1
|
Liu Y, Ding X, Jia S, Gu X. Current understanding and prospects for targeting neurogenesis in the treatment of cognitive impairment. Neural Regen Res 2026; 21:141-155. [PMID: 39820472 PMCID: PMC12094536 DOI: 10.4103/nrr.nrr-d-24-00802] [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: 07/21/2024] [Revised: 07/24/2024] [Accepted: 10/31/2024] [Indexed: 01/19/2025] Open
Abstract
Adult hippocampal neurogenesis is linked to memory formation in the adult brain, with new neurons in the hippocampus exhibiting greater plasticity during their immature stages compared to mature neurons. Abnormal adult hippocampal neurogenesis is closely associated with cognitive impairment in central nervous system diseases. Targeting and regulating adult hippocampal neurogenesis have been shown to improve cognitive deficits. This review aims to expand the current understanding and prospects of targeting neurogenesis in the treatment of cognitive impairment. Recent research indicates the presence of abnormalities in AHN in several diseases associated with cognitive impairment, including cerebrovascular diseases, Alzheimer's disease, aging-related conditions, and issues related to anesthesia and surgery. The role of these abnormalities in the cognitive deficits caused by these diseases has been widely recognized, and targeting AHN is considered a promising approach for treating cognitive impairment. However, the underlying mechanisms of this role are not yet fully understood, and the effectiveness of targeting abnormal adult hippocampal neurogenesis for treatment remains limited, with a need for further development of treatment methods and detection techniques. By reviewing recent studies, we classify the potential mechanisms of adult hippocampal neurogenesis abnormalities into four categories: immunity, energy metabolism, aging, and pathological states. In immunity-related mechanisms, abnormalities in meningeal, brain, and peripheral immunity can disrupt normal adult hippocampal neurogenesis. Lipid metabolism and mitochondrial function disorders are significant energy metabolism factors that lead to abnormal adult hippocampal neurogenesis. During aging, the inflammatory state of the neurogenic niche and the expression of aging-related microRNAs contribute to reduced adult hippocampal neurogenesis and cognitive impairment in older adult patients. Pathological states of the body and emotional disorders may also result in abnormal adult hippocampal neurogenesis. Among the current strategies used to enhance this form of neurogenesis, physical therapies such as exercise, transcutaneous electrical nerve stimulation, and enriched environments have proven effective. Dietary interventions, including energy intake restriction and nutrient optimization, have shown efficacy in both basic research and clinical trials. However, drug treatments, such as antidepressants and stem cell therapy, are primarily reported in basic research, with limited clinical application. The relationship between abnormal adult hippocampal neurogenesis and cognitive impairment has garnered widespread attention, and targeting the former may be an important strategy for treating the latter. However, the mechanisms underlying abnormal adult hippocampal neurogenesis remain unclear, and treatments are lacking. This highlights the need for greater focus on translating research findings into clinical practice.
Collapse
Affiliation(s)
- Ye Liu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
- Second School of Clinical Medicine of Binzhou Medical University, Yantai, Shandong Province, China
| | - Xibing Ding
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Shushan Jia
- Second School of Clinical Medicine of Binzhou Medical University, Yantai, Shandong Province, China
| | - Xiyao Gu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
- Second School of Clinical Medicine of Binzhou Medical University, Yantai, Shandong Province, China
| |
Collapse
|
2
|
Xiao B, Chu C, Lin Z, Fang T, Zhou Y, Zhang C, Shan J, Chen S, Li L. Treadmill exercise in combination with acousto-optic and olfactory stimulation improves cognitive function in APP/PS1 mice through the brain-derived neurotrophic factor- and Cygb-associated signaling pathways. Neural Regen Res 2025; 20:2706-2726. [PMID: 39105365 PMCID: PMC11801291 DOI: 10.4103/nrr.nrr-d-23-01681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/30/2024] [Accepted: 03/23/2024] [Indexed: 08/07/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202509000-00031/figure1/v/2024-11-05T132919Z/r/image-tiff A reduction in adult neurogenesis is associated with behavioral abnormalities in patients with Alzheimer's disease. Consequently, enhancing adult neurogenesis represents a promising therapeutic approach for mitigating disease symptoms and