1
|
Vollhardt A, Frölich L, Stockbauer AC, Danek A, Schmitz C, Wahl AS. Towards a better diagnosis and treatment of dementia: Identifying common and distinct neuropathological mechanisms in Alzheimer's and vascular dementia. Neurobiol Dis 2025; 208:106845. [PMID: 39999928 DOI: 10.1016/j.nbd.2025.106845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 02/05/2025] [Accepted: 02/15/2025] [Indexed: 02/27/2025] Open
Abstract
Alzheimer's disease (AD) and vascular dementia (VaD) together contribute to almost 90 % of all dementia cases leading to major health challenges of our time with a substantial global socioeconomic burden. While in AD, the improved understanding of Amyloid beta (Aß) mismetabolism and tau hyperphosphorylation as pathophysiological hallmarks has led to significant clinical breakthroughs, similar advances in VaD are lacking. After comparing the clinical presentation, including risk factors, disease patterns, course of diseases and further diagnostic parameters for both forms of dementia, we highlight the importance of shared pathomechanisms found in AD and VaD: Endothelial damage, blood brain barrier (BBB) breakdown and hypoperfusion inducing oxidative stress and inflammation and thus trophic uncoupling in the neurovascular unit. A dysfunctional endothelium and BBB lead to the accumulation of neurotoxic molecules and Aß through impaired clearance, which in turn leads to neurodegeneration. In this context we discuss possible neuropathological parameters, which might serve as biomarkers and thus improve diagnostic accuracy or reveal targets for novel therapeutic strategies for both forms of dementia.
Collapse
Affiliation(s)
- Alisa Vollhardt
- Department of Neuroanatomy, Institute of Anatomy, Ludwigs-Maximilians-University, Pettikoferstrasse 11, 80336 Munich, Germany
| | - Lutz Frölich
- Central Institute of Mental Health, University of Heidelberg, J5, 68159 Mannheim, Germany
| | - Anna Christina Stockbauer
- Department of Neurology, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Adrian Danek
- Department of Neurology, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Christoph Schmitz
- Department of Neuroanatomy, Institute of Anatomy, Ludwigs-Maximilians-University, Pettikoferstrasse 11, 80336 Munich, Germany
| | - Anna-Sophia Wahl
- Department of Neuroanatomy, Institute of Anatomy, Ludwigs-Maximilians-University, Pettikoferstrasse 11, 80336 Munich, Germany; Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, Feodor-Lynen-Strasse 17, 81377 Munich, Germany.
| |
Collapse
|
2
|
Song Z, Li M, Zhou Z, Guo X, Wang Q, Zhang Z, Wang K, Zheng Q, Gou W, Wu S, Zhao H, Gong M. Wuzhuyu Decoction Relieves Chronic Migraine by Regulating 5-HT1A and 3A Receptors-Mediated CREB Signaling Pathway in Brain and Intestine. Pharmaceuticals (Basel) 2025; 18:426. [PMID: 40143202 PMCID: PMC11944717 DOI: 10.3390/ph18030426] [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: 02/11/2025] [Revised: 03/04/2025] [Accepted: 03/08/2025] [Indexed: 03/28/2025] Open
Abstract
Background: Chronic migraine (CM) is a common complex nervous system disease, often accompanied by symptoms of the digestive tract that interact with each other, leading to prolonged and difficult-to-cure migraines. These symptoms are associated with abnormalities in 5-HT and its receptors. Wuzhuyu decoction (WZYD) is a traditional Chinese medicine prescription commonly used in clinics to treat CM; it relieves gastrointestinal symptoms, such as nausea and vomiting; however, its mechanism is still unclear. Investigating the differences in the role of WZYD compared to existing drugs targeting 5-HT receptors in the treatment of CM not only helps elucidate its pathogenesis but also provides possibilities for the development of new therapeutic approaches. Methods: An inflammation soup (IS)-induced CM male rat model was established. Based on a preliminary experiment, the target of WZYD in treating CM was determined by network pharmacology, and verified by molecular docking. ELISA, immunofluorescence, western blot, and real-time quantitative polymerase chain reaction (RT-qPCR) were used to evaluate the expression levels of CM-related indicators (5-HT, calcitonin gene-related peptide (CGRP), and c-Fos) to ensure the successful establishment of the CM model and the effectiveness of the drug. On this basis, the protein expression levels of 5-HT1A/3A receptors and their cAMP-response element binding protein (CREB) signaling pathway were detected by western blot and immunohistochemistry. The role of 5-HT1A/3A receptors in the treatment of CM by WZYD was validated using a 5-HT1A receptor antagonist (WAY 100635) and a 5-HT3A receptor agonist (SR 57227). Results: The results showed that WZYD increased the expression of 5-HT in the brain, decreased the expression of CGRP, c-Fos, ionized calcium-binding adapter molecule 1 (Iba1), and relieved CM. At the same time, WZYD also increased the expression of the 5-HT1A receptor and decreased the expression of the 5-HT3A receptor in the brain and colon of CM rats. Subsequently, WZYD further exerted its brain-gut integrated therapeutic effects by regulating the CREB signaling pathway mediated by 5-HT1A/3A receptors in the brain and colon of CM rats. Conclusions: WZYD not only regulates neurotransmitters in the brain and colon at the same time, but also specifically regulates 5-HT1A/3A receptors in the brain and colon, which explains the characteristics and advantages of WZYD from a new perspective. While effectively relieving headache symptoms, it also improves related gastrointestinal symptoms, which is more conducive to the treatment of CM.
Collapse
Affiliation(s)
- Zhimin Song
- School of Traditional Chinese Medicine, Capital Medical University, No. 10 of Xitoutiao, Youanmenwai, Fengtai District, Beijing 100069, China; (Z.S.); (M.L.); (Z.Z.); (X.G.); (Q.W.); (Z.Z.); (K.W.); (Q.Z.); (W.G.); (S.W.); (H.Z.)
- Beijing Key Laboratory of Traditional Chinese Medicine Collateral Disease Theory Research, Beijing 100069, China
| | - Meijing Li
- School of Traditional Chinese Medicine, Capital Medical University, No. 10 of Xitoutiao, Youanmenwai, Fengtai District, Beijing 100069, China; (Z.S.); (M.L.); (Z.Z.); (X.G.); (Q.W.); (Z.Z.); (K.W.); (Q.Z.); (W.G.); (S.W.); (H.Z.)
- Beijing Key Laboratory of Traditional Chinese Medicine Collateral Disease Theory Research, Beijing 100069, China
| | - Ziwei Zhou
- School of Traditional Chinese Medicine, Capital Medical University, No. 10 of Xitoutiao, Youanmenwai, Fengtai District, Beijing 100069, China; (Z.S.); (M.L.); (Z.Z.); (X.G.); (Q.W.); (Z.Z.); (K.W.); (Q.Z.); (W.G.); (S.W.); (H.Z.)
- Beijing Key Laboratory of Traditional Chinese Medicine Collateral Disease Theory Research, Beijing 100069, China
| | - Xiaomeng Guo
- School of Traditional Chinese Medicine, Capital Medical University, No. 10 of Xitoutiao, Youanmenwai, Fengtai District, Beijing 100069, China; (Z.S.); (M.L.); (Z.Z.); (X.G.); (Q.W.); (Z.Z.); (K.W.); (Q.Z.); (W.G.); (S.W.); (H.Z.)
- Beijing Key Laboratory of Traditional Chinese Medicine Collateral Disease Theory Research, Beijing 100069, China
| | - Qi Wang
- School of Traditional Chinese Medicine, Capital Medical University, No. 10 of Xitoutiao, Youanmenwai, Fengtai District, Beijing 100069, China; (Z.S.); (M.L.); (Z.Z.); (X.G.); (Q.W.); (Z.Z.); (K.W.); (Q.Z.); (W.G.); (S.W.); (H.Z.)
- Beijing Key Laboratory of Traditional Chinese Medicine Collateral Disease Theory Research, Beijing 100069, China
| | - Zekuan Zhang
- School of Traditional Chinese Medicine, Capital Medical University, No. 10 of Xitoutiao, Youanmenwai, Fengtai District, Beijing 100069, China; (Z.S.); (M.L.); (Z.Z.); (X.G.); (Q.W.); (Z.Z.); (K.W.); (Q.Z.); (W.G.); (S.W.); (H.Z.)
- Beijing Key Laboratory of Traditional Chinese Medicine Collateral Disease Theory Research, Beijing 100069, China
| | - Keshu Wang
- School of Traditional Chinese Medicine, Capital Medical University, No. 10 of Xitoutiao, Youanmenwai, Fengtai District, Beijing 100069, China; (Z.S.); (M.L.); (Z.Z.); (X.G.); (Q.W.); (Z.Z.); (K.W.); (Q.Z.); (W.G.); (S.W.); (H.Z.)
- Beijing Key Laboratory of Traditional Chinese Medicine Collateral Disease Theory Research, Beijing 100069, China
| | - Qixiang Zheng
- School of Traditional Chinese Medicine, Capital Medical University, No. 10 of Xitoutiao, Youanmenwai, Fengtai District, Beijing 100069, China; (Z.S.); (M.L.); (Z.Z.); (X.G.); (Q.W.); (Z.Z.); (K.W.); (Q.Z.); (W.G.); (S.W.); (H.Z.)
- Beijing Key Laboratory of Traditional Chinese Medicine Collateral Disease Theory Research, Beijing 100069, China
| | - Wenjing Gou
- School of Traditional Chinese Medicine, Capital Medical University, No. 10 of Xitoutiao, Youanmenwai, Fengtai District, Beijing 100069, China; (Z.S.); (M.L.); (Z.Z.); (X.G.); (Q.W.); (Z.Z.); (K.W.); (Q.Z.); (W.G.); (S.W.); (H.Z.)
- Beijing Key Laboratory of Traditional Chinese Medicine Collateral Disease Theory Research, Beijing 100069, China
| | - Sha Wu
- School of Traditional Chinese Medicine, Capital Medical University, No. 10 of Xitoutiao, Youanmenwai, Fengtai District, Beijing 100069, China; (Z.S.); (M.L.); (Z.Z.); (X.G.); (Q.W.); (Z.Z.); (K.W.); (Q.Z.); (W.G.); (S.W.); (H.Z.)
- Beijing Key Laboratory of Traditional Chinese Medicine Collateral Disease Theory Research, Beijing 100069, China
| | - Hui Zhao
- School of Traditional Chinese Medicine, Capital Medical University, No. 10 of Xitoutiao, Youanmenwai, Fengtai District, Beijing 100069, China; (Z.S.); (M.L.); (Z.Z.); (X.G.); (Q.W.); (Z.Z.); (K.W.); (Q.Z.); (W.G.); (S.W.); (H.Z.)
| | - Muxin Gong
- School of Traditional Chinese Medicine, Capital Medical University, No. 10 of Xitoutiao, Youanmenwai, Fengtai District, Beijing 100069, China; (Z.S.); (M.L.); (Z.Z.); (X.G.); (Q.W.); (Z.Z.); (K.W.); (Q.Z.); (W.G.); (S.W.); (H.Z.)
- Beijing Key Laboratory of Traditional Chinese Medicine Collateral Disease Theory Research, Beijing 100069, China
| |
Collapse
|
3
|
Zhu G, Li X, Ma Q, Hong Z, Zhong D, Sun X. Construction and application of magnetic surface molecularly imprinted solid-phase extraction for the detection of 5-hydroxytryptamine in peripheral blood. Mikrochim Acta 2025; 192:220. [PMID: 40067417 DOI: 10.1007/s00604-025-07058-4] [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: 10/04/2024] [Accepted: 02/18/2025] [Indexed: 03/18/2025]
Abstract
The 5-Hydroxytryptamine (5-HT) level determination is crucial for predicting, pathogenesis, diagnosis, and pharmacological treatment of schizophrenia. To realize the extraction of trace 5-HT in complex matrix samples, a magnetic molecularly imprinted solid-phase extraction (MMISPE) pretreatment method was developed. In combination with UPLC-MS/MS, the method was possible to achieve the quantification of 1 ng/mL 5-HT in tissue samples. The type of adsorbent, magnetic surface molecularly imprinted polymers (MMIPs) adsorbent dosage, adsorption temperature, and adsorption method were screened to obtain the better extraction of 5-HT. After optimizing the extraction and separation method, we applied the MMISPE method to the detection of 5-HT in peripheral blood of clinical schizophrenia patients. The results showed that the average concentration of 5-HT in the peripheral blood of healthy controls was 74.30 ng/mL, whereas the 5-HT content in the samples from schizophrenic patients was 1.17 ng/mL.
Collapse
Affiliation(s)
- Gege Zhu
- Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Xinyu Li
- Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Qianjie Ma
- Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Zhanying Hong
- School of Pharmacy, Second Military Medical University, Shanghai, 200433, China.
| | - Dan Zhong
- Shanghai Key Laboratory of Bio-Energy Crops, School of Life Sciences, Shanghai University, Shanghai, 200444, China.
| | - Xiaodong Sun
- Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Medicine, Shanghai University, Shanghai, 200444, China.
- Shanghai Key Laboratory of Bio-Energy Crops, School of Life Sciences, Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
4
|
Fu J, Liang Z, Chen Z, Zhou Y, Xiong F, Liang Q, Gao H. Deciphering the Therapeutic Efficacy and Underlying Mechanisms of Dendrobium officinale Polysaccharides in the Intervention of Alzheimer's Disease Mice: Insights from Metabolomics and Microbiome. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:5635-5648. [PMID: 39536176 DOI: 10.1021/acs.jafc.4c07913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
As a traditional drug-food homologous plant, Dendrobium officinale is widely recognized for its nutritional and medicinal value. Specifically, D. officinale polysaccharide (DOP) has garnered attention as a potential prebiotic for its protective effects on gut microbiota and the nervous system. However, the underlying mechanism by which DOP improves cognitive dysfunction in Alzheimer's disease (AD) remains unclear. This study intends to elucidate the beneficial effects of DOP on AD mice from the perspectives of metabolomics and the intestinal microbiome. The results showed that DOP significantly ameliorated cognitive dysfunction, attenuated hippocampal neuronal damage and Aβ plaque deposition, and restored intestinal barrier integrity in AD mice. The antibiotic-cocktail-induced germ-free mouse model confirmed that the neuroprotective effect of DOP was dependent on gut microbiota. Further investigations demonstrated that DOP influenced the composition of gut microbiota and restored its diversity. Additionally, DOP reshaped metabolic profile disorders in AD mice and increased the short-chain fatty acids (SCFAs) content. Correlation analysis further highlighted that specific gut microbiota was associated with the metabolism of AD mice. In conclusion, this study sheds light on the positive impact of DOP in reshaping the gut microbiota and enhancing cognitive function, offering important perspectives for the possible advancement and utilization of DOP.
Collapse
Affiliation(s)
- Jun Fu
- Innovation Academy of Testing Technology, Scientific Research Center, Wenzhou Medical University, Wenzhou 325035, China
- Key Laboratory of Efficacy Evaluation of Traditional Chinese Medicine and Encephalopathy Research of Zhejiang Province, Wenzhou Medical University, Wenzhou 325035, China
| | - Zhaohan Liang
- Innovation Academy of Testing Technology, Scientific Research Center, Wenzhou Medical University, Wenzhou 325035, China
| | - Zihao Chen
- Innovation Academy of Testing Technology, Scientific Research Center, Wenzhou Medical University, Wenzhou 325035, China
| | - Yiyang Zhou
- Innovation Academy of Testing Technology, Scientific Research Center, Wenzhou Medical University, Wenzhou 325035, China
| | - Fen Xiong
- Innovation Academy of Testing Technology, Scientific Research Center, Wenzhou Medical University, Wenzhou 325035, China
| | - Qian Liang
- Innovation Academy of Testing Technology, Scientific Research Center, Wenzhou Medical University, Wenzhou 325035, China
- Key Laboratory of Efficacy Evaluation of Traditional Chinese Medicine and Encephalopathy Research of Zhejiang Province, Wenzhou Medical University, Wenzhou 325035, China
| | - Hongchang Gao
- Innovation Academy of Testing Technology, Scientific Research Center, Wenzhou Medical University, Wenzhou 325035, China
- Key Laboratory of Efficacy Evaluation of Traditional Chinese Medicine and Encephalopathy Research of Zhejiang Province, Wenzhou Medical University, Wenzhou 325035, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health); Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| |
Collapse
|
5
|
Wang M, Liu Y, Zhong L, Wu F, Wang J. Advancements in the investigation of gut microbiota-based strategies for stroke prevention and treatment. Front Immunol 2025; 16:1533343. [PMID: 40103814 PMCID: PMC11914130 DOI: 10.3389/fimmu.2025.1533343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 02/11/2025] [Indexed: 03/20/2025] Open
Abstract
Stroke represents a predominant cause of mortality and disability on a global scale, impacting millions annually and exerting a considerable strain on healthcare systems. The incidence of stroke exhibits regional variability, with ischemic stroke accounting for the majority of occurrences. Post-stroke complications, such as cognitive impairment, motor dysfunction, and recurrent stroke, profoundly affect patients' quality of life. Recent advancements have elucidated the microbiota-gut-brain axis (MGBA), underscoring the complex interplay between gut health and brain function. Dysbiosis, characterized by an imbalance in gut microbiota, is significantly linked to an elevated risk of stroke and unfavorable outcomes. The MGBA plays a crucial role in modulating immune function, neurotransmitter levels, and metabolic byproducts, which may intensify neuroinflammation and impair cerebral health. This review elucidates the role of MGBA in stroke pathophysiology and explores potential gut-targeted therapeutic strategies to reduce stroke risk and promote recovery, including probiotics, prebiotics, pharmacological interventions, and dietary modifications. However, the current prevention and treatment strategies based on intestinal flora still face many problems, such as the large difference of individual intestinal flora, the stability of efficacy, and the long-term safety need to be considered. Further research needs to be strengthened to promote its better application in clinical practice.
