assist with biomarker selection for AD clinical trials and drug development, and may eventually be applied to implementing biomarkers in patient care. Highlights The Common Alzheimer's Disease Research Ontology (CADRO) system can be used to categorize biomarkers for drug development.We demonstrate the use of CADRO with Alzheimer's disease (AD) biomarkers.We identified AD biomarkers in each of the CADRO categories.CADRO can be incorporated into current AD drug development and clinical trial systems.
Collapse
Affiliation(s)
- Amanda M. Leisgang Osse
- Department of Brain Health, Kirk Kerkorian School of MedicineUniversity of Nevada Las Vegas (UNLV)Las VegasNevadaUSA
| | - Jefferson W. Kinney
- Department of Brain Health, Kirk Kerkorian School of MedicineUniversity of Nevada Las Vegas (UNLV)Las VegasNevadaUSA
| | - Jeffrey L. Cummings
- Department of Brain Health, Kirk Kerkorian School of MedicineUniversity of Nevada Las Vegas (UNLV)Las VegasNevadaUSA
| |
Collapse
|
37
|
Timmis K, Karahan ZC, Ramos JL, Koren O, Pérez‐Cobas AE, Steward K, de Lorenzo V, Caselli E, Douglas M, Schwab C, Rivero V, Giraldo R, Garmendia J, Turner RJ, Perlmutter J, Borrero de Acuña JM, Nikel PI, Bonnet J, Sessitsch A, Timmis JK, Pruzzo C, Prieto MA, Isazadeh S, Huang WE, Clarke G, Ercolini D, Häggblom M. Microbes Saving Lives and Reducing Suffering. Microb Biotechnol 2025; 18:e70068. [PMID: 39844583 PMCID: PMC11754571 DOI: 10.1111/1751-7915.70068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 11/25/2024] [Indexed: 01/24/2025] Open
Affiliation(s)
- Kenneth Timmis
- Institute of MicrobiologyTechnical University BraunschweigBraunschweigGermany
| | - Zeynep Ceren Karahan
- Department of Medical Microbiology and Ibn‐i Sina Hospital Central Microbiology LaboratoryAnkara University School of MedicineAnkaraTurkey
| | - Juan Luis Ramos
- Consejo Superior de Investigaciones Científicas, Estación Experimental del ZaidínGranadaSpain
| | - Omry Koren
- Azrieli Faculty of MedicineBar‐Ilan UniversitySafedIsrael
| | - Ana Elena Pérez‐Cobas
- Department of Microbiology, Ramón y Cajal Institute for Health Research (IRYCIS)Ramón y Cajal University HospitalMadridSpain
- CIBER in Infectious Diseases (CIBERINFEC)MadridSpain
| | | | - Victor de Lorenzo
- Department of Systems BiologyNational Centre of Biotechnology CSICMadridSpain
| | - Elisabetta Caselli
- Section of Microbiology, Department of Environmental and Prevention SciencesUniversity of FerraraFerraraItaly
| | - Margaret Douglas
- Usher InstituteUniversity of Edinburgh Medical School, and Public Health ScotlandEdinburghUK
| | - Clarissa Schwab
- Department of Biological and Chemical EngineeringAarhus UniversityAarhusDenmark
| | - Virginia Rivero
- Polymer Biotechnology Lab, Biological Research Center Margarita SalasSpanish National Research Council (CIB‐CSIC)MadridSpain
| | - Rafael Giraldo
- Department of Microbial BiotechnologyNational Centre for Biotechnology (CNB‐CSIC)MadridSpain
| | - Junkal Garmendia
- Instituto de AgrobiotecnologíaConsejo Superior de Investigaciones Científicas (IdAB‐CSIC)‐Gobierno de Navarra, MutilvaMadridSpain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES)MadridSpain
| | - Raymond J. Turner
- Department of Biological SciencesUniversity of CalgaryCalgaryAlbertaCanada
| | | | | | - Pablo Ivan Nikel
- The Novo Nordisk Foundation Center for BiosustainabilityTechnical University of DenmarkLyngbyDenmark
| | - Jerome Bonnet
- Centre de Biochimie Structurale, INSERM/CNRSUniversity of MontpellierMontpellierFrance
| | - Angela Sessitsch
- Bioresources UnitAIT Austrian Institute of TechnologyViennaAustria
| | - James K. Timmis
- Department of Political ScienceUniversity of FreiburgFreiburgGermany
- Athena Institute for Research on Innovation and Communication in Health and Life SciencesVrije UniversiteitAmsterdamThe Netherlands
| | - Carla Pruzzo
- Department of Earth, Environmental and Life Sciences (DISTAV)University of GenoaGenovaItaly
| | - M. Auxiliadora Prieto
- Polymer Biotechnology Lab, Biological Research Center Margarita SalasSpanish National Research Council (CIB‐CSIC)MadridSpain
| | - Siavash Isazadeh
- Corporate Technical & PerformanceVeolia North AmericaParamusNew JerseyUSA
| | - Wei E. Huang
- Department of Engineering ScienceUniversity of OxfordOxfordUK
| | - Gerard Clarke
- APC Microbiome IrelandUniversity College CorkCorkIreland
- Department of Psychiatry & Neurobehavioral SciencesUniversity College CorkCorkIreland
| | - Danilo Ercolini
- Department of Agricultural SciencesUniversity of Naples Federico IINaplesItaly
| | - Max Häggblom
- Department of Biochemistry and Microbiology, RutgersThe State University of New JerseyNew BrunswickNew JerseyUSA
| |
Collapse
|
38
|