progression. Nonetheless, non-pharmacological interventions aimed at inducing adult neurogenesis are currently limited. Although individual non-pharmacological interventions, such as aerobic exercise, acousto-optic stimulation, and olfactory stimulation, have shown limited capacity to improve neurogenesis and cognitive function in patients with Alzheimer's disease, the therapeutic effect of a strategy that combines these interventions has not been fully explored. In this study, we observed an age-dependent decrease in adult neurogenesis and a concurrent increase in amyloid-beta accumulation in the hippocampus of amyloid precursor protein/presenilin 1 mice aged 2-8 months. Amyloid deposition became evident at 4 months, while neurogenesis declined by 6 months, further deteriorating as the disease progressed. However, following a 4-week multifactor stimulation protocol, which encompassed treadmill running (46 min/d, 10 m/min, 6 days per week), 40 Hz acousto-optic stimulation (1 hour/day, 6 days/week), and olfactory stimulation (1 hour/day, 6 days/week), we found a significant increase in the number of newborn cells (5'-bromo-2'-deoxyuridine-positive cells), immature neurons (doublecortin-positive cells), newborn immature neurons (5'-bromo-2'-deoxyuridine-positive/doublecortin-positive cells), and newborn astrocytes (5'-bromo-2'-deoxyuridine-positive/glial fibrillary acidic protein-positive cells). Additionally, the amyloid-beta load in the hippocampus decreased. These findings suggest that multifactor stimulation can enhance adult hippocampal neurogenesis and mitigate amyloid-beta neuropathology in amyloid precursor protein/presenilin 1 mice. Furthermore, cognitive abilities were improved, and depressive symptoms were alleviated in amyloid precursor protein/presenilin 1 mice following multifactor stimulation, as evidenced by Morris water maze, novel object recognition, forced swimming test, and tail suspension test results. Notably, the efficacy of multifactor stimulation in consolidating immature neurons persisted for at least 2 weeks after treatment cessation. At the molecular level, multifactor stimulation upregulated the expression of neuron-related proteins (NeuN, doublecortin, postsynaptic density protein-95, and synaptophysin), anti-apoptosis-related proteins (Bcl-2 and PARP), and an autophagy-associated protein (LC3B), while decreasing the expression of apoptosis-related proteins (BAX and caspase-9), in the hippocampus of amyloid precursor protein/presenilin 1 mice. These observations might be attributable to both the brain-derived neurotrophic factor-mediated signaling pathway and antioxidant pathways. Furthermore, serum metabolomics analysis indicated that multifactor stimulation regulated differentially expressed metabolites associated with cell apoptosis, oxidative damage, and cognition. Collectively, these findings suggest that multifactor stimulation is a novel non-invasive approach for the prevention and treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Biao Xiao
- Department of Physiology and Pharmacology, Health Science Center, Ningbo University, Ningbo, Zhejiang Province, China
| | - Chaoyang Chu
- Department of Physiology and Pharmacology, Health Science Center, Ningbo University, Ningbo, Zhejiang Province, China
| | - Zhicheng Lin
- Department of Physiology and Pharmacology, Health Science Center, Ningbo University, Ningbo, Zhejiang Province, China
| | - Tianyuan Fang
- Department of Physiology and Pharmacology, Health Science Center, Ningbo University, Ningbo, Zhejiang Province, China
| | - Yuyu Zhou
- Department of Physiology and Pharmacology, Health Science Center, Ningbo University, Ningbo, Zhejiang Province, China
| | - Chuxia Zhang
- Department of Physiology and Pharmacology, Health Science Center, Ningbo University, Ningbo, Zhejiang Province, China
| | - Jianghui Shan
- Department of Physiology and Pharmacology, Health Science Center, Ningbo University, Ningbo, Zhejiang Province, China
| | - Shiyu Chen
- Department of Physiology and Pharmacology, Health Science Center, Ningbo University, Ningbo, Zhejiang Province, China
| | - Liping Li
- Department of Physiology and Pharmacology, Health Science Center, Ningbo University, Ningbo, Zhejiang Province, China
- Ningbo Key Laboratory of Behavioral Neuroscience, Health Science Center, Ningbo University, Ningbo, Zhejiang Province, China
- Key Laboratory of Addiction Research of Zhejiang Province, Ningbo, Zhejiang Province, China
| |
Collapse
|
3
|