Collapse
Affiliation(s)
- Min Wang
- Department of Gastroenterology, The First People's Hospital of Xiaoshan District, Hangzhou, Zhejiang, China
| | - Yan Liu
- Department of Gastroenterology, The First People's Hospital of Xiaoshan District, Hangzhou, Zhejiang, China
| | - Li Zhong
- Department of Gastroenterology, The First People's Hospital of Xiaoshan District, Hangzhou, Zhejiang, China
| | - Fang Wu
- Department of Gastroenterology, The First People's Hospital of Xiaoshan District, Hangzhou, Zhejiang, China
| | - Jinjin Wang
- Department of Gastroenterology, The First People's Hospital of Xiaoshan District, Hangzhou, Zhejiang, China
| |
Collapse
|
6
|
Sharma A, Rudrawar S, Bharate SB, Jadhav HR. Recent advancements in the therapeutic approaches for Alzheimer's disease treatment: current and future perspective. RSC Med Chem 2025; 16:652-693. [PMID: 39790124 PMCID: PMC11707861 DOI: 10.1039/d4md00630e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 12/05/2024] [Indexed: 01/12/2025] Open
Abstract
Alzheimer's disease (AD) is a complex, incurable neurological condition characterized by cognitive decline, cholinergic neuron reduction, and neuronal loss. Its exact pathology remains uncertain, but multiple treatment hypotheses have emerged. The current treatments, single or combined, alleviate only symptoms and struggle to manage AD due to its multifaceted pathology. The developmental drugs target pivotal disease factors involved in the envisaged hypotheses and include targets such as amyloid aggregation, hyperphosphorylated tau proteins, and receptors like cholinergic, adrenergic, etc. Present-day research focuses on multi-target directed ligands (MTDLs), which inhibit multiple factors simultaneously, helping slow the disease's progression. This review attempts to collate the recent information related to proposed hypotheses for AD etiology. It systematically organizes the advances in various therapeutic options for AD, with a particular emphasis on clinical candidates. Also, it is expected to help medicinal chemists design novel AD treatments based on available information, which could be helpful to AD patients.
Collapse
Affiliation(s)
- Amit Sharma
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Sciences Pilani Pilani Campus, Vidya Vihar Pilani 333031 RJ India +91 1596 244183 +91 1596 255 506
| | - Santosh Rudrawar
- The Institute for Biomedicine and Glycomics, Griffith University Gold Coast 4222 Australia
- School of Pharmacy and Medical Sciences, Griffith University Gold Coast 4222 Australia
| | - Sandip B Bharate
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine Canal Road Jammu 181110 India
| | - Hemant R Jadhav
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Sciences Pilani Pilani Campus, Vidya Vihar Pilani 333031 RJ India +91 1596 244183 +91 1596 255 506
| |
Collapse
|
7
|
Ogunro OB, Karigidi ME, Gyebi GA, Turkistani A, Almehmadi AH. Tangeretin offers neuroprotection against colchicine-induced memory impairment in Wistar rats by modulating the antioxidant milieu, inflammatory mediators and oxidative stress in the brain tissue. BMC Complement Med Ther 2025; 25:40. [PMID: 39905390 DOI: 10.1186/s12906-025-04769-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 01/17/2025] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND Tangeretin, a flavone compound (O-polymethoxylated) naturally present in tangerine and other citrus peels has demonstrated effectiveness as an anti-inflammatory and neuroprotective agent in several disease model. This study evaluated the impact of tangeretin in mitigating cognitive dysfunction and oxidative stress induced by colchicine in rats, comparing its efficacy with donepezil hydrochloride. METHODS Cognitive dysfunction was induced by administering colchicine (15 µg/rat) intracerebroventricularly (ICV) via a stereotaxic apparatus in male Wistar rats. Colchicine resulted in poor memory retention in acquiring and retaining a spatial navigation task, passive avoidance apparatus, and Morris water maze paradigms. Chronic treatment with tangeretin (at doses of 50, 100, and 200 mg/kg, p.o. once daily) and donepezil hydrochloride (at a dose of 10 mg/kg, p.o. daily) for 28 days, starting seven days before colchicine injection, significantly ameliorated colchicine-induced cognitive impairment. RESULTS The biochemical analysis showed that chronic administration of tangeretin effectively reversed the colchicine-induced increase in the level/activity of lipid peroxidation, hydrogen peroxide (H2O2), myeloperoxidase (MPO), nitrite, reactive oxygen species (ROS), tumour necrosis factor-α (TNF-α), nuclear factor kappa B (NF-κB), interleukin-1β (IL-1β), interleukin-6 (IL-6), interleukin-10 (IL-10), serotonin, dopamine, glutamate, amyloid beta (Aβ) peptide, and caspase-3. Tangeretin also reversed the colchicine-induced reduction in the level/activity of brain-derived neurotrophic factor (BDNF), amma-aminobutyric acid (GABA), acetylcholinesterase (AChE), glutathione S-Transferase (GST), glutathione peroxidase (GPx), glutathione reductase (GR), catalase (CAT), superoxide dismutase (SOD), reduced glutathione (GSH), and total thiol (T-SH) in rat brains. However, donepezil hydrochloride did not prevent oxidative stress. CONCLUSIONS These findings suggest that chronic administration of tangeretin at 50, 100, and 200 mg/kg, p.o. once daily, was protective in mitigating colchicine-induced cognitive impairment and associated oxidative stress. At the same time, donepezil hydrochloride did not demonstrate similar effects.
Collapse
Affiliation(s)
- Olalekan Bukunmi Ogunro
- Pharmacology, Drug Discovery and Toxicology Research Laboratory, Department of Biological Sciences, KolaDaisi University, Ibadan, 200213, Nigeria.
| | - Mojisola Esther Karigidi
- Pharmacology, Drug Discovery and Toxicology Research Laboratory, Department of Biological Sciences, KolaDaisi University, Ibadan, 200213, Nigeria
| | - Gideon Ampoma Gyebi
- Department of Biotechnology and Food Science, Faculty of Applied Sciences, Durban University of Technology, P.O. Box 1334, Durban, 4000, South Africa
| | - Areej Turkistani
- Department of Pharmacology and Toxicology, College of Medicine, Taif University, Taif, 21944, Kingdom of Saudi Arabia
| | - Ahmad H Almehmadi
- Oral Biology Department, Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
8
|
Jiao W, Lin J, Deng Y, Ji Y, Liang C, Wei S, Jing X, Yan F. The immunological perspective of major depressive disorder: unveiling the interactions between central and peripheral immune mechanisms. J Neuroinflammation 2025; 22:10. [PMID: 39828676 PMCID: PMC11743025 DOI: 10.1186/s12974-024-03312-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 11/26/2024] [Indexed: 01/22/2025] Open
Abstract
Major depressive disorder is a prevalent mental disorder, yet its pathogenesis remains poorly understood. Accumulating evidence implicates dysregulated immune mechanisms as key contributors to depressive disorders. This review elucidates the complex interplay between peripheral and central immune components underlying depressive disorder pathology. Peripherally, systemic inflammation, gut immune dysregulation, and immune dysfunction in organs including gut, liver, spleen and adipose tissue influence brain function through neural and molecular pathways. Within the central nervous system, aberrant microglial and astrocytes activation, cytokine imbalances, and compromised blood-brain barrier integrity propagate neuroinflammation, disrupting neurotransmission, impairing neuroplasticity, and promoting neuronal injury. The crosstalk between peripheral and central immunity creates a vicious cycle exacerbating depressive neuropathology. Unraveling these multifaceted immune-mediated mechanisms provides insights into major depressive disorder's pathogenic basis and potential biomarkers and targets. Modulating both peripheral and central immune responses represent a promising multidimensional therapeutic strategy.
Collapse
Affiliation(s)
- Wenli Jiao
- School of Nursing, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, Guangdong, China
| | - Jiayi Lin
- School of Nursing, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, Guangdong, China
| | - Yanfang Deng
- Department of Psychiatry, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Yelin Ji
- School of Nursing, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, Guangdong, China
| | - Chuoyi Liang
- School of Nursing, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, Guangdong, China
| | - Sijia Wei
- School of Nursing, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, Guangdong, China
| | - Xi Jing
- School of Nursing, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, Guangdong, China.
- Guangdong-Hong Kong-Macau Great Bay Area Geoscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou, Guangdong, China.
| | - Fengxia Yan
- School of Nursing, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, Guangdong, China.
| |
Collapse
|
9
|
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
|
10
|
Cui J, Xiao S, Cao Y, Zhang Y, Yang J, Zheng L, Zhao F, Liu X, Liu D, Zhou Z, Wang P. Organophosphate Insecticide Malathion Induces Alzheimer's Disease-Like Cognitive Impairment in Mice: Evidence of the Microbiota-Gut-Brain Axis. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:21966-21977. [PMID: 39545844 DOI: 10.1021/acs.est.4c07427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
Evidence suggests that exposure to organophosphate pesticides increases the risk of neurodegenerative diseases, but the mechanisms remain unclear. This study investigated the effects of malathion on Alzheimer's disease (AD)-like symptoms at environmentally relevant concentrations using wild-type (WT) and APP/PS1 transgenic mouse models. Results showed that malathion exposure induced AD-like cognitive impairment, amyloid-β (Aβ) accumulation, and neuroinflammation in WT mice, with worsened symptoms in APP/PS1 mice. Mechanistic studies revealed that malathion induced AD-like gut microbiota dysbiosis (reduced Lactobacillus and Akkermansia, and increased Dubosiella), causing gut barrier impairment and tryptophan metabolism disruptions. This resulted in a significant increase in indole derivatives and activation of the colonic aryl hydrocarbon receptor (AhR), promoting the kynurenine (KYN) pathway while inhibiting the serotonin (5-HT) pathway. Increased neurotoxic KYN metabolites (3-hydroxykynurenine and quinolinic acid) triggered gut and systemic inflammation, upregulating hippocampal IL-6 and IL-1β mRNA levels and thereby causing neuroinflammation. Gut tryptophan metabolism disruptions caused hippocampal neurotransmitter imbalances, reducing the levels of 5-HT and its derivatives. These effects promoted AD progression in both WT and APP/PS1 mice. This study highlights the crucial role of the microbiota-gut-brain axis in AD-like cognitive impairment induced by malathion exposure, providing insights into the neurodegenerative disease risks posed by organophosphate pesticides.
Collapse
Affiliation(s)
- Jingna Cui
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P. R. China
| | - Shouchun Xiao
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P. R. China
| | - Yue Cao
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P. R. China
| | - Yaru Zhang
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P. R. China
| | - Jiaxing Yang
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P. R. China
| | - Li Zheng
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P. R. China
| | - Fanrong Zhao
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P. R. China
| | - Xueke Liu
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P. R. China
| | - Donghui Liu
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P. R. China
| | - Zhiqiang Zhou
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P. R. China
| | - Peng Wang
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P. R. China
| |
Collapse
|
11
|
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
|
12
|
Sighencea MG, Popescu RȘ, Trifu SC. From Fundamentals to Innovation in Alzheimer's Disease: Molecular Findings and Revolutionary Therapies. Int J Mol Sci 2024; 25:12311. [PMID: 39596378 PMCID: PMC11594972 DOI: 10.3390/ijms252212311] [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: 10/07/2024] [Revised: 11/11/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Alzheimer's disease (AD) is a global health concern and the leading cause of dementia in the elderly. The prevalence of this neurodegenerative condition is projected to increase concomitantly with increased life expectancy, resulting in a significant economic burden. With very few FDA-approved disease-modifying drugs available for AD, there is an urgent need to develop new compounds capable of impeding the progression of the disease. Given the unclear etiopathogenesis of AD, this review emphasizes the underlying mechanisms of this condition. It explores not only well-studied aspects, such as the accumulation of Aβ plaques and neurofibrillary tangles, but also novel areas, including glymphatic and lymphatic pathways, microbiota and the gut-brain axis, serotoninergic and autophagy alterations, vascular dysfunction, the metal hypothesis, the olfactory pathway, and oral health. Furthermore, the potential molecular targets arising from all these mechanisms have been reviewed, along with novel promising approaches such as nanoparticle-based therapy, neural stem cell transplantation, vaccines, and CRISPR-Cas9-mediated genome editing techniques. Taking into account the overlap of these various mechanisms, individual and combination therapies emerge as the future direction in the AD strategy.
Collapse
Affiliation(s)
| | - Ramona Ștefania Popescu
- Department of Infectious Diseases, “Carol Davila” University of Medicine and Pharmacy Bucharest, 020021 Bucharest, Romania;
| | - Simona Corina Trifu
- Department of Psychiatry, “Carol Davila” University of Medicine and Pharmacy Bucharest, 020021 Bucharest, Romania
| |
Collapse
|
13
|
Miyamoto K, Sujino T, Kanai T. The tryptophan metabolic pathway of the microbiome and host cells in health and disease. Int Immunol 2024; 36:601-616. [PMID: 38869080 PMCID: PMC11562643 DOI: 10.1093/intimm/dxae035] [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: 02/22/2024] [Accepted: 06/06/2024] [Indexed: 06/14/2024] Open
Abstract
The intricate and dynamic tryptophan (Trp) metabolic pathway in both the microbiome and host cells highlights its profound implications for health and disease. This pathway involves complex interactions between host cellular and bacteria processes, producing bioactive compounds such as 5-hydroxytryptamine (5-HT) and kynurenine derivatives. Immune responses to Trp metabolites through specific receptors have been explored, highlighting the role of the aryl hydrocarbon receptor in inflammation modulation. Dysregulation of this pathway is implicated in various diseases, such as Alzheimer's and Parkinson's diseases, mood disorders, neuronal diseases, autoimmune diseases such as multiple sclerosis (MS), and cancer. In this article, we describe the impact of the 5-HT, Trp, indole, and Trp metabolites on health and disease. Furthermore, we review the impact of microbiome-derived Trp metabolites that affect immune responses and contribute to maintaining homeostasis, especially in an experimental autoimmune encephalitis model of MS.
Collapse
Affiliation(s)
- Kentaro Miyamoto
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
- Miyarisan Pharmaceutical Co., Research Laboratory, Tokyo, Japan
| | - Tomohisa Sujino
- Center for Diagnostic and Therapeutic Endoscopy, Keio University School of Medicine, Tokyo, Japan
- Keio Global Research Institute, Keio University, Tokyo, Japan
| | - Takanori Kanai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
14
|
Kustrimovic N, Balkhi S, Bilato G, Mortara L. Gut Microbiota and Immune System Dynamics in Parkinson's and Alzheimer's Diseases. Int J Mol Sci 2024; 25:12164. [PMID: 39596232 PMCID: PMC11595203 DOI: 10.3390/ijms252212164] [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: 09/03/2024] [Revised: 10/25/2024] [Accepted: 10/27/2024] [Indexed: 11/28/2024] Open
Abstract
The gut microbiota, a diverse collection of microorganisms in the gastrointestinal tract, plays a critical role in regulating metabolic, immune, and cognitive functions. Disruptions in the composition of these microbial communities, termed dysbiosis, have been linked to various neurodegenerative diseases (NDs), such as Parkinson's disease (PD) and Alzheimer's disease (AD). One of the key pathological features of NDs is neuroinflammation, which involves the activation of microglia and peripheral immune cells. The gut microbiota modulates immune responses through the production of metabolites and interactions with immune cells, influencing the inflammatory processes within the central nervous system. This review explores the impact of gut dysbiosis on neuroinflammation, focusing on the roles of microglia, immune cells, and potential therapeutic strategies targeting the gut microbiota to alleviate neuroinflammatory processes in NDs.
Collapse
Affiliation(s)
- Natasa Kustrimovic
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy;
| | - Sahar Balkhi
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (S.B.); (G.B.)
| | - Giorgia Bilato
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (S.B.); (G.B.)
- Unit of Molecular Pathology, Biochemistry and Immunology, IRCCS MultiMedica, 20138 Milan, Italy
| | - Lorenzo Mortara
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (S.B.); (G.B.)
