1
|
Gao B, Wang H, Hu S, Zhong K, Liu X, Deng Z, Li Y, Tong A, Zhou L. Sox2-overexpressing neural stem cells alleviate ventricular enlargement and neurological dysfunction in posthemorrhagic hydrocephalus. Neural Regen Res 2026; 21:769-779. [PMID: 40326987 DOI: 10.4103/nrr.nrr-d-24-01491] [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: 11/29/2024] [Accepted: 03/17/2025] [Indexed: 05/07/2025] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202602000-00045/figure1/v/2025-05-05T160104Z/r/image-tiff Neural stem cells (NSCs) have the potential for self-renewal and multidirectional differentiation, and their transplantation has achieved good efficacy in a variety of diseases. However, only 1%-10% of transplanted NSCs survive in the ischemic and hypoxic microenvironment of posthemorrhagic hydrocephalus. Sox2 is an important factor for NSCs to maintain proliferation. Therefore, Sox2-overexpressing NSCs (NSCSox2) may be more successful in improving neurological dysfunction after posthemorrhagic hydrocephalus. In this study, human NSCSox2 was transplanted into a posthemorrhagic hydrocephalus mouse model, and retinoic acid was administered to further promote NSC differentiation. The results showed that NSCSox2 attenuated the ventricular enlargement caused by posthemorrhagic hydrocephalus and improved neurological function. NSCSox2 also promoted nerve regeneration, inhibited neuroinflammation and promoted M2 polarization (anti-inflammatory phenotype), thereby reducing cerebrospinal fluid secretion in choroid plexus. These findings suggest that NSCSox2 rescued ventricular enlargement and neurological dysfunction induced by posthemorrhagic hydrocephalus through neural regeneration and modulation of inflammation.
Collapse
Affiliation(s)
- Baocheng Gao
- Department of Neurosurgery, West China Medical School, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Department of Neurosurgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan Province, China
| | - Haoxiang Wang
- Department of Neurosurgery, West China Medical School, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Shuang Hu
- Department of Otolaryngology & Head and Neck Surgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan Province, China
| | - Kunhong Zhong
- Department of Neurosurgery, West China Medical School, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Xiaoyin Liu
- Department of Neurosurgery, West China Medical School, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ziang Deng
- Department of Neurosurgery, West China Medical School, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yuanyou Li
- Department of Neurosurgery, West China Medical School, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Aiping Tong
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Chengdu, Sichuan Province, China
| | - Liangxue Zhou
- Department of Neurosurgery, West China Medical School, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Department of Neurosurgery, NHC Key Laboratory of Nuclear Technology Medical Transformation (Mianyang Central Hospital),School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan Province, China
- Department of Neurosurgery, Fifth People's Hospital of Ningxia Hui Autonomous Region, Shizuishan, Ningxia Hui Autonomous Region, China
| |
Collapse
|
2
|
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
|
3
|
Geng R, Wang Y, Wang R, Wu J, Bao X. Enhanced neurogenesis after ischemic stroke: The interplay between endogenous and exogenous stem cells. Neural Regen Res 2026; 21:212-223. [PMID: 39820432 PMCID: PMC12094570 DOI: 10.4103/nrr.nrr-d-24-00879] [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: 08/28/2024] [Revised: 09/02/2024] [Accepted: 11/26/2024] [Indexed: 01/19/2025] Open
Abstract
Ischemic stroke is a significant global health crisis, frequently resulting in disability or death, with limited therapeutic interventions available. Although various intrinsic reparative processes are initiated within the ischemic brain, these mechanisms are often insufficient to restore neuronal functionality. This has led to intensive investigation into the use of exogenous stem cells as a potential therapeutic option. This comprehensive review outlines the ontogeny and mechanisms of activation of endogenous neural stem cells within the adult brain following ischemic events, with focus on the impact of stem cell-based therapies on neural stem cells. Exogenous stem cells have been shown to enhance the proliferation of endogenous neural stem cells via direct cell-to-cell contact and through the secretion of growth factors and exosomes. Additionally, implanted stem cells may recruit host stem cells from their niches to the infarct area by establishing so-called "biobridges." Furthermore, xenogeneic and allogeneic stem cells can modify the microenvironment of the infarcted brain tissue through immunomodulatory and angiogenic effects, thereby supporting endogenous neuroregeneration. Given the convergence of regulatory pathways between exogenous and endogenous stem cells and the necessity for a supportive microenvironment, we discuss three strategies to simultaneously enhance the therapeutic efficacy of both cell types. These approaches include: (1) co-administration of various growth factors and pharmacological agents alongside stem cell transplantation to reduce stem cell apoptosis; (2) synergistic administration of stem cells and their exosomes to amplify paracrine effects; and (3) integration of stem cells within hydrogels, which provide a protective scaffold for the implanted cells while facilitating the regeneration of neural tissue and the reconstitution of neural circuits. This comprehensive review highlights the interactions and shared regulatory mechanisms between endogenous neural stem cells and exogenously implanted stem cells and may offer new insights for improving the efficacy of stem cell-based therapies in the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Ruxu Geng
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yuhe Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Renzhi Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jun Wu
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| | - Xinjie Bao
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing, China
| |
Collapse
|
4
|
Zhou HY, Wang X, Li Y, Wang D, Zhou XZ, Xiao N, Li GX, Li G. Dynamic development of microglia and macrophages after spinal cord injury. Neural Regen Res 2025; 20:3606-3619. [PMID: 39101644 PMCID: PMC11974661 DOI: 10.4103/nrr.nrr-d-24-00063] [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: 01/17/2024] [Revised: 04/09/2024] [Accepted: 05/28/2024] [Indexed: 08/06/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202512000-00029/figure1/v/2025-01-31T122243Z/r/image-tiff Secondary injury following spinal cord injury is primarily characterized by a complex inflammatory response, with resident microglia and infiltrating macrophages playing pivotal roles. While previous studies have grouped these two cell types together based on similarities in structure and function, an increasing number of studies have demonstrated that microglia and macrophages exhibit differences in structure and function and have different effects on disease processes. In this study, we used single-cell RNA sequencing and spatial transcriptomics to identify the distinct evolutionary paths of microglia and macrophages following spinal cord injury. Our results showed that microglia were activated to a pro-inflammatory phenotype immediately after spinal cord injury, gradually transforming to an anti-inflammatory steady state phenotype as the disease progressed. Regarding macrophages, our findings highlighted abundant communication with other cells, including fibroblasts and neurons. Both pro-inflammatory and neuroprotective effects of macrophages were also identified; the pro-inflammatory effect may be related to integrin β2 ( Itgb2 ) and the neuroprotective effect may be related to the oncostatin M pathway. These findings were validated by in vivo experiments. This research underscores differences in the cellular dynamics of microglia and macrophages following spinal cord injury, and may offer new perspectives on inflammatory mechanisms and potential therapeutic targets.