Zheng Q, Nan P, Cui Y, Li L. ConnectomeAE: Multimodal brain connectome-based dual-branch autoencoder and its application in the diagnosis of brain diseases. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2025; 267:108801. [PMID: 40294455 DOI: 10.1016/j.cmpb.2025.108801] [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: 12/19/2024] [Revised: 04/07/2025] [Accepted: 04/22/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND AND OBJECTIVE Exploring the dependencies between multimodal brain networks and integrating node features to enhance brain disease diagnosis remains a significant challenge. Some work has examined only brain connectivity changes in patients, ignoring important information about radiomics features such as shape and texture of individual brain regions in structural images. To this end, this study proposed a novel deep learning approach to integrate multimodal brain connectome information and regional radiomics features for brain disease diagnosis. METHODS A dual-branch autoencoder (ConnectomeAE) based on multimodal brain connectomes was proposed for brain disease diagnosis. Specifically, a matrix of radiomics feature extracted from structural magnetic resonance image (MRI) was used as Rad_AE branch inputs for learning important brain region features. Functional brain network built from functional MRI image was used as inputs to Cycle_AE for capturing brain disease-related connections. By separately learning node features and connection features from multimodal brain networks, the method demonstrates strong adaptability in diagnosing different brain diseases. RESULTS ConnectomeAE was validated on two publicly available datasets. The experimental results show that ConnectomeAE achieved excellent diagnostic performance with an accuracy of 70.7 % for autism spectrum disorder and 90.5 % for Alzheimer's disease. A comparison of training time with other methods indicated that ConnectomeAE exhibits simplicity and efficiency suitable for clinical applications. Furthermore, the interpretability analysis of the model aligned with previous studies, further supporting the biological basis of ConnectomeAE. CONCLUSIONS ConnectomeAE could effectively leverage the complementary information between multimodal brain connectomes for brain disease diagnosis. By separately learning radiomic node features and connectivity features, ConnectomeAE demonstrated good adaptability to different brain disease classification tasks.
Collapse
Affiliation(s)
- Qiang Zheng
- School of Computer and Control Engineering, Yantai University, Yantai, 264005, China
| | - Pengzhi Nan
- School of Computer and Control Engineering, Yantai University, Yantai, 264005, China
| | - Yongchao Cui
- Faculty of Computing and Information Technology, Tunku Abdul Rahman University of Management and Technology, 53300, Kuala Lumpur, Malaysia
| | - Lin Li
- Department of Radiology, Yantaishan Hospital Affiliated to Binzhou Medical University, Yantai 264003, China.
| |
Collapse
|
4
|
Yadava S, Reddy DH, Nakka VP, Anusha VL, Dumala N, Viswanadh MK, Chakravarthi G, Nalluri BN, Ramakrishna K. Unravelling neuroregenerative and neuroprotective roles of Wnt/β-catenin pathway in ischemic stroke: Insights into molecular mechanisms. Neuroscience 2025; 565:527-547. [PMID: 39681254 DOI: 10.1016/j.neuroscience.2024.12.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/07/2024] [Accepted: 12/12/2024] [Indexed: 12/18/2024]
Abstract
Stroke is a serious condition often resulting in mortality or long-term disability, causing cognitive, memory, and motor impairments. A reduction in cerebral blood flow below critical levels defines the ischemic core and penumbra: the core undergoes irreversible damage, while the penumbra remains viable but functionally impaired. This functional impairment activates complex cell signaling pathways that determine cell survival or death, making the penumbra a key target for therapeutic interventions to prevent further damage. The Wnt/β-catenin (WβC) signaling pathway has emerged as a potential neuroprotective mechanism, promoting neurogenesis, angiogenesis, neuronal connectivity, and maintaining blood-brain barrier integrity after stroke. Activation of the WβC pathway also mitigates oxidative stress, inflammation, and apoptosis in ischemic regions, enhancing its neuroprotective effects. However, the overexpression of GSK3β and DKK1, or the presence of their agonists, can counteract these benefits. This review explores the therapeutic potential of WβC signaling, highlighting the effects of pharmacological modulation through antagonists, agonists, synthetic chemicals, natural products, stem cells, and macromolecules in preclinical models of ischemic stroke. While preclinical evidence supports the benefits of WβC activation, its role in human stroke requires further investigation. Additionally, the review discusses the potential adverse effects of prolonged WβC activation and suggests strategies to mitigate them. Overall, WβC signaling holds promise as a therapeutic target, offering insights into stroke pathophysiology and informing the development of novel treatment strategies.