- Unit of Molecular Pathology, Biochemistry and Immunology, IRCCS MultiMedica, 20138 Milan, Italy
| |
Collapse
|
15
|
Bano N, Khan S, Ahamad S, Kanshana JS, Dar NJ, Khan S, Nazir A, Bhat SA. Microglia and gut microbiota: A double-edged sword in Alzheimer's disease. Ageing Res Rev 2024; 101:102515. [PMID: 39321881 DOI: 10.1016/j.arr.2024.102515] [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: 04/23/2024] [Revised: 09/06/2024] [Accepted: 09/19/2024] [Indexed: 09/27/2024]
Abstract
The strong association between gut microbiota (GM) and brain functions such as mood, behaviour, and cognition has been well documented. Gut-brain axis is a unique bidirectional communication system between the gut and brain, in which gut microbes play essential role in maintaining various molecular and cellular processes. GM interacts with the brain through various pathways and processes including, metabolites, vagus nerve, HPA axis, endocrine system, and immune system to maintain brain homeostasis. GM dysbiosis, or an imbalance in GM, is associated with several neurological disorders, including anxiety, depression, and Alzheimer's disease (AD). Conversely, AD is sustained by microglia-mediated neuroinflammation and neurodegeneration. Further, GM and their products also affect microglia-mediated neuroinflammation and neurodegeneration. Despite the evidence connecting GM dysbiosis and AD progression, the involvement of GM in modulating microglia-mediated neuroinflammation in AD remains elusive. Importantly, deciphering the mechanism/s by which GM regulates microglia-dependent neuroinflammation may be helpful in devising potential therapeutic strategies to mitigate AD. Herein, we review the current evidence regarding the involvement of GM dysbiosis in microglia activation and neuroinflammation in AD. We also discuss the possible mechanisms through which GM influences the functioning of microglia and its implications for therapeutic intervention. Further, we explore the potential of microbiota-targeted interventions, such as prebiotics, probiotics, faecal microbiota transplantation, etc., as a novel therapeutic strategy to mitigate neuroinflammation and AD progression. By understanding and exploring the gut-brain axis, we aspire to revolutionize the treatment of neurodegenerative disorders, many of which share a common theme of microglia-mediated neuroinflammation and neurodegeneration.
Collapse
Affiliation(s)
- Nargis Bano
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India
| | - Sameera Khan
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India
| | - Shakir Ahamad
- Department of Chemistry, Aligarh Muslim University, Aligarh 202002, India.
| | - Jitendra Singh Kanshana
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburg, PA, USA.
| | - Nawab John Dar
- CNB, SALK Institute of Biological Sciences, La Jolla, CA 92037, USA.
| | - Sumbul Khan
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India
| | - Aamir Nazir
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow, UP, India; Academy of Scientific and Innovative Research, New Delhi, India.
| | - Shahnawaz Ali Bhat
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India.
| |
Collapse
|
16
|
Akbar M, Toppo P, Nazir A. Ageing, proteostasis, and the gut: Insights into neurological health and disease. Ageing Res Rev 2024; 101:102504. [PMID: 39284418 DOI: 10.1016/j.arr.2024.102504] [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: 07/05/2024] [Revised: 08/28/2024] [Accepted: 09/09/2024] [Indexed: 09/22/2024]
Abstract
Recent research has illuminated the profound bidirectional communication between the gastrointestinal tract and the brain, furthering our understanding of neurological ailments facilitating possible therapeutic strategies. Technological advancements in high-throughput sequencing and multi-omics have unveiled significant alterations in gut microbiota and their metabolites in various neurological disorders. This review provides a thorough analysis of the role of microbiome-gut-brain axis in neurodegenerative disease pathology, linking it to reduced age-associated proteostasis. We discuss evidences that substantiate the existence of a gut-brain cross talk ranging from early clinical accounts of James Parkinson to Braak's hypothesis. In addition to understanding of microbes, the review particularly entails specific metabolites which are altered in neurodegenerative diseases. The regulatory effects of microbial metabolites on protein clearance mechanisms, proposing their potential therapeutic implications, are also discussed. By integrating this information, we advocate for a combinatory therapeutic strategy that targets early intervention, aiming to restore proteostasis and ameliorate disease progression. This approach not only provides a new perspective on the pathogenesis of neurodegenerative diseases but also highlights innovative strategies to combat the increasing burden of these age-related disorders.
Collapse
Affiliation(s)
- Mahmood Akbar
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India; Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Pranoy Toppo
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India; Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Aamir Nazir
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India; Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow 226031, India.
| |
Collapse
|
17
|
Capocchi JK, Figueroa-Romero C, Dunham SJB, Faraci G, Rothman JA, Whiteson KL, Seo DO, Holtzman DM, Grabrucker S, Nolan YM, Kaddurah-Daouk R, Jett DA. Symposium: What Does the Microbiome Tell Us about Prevention and Treatment of AD/ADRD? J Neurosci 2024; 44:e1295242024. [PMID: 39384409 PMCID: PMC11466070 DOI: 10.1523/jneurosci.1295-24.2024] [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: 07/07/2024] [Revised: 07/17/2024] [Accepted: 08/19/2024] [Indexed: 10/11/2024] Open
Abstract
Alzheimer's disease (AD) and Alzheimer's disease-related dementias (ADRDs) are broad-impact multifactorial neurodegenerative diseases. Their complexity presents unique challenges for developing effective therapies. This review highlights research presented at the 2024 Society for Neuroscience meeting which emphasized the gut microbiome's role in AD pathogenesis by influencing brain function and neurodegeneration through the microbiota-gut-brain axis. This emerging evidence underscores the potential for targeting the gut microbiota to treat AD/ADRD.
Collapse
Affiliation(s)
| | | | | | - Gina Faraci
- University of California, Irvine, Irvine, California 92697
| | - Jason A Rothman
- University of California, Irvine, Irvine, California 92697
- University of California, Riverside, Riverside, California 92521
| | | | - Dong-Oh Seo
- Washington University School of Medicine in St. Louis, St. Louis, Missouri 63110
| | - David M Holtzman
- Washington University School of Medicine in St. Louis, St. Louis, Missouri 63110
| | - Stefanie Grabrucker
- Department of Anatomy and Neuroscience, University College Cork, Cork T12 XF62, Ireland
| | - Yvonne M Nolan
- Department of Anatomy and Neuroscience, University College Cork, Cork T12 XF62, Ireland
- APC Microbiome Ireland, University College Cork, Cork T12 YT20, Ireland
| | | | - David A Jett
- National Institute of Neurological Disorders and Stroke, Rockville, Maryland 20852
| |
Collapse
|
18
|
Kang JW, Vemuganti V, Kuehn JF, Ulland TK, Rey FE, Bendlin BB. Gut microbial metabolism in Alzheimer's disease and related dementias. Neurotherapeutics 2024; 21:e00470. [PMID: 39462700 PMCID: PMC11585892 DOI: 10.1016/j.neurot.2024.e00470] [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: 07/29/2024] [Revised: 09/30/2024] [Accepted: 10/04/2024] [Indexed: 10/29/2024] Open
Abstract
Multiple studies over the last decade have established that Alzheimer's disease and related dementias (ADRD) are associated with changes in the gut microbiome. These alterations in organismal composition result in changes in the abundances of functions encoded by the microbial community, including metabolic capabilities, which likely impact host disease mechanisms. Gut microbes access dietary components and other molecules made by the host and produce metabolites that can enter circulation and cross the blood-brain barrier (BBB). In recent years, several microbial metabolites have been associated with or have been shown to influence host pathways relevant to ADRD pathology. These include short chain fatty acids, secondary bile acids, tryptophan derivatives (such as kynurenine, serotonin, tryptamine, and indoles), and trimethylamine/trimethylamine N-oxide. Notably, some of these metabolites cross the BBB and can have various effects on the brain, including modulating the release of neurotransmitters and neuronal function, inducing oxidative stress and inflammation, and impacting synaptic function. Microbial metabolites can also impact the central nervous system through immune, enteroendocrine, and enteric nervous system pathways, these perturbations in turn impact the gut barrier function and peripheral immune responses, as well as the BBB integrity, neuronal homeostasis and neurogenesis, and glial cell maturation and activation. This review examines the evidence supporting the notion that ADRD is influenced by gut microbiota and its metabolites. The potential therapeutic advantages of microbial metabolites for preventing and treating ADRD are also discussed, highlighting their potential role in developing new treatments.
Collapse
Affiliation(s)
- Jea Woo Kang
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Vaibhav Vemuganti
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Jessamine F Kuehn
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Tyler K Ulland
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Federico E Rey
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Barbara B Bendlin
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
19
|
Que M, Li S, Xia Q, Li X, Luo X, Zhan G, Luo A. Microbiota-gut-brain axis in perioperative neurocognitive and depressive disorders: Pathogenesis to treatment. Neurobiol Dis 2024; 200:106627. [PMID: 39111702 DOI: 10.1016/j.nbd.2024.106627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/01/2024] [Accepted: 08/02/2024] [Indexed: 08/28/2024] Open
Abstract
An increasing number of people undergo anesthesia and surgery. Perioperative neurocognitive and depressive disorders are common central nervous system complications with similar pathogeneses. These conditions pose a deleterious threat to human health and a significant societal burden. In recent years, numerous studies have focused on the role of the gut microbiota and its metabolites in the central nervous system via the gut-brain axis. Its involvement in perioperative neurocognitive and depressive disorders has attracted considerable attention. This review aimed to elucidate the role of the gut microbiota and its metabolites in the pathogenesis of perioperative neurocognitive and depressive disorders, as well as the value of targeted interventions and treatments.
Collapse
Affiliation(s)
- Mengxin Que
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health; Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shiyong Li
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health; Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Xia
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health; Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xing Li
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health; Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoxiao Luo
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Gaofeng Zhan
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health; Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Ailin Luo
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health; Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
20
|
Wang L, Tian M, Sun H, Gao J, Qi W, Xu J, An Y, Xu W. Association between bowel movement disorders and depressive symptoms: a cross-sectional study. Front Psychiatry 2024; 15:1449948. [PMID: 39355376 PMCID: PMC11442234 DOI: 10.3389/fpsyt.2024.1449948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 08/28/2024] [Indexed: 10/03/2024] Open
Abstract
Objectives This study aimed to explore the association between bowel movement disorders and depression in adults. Method A cross-sectional study was conducted using data from the National Health and Nutritional Examination Survey (NHANES), 2005-2010. Depression, constipation, diarrhea, and fecal incontinence were self-reported via questionnaires. Weighted logistic regression and subgroup analyses were performed to explore the association between bowel movement disorders and the risk of depression. Restricted cubic spline (RCS) was also conducted to investigate the association between bowel movements disorder and depression. Results A total of 13,820 participants were collected. Compared to the participants with normal bowel movements, the full-adjusted depression model ORs for constipation and diarrhea were 2.28 (95%CI,1.78-2.92), 1.75 (95%CI,1.31-2.31), respectively. Any kind of bowel leakage were associated with depression. The RCS showed the possible nonlinear association between bowel movement frequency/stool shape and depression. Conclusions Constipation, diarrhea, and bowel leakage are associated with an increased risk of depression.
Collapse
Affiliation(s)
- Linyue Wang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, China
- Anorectal Surgery Department, The First Affiliated Hospital of Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Maosheng Tian
- Anorectal Surgery Department, The First Affiliated Hospital of Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Hongyuan Sun
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, China
- Anorectal Surgery Department, The First Affiliated Hospital of Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Jihua Gao
- Anorectal Surgery Department, The First Affiliated Hospital of Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Wenyue Qi
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Jiancheng Xu
- Anorectal Surgery Department, The First Affiliated Hospital of Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Yongkang An
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Wencong Xu
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, China
- Anorectal Surgery Department, The First Affiliated Hospital of Hebei University of Chinese Medicine, Shijiazhuang, China
| |
Collapse
|
21
|
El Baassiri MG, Raouf Z, Jang HS, Scheese D, Duess JW, Fulton WB, Sodhi CP, Hackam DJ, Nasr IW. Ccr2-dependent monocytes exacerbate intestinal inflammation and modulate gut serotonergic signaling following traumatic brain injury. J Trauma Acute Care Surg 2024; 97:356-364. [PMID: 38189659 DOI: 10.1097/ta.0000000000004246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
BACKGROUND Traumatic brain injury (TBI) leads to acute gastrointestinal dysfunction and mucosal damage, resulting in feeding intolerance. C-C motif chemokine receptor 2 (Ccr2 + ) monocytes are crucial immune cells that regulate the gut's inflammatory response via the brain-gut axis. Using Ccr2 ko mice, we investigated the intricate interplay between these cells to better elucidate the role of systemic inflammation after TBI. METHODS A murine-controlled cortical impact model was used, and results were analyzed on postinjury days 1 and 3. The experimental groups included (1) sham C57Bl/6 wild type (WT), (2) TBI WT, (3) sham Ccr2 ko , and (4) TBI Ccr2 ko . Mice were euthanized on postinjury days 1 and 3 to harvest the ileum and study intestinal dysfunction and serotonergic signaling using a combination of quantitative real-time polymerase chain reaction, immunohistochemistry, fluorescein isothiocyanate-dextran motility assays, and flow cytometry. Student's t test and one-way analysis of variance were used for statistical analysis, with significance achieved when p < 0.05. RESULTS Traumatic brain injury resulted in severe dysfunction and dysmotility of the small intestine in WT mice as established by significant upregulation of inflammatory cytokines iNOS , Lcn2 , TNFα , and IL1β and the innate immunity receptor toll-like receptor 4 ( Tlr4 ). This was accompanied by disruption of genes related to serotonin synthesis and degradation. Notably, Ccr2 ko mice subjected to TBI showed substantial improvements in intestinal pathology. Traumatic brain injury Ccr2 ko groups demonstrated reduced expression of inflammatory mediators ( iNOS , Lcn2 , IL1β , and Tlr4 ) and improvement in serotonin synthesis genes, including tryptophan hydroxylase 1 ( Tph1 ) and dopa decarboxylase ( Ddc ). CONCLUSION Our study reveals a critical role for Ccr2 + monocytes in modulating intestinal homeostasis after TBI. Ccr2 + monocytes aggravate intestinal inflammation and alter gut-derived serotonergic signaling. Therefore, targeting Ccr2 + monocyte-dependent responses could provide a better understanding of TBI-induced gut inflammation. Further studies are required to elucidate the impact of these changes on brain neuroinflammation and cognitive outcomes.
Collapse
Affiliation(s)
- Mahmoud G El Baassiri
- From the Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Momen YS, Mishra J, Kumar N. Brain-Gut and Microbiota-Gut-Brain Communication in Type-2 Diabetes Linked Alzheimer's Disease. Nutrients 2024; 16:2558. [PMID: 39125436 PMCID: PMC11313915 DOI: 10.3390/nu16152558] [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: 05/27/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 08/12/2024] Open
Abstract
The gastrointestinal (GI) tract, home to the largest microbial population in the human body, plays a crucial role in overall health through various mechanisms. Recent advancements in research have revealed the potential implications of gut-brain and vice-versa communication mediated by gut-microbiota and their microbial products in various diseases including type-2 diabetes and Alzheimer's disease (AD). AD is the most common type of dementia where most of cases are sporadic with no clearly identified cause. However, multiple factors are implicated in the progression of sporadic AD which can be classified as non-modifiable (e.g., genetic) and modifiable (e.g. Type-2 diabetes, diet etc.). Present review focusses on key players particularly the modifiable factors such as Type-2 diabetes (T2D) and diet and their implications in microbiota-gut-brain (MGB) and brain-gut (BG) communication and cognitive functions of healthy brain and their dysfunction in Alzheimer's Disease. Special emphasis has been given on elucidation of the mechanistic aspects of the impact of diet on gut-microbiota and the implications of some of the gut-microbial products in T2D and AD pathology. For example, mechanistically, HFD induces gut dysbiosis with driven metabolites that in turn cause loss of integrity of intestinal barrier with concomitant colonic and systemic chronic low-grade inflammation, associated with obesity and T2D. HFD-induced obesity and T2D parallel neuroinflammation, deposition of Amyloid β (Aβ), and ultimately cognitive impairment. The review also provides a new perspective of the impact of diet on brain-gut and microbiota-gut-brain communication in terms of transcription factors as a commonly spoken language that may facilitates the interaction between gut and brain of obese diabetic patients who are at a higher risk of developing cognitive impairment and AD. Other commonality such as tyrosine kinase expression and functions maintaining intestinal integrity on one hand and the phagocytic clarence by migratory microglial functions in brain are also discussed. Lastly, the characterization of the key players future research that might shed lights on novel potential pharmacological target to impede AD progression are also discussed.