Collapse
Affiliation(s)
- Hu-Yao Zhou
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
- Department of Rehabilitation, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing, China
| | - Xia Wang
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
- Department of Rehabilitation, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing, China
| | - Yi Li
- Department of Rehabilitation, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing, China
| | - Duan Wang
- Department of Rehabilitation, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing, China
| | - Xuan-Zi Zhou
- Department of Rehabilitation, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing, China
| | - Nong Xiao
- Department of Rehabilitation, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing, China
| | - Guo-Xing Li
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Gang Li
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
- Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing, China
| |
Collapse
|
5
|
Spicer MM, Yang J, Fu D, DeVore AN, Lauffer M, Atasoy NS, Atasoy D, Fisher RA. Regulator of G protein signaling 6 mediates exercise-induced recovery of hippocampal neurogenesis, learning, and memory in a mouse model of Alzheimer's disease. Neural Regen Res 2025; 20:2969-2981. [PMID: 39248184 PMCID: PMC11826473 DOI: 10.4103/nrr.nrr-d-23-01993] [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: 12/05/2023] [Revised: 07/16/2024] [Accepted: 08/19/2024] [Indexed: 09/10/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202510000-00027/figure1/v/2024-11-26T163120Z/r/image-tiff Hippocampal neuronal loss causes cognitive dysfunction in Alzheimer's disease. Adult hippocampal neurogenesis is reduced in patients with Alzheimer's disease. Exercise stimulates adult hippocampal neurogenesis in rodents and improves memory and slows cognitive decline in patients with Alzheimer's disease. However, the molecular pathways for exercise-induced adult hippocampal neurogenesis and improved cognition in Alzheimer's disease are poorly understood. Recently, regulator of G protein signaling 6 (RGS6) was identified as the mediator of voluntary running-induced adult hippocampal neurogenesis in mice. Here, we generated novel RGS6 fl/fl ; APP SWE mice and used retroviral approaches to examine the impact of RGS6 deletion from dentate gyrus neuronal progenitor cells on voluntary running-induced adult hippocampal neurogenesis and cognition in an amyloid-based Alzheimer's disease mouse model. We found that voluntary running in APP SWE mice restored their hippocampal cognitive impairments to that of control mice. This cognitive rescue was abolished by RGS6 deletion in dentate gyrus neuronal progenitor cells, which also abolished running-mediated increases in adult hippocampal neurogenesis. Adult hippocampal neurogenesis was reduced in sedentary APP SWE mice versus control mice, with basal adult hippocampal neurogenesis reduced by RGS6 deletion in dentate gyrus neural precursor cells. RGS6 was expressed in neurons within the dentate gyrus of patients with Alzheimer's disease with significant loss of these RGS6-expressing neurons. Thus, RGS6 mediated voluntary running-induced rescue of impaired cognition and adult hippocampal neurogenesis in APP SWE mice, identifying RGS6 in dentate gyrus neural precursor cells as a possible therapeutic target in Alzheimer's disease.
Collapse
Affiliation(s)
- Mackenzie M. Spicer
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Molecular Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Jianqi Yang
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Daniel Fu
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Alison N. DeVore
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Marisol Lauffer
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Neural Circuits and Behavior Core, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Nilufer S. Atasoy
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Deniz Atasoy
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Rory A. Fisher
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| |
Collapse
|
6
|
Zhang Y, Liang Y, Gu Y. The dopaminergic system and Alzheimer's disease. Neural Regen Res 2025; 20:2495-2512. [PMID: 39314145 PMCID: PMC11801300 DOI: 10.4103/nrr.nrr-d-24-00230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 06/21/2024] [Accepted: 07/31/2024] [Indexed: 09/25/2024] Open
Abstract
Alzheimer's disease is a common neurodegenerative disorder in older adults. Despite its prevalence, its pathogenesis remains unclear. In addition to the most widely accepted causes, which include excessive amyloid-beta aggregation, tau hyperphosphorylation, and deficiency of the neurotransmitter acetylcholine, numerous studies have shown that the dopaminergic system is also closely associated with the occurrence and development of this condition. Dopamine is a crucial catecholaminergic neurotransmitter in the human body. Dopamine-associated treatments, such as drugs that target dopamine receptor D and dopamine analogs, can improve cognitive function and alleviate psychiatric symptoms as well as ameliorate other clinical manifestations. However, therapeutics targeting the dopaminergic system are associated with various adverse reactions, such as addiction and exacerbation of cognitive impairment. This review summarizes the role of the dopaminergic system in the pathology of Alzheimer's disease, focusing on currently available dopamine-based therapies for this disorder and the common side effects associated with dopamine-related drugs. The aim of this review is to provide insights into the potential connections between the dopaminergic system and Alzheimer's disease, thus helping to clarify the mechanisms underlying the condition and exploring more effective therapeutic options.