Dhanawat M, Malik G, Wilson K, Gupta S, Gupta N, Sardana S. The Gut Microbiota-Brain Axis: A New Frontier in Alzheimer's Disease Pathology. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2025; 24:7-20. [PMID: 38967078 DOI: 10.2174/0118715273302508240613114103] [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: 01/31/2024] [Revised: 05/12/2024] [Accepted: 05/17/2024] [Indexed: 07/06/2024]
Abstract
Dr. Aloysius Alzheimer, a German neuropathologist and psychiatrist, recognized the primary instance of Alzheimer's disease (AD) for a millennium, and this ailment, along with its related dementias, remains a severe overall community issue related to health. Nearly fifty million individuals worldwide suffer from dementia, with Alzheimer's illness contributing to between 60 and 70% of the instances, estimated through the World Health Organization. In addition, 82 million individuals are anticipated to be affected by the global dementia epidemic by 2030 and 152 million by 2050. Furthermore, age, environmental circumstances, and inherited variables all increase the likelihood of acquiring neurodegenerative illnesses. Most recent pharmacological treatments are found in original hypotheses of disease, which include cholinergic (drugs that show affective cholinergic system availability) as well as amyloid-accumulation (a single drug is an antagonist receptor of Nmethyl D-aspartate). In 2020, the FDA provided approval on anti-amyloid drugs. According to mounting scientific data, this gut microbiota affects healthy physiological homeostasis and has a role in the etiology of conditions that range between obesity and neurodegenerative disorders like Alzheimer's. The microbiota-gut-brain axis might facilitate interconnection among gut microbes as well as the central nervous system (CNS). Interaction among the microbiota-gut system as well as the brain occurs through the "two-way" microbiota-gut-brain axis. Along this axis, the stomach as well as the brain develop physiologically and take on their final forms. This contact is constant and is mediated by numerous microbiota-derived products. The gut microbiota, for instance, can act as non-genetic markers to set a threshold for maintaining homeostasis or getting ill. The scientific community has conducted research and found that bowel dysbiosis and gastrointestinal tract dysregulation frequently occur in Alzheimer's disease (AD) patients. In this review, the effects of the microbiota- gut-brain axis on AD pathogenesis will be discussed.
Collapse
Affiliation(s)
- Meenakshi Dhanawat
- Amity Institute of Pharmacy, Amity University Haryana, Gurugram 122413, India
| | - Garima Malik
- Department of Pharmaceutics, M.M College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana 133207, India
| | - Kashish Wilson
- Department of Pharmaceutics, M.M College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana 133207, India
| | - Sumeet Gupta
- Department of Pharmaceutics, M.M College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana 133207, India
| | - Nidhi Gupta
- Department of Pharmaceutics, M.M College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana 133207, India
| | - Satish Sardana
- Amity Institute of Pharmacy, Amity University Haryana, Gurugram 122413, India
| |
Collapse
|
39
|
Mohammadi G, Babaei F, Golpour F, Rashidi FS, Ghafghazi S, Dargahi L, Nassiri-Asl M. Saccharomyces boulardii Ameliorates LPS-Induced Amyloidogenesis in Rats. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10445-7. [PMID: 39739163 DOI: 10.1007/s12602-024-10445-7] [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] [Accepted: 12/19/2024] [Indexed: 01/02/2025]
Abstract
Gut brain axis can affect the incidence of Alzheimer's disease (AD). Probiotics restore the homeostasis of gut dysbiosis and prevent AD. Here, we evaluated the impact of Saccharomyces boulardii on rats with lipopolysaccharide (LPS)-induced amyloidogenesis. Rats were classified into four groups: (1) Control (saline), (2) LPS 250 µg/kg (saline + LPS), (3) S. boulardii (1010 CFU/mL/rat), and (4) S. boulardii (1010 CFU/mL/rat) + LPS (250 μg/kg). The passive behavioral test, Western blotting, and immunohistochemistry were done using the animal hippocampi. Step-through latency (STL) indicated that the LPS-treated group had decreased memory retrieval compared to the control group. The LPS group had increased hippocampal levels of amyloid-β peptide, amyloid-β precursor protein (APP), and β-secretase (BACE). Administration of the S. boulardii before LPS prolonged STL which has been shortened in the LPS group (P < 0.05). In the LPS + S group, S. boulardii reduced the levels of APP significantly compared to the LPS group (P < 0.01). S. boulardii mitigated Aβ buildup and memory dysfunction caused by LPS through modulating the APP, BACE1, and Aβ pathways. Future studies are required to explain the neuroprotective effects of S. boulardii, since it could be a novel therapy or prevention strategy for AD.