Collapse
Affiliation(s)
- Srikanth Yadava
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram, India.
| | | | - Venkata Prasuja Nakka
- Department of Systems and Computational Biology, School of Life Sciences, University of Hyderabad, 500046, India.
| | | | - Naresh Dumala
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram, India.
| | - Matte Kasi Viswanadh
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram, India.
| | | | - Buchi N Nalluri
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram, India
| | - Kakarla Ramakrishna
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram, India.
| |
Collapse
|
5
|
Chen P, Wu L, Lei J, Chen F, Feng L, Liu G, Zhou B. The ellagitannin metabolite urolithin C attenuated cognitive impairment by inhibiting neuroinflammation via downregulation of MAPK/NF-kB signaling pathways in aging mice. Int Immunopharmacol 2024; 142:113151. [PMID: 39303538 DOI: 10.1016/j.intimp.2024.113151] [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: 05/13/2024] [Revised: 09/09/2024] [Accepted: 09/09/2024] [Indexed: 09/22/2024]
Abstract
The current study aimed to evaluate the preventive effects of urolithin C (Uro C), a gut microbial metabolite of ellagitannins on D-galactose (D-gal)-induced brain damage during the aging process and to elucidate the underlying mechanisms. In our study, the protective effect of Uro C on D-gal-induced BV2 microglia cell-mediated neuroinflammation damage in primary cortical neurons in vitro was confirmed. The results in an aging model in vivo induced by D-gal demonstrated that Uro C prevented D-gal-induced memory impairment, long-term potentiation (LTP) damage, and synaptic dysfunction through behavioral, electrophysiological, and histological examinations. Additionally, amyloidogenesis was observed in the central nervous system. The findings indicated that Uro C exhibited a preventive effect on the D-gal-induced elevation of β-amyloid (1-42 specific) (Aβ1-42) accumulation, APP levels, ABCE1 levels, and the equilibrium of the cholinergic system in the aging mouse brain. Moreover, Uro C demonstrated downregulation of D-gal-induced glial overactivation through inhibition of the MAPK/NF-kB pathway. This resulted in the regulation of inflammatory mediators and cytokines, including iNOS, IL-6, IL-1β, and TNF-ɑ, in the mouse brain and BV2 microglial cells. Taken together, our results suggested that Uro C treatment could effectively mitigate the D-gal-induced memory impairment and amyloidogenesis, and the underlying mechanism might be tightly related to the improvement of neuroinflammation by suppressing the MAPK/NF-kB pathway, indicating Uro C might be an alternative and promising agent for the treatment of aging and age-associated brain diseases.
Collapse
Affiliation(s)
- Peng Chen
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, Hubei.
| | - Lining Wu
- Department of Pharmacy, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi
| | - Jiexin Lei
- Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Fuchao Chen
- Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, PR China
| | - Lihua Feng
- School of Pharmacy, Hubei University of Traditional Chinese Medicine, Wuhan, PR China
| | - Gang Liu
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, Hubei
| | - Benhong Zhou
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, Hubei
| |
Collapse
|
6
|
Li J, Ding Y, Zhang J, Zhang Y, Cui Y, Zhang Y, Chang S, Chang Y, Gao G. Iron overload suppresses hippocampal neurogenesis in adult mice: Implication for iron dysregulation-linked neurological diseases. CNS Neurosci Ther 2024; 30:e14394. [PMID: 37545321 PMCID: PMC10848078 DOI: 10.1111/cns.14394] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 06/24/2023] [Accepted: 07/26/2023] [Indexed: 08/08/2023] Open
Abstract
AIMS Adult hippocampal neurogenesis is an important player in brain homeostasis and its impairment participates in neurological diseases. Iron overload has emerged as an irreversible factor of brain aging, and is also closely related to degenerative disorders, including cognitive dysfunction. However, whether brain iron overload alters hippocampal neurogenesis has not been reported. We investigated the effect of elevated iron content on adult hippocampal neurogenesis and explored the underlying mechanism. METHODS Mouse models with hippocampal iron overload were generated. Neurogenesis in hippocampus and expression levels of related molecules were assessed. RESULTS Iron accumulation in hippocampus remarkably impaired the differentiation of neural stem cells, resulting in a significant decrease in newborn neurons. The damage was possibly attributed to iron-induced downregulation of proprotein convertase furin and subsequently decreased maturation of brain-derived neurotrophic factor (BDNF), thus contributing to memory decline and anxiety-like behavior of mice. Supportively, knockdown of furin indeed suppressed hippocampal neurogenesis, while furin overexpression restored the impairment. CONCLUSION These findings demonstrated that iron overload damaged hippocampal neurogenesis likely via iron-furin-BDNF pathway. This study provides new insights into potential mechanisms on iron-induced neurotoxicity and the causes of neurogenesis injury and renders modulating iron homeostasis and furin expression as novel therapeutic strategies for treatment of neurological diseases.