Collapse
Affiliation(s)
- Yuhan Zhang
- International Medical College, Chongqing Medical University, Chongqing, China
| | - Yuan Liang
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Yixue Gu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| |
Collapse
|
7
|
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: 3] [Impact Index Per Article: 3.0] [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
|
8
|
Ishihara Y, Ando M, Goto Y, Kotani S, Watanabe N, Nakatani Y, Ishii S, Miyamoto N, Mano Y, Ishikawa Y. A novel selective phosphodiesterase 9 inhibitor, irsenontrine (E2027), enhances GluA1 phosphorylation in neurons and improves learning and memory via cyclic GMP elevation. Neuropharmacology 2025; 273:110428. [PMID: 40147639 DOI: 10.1016/j.neuropharm.2025.110428] [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: 12/06/2024] [Revised: 03/22/2025] [Accepted: 03/22/2025] [Indexed: 03/29/2025]
Abstract
Phosphodiesterase 9 (PDE9) plays a critical role in synaptic plasticity and cognitive function by modulating cyclic GMP (cGMP). Many reports have shown that PDE9 inhibition improves cognitive function and synaptic plasticity in rodents. Several studies have found that the NO/cGMP/PKG pathway is downregulated in patients with Alzheimer's disease (AD) or dementia with Lewy bodies (DLB) and in older individuals. A PDE9 inhibitor could therefore be a potential therapeutic approach for improving cognitive dysfunction in dementia, including in AD and DLB. We previously discovered a novel PDE9 inhibitor, irsenontrine (E2027). In the current study, irsenontrine showed highly selective affinity for PDE9 with more than 1800-fold selectivity over other PDEs. Irsenontrine maleate significantly increased intracellular cGMP levels in rat cortical primary neurons, and phosphorylation of AMPA receptor subunit GluA1 was induced following cGMP elevation. Oral administration of irsenontrine significantly upregulated cGMP levels in the hippocampus and cerebrospinal fluid (CSF) of naïve rats, and a novel object recognition test showed that irsenontrine administration also significantly improved learning and memory. The effects of irsenontrine were confirmed in rats treated with Nω-nitro-l-arginine methyl ester hydrochloride (l-NAME), a model of learning and memory impairment due to downregulation of the cGMP pathway. l-NAME downregulated cGMP in the CSF and hippocampus and impaired novel object recognition, but oral administration of irsenontrine clearly attenuated these phenotypes. These results indicate that irsenontrine improves learning and memory via the elevation of cGMP levels, and they strongly suggest that irsenontrine could be a novel therapeutic approach against cognitive dysfunction.
Collapse
Affiliation(s)
- Yasuharu Ishihara
- Deep Human Biology Learning, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba, Ibaraki, 300-2635, Japan; Laboratory of Genomics-based Drug Discovery, Doctoral Program in Medical Sciences, Graduate School of Comprehensive Human Sciences, Degree Program in Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan.
| | - Mai Ando
- Deep Human Biology Learning, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba, Ibaraki, 300-2635, Japan
| | - Yasuaki Goto
- Deep Human Biology Learning, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba, Ibaraki, 300-2635, Japan
| | - Sadaharu Kotani
- Eisai Co., Ltd., 4-6-10 Koishikawa, Bunkyo-ku, Tokyo, 112-8088, Japan
| | - Naoto Watanabe
- Deep Human Biology Learning, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba, Ibaraki, 300-2635, Japan
| | - Yosuke Nakatani
- Deep Human Biology Learning, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba, Ibaraki, 300-2635, Japan
| | - Satoko Ishii
- Deep Human Biology Learning, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba, Ibaraki, 300-2635, Japan
| | - Norimasa Miyamoto
- Deep Human Biology Learning, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba, Ibaraki, 300-2635, Japan; Laboratory of Genomics-based Drug Discovery, Faculty of Medicine and Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan
| | - Yuji Mano
- Deep Human Biology Learning, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba, Ibaraki, 300-2635, Japan; Laboratory of Genomics-based Drug Discovery, Faculty of Medicine and Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan
| | - Yukio Ishikawa
- Deep Human Biology Learning, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba, Ibaraki, 300-2635, Japan
| |
Collapse
|
9
|
Li R, Dai Q, Yu T, Sun Y, Li Y, Zhao T, Xu H, Wang L, Wang Y, Gao X, Liu X. Adolescent marginal zinc deficiency upregulated BDNF and TrkB expression, impaired hippocampal and cortical development, and induced abnormal behaviors in male mice. Comp Biochem Physiol C Toxicol Pharmacol 2025; 294:110197. [PMID: 40154589 DOI: 10.1016/j.cbpc.2025.110197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/07/2025] [Accepted: 03/23/2025] [Indexed: 04/01/2025]
Abstract
Zinc deficiency during adolescence poses a significant yet understudied risk to brain development. The study aimed to investigate the effects of marginal zinc deficiency during adolescence on emotion and cognition, morphological changes and neuronal arrangement of hippocampus and cortical, and proBDNF, mBDNF and TrkB expression levels. The emotion was assessed using the open-field test and three-chamber test. Additionally, cognition was evaluated using the Morris water maze test and novel object recognition test. Morphological changes were evaluated using H&E staining, while Nissl staining was employed to analyze neuronal arrangement. Additionally, proBDNF, mBDNF and TrkB expression levels were quantified by western blot. The results showed that adolescent marginal zinc deficiency induced risk-taking behavior, impaired spatial learning and memory, and caused new object recognition deficits without affecting sociability. Moreover, marginal zinc deficiency critically disrupted hippocampal and cortical development, and aberrant neuronal arrangement. The expression levels of BDNF for both form states were not statistically significant upregulation in marginal zinc deficiency mice compared to controls, along with significantly increased TrkB expression. These findings suggested that adolescent marginal zinc deficiency increased the expression of BDNF and TrkB, as well as abnormal hippocampal and cortical development. These alterations may explain the observed abnormal behavior, including risk-taking behavior, impaired spatial learning and memory, and new object recognition decay.