Collapse
Affiliation(s)
- Ghazaleh Mohammadi
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
- Department of Molecular Medicine, School of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Fatemeh Babaei
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, P.O. Box, Tehran, 19839-63113, Iran
| | - Faezeh Golpour
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Sadat Rashidi
- Neuroscience Research Center, Institute of Neuroscience and Cognition, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shiva Ghafghazi
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, P.O. Box, Tehran, 19839-63113, Iran
| | - Leila Dargahi
- Neuroscience Research Center, Institute of Neuroscience and Cognition, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Marjan Nassiri-Asl
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, P.O. Box, Tehran, 19839-63113, Iran.
- Neuroscience Research Center, Institute of Neuroscience and Cognition, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
40
|
Zhuang X, Lin J, Song Y, Ban R, Zhao X, Xia Z, Wang Z, Zhang G. The Interplay Between Accumulation of Amyloid-Beta and Tau Proteins, PANoptosis, and Inflammation in Alzheimer's Disease. Neuromolecular Med 2024; 27:2. [PMID: 39751702 DOI: 10.1007/s12017-024-08815-z] [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: 07/15/2024] [Accepted: 11/01/2024] [Indexed: 01/04/2025]
Abstract
Alzheimer's disease (AD) is a common progressive neurodegenerative disorder, and the vast majority of cases occur in elderly patients. Recently, the accumulation of Aβ and tau proteins has drawn considerable attention in AD research. This review explores the multifaceted interactions between these proteins and their contribution to the pathological landscape of AD, encompassing synaptic dysfunction, neuroinflammation, and PANoptosis. PANoptosis is a collective term for programmed cell death (PCD) modalities that encompass elements of apoptosis, pyroptosis, and necroptosis. The accumulation of Aβ peptides and tau proteins, along with the immune response in brain cells, may trigger PANoptosis, thus advancing the progression of the disease. Recent advancements in molecular imaging and genetics have provided deeper insights into the interactions between Aβ peptides, tau proteins, and the immune response. The review also discusses the role of mitochondrial dysregulation in AD. The exploration of the interplay between neurodegeneration, immune responses, and cell death offers promising avenues for the development of innovative treatments.
Collapse
Affiliation(s)
- Xianbo Zhuang
- Department of Neurology, Liaocheng People's Hospital and Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, 252000, China
| | - Jie Lin
- School of Basic Medicine Sciences, Shandong University, Jinan, China
- Department of Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, China
| | - Yamin Song
- Department of Neurology, Liaocheng People's Hospital and Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, 252000, China
| | - Ru Ban
- Department of Neurology, Liaocheng People's Hospital and Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, 252000, China
| | - Xin Zhao
- Department of Neurology, Liaocheng People's Hospital and Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, 252000, China
| | - Zhangyong Xia
- Department of Neurology, Liaocheng People's Hospital and Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, 252000, China.
- Department of Neurology, Liaocheng People's Hospital, Shandong University, Jinan, 250012, China.
- Department of Neurology, the Second People's Hospital of Liaocheng, Liaocheng, China.
| | - Zheng Wang
- Department of Neurosurgery, Liaocheng Traditional Chinese Medicine Hospital, Liaocheng, 252000, China.
| | - Guifeng Zhang
- Department of Neurology, Liaocheng People's Hospital and Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, 252000, China.