Collapse
Affiliation(s)
| | | | - Narendra Kumar
- Department of Pharmaceutical Sciences, ILR College of Pharmacy, Texas A&M Health Science Center, Kingsville, TX 78363, USA
| |
Collapse
|
23
|
Jung YH, Chae CW, Han HJ. The potential role of gut microbiota-derived metabolites as regulators of metabolic syndrome-associated mitochondrial and endolysosomal dysfunction in Alzheimer's disease. Exp Mol Med 2024; 56:1691-1702. [PMID: 39085351 PMCID: PMC11372123 DOI: 10.1038/s12276-024-01282-3] [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: 03/12/2024] [Revised: 04/20/2024] [Accepted: 05/10/2024] [Indexed: 08/02/2024] Open
Abstract
Although the role of gut microbiota (GMB)-derived metabolites in mitochondrial and endolysosomal dysfunction in Alzheimer's disease (AD) under metabolic syndrome remains unclear, deciphering these host-metabolite interactions represents a major public health challenge. Dysfunction of mitochondria and endolysosomal networks (ELNs) plays a crucial role in metabolic syndrome and can exacerbate AD progression, highlighting the need to study their reciprocal regulation for a better understanding of how AD is linked to metabolic syndrome. Concurrently, metabolic disorders are associated with alterations in the composition of the GMB. Recent evidence suggests that changes in the composition of the GMB and its metabolites may be involved in AD pathology. This review highlights the mechanisms of metabolic syndrome-mediated AD development, focusing on the interconnected roles of mitochondrial dysfunction, ELN abnormalities, and changes in the GMB and its metabolites. We also discuss the pathophysiological role of GMB-derived metabolites, including amino acids, fatty acids, other metabolites, and extracellular vesicles, in mediating their effects on mitochondrial and ELN dysfunction. Finally, this review proposes therapeutic strategies for AD by directly modulating mitochondrial and ELN functions through targeting GMB metabolites under metabolic syndrome.
Collapse
Affiliation(s)
- Young Hyun Jung
- Department of Physiology, College of Medicine, Soonchunhyang University, Cheonan, 31151, Korea
| | - Chang Woo Chae
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 FOUR Future Veterinary Medicine Leading Education & Research Center, Seoul National University, Seoul, South Korea
| | - Ho Jae Han
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 FOUR Future Veterinary Medicine Leading Education & Research Center, Seoul National University, Seoul, South Korea.
| |
Collapse
|
24
|
Dunham SJB, Avelar‐Barragan J, Rothman JA, Adams ED, Faraci G, Forner S, Kawauchi S, Tenner AJ, Green KN, LaFerla FM, MacGregor GR, Mapstone M, Whiteson KL. Sex-specific associations between AD genotype and the microbiome of human amyloid beta knock-in (hAβ-KI) mice. Alzheimers Dement 2024; 20:4935-4950. [PMID: 38572865 PMCID: PMC11247698 DOI: 10.1002/alz.13794] [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: 10/24/2023] [Revised: 02/20/2024] [Accepted: 02/23/2024] [Indexed: 04/05/2024]
Abstract
INTRODUCTION Emerging evidence links changes in the gut microbiome to late-onset Alzheimer's disease (LOAD), necessitating examination of AD mouse models with consideration of the microbiome. METHODS We used shotgun metagenomics and untargeted metabolomics to study the human amyloid beta knock-in (hAβ-KI) murine model for LOAD compared to both wild-type (WT) mice and a model for early-onset AD (3xTg-AD). RESULTS Eighteen-month female (but not male) hAβ-KI microbiomes were distinct from WT microbiomes, with AD genotype accounting for 18% of the variance by permutational multivariate analysis of variance (PERMANOVA). Metabolomic diversity differences were observed in females, however no individual metabolites were differentially abundant. hAβ-KI mice microbiomes were distinguishable from 3xTg-AD animals (81% accuracy by random forest modeling), with separation primarily driven by Romboutsia ilealis and Turicibacter species. Microbiomes were highly cage specific, with cage assignment accounting for more than 40% of the PERMANOVA variance between the groups. DISCUSSION These findings highlight a sex-dependent variation in the microbiomes of hAβ-KI mice and underscore the importance of considering the microbiome when designing studies that use murine models for AD. HIGHLIGHTS Microbial diversity and the abundance of several species differed in human amyloid beta knock-in (hAβ-KI) females but not males. Correlations to Alzheimer's disease (AD) genotype were stronger for the microbiome than the metabolome. Microbiomes from hAβ-KI mice were distinct from 3xTg-AD mice. Cage effects accounted for most of the variance in the microbiome and metabolome.
Collapse
Affiliation(s)
- Sage J. B. Dunham
- Department of Molecular Biology & BiochemistryUniversity of California IrvineIrvineCaliforniaUSA
| | - Julio Avelar‐Barragan
- Department of Molecular Biology & BiochemistryUniversity of California IrvineIrvineCaliforniaUSA
| | - Jason A. Rothman
- Department of Molecular Biology & BiochemistryUniversity of California IrvineIrvineCaliforniaUSA
| | - Eric D. Adams
- Department of Molecular Biology & BiochemistryUniversity of California IrvineIrvineCaliforniaUSA
| | - Gina Faraci
- Department of Molecular Biology & BiochemistryUniversity of California IrvineIrvineCaliforniaUSA
| | - Stefania Forner
- Institute for Memory Impairments and Neurological Disorders (UCI MIND)University of California IrvineIrvineCaliforniaUSA
| | - Shimako Kawauchi
- Department of Anatomy and NeurobiologyDevelopmental Biology CenterUniversity of California IrvineCollege of MedicineIrvineCaliforniaUSA
| | - Andrea J. Tenner
- Department of Molecular Biology & BiochemistryDepartment of Neurobiology and BehaviorDepartment of Pathology and Laboratory MedicineSchool of MedicineInstitute for Memory Impairments and Neurological Disorders (UCI MIND)University of California IrvineIrvineCaliforniaUSA
| | - Kim N. Green
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), Department of Neurobiology and BehaviorSchool of Biological SciencesCenter for Neural Circuit MappingUniversity of California IrvineIrvineCaliforniaUSA
| | - Frank M. LaFerla
- Institute for Memory Impairments and Neurological Disorders (UCI MIND)Department of Neurobiology and BehaviorUniversity of California IrvineIrvineCaliforniaUSA
| | - Grant R. MacGregor
- Department of Developmental and Cell BiologyUniversity of California IrvineIrvineCaliforniaUSA
| | - Mark Mapstone
- Department of NeurologyUniversity of California IrvineIrvineCaliforniaUSA
| | - Katrine L. Whiteson
- Department of Molecular Biology & BiochemistryUniversity of California IrvineIrvineCaliforniaUSA
| |
Collapse
|
25
|
Hernández-Rodríguez JF, Trachioti MG, Hrbac J, Rojas D, Escarpa A, Prodromidis MI. Spark-Discharge-Activated 3D-Printed Electrochemical Sensors. Anal Chem 2024; 96:10127-10133. [PMID: 38867513 PMCID: PMC11209655 DOI: 10.1021/acs.analchem.4c01249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/11/2024] [Accepted: 06/04/2024] [Indexed: 06/14/2024]
Abstract
3D printing technology is a tremendously powerful technology to fabricate electrochemical sensing devices. However, current conductive filaments are not aimed at electrochemical applications and therefore require intense activation protocols to unleash a suitable electrochemical performance. Current activation methods based on (electro)chemical activation (using strong alkaline solutions and organic solvents and/or electrochemical treatments) or combined approaches are time-consuming and require hazardous chemicals and dedicated operator intervention. Here, pioneering spark-discharge-activated 3D-printed electrodes were developed and characterized, and it was demonstrated that their electrochemical performance was greatly improved by the effective removal of the thermoplastic support polylactic acid (PLA) as well as the formation of sponge-like and low-dimensional carbon nanostructures. This reagent-free approach consists of a direct, fast, and automatized spark discharge between the 3D-electrode and the respective graphite pencil electrode tip using a high-voltage power supply. Activated electrodes were challenged toward the simultaneous voltammetric determination of dopamine (DP) and serotonin (5-HT) in cell culture media. Spark discharge has been demonstrated as a promising approach for conductive filament activation as it is a fast, green (0.94 GREEnness Metric Approach), and automatized procedure that can be integrated into the 3D printing pipeline.
Collapse
Affiliation(s)
- Juan F. Hernández-Rodríguez
- Department
of Analytical Chemistry, Physical Chemistry and Chemical Engineering, University of Alcalá, Alcalá de Henares 28802, Madrid, Spain
| | - Maria G. Trachioti
- Department
of Chemistry, University of Ioannina, 45 110 Ioannina, Greece
| | - Jan Hrbac
- Department
of Chemistry, Masaryk University, 625 00 Brno, Czech Republic
| | - Daniel Rojas
- Department
of Analytical Chemistry, Physical Chemistry and Chemical Engineering, University of Alcalá, Alcalá de Henares 28802, Madrid, Spain
| | - Alberto Escarpa
- Department
of Analytical Chemistry, Physical Chemistry and Chemical Engineering, University of Alcalá, Alcalá de Henares 28802, Madrid, Spain
- Chemical
Research Institute “Andres M. Del Rio”, University of Alcalá, Alcalá
de Henares 28802, Madrid, Spain
| | | |
Collapse
|
26
|
Guo J, Cao Y, Zhang T, Xu C, Liu Z, Li W, Wang Q. Multisensory Fusion Training and 7, 8-Dihydroxyflavone Improve Amyloid-β-Induced Cognitive Impairment, Anxiety, and Depression-Like Behavior in Mice Through Multiple Mechanisms. Neuropsychiatr Dis Treat 2024; 20:1247-1270. [PMID: 38883414 PMCID: PMC11180438 DOI: 10.2147/ndt.s459891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 06/05/2024] [Indexed: 06/18/2024] Open
Abstract
Background There is growing interest in the role of physical activity in patients with of Alzheimer's disease (AD), particularly regarding its impact of cognitive function, gut microbiota, metabolites, and neurotrophic factors. Objective To investigate the impact of multisensory fusion training (MSFT) combined with 7, 8-dihydroxyflavone (DHF) on the behavioral characteristics, protein expression, microbiome, and serum metabolome using the AD model in mice induced with amyloid-β (Aβ). Methods We assessed cognitive ability, anxiety-like and depression-like behaviors in Aβ mice using behavioral measures. Western blotting was employed to detect the expression of relevant proteins. The 16S rRNA gene sequencing and metabolomics were used to analyze changes in the intestinal microbial composition and serum metabolic profile, respectively, of Aβ mice. Results The behavioral outcomes indicated that a 4-week intervention combining DHF and MSFT yielded remarkable improvements in cognitive function and reduced anxiety and depression-like behaviors in Aβ mice. In the hippocampus of Aβ mice, the combined intervention increased the levels of BDNF, VGF, PSD-95, Nrf2, p-GSK3β and p-CREB proteins. Analyses of sequence and metabolomic data revealed that Bacteroides and Ruminococcaceae were remarkably more abundant following the combined intervention, influencing the expression of specific metabolites directly linked to the maintenance of neuronal and neurobehavioral functions. These metabolites play a crucial role in vital processes, such as amino acid metabolism, lipid metabolism, and neurotransmitter metabolism in mice. Conclusion Our study highlighted that MSFT combined with DHF improves cognitive impairment, anxiety, and depression-like behavior in Aβ mice through multiple mechanisms, and further validated the correlation between the gut microbiome and serum metabolome. These findings open up a promising avenue for future investigations into potential treatment strategies for AD.
Collapse
Affiliation(s)
- Jiejie Guo
- Zhejiang Key Laboratory of Pathophysiology, NBU Health Science Center, Ningbo University, Ningbo, People's Republic of China
- Department of Clinical Laboratory, The First People's Hospital of Wenling, Taizhou, People's Republic of China
- Department of Neurology, The First People's Hospital of Wenling, Taizhou, People's Republic of China
| | - Yanzi Cao
- Zhejiang Key Laboratory of Pathophysiology, NBU Health Science Center, Ningbo University, Ningbo, People's Republic of China
| | - Ting Zhang
- Department of Clinical Laboratory, The First People's Hospital of Wenling, Taizhou, People's Republic of China
| | - Chunshuang Xu
- Zhejiang Key Laboratory of Pathophysiology, NBU Health Science Center, Ningbo University, Ningbo, People's Republic of China
| | - Zhitao Liu
- Zhejiang Key Laboratory of Pathophysiology, NBU Health Science Center, Ningbo University, Ningbo, People's Republic of China
- Fujian Normal University, Fuzhou, People's Republic of China
| | - Wanyi Li
- Zhejiang Key Laboratory of Pathophysiology, NBU Health Science Center, Ningbo University, Ningbo, People's Republic of China
| | - Qinwen Wang
- Zhejiang Key Laboratory of Pathophysiology, NBU Health Science Center, Ningbo University, Ningbo, People's Republic of China
| |
Collapse
|
27
|
Sun Z, Aschalew ND, Cheng L, Xia Y, Zhang L, Yin G, Wang S, Wang Z, Dong J, Zhang W, Zhao W, Qin G, Zhang X, Zhong R, Wang T, Zhen Y. Dietary 5-hydroxytryptophan improves sheep growth performance by enhancing ruminal functions, antioxidant capacity, and tryptophan metabolism: in vitro and in vivo studies. Front Immunol 2024; 15:1398310. [PMID: 38835767 PMCID: PMC11148369 DOI: 10.3389/fimmu.2024.1398310] [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: 03/09/2024] [Accepted: 05/03/2024] [Indexed: 06/06/2024] Open
Abstract
Background Hydroxytryptophan (5-HTP) can regulate the synthesis of 5-Hydroxytryptamine (5-HT) and melatonin (MT). In a previous metabolome analysis, we found that 5-HTP is an effective ingredient in yeast culture for regulating rumen fermentation. However, research on the effect of this microbial product (5-HTP) as a functional feed additive in sheep production is still not well explained. Therefore, this study examined the effects of 5-HTP on sheep rumen function and growth performance using in vitro and in vivo models. Methods A two-factor in vitro experiment involving different 5-HTP doses and fermentation times was conducted. Then, in the in vivo experiment, 10 sheep were divided into a control group which was fed a basal diet, and a 5-HTP group supplemented with 8 mg/kg 5-HTP for 60 days. Results The results showed that 5-HTP supplementation had a significant effect on in vitro DMD, pH, NH3-N, acetic acid, propionic acid, and TVFA concentrations. 5-HTP altered rumen bacteria composition and diversity indices including Chao1, Shannon, and Simpson. Moreover, the in vivo study on sheep confirmed that supplementing with 8 mg/kg of 5-HTP improved rumen fermentation efficiency and microbial composition. This led to enhanced sheep growth performance and increased involvement in the tryptophan metabolic pathway, suggesting potential benefits. Conclusion Dietary 5-HTP (8 mg/kg DM) improves sheep growth performance by enhancing ruminal functions, antioxidant capacity, and tryptophan metabolism. This study can provide a foundation for the development of 5-HTP as a functional feed additive in ruminants' production.