Collapse
Affiliation(s)
- Rou Li
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, PR China; Central Laboratory, Tianjin Fifth Central Hospital, Tianjin 300450, PR China
| | - Qiwei Dai
- Department of Stroke Center, The First Hospital of China Medical University, Shenyang 110001, Liaoning, PR China
| | - Tian Yu
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, PR China; Central Laboratory, Tianjin Fifth Central Hospital, Tianjin 300450, PR China
| | - Yajing Sun
- Department of Pathology, Tianjin Fifth Central Hospital, Tianjin 300450, PR China
| | - Yanxia Li
- Central Laboratory, Tianjin Fifth Central Hospital, Tianjin 300450, PR China; Tianjin Key Laboratory of Epigenetic for Organ Development of Preterm Infants, Tianjin Fifth Central Hospital, Tianjin 300450, PR China
| | - Tianyang Zhao
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Hongbin Xu
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang 110001, Liaoning, PR China
| | - Liang Wang
- Neurosurgery department, Tianjin Fifth Central Hospital, Tianjin 300450, PR China
| | - Yuxiang Wang
- Central Laboratory, Tianjin Fifth Central Hospital, Tianjin 300450, PR China; Tianjin Key Laboratory of Epigenetic for Organ Development of Preterm Infants, Tianjin Fifth Central Hospital, Tianjin 300450, PR China; School of Basic Medical Sciences, Hebei University of Chinese Medicine, Shijiazhuang 050200, Hebei, PR China.
| | - Xia Gao
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, PR China.
| | - Xiaozhi Liu
- Tianjin Key Laboratory of Epigenetic for Organ Development of Preterm Infants, Tianjin Fifth Central Hospital, Tianjin 300450, PR China; The Emergency Center, Tianjin Fifth Central Hospital, Tianjin 300450, PR China; Tianjin Binhai Huangnan Plateau Medical Research Institute, Huangnan Tibetan Autonomous Prefecture People's Hospital, Huangnan Prefecture 811399, Qinghai Province, PR China.
| |
Collapse
|
10
|
Ma Y, Dong T, Luan F, Yang J, Miao F, Wei P. Interaction of major facilitator superfamily domain containing 2A with the blood-brain barrier. Neural Regen Res 2025; 20:2133-2152. [PMID: 39248155 PMCID: PMC11759009 DOI: 10.4103/nrr.nrr-d-24-00191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 06/02/2024] [Accepted: 07/08/2024] [Indexed: 09/10/2024] Open
Abstract
The functional and structural integrity of the blood-brain barrier is crucial in maintaining homeostasis in the brain microenvironment; however, the molecular mechanisms underlying the formation and function of the blood-brain barrier remain poorly understood. The major facilitator superfamily domain containing 2A has been identified as a key regulator of blood-brain barrier function. It plays a critical role in promoting and maintaining the formation and functional stability of the blood-brain barrier, in addition to the transport of lipids, such as docosahexaenoic acid, across the blood-brain barrier. Furthermore, an increasing number of studies have suggested that major facilitator superfamily domain containing 2A is involved in the molecular mechanisms of blood-brain barrier dysfunction in a variety of neurological diseases; however, little is known regarding the mechanisms by which major facilitator superfamily domain containing 2A affects the blood-brain barrier. This paper provides a comprehensive and systematic review of the close relationship between major facilitator superfamily domain containing 2A proteins and the blood-brain barrier, including their basic structures and functions, cross-linking between major facilitator superfamily domain containing 2A and the blood-brain barrier, and the in-depth studies on lipid transport and the regulation of blood-brain barrier permeability. This comprehensive systematic review contributes to an in-depth understanding of the important role of major facilitator superfamily domain containing 2A proteins in maintaining the structure and function of the blood-brain barrier and the research progress to date. This will not only help to elucidate the pathogenesis of neurological diseases, improve the accuracy of laboratory diagnosis, and optimize clinical treatment strategies, but it may also play an important role in prognostic monitoring. In addition, the effects of major facilitator superfamily domain containing 2A on blood-brain barrier leakage in various diseases and the research progress on cross-blood-brain barrier drug delivery are summarized. This review may contribute to the development of new approaches for the treatment of neurological diseases.
Collapse
Affiliation(s)
- Yilun Ma
- College of Pharmacy and First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Taiwei Dong
- College of Pharmacy and First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Fei Luan
- College of Pharmacy and First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Juanjuan Yang
- National Drug Clinical Trial Agency, The Second Affiliated Hospital of Shaanxi University of Chinese Medicine/Xixian New District Central Hospital, Xi′an, Shaanxi Province, China
| | - Feng Miao
- College of Pharmacy and First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Peifeng Wei
- National Drug Clinical Trial Agency, The Second Affiliated Hospital of Shaanxi University of Chinese Medicine/Xixian New District Central Hospital, Xi′an, Shaanxi Province, China
| |
Collapse
|
11
|
Chen G, Zhang K, Sun M, Xie N, Wu L, Zhang G, Guo B, Huang C, Man Hoi MP, Zhang G, Shi C, Sun Y, Zhang Z, Wang Y. Multi-functional memantine nitrate attenuated cognitive impairment in models of vascular dementia and Alzheimer's disease through neuroprotection and increased cerebral blood flow. Neuropharmacology 2025; 272:110410. [PMID: 40081796 DOI: 10.1016/j.neuropharm.2025.110410] [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: 12/07/2024] [Revised: 02/26/2025] [Accepted: 03/09/2025] [Indexed: 03/16/2025]
Abstract
Alzheimer's disease (AD) and vascular dementia (VaD) are two prevalent forms of dementia. VaD is linked to cerebrovascular lesions, such as those from white matter ischemia and chronic cerebral hypoperfusion, which can also occur in AD. Nitric oxide (NO) regulates cerebral blood flow (CBF) in the central nervous system. Memantine is an NMDA receptor antagonist approved for AD treatment. This study investigated the efficacy and molecular mechanism of MN-08, a novel memantine nitrate, in one VaD model (2VO) and two AD models (APP/PS1 mice and Aβ1-42-induced mice). MN-08 increased CBF, ameliorated cognitive and memory functions in VaD and AD, and was more effective than memantine. MN-08 increased the survival rate of CA1 neurons and mitigated white matter lesions and axonal damage. Moreover, MN-08 protected neurons from OGD-induced loss and promoted axonal outgrowth in the hippocampus by upregulating phosphorylated Akt (p-Akt), glycogen synthase kinase-3β (p-GSK3β), and high-molecular-weight neurofilaments (p-NFH). The beneficial effects of MN-08 were attenuated by carboxy-PTIO, a potent NO scavenger, suggesting that MN-08-derived NO may alleviate cognitive impairment from cerebral hypoperfusion. Taken together, our studies demonstrate that MN-08 is a promising therapeutic agent for the treatment of dementia including VaD and AD.