| |
Collapse
|
41
|
Snyder JR, Ahmed M, Bhave S, Hotta R, Koppes RA, Goldstein AM, Koppes AN. Enteroendocrine Cells Sense Sucrose and Alter Enteric Neuron Excitability via Insulin Signaling. Adv Biol (Weinh) 2024:e2300566. [PMID: 39703141 DOI: 10.1002/adbi.202300566] [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: 07/19/2024] [Revised: 11/25/2024] [Indexed: 12/21/2024]
Abstract
Neurosensory circuits of the gastrointestinal tract sense microbial and nutrient changes in the gut; however, studying these circuits in vivo is hindered by invasive techniques and ethical concerns. Here, an in vitro model of enteroendocrine cells (EECs) and calcium reporting enteric neurons (ENs) is established and validated for functional signaling. Both mechanical and sucrose stimulation of co-cultures increased the percentage of neurons undergoing a calcium flux, indicating an action potential. Neuronal activation is blocked with either a piezo or insulin receptor blocker. At baseline, a flow only stimulus elicited 51.9% of neurons to activate in co-culture, which is decreased to 15.1% with a piezo blocker. Piezo blocked and sucrose stimulated EECs increased neuronal activation to 43.9%, and an insulin blocker reduced response to 12.4%. Since a cell line is used to model the EEC in the previous experiments, primary rat duodenal epithelium enriched for EECs are also stimulated and found to produced measurable insulin. This work shows the ability of EECs to produce insulin and for ENs to sense insulin. These results inspire further work on how insulin production outside the pancreas effects diabetes, insulin as a neurotransmitter, and exploration of additional nutritional and microbiotic stimuli on enteroendocrine-to-neuronal signaling.
Collapse
Affiliation(s)
- Jessica R Snyder
- Department of Bioengineering, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA
| | - Minhal Ahmed
- Harvard Medical School, 25 Shattuck St, Boston, MA, 02115, USA
| | - Sukhada Bhave
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, 185 Cambridge St, CPZN 6-215, Boston, MA, 02114, USA
| | - Ryo Hotta
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, 185 Cambridge St, CPZN 6-215, Boston, MA, 02114, USA
| | - Ryan A Koppes
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA
| | - Allan M Goldstein
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, 185 Cambridge St, CPZN 6-215, Boston, MA, 02114, USA
| | - Abigail N Koppes
- Department of Bioengineering, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA
- Department of Biology, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA
| |
Collapse
|
42
|
Castaño-Joaqui OG, Jiménez Ortega L, Cerero Lapiedra R, Domínguez Gordillo AÁ. Burning Mouth Syndrome Underlying Factors: A Roadmap From a Network Perspective. Oral Dis 2024. [PMID: 39673150 DOI: 10.1111/odi.15219] [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: 07/20/2024] [Revised: 11/25/2024] [Accepted: 11/29/2024] [Indexed: 12/16/2024]
Abstract
OBJECTIVE To investigate the relationship between biological, psychological, and social factors underlying Burning Mouth Syndrome (BMS). SUBJECTS AND METHODS A case (n = 40) and control (n = 42) study containing 80 variables was examined using two network models based on regularized partial correlations (n = 82). RESULTS The structure of the associative pathways with the BMS was revealed. Direct associations involved Gastrointestinal Alterations (0.23), Vitamin D Deficiency (0.29), Musculoskeletal Alterations (0.29), Symptom Severity Score 2 (SSS2) (0.22), Cortisol Variation (0.10), Interpersonal Sensitivity (0.04), Hostility (0.03). Global Severity Index, Symptom Severity Score 1, Psychoticism, Obsession-Compulsion, Depression, Anxiety, and Somatization were indirectly related. The SSS2 was the most influential on BMS accuracy. CONCLUSIONS Gastrointestinal alterations and vitamin D deficiency show a significant influence on BMS while cortisol mediates in multiple associative pathways between musculoskeletal alterations, gastrointestinal alterations, vitamin D deficiency, non-restorative sleep, fatigue, and cognitive problems. In addition to anxiety and depression, psychoticism, interpersonal sensitivity, and hostility stand out as psychological factors that seem to be related to a lack of vitamin D. None of the factors studied seem to have a relevant predictive potential for BMS, except for nonspecific symptoms of central sensitization.
Collapse
Affiliation(s)
- Oscar Gabriel Castaño-Joaqui
- Department of Conservative Dentistry and Bucofacial Prosthesis, Faculty of Odontology, Complutense University of Madrid, Madrid, Spain
| | - Laura Jiménez Ortega
- Department of Psychobiology and Behavioral Sciences Methods, Faculty of Odontology, Complutense University of Madrid, Madrid, Spain
- Center of Human Evolution and Behavior, UCM-ISCIII, Madrid, Spain
- Psychology and Orofacial Pain Working Group, Sociedad Española de Disfunción Craneomandibular y Dolor Orofacial (SEDCYDO), Madrid, Spain
| | - Rocío Cerero Lapiedra
- Department of Odontologic Clinic Specialty, Faculty of Odontology, Complutense University of Madrid, Madrid, Spain
| | | |
Collapse
|
43
|
Xu X, Wei S, Lin M, Chen F, Zhang X, Zhu Y. The relationship between acrylamide and neurodegenerative diseases: gut microbiota as a new intermediate cue. Crit Rev Food Sci Nutr 2024:1-13. [PMID: 39668759 DOI: 10.1080/10408398.2024.2440602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2024]
Abstract
Acrylamide (AA), a compound formed during the thermal processing of high-carbohydrate foods, has been implicated in the onset and progression of neurodegenerative diseases. An increasing number of reports support that gut microbiota plays a significant role in brain function and diseases, suggesting it may act as a mediator between AA exposure and the development of neurodegenerative diseases. Available studies have shown that AA intake affects the composition of the gut microbiota and the integrity of the intestinal barrier, both of which are often thought to be associated with the pathogenesis of neurodegenerative diseases, given the numerous evidences linking gut microbiota with the brain. Based on the current understanding, this paper discusses that AA induces the onset and progression of neurodegenerative diseases by disrupting the composition of the gut microbiota and the structure of the intestinal barrier. Furthermore, it explores the interaction between probiotics and AA exposure, as well as the potential for polysaccharides and polyphenols to improve the gut microenvironment, which provides novel perspectives on modulating the neurodegenerative diseases caused by AA exposure through diet.