Collapse
Affiliation(s)
- Jie Li
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Molecular and Cellular Biology of the Ministry of Education, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life SciencesHebei Normal UniversityShijiazhuangChina
| | - Yiqian Ding
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Molecular and Cellular Biology of the Ministry of Education, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life SciencesHebei Normal UniversityShijiazhuangChina
| | - Jianhua Zhang
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Molecular and Cellular Biology of the Ministry of Education, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life SciencesHebei Normal UniversityShijiazhuangChina
| | - Yating Zhang
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Molecular and Cellular Biology of the Ministry of Education, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life SciencesHebei Normal UniversityShijiazhuangChina
| | - Yiduo Cui
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Molecular and Cellular Biology of the Ministry of Education, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life SciencesHebei Normal UniversityShijiazhuangChina
| | - Yi Zhang
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Molecular and Cellular Biology of the Ministry of Education, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life SciencesHebei Normal UniversityShijiazhuangChina
| | - Shiyang Chang
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Molecular and Cellular Biology of the Ministry of Education, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life SciencesHebei Normal UniversityShijiazhuangChina
- College of Basic MedicineHebei Medical UniversityShijiazhuangChina
| | - Yan‐Zhong Chang
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Molecular and Cellular Biology of the Ministry of Education, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life SciencesHebei Normal UniversityShijiazhuangChina
| | - Guofen Gao
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Molecular and Cellular Biology of the Ministry of Education, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life SciencesHebei Normal UniversityShijiazhuangChina
| |
Collapse
|
7
|
Liu Y, Tan Y, Zhang Z, Yi M, Zhu L, Peng W. The interaction between ageing and Alzheimer's disease: insights from the hallmarks of ageing. Transl Neurodegener 2024; 13:7. [PMID: 38254235 PMCID: PMC10804662 DOI: 10.1186/s40035-024-00397-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 12/31/2023] [Accepted: 01/02/2024] [Indexed: 01/24/2024] Open
Abstract
Ageing is a crucial risk factor for Alzheimer's disease (AD) and is characterised by systemic changes in both intracellular and extracellular microenvironments that affect the entire body instead of a single organ. Understanding the specific mechanisms underlying the role of ageing in disease development can facilitate the treatment of ageing-related diseases, such as AD. Signs of brain ageing have been observed in both AD patients and animal models. Alleviating the pathological changes caused by brain ageing can dramatically ameliorate the amyloid beta- and tau-induced neuropathological and memory impairments, indicating that ageing plays a crucial role in the pathophysiological process of AD. In this review, we summarize the impact of several age-related factors on AD and propose that preventing pathological changes caused by brain ageing is a promising strategy for improving cognitive health.
Collapse
Affiliation(s)
- Yuqing Liu
- Department of Integrated Traditional Chinese and Western Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China
- National Clinical Research Center for Metabolic Diseases, Changsha, 410011, People's Republic of China
| | - Yejun Tan
- School of Mathematics, University of Minnesota Twin Cities, Minneapolis, MN, 55455, USA
| | - Zheyu Zhang
- Department of Integrated Traditional Chinese and Western Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China
- National Clinical Research Center for Metabolic Diseases, Changsha, 410011, People's Republic of China
| | - Min Yi
- Department of Integrated Traditional Chinese and Western Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China
- National Clinical Research Center for Metabolic Diseases, Changsha, 410011, People's Republic of China
| | - Lemei Zhu
- Academician Workstation, Changsha Medical University, Changsha, 410219, People's Republic of China
| | - Weijun Peng
- Department of Integrated Traditional Chinese and Western Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China.
- National Clinical Research Center for Metabolic Diseases, Changsha, 410011, People's Republic of China.
| |
Collapse
|
8
|
Bou Ghanem A, Hussayni Y, Kadbey R, Ratel Y, Yehya S, Khouzami L, Ghadieh HE, Kanaan A, Azar S, Harb F. Exploring the complexities of 1C metabolism: implications in aging and neurodegenerative diseases. Front Aging Neurosci 2024; 15:1322419. [PMID: 38239489 PMCID: PMC10794399 DOI: 10.3389/fnagi.2023.1322419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 12/11/2023] [Indexed: 01/22/2024] Open
Abstract
The intricate interplay of one-carbon metabolism (OCM) with various cellular processes has garnered substantial attention due to its fundamental implications in several biological processes. OCM serves as a pivotal hub for methyl group donation in vital biochemical reactions, influencing DNA methylation, protein synthesis, and redox balance. In the context of aging, OCM dysregulation can contribute to epigenetic modifications and aberrant redox states, accentuating cellular senescence and age-associated pathologies. Furthermore, OCM's intricate involvement in cancer progression is evident through its capacity to provide essential one-carbon units crucial for nucleotide synthesis and DNA methylation, thereby fueling uncontrolled cell proliferation and tumor development. In neurodegenerative disorders like Alzheimer's and Parkinson's, perturbations in OCM pathways are implicated in the dysregulation of neurotransmitter synthesis and mitochondrial dysfunction, contributing to disease pathophysiology. This review underscores the profound impact of OCM in diverse disease contexts, reinforcing the need for a comprehensive understanding of its molecular complexities to pave the way for targeted therapeutic interventions across inflammation, aging and neurodegenerative disorders.