Collapse
Affiliation(s)
- Guangying Chen
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, and Institute of New Drug Research, Jinan University College of Pharmacy, Guangzhou, 510632, China
| | - Kexin Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, and Institute of New Drug Research, Jinan University College of Pharmacy, Guangzhou, 510632, China
| | - Minghua Sun
- Department of Medical Imaging Center, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China; Department of Radiology, The Fuyang Hospital of Anhui Medical University, Fuyang, 236000, China
| | - Ningqing Xie
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, and Institute of New Drug Research, Jinan University College of Pharmacy, Guangzhou, 510632, China
| | - Liangmiao Wu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, and Institute of New Drug Research, Jinan University College of Pharmacy, Guangzhou, 510632, China; Department of Medical Imaging Center, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Guiliang Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao Special Administrative Region, China
| | - Baojian Guo
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, and Institute of New Drug Research, Jinan University College of Pharmacy, Guangzhou, 510632, China
| | - Chunhui Huang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, and Institute of New Drug Research, Jinan University College of Pharmacy, Guangzhou, 510632, China
| | - Maggie Pui Man Hoi
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao Special Administrative Region, China
| | - Gaoxiao Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, and Institute of New Drug Research, Jinan University College of Pharmacy, Guangzhou, 510632, China
| | - Changzheng Shi
- Department of Medical Imaging Center, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China.
| | - Yewei Sun
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, and Institute of New Drug Research, Jinan University College of Pharmacy, Guangzhou, 510632, China.
| | - Zaijun Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, and Institute of New Drug Research, Jinan University College of Pharmacy, Guangzhou, 510632, China.
| | - Yuqiang Wang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, and Institute of New Drug Research, Jinan University College of Pharmacy, Guangzhou, 510632, China
| |
Collapse
|
12
|
Zhou BY, Li ZX, Li YW, Li JN, Liu WT, Liu XY, Hu ZB, Zhao L, Chen JY, Hu L, Song NN, Feng X, Wang G, Xu L, Ding YQ. Central Med23 deficiency leads to malformation of dentate gyrus and ADHD-like behaviors in mice. Neuropsychopharmacology 2025; 50:1224-1236. [PMID: 40114018 DOI: 10.1038/s41386-025-02088-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 02/25/2025] [Accepted: 03/06/2025] [Indexed: 03/22/2025]
Abstract
Attention-deficit hyperactivity disorder (ADHD) is a prevalent psychiatric disorder with high heritability, while its etiology and pathophysiology remain unclear. Med23 is a subunit of the Mediator complex, a key regulator of gene expression by linking transcription factors to RNA polymerase II. The mutations of Med23 are associated with several brain diseases including microcephaly, epilepsy and intellectual disability, but its biological roles in brain development and possible behavioral consequence have not been explored in the animal model. In this study, Emx1-Cre mice were used to generate Med23 conditional knockout (Med23 CKO) mice that showed severe hypoplasia of the dentate gyrus (DG) with malformation of the dendritic tree and spines along with impaired short-term synaptic plasticity. Interestingly, Med23 CKO mice exhibited ADHD-like behaviors as shown by hyperactivity, inattention and impulsivity, as well as impaired sensory gating and working memory. Importantly, methylphenidate (MPH), a common drug for ADHD ameliorated these deficits in the CKO mice. Furthermore, we also revealed that the impaired synaptic plasticity was partially restored by MPH in an N-methyl-d-aspartate (NMDA) receptor-dependent way. Collectively, our data demonstrate Med23 deficiency causes DG malformation and ADHD-like behaviors, suggesting a novel mechanism underlying relevant brain diseases.
Collapse
Affiliation(s)
- Bing-Yao Zhou
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Brain Science, Fudan University, Shanghai, 200032, China
| | - Ze-Xuan Li
- Laboratory Animal Center, Fudan University, Shanghai, 200032, China
| | - Yi-Wei Li
- Laboratory Animal Center, Fudan University, Shanghai, 200032, China
| | - Jin-Nan Li
- Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, the Chinese Academy of Science, Kunming, 650223, China
| | - Wei-Tang Liu
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200032, China
| | - Xi-Yue Liu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Brain Science, Fudan University, Shanghai, 200032, China
| | - Zhi-Bin Hu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Brain Science, Fudan University, Shanghai, 200032, China
| | - Li Zhao
- Laboratory Animal Center, Fudan University, Shanghai, 200032, China
| | - Jia-Yin Chen
- Laboratory Animal Center, Fudan University, Shanghai, 200032, China
| | - Ling Hu
- Laboratory Animal Center, Fudan University, Shanghai, 200032, China
| | - Ning-Ning Song
- Laboratory Animal Center, Fudan University, Shanghai, 200032, China
| | - Xue Feng
- Laboratory Animal Center, Fudan University, Shanghai, 200032, China
| | - Gang Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Zhongshan Hospital, Fudan University, Shanghai, 200438, China
| | - Lin Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, the Chinese Academy of Science, Kunming, 650223, China.
| | - Yu-Qiang Ding
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Brain Science, Fudan University, Shanghai, 200032, China.
- Laboratory Animal Center, Fudan University, Shanghai, 200032, China.
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200032, China.