Collapse
Affiliation(s)
- Xinrui Xu
- College of Food Science and Nutritional Engineering, National Engineering Research Centre for Fruits and Vegetables Processing, Key Laboratory of Storage and Processing of Fruits and Vegetables, Ministry of Agriculture, Engineering Research Centre for Fruits and Vegetables Processing, Ministry of Education, China Agricultural University, Beijing, P. R. China
| | - Siyu Wei
- College of Food Science and Nutritional Engineering, National Engineering Research Centre for Fruits and Vegetables Processing, Key Laboratory of Storage and Processing of Fruits and Vegetables, Ministry of Agriculture, Engineering Research Centre for Fruits and Vegetables Processing, Ministry of Education, China Agricultural University, Beijing, P. R. China
| | - Mengyi Lin
- College of Food Science and Nutritional Engineering, National Engineering Research Centre for Fruits and Vegetables Processing, Key Laboratory of Storage and Processing of Fruits and Vegetables, Ministry of Agriculture, Engineering Research Centre for Fruits and Vegetables Processing, Ministry of Education, China Agricultural University, Beijing, P. R. China
| | - Fang Chen
- College of Food Science and Nutritional Engineering, National Engineering Research Centre for Fruits and Vegetables Processing, Key Laboratory of Storage and Processing of Fruits and Vegetables, Ministry of Agriculture, Engineering Research Centre for Fruits and Vegetables Processing, Ministry of Education, China Agricultural University, Beijing, P. R. China
| | - Xin Zhang
- Department of Food Science and Engineering, Ningbo University, Ningbo, P. R. China
| | - Yuchen Zhu
- College of Food Science and Nutritional Engineering, National Engineering Research Centre for Fruits and Vegetables Processing, Key Laboratory of Storage and Processing of Fruits and Vegetables, Ministry of Agriculture, Engineering Research Centre for Fruits and Vegetables Processing, Ministry of Education, China Agricultural University, Beijing, P. R. China
| |
Collapse
|
44
|
Pasupalak JK, Rajput P, Gupta GL. Gut microbiota and Alzheimer's disease: Exploring natural product intervention and the Gut-Brain axis for therapeutic strategies. Eur J Pharmacol 2024; 984:177022. [PMID: 39362390 DOI: 10.1016/j.ejphar.2024.177022] [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: 07/11/2024] [Revised: 09/14/2024] [Accepted: 09/30/2024] [Indexed: 10/05/2024]
Abstract
Numerous studies conducted over the last ten years have shown a strong correlation between the gut microbiota and the onset and progression of Alzheimer's disease (AD). However, the exact underlying mechanism is still unknown. An ongoing communication mechanism linking the gut and the brain is highlighted by the term "microbiota-gut-brain axis," which was originally coined the "gut-brain axis." Key metabolic, endocrine, neurological, and immunological mechanisms are involved in the microbiota‒gut‒brain axis and are essential for preserving brain homeostasis. Thus, the main emphasis of this review is how the gut microbiota contributes to the development of AD and how various natural products intervene in this disease. The first part of the review provides an outline of various pathways and relationships between the brain and gut microbiota, and the second part provides various mechanisms involved in the gut microbiota and AD. Finally, this review provides knowledge about natural products and their effectiveness in treating gut microbiota-induced AD. AD may be treated in the future by altering the gut microbiota with a customized diet, probiotics/prebiotics, plant products, and natural products. This entails altering the microbiological partners and products (such as amyloid protein) that these partners generate.