Collapse
Affiliation(s)
- Zhe Sun
- Jilin Agricultural University (JLAU)-Borui Dairy Science and Technology R&D Center, Key Laboratory of Animal Nutrition and Feed Science of Jilin Province, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Key Laboratory of Animal Production Product Quality and Security Ministry of Education, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- College of Life Sciences, Engineering Research Center of Bioreactor and Pharmaceutical Development, Ministry of Education, Jilin Agricultural University, Changchun, China
- Postdoctoral Scientific Research Workstation, Feed Engineering Technology Research Center of Jilin Province, Changchun Borui Science & Technology Co. Ltd., Changchun, China
| | - Natnael D Aschalew
- Jilin Agricultural University (JLAU)-Borui Dairy Science and Technology R&D Center, Key Laboratory of Animal Nutrition and Feed Science of Jilin Province, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Key Laboratory of Animal Production Product Quality and Security Ministry of Education, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- College of Agriculture and Environmental Science, Dilla University, Dilla, Ethiopia
| | - Long Cheng
- Jilin Agricultural University (JLAU)-Borui Dairy Science and Technology R&D Center, Key Laboratory of Animal Nutrition and Feed Science of Jilin Province, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Key Laboratory of Animal Production Product Quality and Security Ministry of Education, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Yuanhong Xia
- Jilin Agricultural University (JLAU)-Borui Dairy Science and Technology R&D Center, Key Laboratory of Animal Nutrition and Feed Science of Jilin Province, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Key Laboratory of Animal Production Product Quality and Security Ministry of Education, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Longyu Zhang
- Jilin Agricultural University (JLAU)-Borui Dairy Science and Technology R&D Center, Key Laboratory of Animal Nutrition and Feed Science of Jilin Province, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Key Laboratory of Animal Production Product Quality and Security Ministry of Education, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Guopei Yin
- Jilin Agricultural University (JLAU)-Borui Dairy Science and Technology R&D Center, Key Laboratory of Animal Nutrition and Feed Science of Jilin Province, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Key Laboratory of Animal Production Product Quality and Security Ministry of Education, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Shikun Wang
- College of Life Sciences, Engineering Research Center of Bioreactor and Pharmaceutical Development, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Ziyuan Wang
- Jilin Agricultural University (JLAU)-Borui Dairy Science and Technology R&D Center, Key Laboratory of Animal Nutrition and Feed Science of Jilin Province, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Key Laboratory of Animal Production Product Quality and Security Ministry of Education, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Jianan Dong
- Jilin Agricultural University (JLAU)-Borui Dairy Science and Technology R&D Center, Key Laboratory of Animal Nutrition and Feed Science of Jilin Province, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Key Laboratory of Animal Production Product Quality and Security Ministry of Education, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Weigang Zhang
- Postdoctoral Scientific Research Workstation, Feed Engineering Technology Research Center of Jilin Province, Changchun Borui Science & Technology Co. Ltd., Changchun, China
| | - Wei Zhao
- Postdoctoral Scientific Research Workstation, Feed Engineering Technology Research Center of Jilin Province, Changchun Borui Science & Technology Co. Ltd., Changchun, China
| | - Guixin Qin
- Jilin Agricultural University (JLAU)-Borui Dairy Science and Technology R&D Center, Key Laboratory of Animal Nutrition and Feed Science of Jilin Province, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Key Laboratory of Animal Production Product Quality and Security Ministry of Education, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Xuefeng Zhang
- Jilin Agricultural University (JLAU)-Borui Dairy Science and Technology R&D Center, Key Laboratory of Animal Nutrition and Feed Science of Jilin Province, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Key Laboratory of Animal Production Product Quality and Security Ministry of Education, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Rongzhen Zhong
- Jilin Province Feed Processing and Ruminant Precision Breeding Cross-Regional Cooperation Technology Innovation Center, Jilin Provincial Key Laboratory of Grassland Farming, Northeast Institute of Geography and Agroecology, Chinese Academy of Sciences, Changchun, China
| | - Tao Wang
- Jilin Agricultural University (JLAU)-Borui Dairy Science and Technology R&D Center, Key Laboratory of Animal Nutrition and Feed Science of Jilin Province, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Key Laboratory of Animal Production Product Quality and Security Ministry of Education, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Postdoctoral Scientific Research Workstation, Feed Engineering Technology Research Center of Jilin Province, Changchun Borui Science & Technology Co. Ltd., Changchun, China
| | - Yuguo Zhen
- Jilin Agricultural University (JLAU)-Borui Dairy Science and Technology R&D Center, Key Laboratory of Animal Nutrition and Feed Science of Jilin Province, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Key Laboratory of Animal Production Product Quality and Security Ministry of Education, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Postdoctoral Scientific Research Workstation, Feed Engineering Technology Research Center of Jilin Province, Changchun Borui Science & Technology Co. Ltd., Changchun, China
| |
Collapse
|
28
|
El Baassiri MG, Raouf Z, Badin S, Escobosa A, Sodhi CP, Nasr IW. Dysregulated brain-gut axis in the setting of traumatic brain injury: review of mechanisms and anti-inflammatory pharmacotherapies. J Neuroinflammation 2024; 21:124. [PMID: 38730498 PMCID: PMC11083845 DOI: 10.1186/s12974-024-03118-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 04/30/2024] [Indexed: 05/13/2024] Open
Abstract
Traumatic brain injury (TBI) is a chronic and debilitating disease, associated with a high risk of psychiatric and neurodegenerative diseases. Despite significant advancements in improving outcomes, the lack of effective treatments underscore the urgent need for innovative therapeutic strategies. The brain-gut axis has emerged as a crucial bidirectional pathway connecting the brain and the gastrointestinal (GI) system through an intricate network of neuronal, hormonal, and immunological pathways. Four main pathways are primarily implicated in this crosstalk, including the systemic immune system, autonomic and enteric nervous systems, neuroendocrine system, and microbiome. TBI induces profound changes in the gut, initiating an unrestrained vicious cycle that exacerbates brain injury through the brain-gut axis. Alterations in the gut include mucosal damage associated with the malabsorption of nutrients/electrolytes, disintegration of the intestinal barrier, increased infiltration of systemic immune cells, dysmotility, dysbiosis, enteroendocrine cell (EEC) dysfunction and disruption in the enteric nervous system (ENS) and autonomic nervous system (ANS). Collectively, these changes further contribute to brain neuroinflammation and neurodegeneration via the gut-brain axis. In this review article, we elucidate the roles of various anti-inflammatory pharmacotherapies capable of attenuating the dysregulated inflammatory response along the brain-gut axis in TBI. These agents include hormones such as serotonin, ghrelin, and progesterone, ANS regulators such as beta-blockers, lipid-lowering drugs like statins, and intestinal flora modulators such as probiotics and antibiotics. They attenuate neuroinflammation by targeting distinct inflammatory pathways in both the brain and the gut post-TBI. These therapeutic agents exhibit promising potential in mitigating inflammation along the brain-gut axis and enhancing neurocognitive outcomes for TBI patients.
Collapse
Affiliation(s)
- Mahmoud G El Baassiri
- Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Zachariah Raouf
- Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Sarah Badin
- Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Alejandro Escobosa
- Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Chhinder P Sodhi
- Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Isam W Nasr
- Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
| |
Collapse
|
29
|
Tang X, Guo Z, Chen G, Sun S, Xiao S, Chen P, Tang G, Huang L, Wang Y. A Multimodal Meta-Analytical Evidence of Functional and Structural Brain Abnormalities Across Alzheimer's Disease Spectrum. Ageing Res Rev 2024; 95:102240. [PMID: 38395200 DOI: 10.1016/j.arr.2024.102240] [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: 01/05/2024] [Accepted: 02/18/2024] [Indexed: 02/25/2024]
Abstract
BACKGROUND Numerous neuroimaging studies have reported that Alzheimer's disease (AD) spectrum have been linked to alterations in intrinsic functional activity and cortical thickness (CT) of some brain areas. However, the findings have been inconsistent and the correlation with the transcriptional profile and neurotransmitter systems remain largely unknown. METHODS We conducted a meta-analysis to identify multimodal differences in the amplitude of low-frequency fluctuation (ALFF)/fractional ALFF (fALFF) and CT in patients with AD and preclinical AD compared to healthy controls (HCs), using the Seed-based d Mapping with Permutation of Subject Images software. Transcriptional data were retrieved from the Allen Human Brain Atlas. The atlas-based nuclear imaging-derived neurotransmitter maps were investigated by JuSpace toolbox. RESULTS We included 26 ALFF/fALFF studies comprising 884 patients with AD and 1,020 controls, along with 52 studies comprising 2,046 patients with preclinical AD and 2,336 controls. For CT, we included 11 studies comprising 353 patients with AD and 330 controls. Overall, compared to HCs, patients with AD showed decreased ALFF/fALFF in the bilateral posterior cingulate gyrus (PCC)/precuneus and right angular gyrus, as well as increased ALFF/fALFF in the bilateral parahippocampal gyrus (PHG). Patients with peclinical AD showed decreased ALFF/fALFF in the left precuneus. Additionally, patients with AD displayed decreased CT in the bilateral PHG, left PCC, bilateral orbitofrontal cortex, sensorimotor areas and temporal lobe. Furthermore, gene sets related to brain structural and functional changes in AD and preclincal AD were enriched for G protein-coupled receptor signaling pathway, ion gated channel activity, and components of biological membrane. Functional and structural alterations in AD and preclinical AD were spatially associated with dopaminergic, serotonergic, and GABAergic neurotransmitter systems. CONCLUSIONS The multimodal meta-analysis demonstrated that patients with AD exhibited convergent functional and structural alterations in the PCC/precuneus and PHG, as well as cortical thinning in the primary sensory and motor areas. Furthermore, patients with preclinical AD showed reduced functional activity in the precuneus. AD and preclinical AD showed genetic modulations/neurotransmitter deficits of brain functional and structural impairments. These findings may provide new insights into the pathophysiology of the AD spectrum.
Collapse
Affiliation(s)
- Xinyue Tang
- Medical Imaging Center, First Affiliated Hospital of Jinan University, Guangzhou 510630, China; Institute of Molecular and Functional Imaging, Jinan University, Guangzhou 510630, China
| | - Zixuan Guo
- Medical Imaging Center, First Affiliated Hospital of Jinan University, Guangzhou 510630, China; Institute of Molecular and Functional Imaging, Jinan University, Guangzhou 510630, China
| | - Guanmao Chen
- Medical Imaging Center, First Affiliated Hospital of Jinan University, Guangzhou 510630, China; Institute of Molecular and Functional Imaging, Jinan University, Guangzhou 510630, China
| | - Shilin Sun
- Medical Imaging Center, First Affiliated Hospital of Jinan University, Guangzhou 510630, China; Institute of Molecular and Functional Imaging, Jinan University, Guangzhou 510630, China
| | - Shu Xiao
- Medical Imaging Center, First Affiliated Hospital of Jinan University, Guangzhou 510630, China; Institute of Molecular and Functional Imaging, Jinan University, Guangzhou 510630, China
| | - Pan Chen
- Medical Imaging Center, First Affiliated Hospital of Jinan University, Guangzhou 510630, China; Institute of Molecular and Functional Imaging, Jinan University, Guangzhou 510630, China
| | - Guixian Tang
- Medical Imaging Center, First Affiliated Hospital of Jinan University, Guangzhou 510630, China; Institute of Molecular and Functional Imaging, Jinan University, Guangzhou 510630, China
| | - Li Huang
- Medical Imaging Center, First Affiliated Hospital of Jinan University, Guangzhou 510630, China; Institute of Molecular and Functional Imaging, Jinan University, Guangzhou 510630, China
| | - Ying Wang
- Medical Imaging Center, First Affiliated Hospital of Jinan University, Guangzhou 510630, China; Institute of Molecular and Functional Imaging, Jinan University, Guangzhou 510630, China.
| |
Collapse
|
30
|
Zhuang H, Cao X, Tang X, Zou Y, Yang H, Liang Z, Yan X, Chen X, Feng X, Shen L. Investigating metabolic dysregulation in serum of triple transgenic Alzheimer's disease male mice: implications for pathogenesis and potential biomarkers. Amino Acids 2024; 56:10. [PMID: 38315232 PMCID: PMC10844422 DOI: 10.1007/s00726-023-03375-1] [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: 05/15/2023] [Accepted: 11/11/2023] [Indexed: 02/07/2024]
Abstract
Alzheimer's disease (AD) is a multifactorial neurodegenerative disease that lacks convenient and accessible peripheral blood diagnostic markers and effective drugs. Metabolic dysfunction is one of AD risk factors, which leaded to alterations of various metabolites in the body. Pathological changes of the brain can be reflected in blood metabolites that are expected to explain the disease mechanisms or be candidate biomarkers. The aim of this study was to investigate the changes of targeted metabolites within peripheral blood of AD mouse model, with the purpose of exploring the disease mechanism and potential biomarkers. Targeted metabolomics was used to quantify 256 metabolites in serum of triple transgenic AD (3 × Tg-AD) male mice. Compared with controls, 49 differential metabolites represented dysregulation in purine, pyrimidine, tryptophan, cysteine and methionine and glycerophospholipid metabolism. Among them, adenosine, serotonin, N-acetyl-5-hydroxytryptamine, and acetylcholine play a key role in regulating neural transmitter network. The alteration of S-adenosine-L-homocysteine, S-adenosine-L-methionine, and trimethylamine-N-oxide in AD mice serum can served as indicator of AD risk. The results revealed the changes of metabolites in serum, suggesting that metabolic dysregulation in periphery in AD mice may be related to the disturbances in neuroinhibition, the serotonergic system, sleep function, the cholinergic system, and the gut microbiota. This study provides novel insights into the dysregulation of several key metabolites and metabolic pathways in AD, presenting potential avenues for future research and the development of peripheral biomarkers.
Collapse
Affiliation(s)
- Hongbin Zhuang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Xueshan Cao
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Xiaoxiao Tang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Yongdong Zou
- Center for Instrumental Analysis, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Hongbo Yang
- Center for Instrumental Analysis, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Zhiyuan Liang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Xi Yan
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025, People's Republic of China
| | - Xiaolu Chen
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025, People's Republic of China
| | - Xingui Feng
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Liming Shen
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China.
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, People's Republic of China.
| |
Collapse
|
31
|
Yue M, Zhang L. Exploring the Mechanistic Interplay between Gut Microbiota and Precocious Puberty: A Narrative Review. Microorganisms 2024; 12:323. [PMID: 38399733 PMCID: PMC10892899 DOI: 10.3390/microorganisms12020323] [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: 01/02/2024] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
The gut microbiota has been implicated in the context of sexual maturation during puberty, with discernible differences in its composition before and after this critical developmental stage. Notably, there has been a global rise in the prevalence of precocious puberty in recent years, particularly among girls, where approximately 90% of central precocious puberty cases lack a clearly identifiable cause. While a link between precocious puberty and the gut microbiota has been observed, the precise causality and underlying mechanisms remain elusive. This narrative review aims to systematically elucidate the potential mechanisms that underlie the intricate relationship between the gut microbiota and precocious puberty. Potential avenues of exploration include investigating the impact of the gut microbiota on endocrine function, particularly in the regulation of hormones, such as gonadotropin-releasing hormone (GnRH), luteinizing hormone (LH), and follicle-stimulating hormone (FSH). Additionally, this review will delve into the intricate interplay between the gut microbiome, metabolism, and obesity, considering the known association between obesity and precocious puberty. This review will also explore how the microbiome's involvement in nutrient metabolism could impact precocious puberty. Finally, attention is given to the microbiota's ability to produce neurotransmitters and neuroactive compounds, potentially influencing the central nervous system components involved in regulating puberty. By exploring these mechanisms, this narrative review seeks to identify unexplored targets and emerging directions in understanding the role of the gut microbiome in relation to precocious puberty. The ultimate goal is to provide valuable insights for the development of non-invasive diagnostic methods and innovative therapeutic strategies for precocious puberty in the future, such as specific probiotic therapy.
Collapse
Affiliation(s)
- Min Yue
- Microbiome-X, National Institute of Health Data Science of China & Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Lei Zhang
- Microbiome-X, National Institute of Health Data Science of China & Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| |
Collapse
|
32
|
Dissanayaka DMS, Jayasena V, Rainey-Smith SR, Martins RN, Fernando WMADB. The Role of Diet and Gut Microbiota in Alzheimer's Disease. Nutrients 2024; 16:412. [PMID: 38337696 PMCID: PMC10857293 DOI: 10.3390/nu16030412] [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: 12/30/2023] [Revised: 01/22/2024] [Accepted: 01/26/2024] [Indexed: 02/12/2024] Open
Abstract
Alzheimer's disease (AD), the most prevalent form of dementia, is characterized by the accumulation of amyloid-beta (Aβ) plaques and hyperphosphorylated tau tangles. Currently, Alzheimer's disease (AD) impacts 50 million individuals, with projections anticipating an increase to 152 million by the year 2050. Despite the increasing global prevalence of AD, its underlying pathology remains poorly understood, posing challenges for early diagnosis and treatment. Recent research suggests a link between gut dysbiosis and the aggregation of Aβ, the development of tau proteins, and the occurrence of neuroinflammation and oxidative stress are associated with AD. However, investigations into the gut-brain axis (GBA) in the context of AD progression and pathology have yielded inconsistent findings. This review aims to enhance our understanding of microbial diversity at the species level and the role of these species in AD pathology. Additionally, this review addresses the influence of confounding elements, including diet, probiotics, and prebiotics, on AD throughout different stages (preclinical, mild cognitive impairment (MCI), and AD) of its progression.
Collapse
Affiliation(s)
- D. M. Sithara Dissanayaka
- Centre of Excellence for Alzheimer’s Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia; (D.M.S.D.); (S.R.R.-S.); (R.N.M.)
- Alzheimer’s Research Australia, Ralph and Patricia Sarich Neuroscience Research Institute, Nedlands, WA 6009, Australia
| | - Vijay Jayasena
- School of Science and Health, Western Sydney University, M15, Rm. G54, Locked Bag 1797, Penrith, NSW 2751, Australia;
| | - Stephanie R. Rainey-Smith
- Centre of Excellence for Alzheimer’s Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia; (D.M.S.D.); (S.R.R.-S.); (R.N.M.)
- Alzheimer’s Research Australia, Ralph and Patricia Sarich Neuroscience Research Institute, Nedlands, WA 6009, Australia
- Centre for Healthy Aging, Murdoch University, Murdoch, WA 6150, Australia
| | - Ralph N. Martins
- Centre of Excellence for Alzheimer’s Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia; (D.M.S.D.); (S.R.R.-S.); (R.N.M.)
- Alzheimer’s Research Australia, Ralph and Patricia Sarich Neuroscience Research Institute, Nedlands, WA 6009, Australia
- Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - W. M. A. D. Binosha Fernando
- Centre of Excellence for Alzheimer’s Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia; (D.M.S.D.); (S.R.R.-S.); (R.N.M.)