- Huashan Institute of Medicine (HS-IOM), Huashan Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
13
|
Lei J, Xin Z, Liu N, Ning T, Jing Y, Qiao Y, He Z, Jiang M, Yang Y, Zhang Z, Zhao L, Li J, Lv D, Yan Y, Zhang H, Xiao L, Zhang B, Huang H, Sun S, Zheng F, Jiang X, Lu H, Dong X, Yue S, Ma C, Shuai J, Ji Z, Liu F, Ye Y, Yan K, Hu Q, Xu G, Zhao Q, Wu R, Cai Y, Fan Y, Jing Y, Wang Q, Reddy P, Lu X, Zheng Z, Liu B, Haghani A, Ma S, Suzuki K, Rodriguez Esteban C, Yang J, Song M, Horvath S, Zhang W, Li W, Xiang AP, Zhu L, Fu X, Zhao G, Belmonte JCI, Qu J, Wang S, Liu GH. Senescence-resistant human mesenchymal progenitor cells counter aging in primates. Cell 2025:S0092-8674(25)00571-9. [PMID: 40516525 DOI: 10.1016/j.cell.2025.05.021] [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: 10/09/2024] [Revised: 03/08/2025] [Accepted: 05/19/2025] [Indexed: 06/16/2025]
Abstract
Aging is characterized by a deterioration of stem cell function, but the feasibility of replenishing these cells to counteract aging remains poorly defined. Our study addresses this gap by developing senescence (seno)-resistant human mesenchymal progenitor cells (SRCs), genetically fortified to enhance cellular resilience. In a 44-week trial, we intravenously delivered SRCs to aged macaques, noting a systemic reduction in aging indicators, such as cellular senescence, chronic inflammation, and tissue degeneration, without any detected adverse effects. Notably, SRC treatment enhanced brain architecture and cognitive function and alleviated the reproductive system decline. The restorative effects of SRCs are partly attributed to their exosomes, which combat cellular senescence. This study provides initial evidence that genetically modified human mesenchymal progenitors can slow primate aging, highlighting the therapeutic potential of regenerative approaches in combating age-related health decline.
Collapse
Affiliation(s)
- Jinghui Lei
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Aging Translational Medicine Center, Beijing Municipal Geriatric Medical Research Center, Beijing Key Laboratory of Environment and Aging, Xuanwu Hospital Capital Medical University, Beijing 100053, China
| | - Zijuan Xin
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Aging Translational Medicine Center, Beijing Municipal Geriatric Medical Research Center, Beijing Key Laboratory of Environment and Aging, Xuanwu Hospital Capital Medical University, Beijing 100053, China; State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Ning Liu
- State Key Laboratory of Cognitive Science and Mental Health, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Taixin Ning
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Aging Translational Medicine Center, Beijing Municipal Geriatric Medical Research Center, Beijing Key Laboratory of Environment and Aging, Xuanwu Hospital Capital Medical University, Beijing 100053, China
| | - Ying Jing
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Aging Translational Medicine Center, Beijing Municipal Geriatric Medical Research Center, Beijing Key Laboratory of Environment and Aging, Xuanwu Hospital Capital Medical University, Beijing 100053, China
| | - Yicheng Qiao
- State Key Laboratory of Cognitive Science and Mental Health, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zan He
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Aging Translational Medicine Center, Beijing Municipal Geriatric Medical Research Center, Beijing Key Laboratory of Environment and Aging, Xuanwu Hospital Capital Medical University, Beijing 100053, China
| | - Mengmeng Jiang
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Yuanhan Yang
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhiyi Zhang
- State Key Laboratory of Cognitive Science and Mental Health, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Liyun Zhao
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Aging Translational Medicine Center, Beijing Municipal Geriatric Medical Research Center, Beijing Key Laboratory of Environment and Aging, Xuanwu Hospital Capital Medical University, Beijing 100053, China
| | - Jingyi Li
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Aging Biomarker Consortium (ABC), Beijing 100101, China
| | - Dongliang Lv
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yupeng Yan
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Hui Zhang
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Lingling Xiao
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Aging Translational Medicine Center, Beijing Municipal Geriatric Medical Research Center, Beijing Key Laboratory of Environment and Aging, Xuanwu Hospital Capital Medical University, Beijing 100053, China
| | - Baohu Zhang
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Haoyan Huang
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Aging Translational Medicine Center, Beijing Municipal Geriatric Medical Research Center, Beijing Key Laboratory of Environment and Aging, Xuanwu Hospital Capital Medical University, Beijing 100053, China
| | - Shuhui Sun
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Fangshuo Zheng
- Chongqing Fifth People's Hospital, Chongqing 400060, China
| | - Xiaoyu Jiang
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Huifen Lu
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Aging Translational Medicine Center, Beijing Municipal Geriatric Medical Research Center, Beijing Key Laboratory of Environment and Aging, Xuanwu Hospital Capital Medical University, Beijing 100053, China
| | - Xueda Dong
- State Key Laboratory of Cognitive Science and Mental Health, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; Sino-Danish College, Sino-Danish Centre for Education and Research, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shasha Yue
- State Key Laboratory of Cognitive Science and Mental Health, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Chencan Ma
- State Key Laboratory of Cognitive Science and Mental Health, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jichen Shuai
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Aging Translational Medicine Center, Beijing Municipal Geriatric Medical Research Center, Beijing Key Laboratory of Environment and Aging, Xuanwu Hospital Capital Medical University, Beijing 100053, China
| | - Zhejun Ji
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Feifei Liu
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Yanxia Ye
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Kaowen Yan
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Qinchao Hu
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou 510060, China
| | - Gang Xu
- Liver Transplant Center, Organ Transplant Center, West China Hospital of Sichuan University, Chengdu 610000, China; Laboratory of Liver Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital of Sichuan University, Chengdu 610000, China
| | - Qian Zhao
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Aging Translational Medicine Center, Beijing Municipal Geriatric Medical Research Center, Beijing Key Laboratory of Environment and Aging, Xuanwu Hospital Capital Medical University, Beijing 100053, China
| | - Ruochen Wu
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yusheng Cai
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Yanling Fan
- Beijing Institute of Genomics, China National Center for Bioinformation, Chinese Academy of Sciences, Beijing 100101, China
| | - Yaobin Jing
- International Center for Aging and Cancer, Hainan Medical University, Haikou 571199, China
| | - Qiaoran Wang
- Beijing Institute of Genomics, China National Center for Bioinformation, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Pradeep Reddy
- Altos Labs San Diego Institute of Science, San Diego, CA, USA
| | - Xiaoyong Lu
- Beijing Institute of Genomics, China National Center for Bioinformation, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zikai Zheng
- Beijing Institute of Genomics, China National Center for Bioinformation, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Beibei Liu
- Beijing Institute of Genomics, China National Center for Bioinformation, Chinese Academy of Sciences, Beijing 100101, China
| | - Amin Haghani
- Altos Labs San Diego Institute of Science, San Diego, CA, USA
| | - Shuai Ma
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Aging Biomarker Consortium (ABC), Beijing 100101, China
| | - Keiichiro Suzuki
- Institute for Advanced Co-Creation Studies, The University of Osaka, Osaka 560-8531, Japan
| | | | - Jiayin Yang
- Liver Transplant Center, Organ Transplant Center, West China Hospital of Sichuan University, Chengdu 610000, China; Laboratory of Liver Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital of Sichuan University, Chengdu 610000, China
| | - Moshi Song
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Aging Biomarker Consortium (ABC), Beijing 100101, China
| | - Steve Horvath
- Altos Labs San Diego Institute of Science, San Diego, CA, USA
| | - Weiqi Zhang
- Beijing Institute of Genomics, China National Center for Bioinformation, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Sino-Danish College, Sino-Danish Centre for Education and Research, University of Chinese Academy of Sciences, Beijing 100049, China; Aging Biomarker Consortium (ABC), Beijing 100101, China
| | - Wei Li
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Andy Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, Guangdong, China; Aging Biomarker Consortium (ABC), Beijing 100101, China
| | - Lan Zhu
- National Clinical Research Center for Obstetric and Gynecologic Diseases, Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Xiaobing Fu
- Tissue Repair and Regeneration Research Center, Medical Innovation Department, PLA General Hospital and Medical College, Beijing 100842, China
| | - Guoguang Zhao
- Department of Neurosurgery, Beijing Municipal Geriatric Medical Research Center, National Medical Center for Neurological Diseases, Xuanwu Hospital Capital Medical University, Beijing 100053, China
| | - Juan Carlos Izpisua Belmonte
- Altos Labs San Diego Institute of Science, San Diego, CA, USA; Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Jing Qu
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China; University of Chinese Academy of Sciences, Beijing 100049, China; Aging Biomarker Consortium (ABC), Beijing 100101, China.