Collapse
Affiliation(s)
- Jajati K Pasupalak
- School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra, 425405, India
| | - Prabha Rajput
- School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra, 425405, India
| | - Girdhari Lal Gupta
- School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra, 425405, India.
| |
Collapse
|
45
|
Wang H, Shi C, Jiang L, Liu X, Tang R, Tang M. Neuroimaging techniques, gene therapy, and gut microbiota: frontier advances and integrated applications in Alzheimer's Disease research. Front Aging Neurosci 2024; 16:1485657. [PMID: 39691161 PMCID: PMC11649678 DOI: 10.3389/fnagi.2024.1485657] [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: 08/24/2024] [Accepted: 11/19/2024] [Indexed: 12/19/2024] Open
Abstract
Alzheimer's Disease (AD) is a neurodegenerative disorder marked by cognitive decline, for which effective treatments remain elusive due to complex pathogenesis. Recent advances in neuroimaging, gene therapy, and gut microbiota research offer new insights and potential intervention strategies. Neuroimaging enables early detection and staging of AD through visualization of biomarkers, aiding diagnosis and tracking of disease progression. Gene therapy presents a promising approach for modifying AD-related genetic expressions, targeting amyloid and tau pathology, and potentially repairing neuronal damage. Furthermore, emerging evidence suggests that the gut microbiota influences AD pathology through the gut-brain axis, impacting inflammation, immune response, and amyloid metabolism. However, each of these technologies faces significant challenges, including concerns about safety, efficacy, and ethical considerations. This article reviews the applications, advantages, and limitations of neuroimaging, gene therapy, and gut microbiota research in AD, with a particular focus on their combined potential for early diagnosis, mechanistic insights, and therapeutic interventions. We propose an integrated approach that leverages these tools to provide a multi-dimensional framework for advancing AD diagnosis, treatment, and prevention.
Collapse
Affiliation(s)
- Haitao Wang
- School of Basic Medicine, Southwest Medical University, Luzhou, Sichuan, China
- The School of Clinical Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Chen Shi
- Department of Gynaecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ling Jiang
- Department of Anorectal, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Xiaozhu Liu
- Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Rui Tang
- School of Basic Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Mingxi Tang
- School of Basic Medicine, Southwest Medical University, Luzhou, Sichuan, China
- Department of Pathology, Yaan People’s Hospital (Yaan Hospital of West China Hospital of Sichuan University), Yaan, Sichuan, China
| |
Collapse
|
46
|
Menezes AA, Shah ZA. A Review of the Consequences of Gut Microbiota in Neurodegenerative Disorders and Aging. Brain Sci 2024; 14:1224. [PMID: 39766423 PMCID: PMC11726757 DOI: 10.3390/brainsci14121224] [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: 10/29/2024] [Revised: 11/25/2024] [Accepted: 11/27/2024] [Indexed: 01/15/2025] Open
Abstract
Age-associated alterations in the brain lead to cognitive deterioration and neurodegenerative disorders (NDDs). This review with a particular focus on Alzheimer's disease (AD), emphasizes the burgeoning significance of the gut microbiota (GMB) in neuroinflammation and its impact on the gut-brain axis (GBA), a communication conduit between the gut and the central nervous system (CNS). Changes in the gut microbiome, including diminished microbial diversity and the prevalence of pro-inflammatory bacteria, are associated with AD pathogenesis. Promising therapies, such as fecal microbiota transplantation (FMT), probiotics, and prebiotics, may restore gut health and enhance cognitive performance. Clinical data remain insufficient, necessitating further research to elucidate causes, enhance therapy, and consider individual variances. This integrative approach may yield innovative therapies aimed at the GMB to improve cognitive function and brain health in older people.