Collapse
Affiliation(s)
- Ayman Bou Ghanem
- Faculty of Medicine and Medical Sciences, University of Balamand, Tripoli, Lebanon
| | - Yaman Hussayni
- Faculty of Medicine and Medical Sciences, University of Balamand, Tripoli, Lebanon
| | - Raghid Kadbey
- Faculty of Medicine and Medical Sciences, University of Balamand, Tripoli, Lebanon
| | - Yara Ratel
- Faculty of Medicine and Medical Sciences, University of Balamand, Tripoli, Lebanon
| | - Shereen Yehya
- Faculty of Medicine and Medical Sciences, University of Balamand, Tripoli, Lebanon
| | - Lara Khouzami
- College of Natural and Health Sciences, Zayed University, Dubai, United Arab Emirates
| | - Hilda E. Ghadieh
- Faculty of Medicine and Medical Sciences, University of Balamand, Tripoli, Lebanon
- AUB Diabetes, American University of Beirut Medical Center, Beirut, Lebanon
| | - Amjad Kanaan
- Faculty of Medicine and Medical Sciences, University of Balamand, Tripoli, Lebanon
| | - Sami Azar
- Faculty of Medicine and Medical Sciences, University of Balamand, Tripoli, Lebanon
| | - Frederic Harb
- Faculty of Medicine and Medical Sciences, University of Balamand, Tripoli, Lebanon
- AUB Diabetes, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
9
|
Echeverria V, Mendoza C, Iarkov A. Nicotinic acetylcholine receptors and learning and memory deficits in Neuroinflammatory diseases. Front Neurosci 2023; 17:1179611. [PMID: 37255751 PMCID: PMC10225599 DOI: 10.3389/fnins.2023.1179611] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 04/07/2023] [Indexed: 06/01/2023] Open
Abstract
Animal survival depends on cognitive abilities such as learning and memory to adapt to environmental changes. Memory functions require an enhanced activity and connectivity of a particular arrangement of engram neurons, supported by the concerted action of neurons, glia, and vascular cells. The deterioration of the cholinergic system is a common occurrence in neurological conditions exacerbated by aging such as traumatic brain injury (TBI), posttraumatic stress disorder (PTSD), Alzheimer's disease (AD), and Parkinson's disease (PD). Cotinine is a cholinergic modulator with neuroprotective, antidepressant, anti-inflammatory, antioxidant, and memory-enhancing effects. Current evidence suggests Cotinine's beneficial effects on cognition results from the positive modulation of the α7-nicotinic acetylcholine receptors (nAChRs) and the inhibition of the toll-like receptors (TLRs). The α7nAChR affects brain functions by modulating the function of neurons, glia, endothelial, immune, and dendritic cells and regulates inhibitory and excitatory neurotransmission throughout the GABA interneurons. In addition, Cotinine acting on the α7 nAChRs and TLR reduces neuroinflammation by inhibiting the release of pro-inflammatory cytokines by the immune cells. Also, α7nAChRs stimulate signaling pathways supporting structural, biochemical, electrochemical, and cellular changes in the Central nervous system during the cognitive processes, including Neurogenesis. Here, the mechanisms of memory formation as well as potential mechanisms of action of Cotinine on memory preservation in aging and neurological diseases are discussed.
Collapse
Affiliation(s)
- Valentina Echeverria
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Concepción, Chile
- Research and Development Department, Bay Pines VAHCS, Bay Pines, FL, United States
| | - Cristhian Mendoza
- Facultad de Odontologia y Ciencias de la Rehabilitacion, Universidad San Sebastián, Concepción, Chile
| | - Alex Iarkov
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Concepción, Chile
| |
Collapse
|
10
|
Gulyaeva NV. Glucocorticoids Orchestrate Adult Hippocampal Plasticity: Growth Points and Translational Aspects. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:565-589. [PMID: 37331704 DOI: 10.1134/s0006297923050012] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/10/2023] [Accepted: 04/10/2023] [Indexed: 06/20/2023]
Abstract
The review analyzes modern concepts about the control of various mechanisms of the hippocampal neuroplasticity in adult mammals and humans by glucocorticoids. Glucocorticoid hormones ensure the coordinated functioning of key components and mechanisms of hippocampal plasticity: neurogenesis, glutamatergic neurotransmission, microglia and astrocytes, systems of neurotrophic factors, neuroinflammation, proteases, metabolic hormones, neurosteroids. Regulatory mechanisms are diverse; along with the direct action of glucocorticoids through their receptors, there are conciliated glucocorticoid-dependent effects, as well as numerous interactions between various systems and components. Despite the fact that many connections in this complex regulatory scheme have not yet been established, the study of the factors and mechanisms considered in the work forms growth points in the field of glucocorticoid-regulated processes in the brain and primarily in the hippocampus. These studies are fundamentally important for the translation into the clinic and the potential treatment/prevention of common diseases of the emotional and cognitive spheres and respective comorbid conditions.