| | - Si Wang
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Aging Translational Medicine Center, Beijing Municipal Geriatric Medical Research Center, Beijing Key Laboratory of Environment and Aging, Xuanwu Hospital Capital Medical University, Beijing 100053, China; Aging Biomarker Consortium (ABC), Beijing 100101, China.
| | - Guang-Hui Liu
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Aging Translational Medicine Center, Beijing Municipal Geriatric Medical Research Center, Beijing Key Laboratory of Environment and Aging, Xuanwu Hospital Capital Medical University, Beijing 100053, China; State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Aging Biomarker Consortium (ABC), Beijing 100101, China.
| |
Collapse
|
14
|
Oliveira MS, Fernandes RA, Pinto LS, Moreira FA, Castro OWD, Santos VR. Balancing efficacy and safety: The dual impact of antiseizure medications on the developing brain. Epilepsy Behav 2025; 167:110400. [PMID: 40187052 DOI: 10.1016/j.yebeh.2025.110400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 02/25/2025] [Accepted: 03/21/2025] [Indexed: 04/07/2025]
Abstract
The number of neurons in the developing brain is greater than typically found in adulthood, and the brain possesses delicate mechanisms to induce the death of excess cells and refine neural circuitry. The correct tuning between the processes of neuronal death and survival generates a mature and functional brain in its complexity and plastic capacity. Epilepsy is a highly prevalent neurological condition worldwide, including among young individuals. However, exposure to the main treatment approaches, the long-term use of Antiseizure Medication (ASM), during the critical period of development can induce a series of changes in this delicate balance. Acting by various mechanisms of action, ASMs may induce an increase in neuronal death, something that translates into deleterious neuropsychiatric effects in adulthood. Several investigations conducted in recent years have brought to light new aspects related to this dynamic, yet many questions, such as the cellular mechanisms of death and the pathophysiology of late effects, still have unresolved elements. In this review, we aimed to explore the mechanisms of action of the most widely used ASMs in the treatment of neonatal epilepsy, the broad aspects of neuronal death in the developing brain and the repercussions of this death and other effects in adulthood. We review the evidence indicating a relationship between exposure to ASMs and the manifestation of associated psychiatric comorbidities in adulthood and discuss some possible mechanisms underlying the induction of this process by morphological and physiological changes in the related behaviors.
Collapse
Affiliation(s)
- M S Oliveira
- Department of Morphology, Institute of Biological Science, Universidade Federal de Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
| | - R A Fernandes
- Department of Morphology, Institute of Biological Science, Universidade Federal de Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
| | - L S Pinto
- Department of Morphology, Institute of Biological Science, Universidade Federal de Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
| | - F A Moreira
- Department of Pharmacology, Institute of Biological Science, Universidade Federal de Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
| | - O W de Castro
- Departament of Physiology, Institute of Biological Science and Health, Universidade Federal de Alagoas - UFAL, Brazil
| | - V R Santos
- Department of Morphology, Institute of Biological Science, Universidade Federal de Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
| |
Collapse
|
15
|
Pedrinolla A, Dorelli G, Porcelli S, Burleigh M, Mendo M, Martignon C, Fonte C, Dalle Carbonare LG, Easton C, Muti E, Schena F, Venturelli M. Increasing nitric oxide availability via ingestion of nitrate-rich beetroot juice improves vascular responsiveness in individuals with Alzheimer's Disease. Nitric Oxide 2025; 156:50-56. [PMID: 40089052 DOI: 10.1016/j.niox.2025.03.001] [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: 10/21/2024] [Revised: 03/03/2025] [Accepted: 03/13/2025] [Indexed: 03/17/2025]
Abstract
Poor vascular function and reduced nitric oxide (NO)-bioavailability have been recognized to be involved in aging and Alzheimer's Disease (AD). A non-pharmacological treatment that is gaining clinical interest in the context of vascular function is dietary inorganic nitrate (NO3-) supplementation which increases NO-bioavailability through the NO3- -nitrite (NO2-) - NO pathway. This treatment has been demonstrated to improve vascular function in several clinical populations, but no study has investigated the effects in individuals with AD. Therefore, changes in plasma NO3- and NO2- and vascular responsiveness (hyperemic response to single-passive leg movement (ΔPLM)) were measured in individuals with AD (n = 10, 76 ± 9 years), healthy elderly (OLD, n = 10, 75 ± 6 years), and young individuals (YN, n = 10, 25 ± 4 years) before (T0) and hourly for 4 h (T1, T2, T3, and T4) after ingestion of either NO3--rich beetroot juice (BR) or a placebo (PLA). No changes in NO3- and NO2-, nor ΔPLM were detected in any group following PLA intake. Plasma NO3- and NO2- increased significantly in all three groups at T1 (p < 0.001) and remained elevated for the rest of the trial. The same trend was found in ΔPLM, which significantly increased in all three groups over the time (p < 0.001). However, AD exhibited significantly lower ΔPLM values at any time point compared to YN (p < 0.001) and OLD (p < 0.001). These data suggest that AD-individuals included in this study were able to reduce NO3- to NO2- and to increase NO-mediated vascular responsiveness as non-AD-individuals. Other mechanisms, beyond NO-bioavailability, may be involved in vascular dysfunction in patients with AD. This research suggests that an acute administration of inorganic nitrate is not enough to revert chronically adapted vascular properties and completely restore vascular responsiveness in AD.