- Alzheimer’s Research Australia, Ralph and Patricia Sarich Neuroscience Research Institute, Nedlands, WA 6009, Australia
| |
Collapse
|
33
|
Hayer SS, Conrin M, French JA, Benson AK, Alvarez S, Cooper K, Fischer A, Alsafwani ZW, Gasper W, Suhr Van Haute MJ, Hassenstab HR, Azadmanesh S, Briardy M, Gerbers S, Jabenis A, Thompson JL, Clayton JB. Antibiotic-induced gut dysbiosis elicits gut-brain axis relevant multi-omic signatures and behavioral and neuroendocrine changes in a nonhuman primate model. Gut Microbes 2024; 16:2305476. [PMID: 38284649 PMCID: PMC10826635 DOI: 10.1080/19490976.2024.2305476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 01/10/2024] [Indexed: 01/30/2024] Open
Abstract
Emerging evidence indicates that antibiotic-induced dysbiosis can play an etiological role in the pathogenesis of neuropsychiatric disorders. However, most of this evidence comes from rodent models. The objective of this study was to evaluate if antibiotic-induced gut dysbiosis can elicit changes in gut metabolites and behavior indicative of gut-brain axis disruption in common marmosets (Callithrix jacchus) - a nonhuman primate model often used to study sociability and stress. We were able to successfully induce dysbiosis in marmosets using a custom antibiotic cocktail (vancomycin, enrofloxacin and neomycin) administered orally for 28 days. This gut dysbiosis altered gut metabolite profiles, behavior, and stress reactivity. Increase in gut Fusobacterium spp. post-antibiotic administration was a novel dysbiotic response and has not been observed in any rodent or human studies to date. There were significant changes in concentrations of several gut metabolites which are either neurotransmitters (e.g., GABA and serotonin) or have been found to be moderators of gut-brain axis communication in rodent models (e.g., short-chain fatty acids and bile acids). There was an increase in affiliative behavior and sociability in antibiotic-administered marmosets, which might be a coping mechanism in response to gut dysbiosis-induced stress. Increase in urinary cortisol levels after multiple stressors provides more definitive proof that this model of dysbiosis may cause disrupted communication between gut and brain in common marmosets. This study is a first attempt to establish common marmosets as a novel model to study the impact of severe gut dysbiosis on gut-brain axis cross-talk and behavior.
Collapse
Affiliation(s)
- Shivdeep S. Hayer
- Department of Biology, University of Nebraska at Omaha, Omaha, NE, USA
- Callitrichid Research Center, University of Nebraska at Omaha, Omaha, NE, USA
- Nebraska Food for Health Center, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Mackenzie Conrin
- Department of Biology, University of Nebraska at Omaha, Omaha, NE, USA
- Callitrichid Research Center, University of Nebraska at Omaha, Omaha, NE, USA
| | - Jeffrey A. French
- Callitrichid Research Center, University of Nebraska at Omaha, Omaha, NE, USA
- Nebraska Food for Health Center, University of Nebraska-Lincoln, Lincoln, NE, USA
- Program in Neuroscience and Behavior, University of Nebraska at Omaha, Omaha, NE, USA
| | - Andrew K. Benson
- Nebraska Food for Health Center, University of Nebraska-Lincoln, Lincoln, NE, USA
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Sophie Alvarez
- Proteomics and Metabolomics Facility, Nebraska Center for Biotechnology, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Kathryn Cooper
- School of Interdisciplinary Informatics, College of Information Science and Technology, University of Nebraska at Omaha, Omaha, NE, USA
| | - Anne Fischer
- Proteomics and Metabolomics Facility, Nebraska Center for Biotechnology, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Zahraa Wajih Alsafwani
- School of Interdisciplinary Informatics, College of Information Science and Technology, University of Nebraska at Omaha, Omaha, NE, USA
| | - William Gasper
- School of Interdisciplinary Informatics, College of Information Science and Technology, University of Nebraska at Omaha, Omaha, NE, USA
| | - Mallory J. Suhr Van Haute
- Nebraska Food for Health Center, University of Nebraska-Lincoln, Lincoln, NE, USA
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Haley R. Hassenstab
- Department of Biology, University of Nebraska at Omaha, Omaha, NE, USA
- Callitrichid Research Center, University of Nebraska at Omaha, Omaha, NE, USA
| | - Shayda Azadmanesh
- Department of Biology, University of Nebraska at Omaha, Omaha, NE, USA
- Callitrichid Research Center, University of Nebraska at Omaha, Omaha, NE, USA
| | - Missy Briardy
- Department of Biology, University of Nebraska at Omaha, Omaha, NE, USA
- Callitrichid Research Center, University of Nebraska at Omaha, Omaha, NE, USA
| | - Skyler Gerbers
- Department of Biology, University of Nebraska at Omaha, Omaha, NE, USA
- Callitrichid Research Center, University of Nebraska at Omaha, Omaha, NE, USA
| | - Aliyah Jabenis
- Department of Biology, University of Nebraska at Omaha, Omaha, NE, USA
- Callitrichid Research Center, University of Nebraska at Omaha, Omaha, NE, USA
| | - Jennifer L. Thompson
- Department of Biology, University of Nebraska at Omaha, Omaha, NE, USA
- Callitrichid Research Center, University of Nebraska at Omaha, Omaha, NE, USA
| | - Jonathan B. Clayton
- Department of Biology, University of Nebraska at Omaha, Omaha, NE, USA
- Callitrichid Research Center, University of Nebraska at Omaha, Omaha, NE, USA
- Nebraska Food for Health Center, University of Nebraska-Lincoln, Lincoln, NE, USA
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, USA
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
- Primate Microbiome Project, University of Nebraska-Lincoln, Lincoln, NE, USA
| |
Collapse
|
34
|
Yan M, Man S, Sun B, Ma L, Guo L, Huang L, Gao W. Gut liver brain axis in diseases: the implications for therapeutic interventions. Signal Transduct Target Ther 2023; 8:443. [PMID: 38057297 PMCID: PMC10700720 DOI: 10.1038/s41392-023-01673-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 09/10/2023] [Accepted: 09/28/2023] [Indexed: 12/08/2023] Open
Abstract
Gut-liver-brain axis is a three-way highway of information interaction system among the gastrointestinal tract, liver, and nervous systems. In the past few decades, breakthrough progress has been made in the gut liver brain axis, mainly through understanding its formation mechanism and increasing treatment strategies. In this review, we discuss various complex networks including barrier permeability, gut hormones, gut microbial metabolites, vagus nerve, neurotransmitters, immunity, brain toxic metabolites, β-amyloid (Aβ) metabolism, and epigenetic regulation in the gut-liver-brain axis. Some therapies containing antibiotics, probiotics, prebiotics, synbiotics, fecal microbiota transplantation (FMT), polyphenols, low FODMAP diet and nanotechnology application regulate the gut liver brain axis. Besides, some special treatments targeting gut-liver axis include farnesoid X receptor (FXR) agonists, takeda G protein-coupled receptor 5 (TGR5) agonists, glucagon-like peptide-1 (GLP-1) receptor antagonists and fibroblast growth factor 19 (FGF19) analogs. Targeting gut-brain axis embraces cognitive behavioral therapy (CBT), antidepressants and tryptophan metabolism-related therapies. Targeting liver-brain axis contains epigenetic regulation and Aβ metabolism-related therapies. In the future, a better understanding of gut-liver-brain axis interactions will promote the development of novel preventative strategies and the discovery of precise therapeutic targets in multiple diseases.
Collapse
Affiliation(s)
- Mengyao Yan
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, 300457, Tianjin, China
| | - Shuli Man
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, 300457, Tianjin, China.
| | - Benyue Sun
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, 300457, Tianjin, China
| | - Long Ma
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, 300457, Tianjin, China
| | - Lanping Guo
- National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, 100700, Beijing, China.
| | - Luqi Huang
- National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, 100700, Beijing, China
| | - Wenyuan Gao
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Weijin Road, 300072, Tianjin, China.
| |
Collapse
|
35
|
Tian J, Du E, Guo L. Mitochondrial Interaction with Serotonin in Neurobiology and Its Implication in Alzheimer's Disease. J Alzheimers Dis Rep 2023; 7:1165-1177. [PMID: 38025801 PMCID: PMC10657725 DOI: 10.3233/adr-230070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 08/16/2023] [Indexed: 12/01/2023] Open
Abstract
Alzheimer's disease (AD) is a lethal neurodegenerative disorder characterized by severe brain pathologies and progressive cognitive decline. While the exact cause of this disease remains unknown, emerging evidence suggests that dysregulation of neurotransmitters contributes to the development of AD pathology and symptoms. Serotonin, a critical neurotransmitter in the brain, plays a pivotal role in regulating various brain processes and is implicated in neurological and psychiatric disorders, including AD. Recent studies have shed light on the interplay between mitochondrial function and serotonin regulation in brain physiology. In AD, there is a deficiency of serotonin, along with impairments in mitochondrial function, particularly in serotoninergic neurons. Additionally, altered activity of mitochondrial enzymes, such as monoamine oxidase, may contribute to serotonin dysregulation in AD. Understanding the intricate relationship between mitochondria and serotonin provides valuable insights into the underlying mechanisms of AD and identifies potential therapeutic targets to restore serotonin homeostasis and alleviate AD symptoms. This review summarizes the recent advancements in unraveling the connection between brain mitochondria and serotonin, emphasizing their significance in AD pathogenesis and underscoring the importance of further research in this area. Elucidating the role of mitochondria in serotonin dysfunction will promote the development of therapeutic strategies for the treatment and prevention of this neurodegenerative disorder.
Collapse
Affiliation(s)
- Jing Tian
- Department of Pharmacology and Toxicology, University of Kansas, Lawrence, KS, USA
| | - Eric Du
- Department of Pharmacology and Toxicology, University of Kansas, Lawrence, KS, USA
- Blue Valley West High School, Overland Park, KS, USA
| | - Lan Guo
- Department of Pharmacology and Toxicology, University of Kansas, Lawrence, KS, USA
| |
Collapse
|
36
|
Tang H, Chen X, Huang S, Yin G, Wang X, Shen G. Targeting the gut-microbiota-brain axis in irritable bowel disease to improve cognitive function - recent knowledge and emerging therapeutic opportunities. Rev Neurosci 2023; 34:763-773. [PMID: 36757367 DOI: 10.1515/revneuro-2022-0155] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 01/21/2023] [Indexed: 02/10/2023]
Abstract
The brain-gut axis forms a bidirectional communication system between the gastrointestinal (GI) tract and cognitive brain areas. Disturbances to this system in disease states such as inflammatory bowel disease have consequences for neuronal activity and subsequent cognitive function. The gut-microbiota-brain axis refers to the communication between gut-resident bacteria and the brain. This circuits exists to detect gut microorganisms and relay information to specific areas of the central nervous system (CNS) that in turn, regulate gut physiology. Changes in both the stability and diversity of the gut microbiota have been implicated in several neuronal disorders, including depression, autism spectrum disorder Parkinson's disease, Alzheimer's disease and multiple sclerosis. Correcting this imbalance with medicinal herbs, the metabolic products of dysregulated bacteria and probiotics have shown hope for the treatment of these neuronal disorders. In this review, we focus on recent advances in our understanding of the intricate connections between the gut-microbiota and the brain. We discuss the contribution of gut microbiota to neuronal disorders and the tangible links between diseases of the GI tract with cognitive function and behaviour. In this regard, we focus on irritable bowel syndrome (IBS) given its strong links to brain function and anxiety disorders. This adds to the growing body of evidence supporting targeted therapeutic strategies to modulate the gut microbiota for the treatment of brain/mental-health-related disease.
Collapse
Affiliation(s)
- Heyong Tang
- School of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, No. 1, Qianjiang Road, 230012 Hefei, Anhui, China
| | - Xiaoqi Chen
- School of Acupuncture and Massage, Anhui University of Chinese Medicine, 230012 Hefei, Anhui, China
| | - Shun Huang
- School of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, No. 1, Qianjiang Road, 230012 Hefei, Anhui, China
| | - Gang Yin
- Xin'an School, Anhui University of Chinese Medicine, 230012 Hefei, Anhui, China
| | - Xiyang Wang
- School of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, No. 1, Qianjiang Road, 230012 Hefei, Anhui, China
| | - Guoming Shen
- School of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, No. 1, Qianjiang Road, 230012 Hefei, Anhui, China
| |
Collapse
|
37
|
Jiang J, Shi H, Jiang S, Wang A, Zou X, Wang Y, Li W, Zhang Y, Sun M, Ren Q, Xu J. Nutrition in Alzheimer's disease: a review of an underappreciated pathophysiological mechanism. SCIENCE CHINA. LIFE SCIENCES 2023; 66:2257-2279. [PMID: 37058185 DOI: 10.1007/s11427-022-2276-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 02/16/2023] [Indexed: 04/15/2023]
Abstract
Alzheimer's disease (AD) is the leading cause of dementia in older individuals and is an escalating challenge to global public health. Pharmacy therapy of AD is one of the well-funded areas; however, little progress has been made due to the complex pathogenesis. Recent evidence has demonstrated that modifying risk factors and lifestyle may prevent or delay the incidence of AD by 40%, which suggests that the management should pivot from single pharmacotherapy toward a multipronged approach because AD is a complex and multifaceted disease. Recently, the gut-microbiota-brain axis has gained tremendous traction in the pathogenesis of AD through bidirectional communication with multiple neural, immune, and metabolic pathways, providing new insights into novel therapeutic strategies. Dietary nutrition is an important and profound environmental factor that influences the composition and function of the microbiota. The Nutrition for Dementia Prevention Working Group recently found that dietary nutrition can affect cognition in AD-related dementia directly or indirectly through complex interactions of behavioral, genetic, systemic, and brain factors. Thus, considering the multiple etiologies of AD, nutrition represents a multidimensional factor that has a profound effect on AD onset and development. However, mechanistically, the effect of nutrition on AD is uncertain; therefore, optimal strategies or the timing of nutritional intervention to prevent or treat AD has not been established.Thus, this review summarizes the current state of knowledge concerning nutritional disorders, AD patient and caregiver burden, and the roles of nutrition in the pathophysiology of AD. We aim to emphasize knowledge gaps to provide direction for future research and to establish optimal nutrition-based intervention strategies for AD.
Collapse
Affiliation(s)
- Jiwei Jiang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China
| | - Hanping Shi
- Department of Gastrointestinal Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
- Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, 100038, China
| | - Shirui Jiang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China
| | - Anxin Wang
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China
| | - Xinying Zou
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China
| | - Yanli Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China
| | - Wenyi Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China
| | - Yuan Zhang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China
| | - Mengfan Sun
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China
| | - Qiwei Ren
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China
| | - Jun Xu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China.
| |
Collapse
|
38
|
Chen H, Wang C, Bai J, Song J, Bu L, Liang M, Suo H. Targeting microbiota to alleviate the harm caused by sleep deprivation. Microbiol Res 2023; 275:127467. [PMID: 37549451 DOI: 10.1016/j.micres.2023.127467] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/23/2023] [Accepted: 07/27/2023] [Indexed: 08/09/2023]
Abstract
Sleep deprivation has become a common health hazard, affecting 37-58% of the population and promoting the occurrence and development of many diseases. To date, effective treatment strategies are still elusive. Accumulating evidence indicates that modulating the intestinal microbiota harbors significant potential for alleviating the deleterious impacts of sleep deprivation. This paper first reviews the effects of sleep deprivation on gastrointestinal diseases, metabolic diseases, and neuropsychiatric diseases, discussing its specific mechanisms of influence. We then focus on summarizing existing interventions, including probiotics, melatonin, prebiotics, diet, and fecal microbiota transplantation (FMT). Finally, we have discussed the advantages and limitations of each strategy. Compared with other strategies, probiotics showed a high potential in alleviating sleep deprivation-related hazards due to their reduced risk and high security. We suggest that future research should focus on the specific mechanisms by which probiotics mitigate the harms of sleep deprivation, such insights may unveil novel pathways for treating diseases exacerbated by insufficient sleep.