Collapse
Affiliation(s)
- Natalia V Gulyaeva
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, 117485, Russia.
- Research and Clinical Center for Neuropsychiatry of Moscow Healthcare Department, Moscow, 115419, Russia
| |
Collapse
|
11
|
Jiang M, Jang SE, Zeng L. The Effects of Extrinsic and Intrinsic Factors on Neurogenesis. Cells 2023; 12:cells12091285. [PMID: 37174685 PMCID: PMC10177620 DOI: 10.3390/cells12091285] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/18/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
In the mammalian brain, neurogenesis is maintained throughout adulthood primarily in two typical niches, the subgranular zone (SGZ) of the dentate gyrus and the subventricular zone (SVZ) of the lateral ventricles and in other nonclassic neurogenic areas (e.g., the amygdala and striatum). During prenatal and early postnatal development, neural stem cells (NSCs) differentiate into neurons and migrate to appropriate areas such as the olfactory bulb where they integrate into existing neural networks; these phenomena constitute the multistep process of neurogenesis. Alterations in any of these processes impair neurogenesis and may even lead to brain dysfunction, including cognitive impairment and neurodegeneration. Here, we first summarize the main properties of mammalian neurogenic niches to describe the cellular and molecular mechanisms of neurogenesis. Accumulating evidence indicates that neurogenesis plays an integral role in neuronal plasticity in the brain and cognition in the postnatal period. Given that neurogenesis can be highly modulated by a number of extrinsic and intrinsic factors, we discuss the impact of extrinsic (e.g., alcohol) and intrinsic (e.g., hormones) modulators on neurogenesis. Additionally, we provide an overview of the contribution of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection to persistent neurological sequelae such as neurodegeneration, neurogenic defects and accelerated neuronal cell death. Together, our review provides a link between extrinsic/intrinsic factors and neurogenesis and explains the possible mechanisms of abnormal neurogenesis underlying neurological disorders.
Collapse
Affiliation(s)
- Mei Jiang
- Department of Human Anatomy, Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Dongguan Campus, Guangdong Medical University, Dongguan 523808, China
| | - Se Eun Jang
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore 308433, Singapore
| | - Li Zeng
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore 308433, Singapore
- Neuroscience and Behavioral Disorders Program, DUKE-NUS Graduate Medical School, Singapore 169857, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technology University, Novena Campus, 11 Mandalay Road, Singapore 308232, Singapore
| |
Collapse
|
12
|
Tan CH, Tan JJX. Low neighborhood deprivation buffers against hippocampal neurodegeneration, white matter hyperintensities, and poorer cognition. GeroScience 2023:10.1007/s11357-023-00780-y. [PMID: 37004594 PMCID: PMC10400521 DOI: 10.1007/s11357-023-00780-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 03/20/2023] [Indexed: 04/04/2023] Open
Abstract
There is increasing recognition that socioeconomic inequalities contribute to disparities in brain and cognitive health in older adults. However, whether neighborhood socioeconomic status (SES) buffers individuals with low individual SES against neurodegeneration, cerebrovascular disease, and poorer cognitive function is not well understood. Here, we evaluated whether neighborhood deprivation (Townsend deprivation index) interacted with individual SES (composite household income and education levels) on hippocampus volume, regional cortical thickness, white matter hyperintensities, and cognition in 19,638 individuals (mean age = 54.8) from the UK Biobank. We found that individuals with low individual SES had the smallest hippocampal volumes, greatest white matter hyperintensity burden, and poorest cognition if they were living in high deprivation neighborhoods but that these deleterious effects on brain and cognitive function were attenuated if they were living in low deprivation neighborhoods (p for interactions < .05). While neighborhood deprivation did not interact with individual SES to influence regional cortical thickness, higher neighborhood deprivation was independently associated with lower cortical thickness in 16 regions (false discovery rate q < .05). Across multiple brain indices and cognitive function analyses, we found converging evidence suggesting that low neighborhood deprivation may have a neuroprotective effect against neurodegeneration, cerebrovascular pathology, and cognitive impairment, particularly in vulnerable individuals with low household income and education levels.