Collapse
Affiliation(s)
| | - Zahoor A. Shah
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, OH 43614, USA;
| |
Collapse
|
47
|
Qi Y, Li J, Tang Y, Cao R, Gao Y, Xu Q, Han Y. Total Alkaloids of Rhizoma Corydalis regulates gut microbiota and restores gut immune barrier to ameliorate cognitive dysfunction in diabetic rats. Front Microbiol 2024; 15:1456406. [PMID: 39687865 PMCID: PMC11647011 DOI: 10.3389/fmicb.2024.1456406] [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: 06/28/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Background and objectives Given the widespread dysbiosis of gut microbiota in patients with T2DM, it has been found that the microbiota-gut-brain axis plays an influential regulatory role in diabetic cognitive dysfunction, and improving gut dysbiosis may be a potential strategy for treating diabetic cognitive dysfunction. Total Alkaloids of Rhizoma Corydalis (TAC) is the main active component extracted from Rhizoma Corydalis. Pharmacological studies have demonstrated its significant pharmacological effects on the cardiovascular and cerebrovascular systems, and berberine, the main component of TAC, has a certain regulatory effect on gut microbiota. Materials and methods Rats were randomly divided into Control group, Model group, TAC-low group, TAC-mid group and TAC-high group. Cognitive function of diabetic rats was evaluated through behavioral testing using the Morris water maze experiment. The relative abundance of gut bacteria in rat feces was determined via 16S rRNA analysis. IHC and Western blot techniques were employed to assess IL-22, IL-23, Reg3g, ZO-1, occludin 1 expression in the colon tissue; GPX4, xCT, NLRP3, Caspase-1 p20, GSDMD-N were detected in the hippocampus. Results The cognitive function of diabetic rats decreased significantly. TAC demonstrated a significant reduction in inflammatory factors in serum, hippocampus, and colon, thus alleviating inflammation. Additionally, it effectively decreased ferroptosis induced by NLRP3 and reduced pathological damage in the hippocampus of diabetic rats. After treatment, the differential microbiota such as Lachnoclotridium and Bacteroides. TAC improved gut barrier permeability and integrity in rats while remodeling gut mucosal homeostasis. Moreover, pyroptosis and ferroptosis caused by the inflammatory cascade in the rat hippocampus were also significantly inhibited. Conclusion The combination of high lipid and high glucose with STZ can result in gut microbiota disturbance, damage gut immune barrier, decreased gut mucosal permeability and integrity, aggravated gut inflammation, further spread inflammatory factors to brain tissue, cause inflammatory cascade reaction of encephalopathy, and ultimately resulting in neuronal ferroptosis and cognitive dysfunction in diabetes mellitus. Our study suggests that TAC may regulate gut microbiota, restore gut immune homeostasis, improve gut barrier permeability and integrity, inhibit brain tissue inflammatory cascade, reduce neuronal ferroptosis, and thus improve diabetes. This provides new targets for its treatment strategy.
Collapse
Affiliation(s)
- Yazhi Qi
- Basic Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jun Li
- Basic Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ya Tang
- Basic Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Rui Cao
- Jiamusi Campus, Heilongjiang University of Chinese Medicine, Jiamusi, China
| | - Yishu Gao
- Basic Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qiang Xu
- Basic Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yusheng Han
- Basic Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
48
|
Berzack S, Galor A. Microbiome-based therapeutics for ocular diseases. Clin Exp Optom 2024:1-8. [PMID: 39617011 DOI: 10.1080/08164622.2024.2422479] [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: 09/16/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 12/08/2024] Open
Abstract
The relationship between the gut microbiome and ocular health has garnered increasing attention within the scientific community. Recent research has focused on the gut-eye axis, examining whether imbalances within the gut microbiome can influence the development, progression and severity of ocular diseases, including dry eye disease, uveitis, and glaucoma. Dysbiosis within the gut microbiome is linked to immune dysregulation, chronic inflammation, and epithelial barrier dysfunction, all of which contribute to ocular pathology. This review synthesises current evidence on these associations, exploring how gut microbiome alterations drive disease mechanisms. Furthermore, it examines the therapeutic potential of microbiome-targeted interventions, including antibiotics, prebiotics, probiotics, and faecal microbiota transplantation, all of which aim to restore microbial balance and modulate immune responses. As the prevalence of these conditions continues to rise, a deeper understanding of the gut-eye axis may facilitate the development of novel, targeted therapies to address unmet needs in the management of ocular diseases.
Collapse
Affiliation(s)
- Shannan Berzack
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Anat Galor
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
49
|
Ma X, Fan M, Hannachi K, Qian H, Li Y, Wang L. Unveiling the microbiota-mediated impact of different dietary proteins on post-digestive processes: A simulated in vitro approach. Food Res Int 2024; 198:115381. [PMID: 39643348 DOI: 10.1016/j.foodres.2024.115381] [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: 08/07/2024] [Revised: 10/19/2024] [Accepted: 11/14/2024] [Indexed: 12/09/2024]
Abstract
Protein digestion and microbial metabolism play crucial roles in overall health. However, the mechanisms that differentiate the digestion and metabolism of dietary proteins from different sources in the organism remain poorly understood. This study investigated the digestive properties and microbial fermentation of various animal proteins (chicken, pork, beef, and casein) and plant proteins (soy bean, mung bean, kidney bean, rice, and wheat) in an in vitro simulation. The results indicated that animal-derived proteins had higher essential amino acid content (33.97-37.12 g/100 g) and digestibility levels (49.15-60.94 %), and provided more small molecule peptides upon digestion. Nevertheless, soy bean and wheat proteins also exhibited higher digestibility (54.70 % and 60.94 %), probably due to the extraction process. The fermentation results showed that distinct metabolic profiles that emerged for different protein sources. Plant-derived proteins (especially kidney bean, rice and wheat) promoted the proliferation of beneficial bacteria and microbial diversification and stimulated short-chain fatty acids (SCFA) production. Conversely, meat proteins (pork, chicken, beef) had significantly lower microbial diversity and SCFA than these plant proteins. These findings provide valuable insights into the effects of dietary protein sources on digestion and gut microbiome, and offer scientific guidance for optimizing dietary choices to improve health.