Collapse
Affiliation(s)
- Hongyu Chen
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Chen Wang
- College of Food Science, Southwest University, Chongqing 400715, China; Food Industry Innovation Research Institute of Modern Sichuan Cuisine & Chongqing Flavor, Chongqing 400715, China
| | - Junying Bai
- Citrus Research Institute, National Citrus Engineering Research Center, Southwest University, Chongqing 400715, China
| | - Jiajia Song
- College of Food Science, Southwest University, Chongqing 400715, China; Food Industry Innovation Research Institute of Modern Sichuan Cuisine & Chongqing Flavor, Chongqing 400715, China
| | - Linli Bu
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Ming Liang
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Huayi Suo
- College of Food Science, Southwest University, Chongqing 400715, China; Food Industry Innovation Research Institute of Modern Sichuan Cuisine & Chongqing Flavor, Chongqing 400715, China.
| |
Collapse
|
39
|
Sharma HS, Feng L, Muresanu DF, Tian ZR, Lafuente JV, Buzoianu AD, Nozari A, Bryukhovetskiy I, Manzhulo I, Wiklund L, Sharma A. Stress induced exacerbation of Alzheimer's disease brain pathology is thwarted by co-administration of nanowired cerebrolysin and monoclonal amyloid beta peptide antibodies with serotonin 5-HT6 receptor antagonist SB-399885. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 171:3-46. [PMID: 37783559 DOI: 10.1016/bs.irn.2023.05.018] [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] [Indexed: 10/04/2023]
Abstract
Alzheimer's disease is one of the devastating neurodegenerative diseases affecting mankind worldwide with advancing age mainly above 65 years and above causing great misery of life. About more than 7 millions are affected with Alzheimer's disease in America in 2023 resulting in huge burden on health care system and care givers and support for the family. However, no suitable therapeutic measures are available at the moment to enhance quality of life to these patients. Development of Alzheimer's disease may reflect the stress burden of whole life inculcating the disease processes of these neurodegenerative disorders of the central nervous system. Thus, new strategies using nanodelivery of suitable drug therapy including antibodies are needed in exploring neuroprotection in Alzheimer's disease brain pathology. In this chapter role of stress in exacerbating Alzheimer's disease brain pathology is explored and treatment strategies are examined using nanotechnology based on our own investigation. Our observations clearly show that restraint stress significantly exacerbate Alzheimer's disease brain pathology and nanodelivery of a multimodal drug cerebrolysin together with monoclonal antibodies (mAb) to amyloid beta peptide (AβP) together with a serotonin 5-HT6 receptor antagonist SB399885 significantly thwarted Alzheimer's disease brain pathology exacerbated by restraint stress, not reported earlier. The possible mechanisms and future clinical significance is discussed.
Collapse
Affiliation(s)
- Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Dept. of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| | - Lianyuan Feng
- Department of Neurology, Bethune International Peace Hospital, Zhongshan Road (West), Shijiazhuang, Hebei Province, P.R. China
| | - Dafin F Muresanu
- Dept. Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania; ''RoNeuro'' Institute for Neurological Research and Diagnostic, Mircea Eliade Street, Cluj-Napoca, Romania
| | - Z Ryan Tian
- Dept. Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - José Vicente Lafuente
- LaNCE, Dept. Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ala Nozari
- Department of Anesthesiology, Boston University, Albany str, Boston, MA, United States
| | - Igor Bryukhovetskiy
- Department of Fundamental Medicine, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia; Laboratory of Pharmacology, National Scientific Center of Marine Biology, Far East Branch of the Russian Academy of Sciences, Vladivostok, Russia
| | - Igor Manzhulo
- Laboratory of Pharmacology, National Scientific Center of Marine Biology, Far East Branch of the Russian Academy of Sciences, Vladivostok, Russia
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Dept. of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Dept. of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
40
|
Li Y, Wu M, Kong M, Sui S, Wang Q, He Y, Gu J. Impact of Donepezil Supplementation on Alzheimer's Disease-like Pathology and Gut Microbiome in APP/PS1 Mice. Microorganisms 2023; 11:2306. [PMID: 37764150 PMCID: PMC10537997 DOI: 10.3390/microorganisms11092306] [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: 07/24/2023] [Revised: 08/31/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
Based on published information, the occurrence and development of Alzheimer's disease (AD) are potentially related to gut microbiota changes. Donepezil hydrochloride (DH), which enhances cholinergic activity by blocking acetylcholinesterase (AChE), is one of the first-line drugs for AD treatment approved by the Food and Drug Administration (FDA) of the USA. However, the potential link between the effects of DH on the pathophysiological processes of AD and the gut microbiota remains unclear. In this study, pathological changes in the brain and colon, the activities of superoxide dismutase (SOD) and AChE, and changes in intestinal flora were observed. The results showed that Aβ deposition in the prefrontal cortex and hippocampus of AD mice was significantly decreased, while colonic inflammation was significantly alleviated by DH treatment. Concomitantly, SOD activity was significantly improved, while AChE was significantly reduced after DH administration. In addition, the gut microbiota community composition of AD mice was significantly altered after DH treatment. The relative abundance of Akkermansia in the AD group was 54.8% higher than that in the N group. The relative abundance of Akkermansia was increased by 18.3% and 53.8% in the AD_G group and the N_G group, respectively. Interestingly, Akkermansia showed a potential predictive value and might be a biomarker for AD. Molecular docking revealed the binding mode and major forces between DH and membrane proteins of Akkermansia. The overall results suggest a novel therapeutic mechanism for treating AD and highlight the critical role of gut microbiota in AD pathology.
Collapse
Affiliation(s)
- Yuan Li
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China; (Y.L.); (M.W.); (M.K.)
| | - Mengyao Wu
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China; (Y.L.); (M.W.); (M.K.)
| | - Mengmeng Kong
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China; (Y.L.); (M.W.); (M.K.)
| | - Shaomei Sui
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China; (S.S.); (Q.W.)
| | - Qi Wang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China; (S.S.); (Q.W.)
| | - Yan He
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China; (S.S.); (Q.W.)
| | - Jinsong Gu
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China; (Y.L.); (M.W.); (M.K.)
| |
Collapse
|
41
|
Choi H, Mook-Jung I. Functional effects of gut microbiota-derived metabolites in Alzheimer's disease. Curr Opin Neurobiol 2023; 81:102730. [PMID: 37236067 DOI: 10.1016/j.conb.2023.102730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 04/28/2023] [Accepted: 04/30/2023] [Indexed: 05/28/2023]
Abstract
The precise causation of Alzheimer's disease (AD) is unknown, and the factors that contribute to its etiology are highly complicated. Numerous research has been conducted to investigate the potential impact of various factors to the risk of AD development or prevention against it. A growing body of evidence suggests to the importance of the gut microbiota-brain axis in the modulation of AD, which is characterized by altered gut microbiota composition. These changes can alter the production of microbial-derived metabolites, which may play a detrimental role in disease progression by being involved in cognitive decline, neurodegeneration, neuroinflammation, and accumulation of Aβ and tau. The focus of this review is on the relationship between the key metabolic products of the gut microbiota and AD pathogenesis in the brain. Understanding the action of microbial metabolites can open up new avenues for the development of AD treatment targets.
Collapse
Affiliation(s)
- Hyunjung Choi
- Genomic Medicine Institute, Medical Research Center, Seoul National University, Seoul, Republic of Korea; Convergence Dementia Research Center, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Inhee Mook-Jung
- Genomic Medicine Institute, Medical Research Center, Seoul National University, Seoul, Republic of Korea; Convergence Dementia Research Center, College of Medicine, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
42
|
Post Z, Manfready RA, Keshavarzian A. Overview of the Gut-Brain Axis: From Gut to Brain and Back Again. Semin Neurol 2023; 43:506-517. [PMID: 37562457 DOI: 10.1055/s-0043-1771464] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
The gut-brain axis refers to a bidirectional communication pathway linking the gastrointestinal system to the central nervous system. The hardware of this multifaceted pathway takes many forms, at once structural (neurons, microglia, intestinal epithelial cell barrier), chemical (neurotransmitters, enteroendocrine hormones, bacterial metabolites), and cellular (immune signaling, inflammatory pathways). The gut-brain axis is exquisitely influenced by our environment, diet, and behaviors. Here, we will describe recent progress in understanding the gut-brain axis in neurological disease, using Parkinson's disease as a guide. We will see that each component of the gut-brain axis is heavily mediated by intestinal microbiota and learn how gut-brain communication can go awry in microbial dysbiosis.
Collapse
Affiliation(s)
- Zoë Post
- Division of Digestive Diseases and Nutrition, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois
| | - Richard A Manfready
- Division of Digestive Diseases and Nutrition, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, Illinois
- Departments of Physiology and Anatomy & Cell Biology, Rush University Medical Center, Chicago, Illinois
| | - Ali Keshavarzian
- Division of Digestive Diseases and Nutrition, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, Illinois
- Departments of Physiology and Anatomy & Cell Biology, Rush University Medical Center, Chicago, Illinois
| |
Collapse
|
43
|
Zhu G, Zhao J, Zhang H, Wang G, Chen W. Gut Microbiota and its Metabolites: Bridge of Dietary Nutrients and Alzheimer's Disease. Adv Nutr 2023; 14:819-839. [PMID: 37075947 PMCID: PMC10334159 DOI: 10.1016/j.advnut.2023.04.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/29/2023] [Accepted: 04/14/2023] [Indexed: 04/21/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by progressive cognitive impairment and neuroinflammation. Recent research has revealed the crucial role of gut microbiota and microbial metabolites in modulating AD. However, the mechanisms by which the microbiome and microbial metabolites affect brain function remain poorly understood. Here, we review the literature on changes in the diversity and composition of the gut microbiome in patients with AD and in animal models of AD. We also discuss the latest progress in understanding the pathways by which the gut microbiota and microbial metabolites from the host or diet regulate AD. By understanding the effects of dietary components on brain function, microbiota composition, and microbial metabolites, we examine the potential for manipulation of the gut microbiota through dietary intervention to delay the progression of AD. Although it is challenging to translate our understanding of microbiome-based approaches to dietary guidelines or clinical therapies, these findings provide an attractive target for promoting brain function.
Collapse
Affiliation(s)
- Guangsu Zhu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China; College of Food Science and Technology, Henan University of Technology, Zhengzhou, Henan, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China; (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou, Jiangsu, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China; (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou, Jiangsu, China; National Engineering Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China
| | - Gang Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China; (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou, Jiangsu, China.
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China; National Engineering Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China
| |
Collapse
|
44
|
López-Villodres JA, Escamilla A, Mercado-Sáenz S, Alba-Tercedor C, Rodriguez-Perez LM, Arranz-Salas I, Sanchez-Varo R, Bermúdez D. Microbiome Alterations and Alzheimer's Disease: Modeling Strategies with Transgenic Mice. Biomedicines 2023; 11:1846. [PMID: 37509487 PMCID: PMC10377071 DOI: 10.3390/biomedicines11071846] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 06/21/2023] [Accepted: 06/23/2023] [Indexed: 07/30/2023] Open
Abstract
In the last decade, the role of the microbiota-gut-brain axis has been gaining momentum in the context of many neurodegenerative and metabolic disorders, including Alzheimer's disease (AD) and diabetes, respectively. Notably, a balanced gut microbiota contributes to the epithelial intestinal barrier maintenance, modulates the host immune system, and releases neurotransmitters and/or neuroprotective short-chain fatty acids. However, dysbiosis may provoke immune dysregulation, impacting neuroinflammation through peripheral-central immune communication. Moreover, lipopolysaccharide or detrimental microbial end-products can cross the blood-brain barrier and induce or at least potentiate the neuropathological progression of AD. Thus, after repeated failure to find a cure for this dementia, a necessary paradigmatic shift towards considering AD as a systemic disorder has occurred. Here, we present an overview of the use of germ-free and/or transgenic animal models as valid tools to unravel the connection between dysbiosis, metabolic diseases, and AD, and to investigate novel therapeutical targets. Given the high impact of dietary habits, not only on the microbiota but also on other well-established AD risk factors such as diabetes or obesity, consistent changes of lifestyle along with microbiome-based therapies should be considered as complementary approaches.
Collapse
Affiliation(s)
- Juan Antonio López-Villodres
- Departamento Fisiologia Humana, Histologia Humana, Anatomia Patologica y Educacion Fisica y Deportiva, Facultad de Medicina, Universidad de Malaga, 29071 Malaga, Spain
| | - Alejandro Escamilla
- Departamento Fisiologia Humana, Histologia Humana, Anatomia Patologica y Educacion Fisica y Deportiva, Facultad de Medicina, Universidad de Malaga, 29071 Malaga, Spain
- Instituto de Investigacion Biomedica de Malaga-IBIMA-Plataforma Bionand, 29071 Malaga, Spain
| | - Silvia Mercado-Sáenz
- Departamento Fisiologia Humana, Histologia Humana, Anatomia Patologica y Educacion Fisica y Deportiva, Facultad de Medicina, Universidad de Malaga, 29071 Malaga, Spain
| | - Carmen Alba-Tercedor
- Departamento Fisiologia Humana, Histologia Humana, Anatomia Patologica y Educacion Fisica y Deportiva, Facultad de Medicina, Universidad de Malaga, 29071 Malaga, Spain
| | - Luis Manuel Rodriguez-Perez
- Departamento Fisiologia Humana, Histologia Humana, Anatomia Patologica y Educacion Fisica y Deportiva, Facultad de Medicina, Universidad de Malaga, 29071 Malaga, Spain
- Instituto de Investigacion Biomedica de Malaga-IBIMA-Plataforma Bionand, 29071 Malaga, Spain
| | - Isabel Arranz-Salas
- Departamento Fisiologia Humana, Histologia Humana, Anatomia Patologica y Educacion Fisica y Deportiva, Facultad de Medicina, Universidad de Malaga, 29071 Malaga, Spain
- Instituto de Investigacion Biomedica de Malaga-IBIMA-Plataforma Bionand, 29071 Malaga, Spain
- Unidad de Anatomia Patologica, Hospital Universitario Virgen de la Victoria, 29010 Malaga, Spain
| | - Raquel Sanchez-Varo
- Departamento Fisiologia Humana, Histologia Humana, Anatomia Patologica y Educacion Fisica y Deportiva, Facultad de Medicina, Universidad de Malaga, 29071 Malaga, Spain
- Instituto de Investigacion Biomedica de Malaga-IBIMA-Plataforma Bionand, 29071 Malaga, Spain
- Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Diego Bermúdez
- Departamento Fisiologia Humana, Histologia Humana, Anatomia Patologica y Educacion Fisica y Deportiva, Facultad de Medicina, Universidad de Malaga, 29071 Malaga, Spain
| |
Collapse
|
45
|
Morrone CD, Raghuraman R, Hussaini SA, Yu WH. Proteostasis failure exacerbates neuronal circuit dysfunction and sleep impairments in Alzheimer's disease. Mol Neurodegener 2023; 18:27. [PMID: 37085942 PMCID: PMC10119020 DOI: 10.1186/s13024-023-00617-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 03/29/2023] [Indexed: 04/23/2023] Open
Abstract
Failed proteostasis is a well-documented feature of Alzheimer's disease, particularly, reduced protein degradation and clearance. However, the contribution of failed proteostasis to neuronal circuit dysfunction is an emerging concept in neurodegenerative research and will prove critical in understanding cognitive decline. Our objective is to convey Alzheimer's disease progression with the growing evidence for a bidirectional relationship of sleep disruption and proteostasis failure. Proteostasis dysfunction and tauopathy in Alzheimer's disease disrupts neurons that regulate the sleep-wake cycle, which presents behavior as impaired slow wave and rapid eye movement sleep patterns. Subsequent sleep loss further impairs protein clearance. Sleep loss is a defined feature seen early in many neurodegenerative disorders and contributes to memory impairments in Alzheimer's disease. Canonical pathological hallmarks, β-amyloid, and tau, directly disrupt sleep, and neurodegeneration of locus coeruleus, hippocampal and hypothalamic neurons from tau proteinopathy causes disruption of the neuronal circuitry of sleep. Acting in a positive-feedback-loop, sleep loss and circadian rhythm disruption then increase spread of β-amyloid and tau, through impairments of proteasome, autophagy, unfolded protein response and glymphatic clearance. This phenomenon extends beyond β-amyloid and tau, with interactions of sleep impairment with the homeostasis of TDP-43, α-synuclein, FUS, and huntingtin proteins, implicating sleep loss as an important consideration in an array of neurodegenerative diseases and in cases of mixed neuropathology. Critically, the dynamics of this interaction in the neurodegenerative environment are not fully elucidated and are deserving of further discussion and research. Finally, we propose sleep-enhancing therapeutics as potential interventions for promoting healthy proteostasis, including β-amyloid and tau clearance, mechanistically linking these processes. With further clinical and preclinical research, we propose this dynamic interaction as a diagnostic and therapeutic framework, informing precise single- and combinatorial-treatments for Alzheimer's disease and other brain disorders.
Collapse
Affiliation(s)
- Christopher Daniel Morrone
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College St., Toronto, ON, M5T 1R8, Canada.
| | - Radha Raghuraman
- Taub Institute, Columbia University Irving Medical Center, 630W 168th Street, New York, NY, 10032, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, 630W 168th Street, New York, NY, 10032, USA
| | - S Abid Hussaini
- Taub Institute, Columbia University Irving Medical Center, 630W 168th Street, New York, NY, 10032, USA.
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, 630W 168th Street, New York, NY, 10032, USA.
| | - Wai Haung Yu
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College St., Toronto, ON, M5T 1R8, Canada.
- Geriatric Mental Health Research Services, Centre for Addiction and Mental Health, 250 College St., Toronto, ON, M5T 1R8, Canada.