Collapse
Affiliation(s)
- Chin Hong Tan
- Department of Psychology, Nanyang Technological University, 48 Nanyang Avenue, Singapore, S639818, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University, 48 Nanyang Avenue, Singapore, S639818, Singapore.
| | - Jacinth J X Tan
- School of Social Sciences, Singapore Management University, Singapore, Singapore
| |
Collapse
|
13
|
Gulino R. Synaptic Dysfunction and Plasticity in Amyotrophic Lateral Sclerosis. Int J Mol Sci 2023; 24:ijms24054613. [PMID: 36902042 PMCID: PMC10003601 DOI: 10.3390/ijms24054613] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 03/03/2023] Open
Abstract
Recent evidence has supported the hypothesis that amyotrophic lateral sclerosis (ALS) is a multi-step disease, as the onset of symptoms occurs after sequential exposure to a defined number of risk factors. Despite the lack of precise identification of these disease determinants, it is known that genetic mutations may contribute to one or more of the steps leading to ALS onset, the remaining being linked to environmental factors and lifestyle. It also appears evident that compensatory plastic changes taking place at all levels of the nervous system during ALS etiopathogenesis may likely counteract the functional effects of neurodegeneration and affect the timing of disease onset and progression. Functional and structural events of synaptic plasticity probably represent the main mechanisms underlying this adaptive capability, causing a significant, although partial and transient, resiliency of the nervous system affected by a neurodegenerative disease. On the other hand, the failure of synaptic functions and plasticity may be part of the pathological process. The aim of this review was to summarize what it is known today about the controversial involvement of synapses in ALS etiopathogenesis, and an analysis of the literature, although not exhaustive, confirmed that synaptic dysfunction is an early pathogenetic process in ALS. Moreover, it appears that adequate modulation of structural and functional synaptic plasticity may likely support function sparing and delay disease progression.
Collapse
Affiliation(s)
- Rosario Gulino
- Department of Biomedical and Biotechnological Sciences, Physiology Section, University of Catania, 95123 Catania, Italy
| |
Collapse
|
14
|
Galvano E, Pandit H, Sepulveda J, Ng CAS, Becher MK, Mandelblatt JS, Van Dyk K, Rebeck GW. Behavioral and transcriptomic effects of the cancer treatment tamoxifen in mice. Front Neurosci 2023; 17:1068334. [PMID: 36845433 PMCID: PMC9951777 DOI: 10.3389/fnins.2023.1068334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 01/24/2023] [Indexed: 02/12/2023] Open
Abstract
Introduction Tamoxifen is a common treatment for estrogen receptor-positive breast cancer. While tamoxifen treatment is generally accepted as safe, there are concerns about adverse effects on cognition. Methods We used a mouse model of chronic tamoxifen exposure to examine the effects of tamoxifen on the brain. Female C57/BL6 mice were exposed to tamoxifen or vehicle control for six weeks; brains of 15 mice were analyzed for tamoxifen levels and transcriptomic changes, and an additional 32 mice were analyzed through a battery of behavioral tests. Results Tamoxifen and its metabolite 4-OH-tamoxifen were found at higher levels in the brain than in the plasma, demonstrating the facile entry of tamoxifen into the CNS. Behaviorally, tamoxifen-exposed mice showed no impairment in assays related to general health, exploration, motor function, sensorimotor gating, and spatial learning. Tamoxifen-treated mice showed a significantly increased freezing response in a fear conditioning paradigm, but no effects on anxiety measures in the absence of stressors. RNA sequencing analysis of whole hippocampi showed tamoxifen-induced reductions in gene pathways related to microtubule function, synapse regulation, and neurogenesis. Discussion These findings of the effects of tamoxifen exposure on fear conditioning and on gene expression related to neuronal connectivity suggest that there may be CNS side effects of this common breast cancer treatment.
Collapse
Affiliation(s)
- Elena Galvano
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, United States
| | - Harshul Pandit
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, United States
| | - Jordy Sepulveda
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States
| | - Christi Anne S. Ng
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, United States
| | - Melanie K. Becher
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, United States
| | - Jeanne S. Mandelblatt
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, United States
| | - Kathleen Van Dyk
- Department of Psychiatry, UCLA Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA, United States
| | - G. William Rebeck
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, United States
| |
Collapse
|