Collapse
Affiliation(s)
- Xuedan Ma
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
| | - Mingcong Fan
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
| | - Kanza Hannachi
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
| | - Haifeng Qian
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
| | - Yan Li
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
| | - Li Wang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China.
| |
Collapse
|
50
|
Martin M, Boulaire M, Lucas C, Peltier A, Pourtau L, Gaudout D, Layé S, Pallet V, Joffre C, Dinel AL. Plant Extracts and ω-3 Improve Short-Term Memory and Modulate the Microbiota-Gut-Brain Axis in D-galactose Model Mice. J Nutr 2024; 154:3704-3717. [PMID: 39332773 DOI: 10.1016/j.tjnut.2024.09.015] [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: 05/22/2024] [Revised: 09/05/2024] [Accepted: 09/16/2024] [Indexed: 09/29/2024] Open
Abstract
BACKGROUND Aging, characterized by a slow and progressive alteration of cognitive functions, is associated with gut microbiota dysbiosis, low-grade chronic inflammation, as well as increased oxidative stress and neurofunctional alterations. Some nutrients, such as polyphenols, carotenoids, and omega (ω)-3 (n-3), are good candidates to prevent age-related cognitive decline, because of their immunomodulatory, antioxidant, and neuroprotective properties. OBJECTIVES The objective of this study was to demonstrate the preventive effect of a combination of plant extracts (PE) containing Memophenol™ (grapes and blueberries polyphenols) and a patented saffron extract (saffron carotenoids and safranal) and ω-3 on cognitive function in a mouse model of accelerated aging and to understand the biological mechanisms involved. METHODS We used an accelerated-aging model by injecting 3-mo-old male C57Bl6/J mice with D-galactose for 8 wk, during which they were fed with a balanced control diet and supplemented or not with PE and/or ω-3 (n = 15-16/group). Short-term memory was evaluated by Y-maze test, following analyses of hippocampal and intestinal RNA expressions, brain fatty acid and oxylipin amounts, and gut microbiota composition (16S rRNA gene sequencing). Statistical analyses were performed (t test, analysis of variance, and Pearson correlation). RESULTS Our results showed that oral administration of PE, ω-3, or both (mix) prevented hippocampus-dependent short-term memory deficits induced by D-galactose (P < 0.05). This effect was accompanied by the modulation of gut microbiota, altered by the treatment. PE and the mix increased the expression of antioxidative and neurogenesis markers, such as catalase and doublecortin, in hippocampus (P < 0.05 for both). Moreover, ω-3 and the mix showed a higher ω-3 amounts (P < 0.05) and EPA-derived 18- hydroxyeicosapentaenoic acid (P < 0.001) in prefrontal cortex. These changes may contribute to the improvement in memory. CONCLUSIONS These results suggest that the mix of PE and ω-3 could be more efficient at attenuating age-related cognitive decline than individual supplementations because it targeted, in mice, the different pathways impaired with aging.
Collapse
Affiliation(s)
- Marie Martin
- Université Bordeaux, INRAE, Bordeaux INP, Nutrineuro, Bordeaux, France; Activ'Inside, 12 route de Beroy, ZA du Grand Cazeau, Beychac-et-Caillau
| | - Milan Boulaire
- Université Bordeaux, INRAE, Bordeaux INP, Nutrineuro, Bordeaux, France
| | - Céline Lucas
- Université Bordeaux, INRAE, Bordeaux INP, Nutrineuro, Bordeaux, France; NutriBrain Research and Technology Transfer, NutriNeuro, Bordeaux, France
| | - Adrien Peltier
- Université Bordeaux, INRAE, Bordeaux INP, Nutrineuro, Bordeaux, France; NutriBrain Research and Technology Transfer, NutriNeuro, Bordeaux, France
| | - Line Pourtau
- Activ'Inside, 12 route de Beroy, ZA du Grand Cazeau, Beychac-et-Caillau
| | - David Gaudout
- Activ'Inside, 12 route de Beroy, ZA du Grand Cazeau, Beychac-et-Caillau
| | - Sophie Layé
- Université Bordeaux, INRAE, Bordeaux INP, Nutrineuro, Bordeaux, France
| | - Véronique Pallet
- Université Bordeaux, INRAE, Bordeaux INP, Nutrineuro, Bordeaux, France
| | - Corinne Joffre
- Université Bordeaux, INRAE, Bordeaux INP, Nutrineuro, Bordeaux, France
| | - Anne-Laure Dinel
- Université Bordeaux, INRAE, Bordeaux INP, Nutrineuro, Bordeaux, France; NutriBrain Research and Technology Transfer, NutriNeuro, Bordeaux, France.
| |
Collapse
|