- Department of Pharmacology and Toxicology, University of Toronto, Medical Sciences Building, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
46
|
Heylen A, Vermeiren Y, Kema IP, van Faassen M, van der Ley C, Van Dam D, De Deyn PP. Brain Kynurenine Pathway Metabolite Levels May Reflect Extent of Neuroinflammation in ALS, FTD and Early Onset AD. Pharmaceuticals (Basel) 2023; 16:ph16040615. [PMID: 37111372 PMCID: PMC10143579 DOI: 10.3390/ph16040615] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/04/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
OBJECTIVES Despite distinct clinical profiles, amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) patients share a remarkable portion of pathological features, with a substantial percentage of patients displaying a mixed disease phenotype. Kynurenine metabolism seems to play a role in dementia-associated neuroinflammation and has been linked to both diseases. We aimed to explore dissimilarities in kynurenine pathway metabolites in these early onset neurodegenerative disorders in a brain-region-specific manner. METHODS Using liquid chromatography mass spectrometry (LC-MS/MS), kynurenine metabolite levels were determined in the brain samples of 98 healthy control subjects (n = 20) and patients with early onset Alzheimer's disease (EOAD) (n = 23), ALS (n = 20), FTD (n = 24) or a mixed FTD-ALS (n = 11) disease profile. RESULTS Overall, the kynurenine pathway metabolite levels were significantly lower in patients with ALS compared to FTD, EOAD and control subjects in the frontal cortex, substantia nigra, hippocampus and neostriatum. Anthranilic acid levels and kynurenine-to-tryptophan ratios were consistently lower in all investigated brain regions in ALS compared to the other diagnostic groups. CONCLUSIONS These results suggest that the contribution of kynurenine metabolism in neuroinflammation is lower in ALS than in FTD or EOAD and may also be traced back to differences in the age of onset between these disorders. Further research is necessary to confirm the potential of the kynurenine system as a therapeutic target in these early onset neurodegenerative disorders.
Collapse
Affiliation(s)
- Annelies Heylen
- Laboratory of Neurochemistry and Behavior, Experimental Neurobiology Unit, University of Antwerp, 2610 Antwerp, Belgium
| | - Yannick Vermeiren
- Division of Human Nutrition and Health, Chair Group of Nutritional Biology, Wageningen University and Research, 6708 Wageningen, The Netherlands
- Faculty of Medicine & Health Sciences, Translational Neurosciences, University of Antwerp, 2000 Antwerp, Belgium
| | - Ido P Kema
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, 9713 Groningen, The Netherlands
| | - Martijn van Faassen
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, 9713 Groningen, The Netherlands
| | - Claude van der Ley
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, 9713 Groningen, The Netherlands
| | - Debby Van Dam
- Laboratory of Neurochemistry and Behavior, Experimental Neurobiology Unit, University of Antwerp, 2610 Antwerp, Belgium
- Department of Neurology and Alzheimer Center Groningen, University of Groningen, University Medical Center Groningen, 9713 Groningen, The Netherlands
| | - Peter P De Deyn
- Laboratory of Neurochemistry and Behavior, Experimental Neurobiology Unit, University of Antwerp, 2610 Antwerp, Belgium
- Department of Neurology and Alzheimer Center Groningen, University of Groningen, University Medical Center Groningen, 9713 Groningen, The Netherlands
| |
Collapse
|
47
|
Wang XN, Zhang JC, Zhang HY, Wang XF, You CX. Ectopic expression of MmSERT, a mouse serotonin transporter gene, regulates salt tolerance and ABA sensitivity in apple and Arabidopsis. PLANT PHYSIOLOGY AND BIOCHEMISTRY : PPB 2023; 197:107627. [PMID: 36940523 DOI: 10.1016/j.plaphy.2023.03.004] [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: 11/15/2022] [Revised: 02/20/2023] [Accepted: 03/01/2023] [Indexed: 06/18/2023]
Abstract
5-hydroxytryptamine (5-HT) is ubiquitously present in animals and plants, playing a vital regulatory role. SERT, a conserved serotonin reuptake transporter in animals, regulates intracellular and extracellular concentrations of 5-HT. Few studies have reported 5-HT transporters in plants. Hence, we cloned MmSERT, a serotonin reuptake transporter, from Mus musculus. Ectopic expression of MmSERT into apple calli, apple roots and Arabidopsis. Because 5-HT plays a momentous role in plant stress tolerance, we used MmSERT transgenic materials for stress treatment. We found that MmSERT transgenic materials, including apple calli, apple roots and Arabidopsis, exhibited a stronger salt tolerance phenotype. The reactive oxygen species (ROS) produced were significantly lower in MmSERT transgenic materials compared with controls under salt stress. Meanwhile, MmSERT induced the expression of SOS1, SOS3, NHX1, LEA5 and LTP1 in response to salt stress. 5-HT is the precursor of melatonin, which regulates plant growth under adversity and effectively scavenges ROS. Detection of MmSERT transgenic apple calli and Arabidopsis revealed higher melatonin levels than controls. Besides, MmSERT decreased the sensitivity of apple calli and Arabidopsis to abscisic acid (ABA). In summary, these results demonstrated that MmSERT plays a vital role in plant stress resistances, which perhaps serves as a reference for the application of transgenic technology to improve crops in the future.
Collapse
Affiliation(s)
- Xiao-Na Wang
- National Key Laboratory of Crop Biology, MOA Key Laboratory of Horticultural Crop Biology and Germplasm Innovation, College of Horticulture Science and Engineering, Shandong Agricultural University, Tai-An, 271018, Shandong, China
| | - Jiu-Cheng Zhang
- National Key Laboratory of Crop Biology, MOA Key Laboratory of Horticultural Crop Biology and Germplasm Innovation, College of Horticulture Science and Engineering, Shandong Agricultural University, Tai-An, 271018, Shandong, China
| | - Hai-Yuan Zhang
- National Key Laboratory of Crop Biology, MOA Key Laboratory of Horticultural Crop Biology and Germplasm Innovation, College of Horticulture Science and Engineering, Shandong Agricultural University, Tai-An, 271018, Shandong, China
| | - Xiao-Fei Wang
- National Key Laboratory of Crop Biology, MOA Key Laboratory of Horticultural Crop Biology and Germplasm Innovation, College of Horticulture Science and Engineering, Shandong Agricultural University, Tai-An, 271018, Shandong, China.
| | - Chun-Xiang You
- National Key Laboratory of Crop Biology, MOA Key Laboratory of Horticultural Crop Biology and Germplasm Innovation, College of Horticulture Science and Engineering, Shandong Agricultural University, Tai-An, 271018, Shandong, China.
| |
Collapse
|
48
|
Borsom EM, Conn K, Keefe CR, Herman C, Orsini GM, Hirsch AH, Palma Avila M, Testo G, Jaramillo SA, Bolyen E, Lee K, Caporaso JG, Cope EK. Predicting Neurodegenerative Disease Using Prepathology Gut Microbiota Composition: a Longitudinal Study in Mice Modeling Alzheimer's Disease Pathologies. Microbiol Spectr 2023; 11:e0345822. [PMID: 36877047 PMCID: PMC10101110 DOI: 10.1128/spectrum.03458-22] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 01/12/2023] [Indexed: 03/07/2023] Open
Abstract
The gut microbiota-brain axis is suspected to contribute to the development of Alzheimer's disease (AD), a neurodegenerative disease characterized by amyloid-β plaque deposition, neurofibrillary tangles, and neuroinflammation. To evaluate the role of the gut microbiota-brain axis in AD, we characterized the gut microbiota of female 3xTg-AD mice modeling amyloidosis and tauopathy and wild-type (WT) genetic controls. Fecal samples were collected fortnightly from 4 to 52 weeks, and the V4 region of the 16S rRNA gene was amplified and sequenced on an Illumina MiSeq. RNA was extracted from the colon and hippocampus, converted to cDNA, and used to measure immune gene expression using reverse transcriptase quantitative PCR (RT-qPCR). Diversity metrics were calculated using QIIME2, and a random forest classifier was applied to predict bacterial features that are important in predicting mouse genotype. Gene expression of glial fibrillary acidic protein (GFAP; indicating astrocytosis) was elevated in the colon at 24 weeks. Markers of Th1 inflammation (il6) and microgliosis (mrc1) were elevated in the hippocampus. Gut microbiota were compositionally distinct early in life between 3xTg-AD mice and WT mice (permutational multivariate analysis of variance [PERMANOVA], 8 weeks, P = 0.001, 24 weeks, P = 0.039, and 52 weeks, P = 0.058). Mouse genotypes were correctly predicted 90 to 100% of the time using fecal microbiome composition. Finally, we show that the relative abundance of Bacteroides species increased over time in 3xTg-AD mice. Taken together, we demonstrate that changes in bacterial gut microbiota composition at prepathology time points are predictive of the development of AD pathologies. IMPORTANCE Recent studies have demonstrated alterations in the gut microbiota composition in mice modeling Alzheimer's disease (AD) pathologies; however, these studies have only included up to 4 time points. Our study is the first of its kind to characterize the gut microbiota of a transgenic AD mouse model, fortnightly, from 4 weeks of age to 52 weeks of age, to quantify the temporal dynamics in the microbial composition that correlate with the development of disease pathologies and host immune gene expression. In this study, we observed temporal changes in the relative abundances of specific microbial taxa, including the genus Bacteroides, that may play a central role in disease progression and the severity of pathologies. The ability to use features of the microbiota to discriminate between mice modeling AD and wild-type mice at prepathology time points indicates a potential role of the gut microbiota as a risk or protective factor in AD.
Collapse
Affiliation(s)
- Emily M. Borsom
- Center for Applied Microbiome Sciences, the Pathogen and Microbiome Institute, Department of Biological Sciences, Northern Arizona University, Flagstaff, Arizona, USA
| | - Kathryn Conn
- Center for Applied Microbiome Sciences, the Pathogen and Microbiome Institute, Department of Biological Sciences, Northern Arizona University, Flagstaff, Arizona, USA
| | - Christopher R. Keefe
- Center for Applied Microbiome Sciences, the Pathogen and Microbiome Institute, Department of Biological Sciences, Northern Arizona University, Flagstaff, Arizona, USA
| | - Chloe Herman
- Center for Applied Microbiome Sciences, the Pathogen and Microbiome Institute, Department of Biological Sciences, Northern Arizona University, Flagstaff, Arizona, USA
| | - Gabrielle M. Orsini
- Center for Applied Microbiome Sciences, the Pathogen and Microbiome Institute, Department of Biological Sciences, Northern Arizona University, Flagstaff, Arizona, USA
| | - Allyson H. Hirsch
- Center for Applied Microbiome Sciences, the Pathogen and Microbiome Institute, Department of Biological Sciences, Northern Arizona University, Flagstaff, Arizona, USA
| | - Melanie Palma Avila
- Center for Applied Microbiome Sciences, the Pathogen and Microbiome Institute, Department of Biological Sciences, Northern Arizona University, Flagstaff, Arizona, USA
| | - George Testo
- Center for Applied Microbiome Sciences, the Pathogen and Microbiome Institute, Department of Biological Sciences, Northern Arizona University, Flagstaff, Arizona, USA
| | - Sierra A. Jaramillo
- Center for Applied Microbiome Sciences, the Pathogen and Microbiome Institute, Department of Biological Sciences, Northern Arizona University, Flagstaff, Arizona, USA
| | - Evan Bolyen
- Center for Applied Microbiome Sciences, the Pathogen and Microbiome Institute, Department of Biological Sciences, Northern Arizona University, Flagstaff, Arizona, USA
| | - Keehoon Lee
- Center for Applied Microbiome Sciences, the Pathogen and Microbiome Institute, Department of Biological Sciences, Northern Arizona University, Flagstaff, Arizona, USA
| | - J. Gregory Caporaso
- Center for Applied Microbiome Sciences, the Pathogen and Microbiome Institute, Department of Biological Sciences, Northern Arizona University, Flagstaff, Arizona, USA
| | - Emily K. Cope
- Center for Applied Microbiome Sciences, the Pathogen and Microbiome Institute, Department of Biological Sciences, Northern Arizona University, Flagstaff, Arizona, USA
| |
Collapse
|
49
|
Tian D, Shi W, Yu Y, Zhou W, Tang Y, Zhang W, Huang L, Han Y, Liu G. Enrofloxacin exposure induces anxiety-like behavioral responses in zebrafish by affecting the microbiota-gut-brain axis. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 858:160094. [PMID: 36372168 DOI: 10.1016/j.scitotenv.2022.160094] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 10/05/2022] [Accepted: 11/05/2022] [Indexed: 06/16/2023]
Abstract
The ubiquitous presence of antibiotic residues in aqueous environments poses a great potential threat to aquatic organisms. Nevertheless, the behavioral effects of environmentally realistic levels of antibiotics remain poorly understood in fish species. In this study, the behavioral impacts of enrofloxacin, one of typical fluoroquinolone antibiotics that is frequently detected in aquatic environments, were evaluated by the classic light-dark test (LDT) and novel tank task (NTT) in zebrafish. Furthermore, the effects of enrofloxacin exposure on the microbiota-gut-brain axis were also assessed to reveal potential affecting mechanisms underlying the behavioral abnormality observed. Our results demonstrated that zebrafish exposed to 60 μg/L enrofloxacin for 28 days took significantly longer to enter the stressful area of the testing tank and spent significantly less time there in both the LDT and NTT, indicating abnormal anxiety-like behaviors induced by the exposure. In addition, exposure to enrofloxacin at 6 and 60 μg/L resulted in a significant elevation in Bacteroidetes and a marked decline in the Firmicutes/Bacteroidetes ratio of the gut microbiota. Moreover, the intestinal contents of interleukin 6 (IL-6), tumor necrosis factor-alpha (TNF-α), glucagon-like peptide 1 (GLP-1), and 5-hydroxytryptamine (5-HT) in zebrafish were significantly upregulated, whereas those of plasma adrenocorticotropic hormone (ACTH) and cortisol (COR) were markedly downregulated upon enrofloxacin exposure. Incubation of zebrafish with a high dose of enrofloxacin (60 μg/L) also resulted in evident increases in the contents of corticotropin-releasing hormone (CRH), brain-derived neurotrophic factor (BDNF), and neuropeptide Y (NPY) in the brain. Fortunately, no significant alteration in the expression of glial fibrillary acidic protein (GFAP) was detected in the brain after enrofloxacin exposure. Our findings suggest that the disruption of the microbiota-gut-brain axis may account for enrofloxacin-induced anxiety-like behaviors in zebrafish. Since the disruption of microbiota-gut-brain axis may give rise to various clinical symptoms, the health risk of antibiotic exposure deserves more attention.
Collapse
Affiliation(s)
- Dandan Tian
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wei Shi
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yihan Yu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Weishang Zhou
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yu Tang
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Weixia Zhang
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Lin Huang
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yu Han
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Guangxu Liu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
50
|
Parolo S, Mariotti F, Bora P, Carboni L, Domenici E. Single-cell-led drug repurposing for Alzheimer's disease. Sci Rep 2023; 13:222. [PMID: 36604493 PMCID: PMC9816180 DOI: 10.1038/s41598-023-27420-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 12/29/2022] [Indexed: 01/06/2023] Open
Abstract
Alzheimer's disease is the most common form of dementia. Notwithstanding the huge investments in drug development, only one disease-modifying treatment has been recently approved. Here we present a single-cell-led systems biology pipeline for the identification of drug repurposing candidates. Using single-cell RNA sequencing data of brain tissues from patients with Alzheimer's disease, genome-wide association study results, and multiple gene annotation resources, we built a multi-cellular Alzheimer's disease molecular network that we leveraged for gaining cell-specific insights into Alzheimer's disease pathophysiology and for the identification of drug repurposing candidates. Our computational approach pointed out 54 candidate drugs, mainly targeting MAPK and IGF1R signaling pathways, which could be further evaluated for their potential as Alzheimer's disease therapy.
Collapse
Affiliation(s)
- Silvia Parolo
- Fondazione the Microsoft Research-University of Trento Centre for Computational and Systems Biology (COSBI), 38068, Rovereto, Italy.
| | - Federica Mariotti
- grid.491181.4Fondazione the Microsoft Research-University of Trento Centre for Computational and Systems Biology (COSBI), 38068 Rovereto, Italy
| | - Pranami Bora
- grid.491181.4Fondazione the Microsoft Research-University of Trento Centre for Computational and Systems Biology (COSBI), 38068 Rovereto, Italy
| | - Lucia Carboni
- grid.6292.f0000 0004 1757 1758Department of Pharmacy and Biotechnology, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy
| | - Enrico Domenici
- grid.491181.4Fondazione the Microsoft Research-University of Trento Centre for Computational and Systems Biology (COSBI), 38068 Rovereto, Italy ,grid.11696.390000 0004 1937 0351Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy
| |
Collapse
|