Collapse
Affiliation(s)
- Anna Pedrinolla
- Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Trento, Italy.
| | - Gianluigi Dorelli
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Simone Porcelli
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Mia Burleigh
- Sport and Physical Activity Research Institute, University of the West of Scotland, Blantyre, Scotland, UK
| | - Martina Mendo
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Camilla Martignon
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Cristina Fonte
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | | | - Chris Easton
- Sport and Physical Activity Research Institute, University of the West of Scotland, Blantyre, Scotland, UK; School of Energy, Geoscience, Infrastructure and Society, Heriot-Watt University, Edinburgh, Scotland, UK
| | | | - Federico Schena
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Massimo Venturelli
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy; Department of Internal Medicine Section of Geriatrics, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
16
|
Fang CW, Hsieh CY, Yang HY, Tsai CF, Sung SF. Comparative effectiveness and safety of direct oral anticoagulants in atrial fibrillation patients with dementia. Thromb Res 2025; 250:109332. [PMID: 40311503 DOI: 10.1016/j.thromres.2025.109332] [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: 02/23/2025] [Revised: 04/05/2025] [Accepted: 04/21/2025] [Indexed: 05/03/2025]
Abstract
INTRODUCTION Patients with atrial fibrillation (AF) and dementia face unique challenges in stroke prevention, particularly in selecting appropriate anticoagulation therapy. Direct oral anticoagulants (DOACs) effectively reduce stroke and embolism risks, but evidence comparing their effectiveness and safety in this population remains limited. METHODS This retrospective, population-based cohort study used data from Taiwan's National Health Insurance Research Database to evaluate outcomes of four DOACs (dabigatran, apixaban, edoxaban, and rivaroxaban) in AF patients with dementia aged 50 years or older. We used propensity score matching to balance baseline characteristics across six DOAC comparison pairs. RESULTS Dabigatran demonstrated superior outcomes, reducing the composite risk of ischemic stroke, acute myocardial infarction, intracranial hemorrhage, major bleeding, and all-cause mortality compared to apixaban (hazard ratio [HR], 0.82; 95 % confidence interval [CI], 0.73-0.92), edoxaban (HR, 0.81; 95 % CI, 0.71-0.92), and rivaroxaban (HR, 0.82; 95 % CI, 0.73-0.91). It also showed lower risks of intracranial hemorrhage and all-cause mortality. Sensitivity analyses excluding patients with nasogastric tubes or severe renal impairment showed smaller differences in overall outcomes but maintained dabigatran's advantage in reducing intracranial hemorrhage risk. CONCLUSIONS This study demonstrates the need for tailored anticoagulation strategies in this vulnerable population, with dabigatran emerging as a potentially safer and more effective option for stroke prevention in AF patients with dementia. Future research should examine individual DOAC effects across diverse clinical settings to optimize treatment outcomes.
Collapse
Affiliation(s)
- Chen-Wen Fang
- Department of Neurology, National Taiwan University Hospital, Yunlin Branch, Douliu City, Taiwan; Department of Biomedical Engineering, National Cheng Kung University, Tainan City, Taiwan
| | - Cheng-Yang Hsieh
- Department of Neurology, Tainan Sin Lau Hospital, Tainan, Taiwan
| | - Hsin-Yi Yang
- Clinical Data Center, Department of Medical Research, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi City, Taiwan
| | - Ching-Fang Tsai
- Clinical Data Center, Department of Medical Research, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi City, Taiwan
| | - Sheng-Feng Sung
- Division of Neurology, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi City, Taiwan; Department of Nursing, Fooyin University, Kaohsiung, Taiwan.
| |
Collapse
|
17
|
Rossetti GMK, Dunster JL, Sohail A, Williams B, Cox KM, Rawlings S, Jewett E, Benford E, Lovegrove JA, Gibbins JM, Christakou A. Evidence for control of cerebral neurovascular function by circulating platelets in healthy older adults. J Physiol 2025; 603:3379-3404. [PMID: 40434152 DOI: 10.1113/jp288405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 04/10/2025] [Indexed: 05/29/2025] Open
Abstract
Platelets play a vital role in preventing haemorrhage through haemostasis, but complications arise when platelets become overly reactive, leading to pathophysiology such as atherothrombosis. Elevated haemostatic markers are linked to dementia and predict its onset in long-term studies. Despite epidemiological evidence, the mechanism linking haemostasis with early brain pathophysiology remains unclear. Here, we aimed to determine whether a mechanistic association exists between platelet function and cerebral neurovascular function in 52 healthy mid- to older-age adults. To do this, we combined, for the first time, magnetic resonance imaging of cerebral neurovascular function, peripheral vascular physiology and in vitro platelet assaying. We show an association between platelet reactivity and cerebral neurovascular function that is both independent of vascular reactivity and mechanistically specific: Distinct platelet signalling mechanisms (ADP, collagen-related peptide, thrombin receptor activator peptide 6) were associated with different physiological components of the haemodynamic response to neuronal (visual) stimulation (full-width half-maximum, time to peak, area under the curve), an association that was not mediated by peripheral vascular effects. This finding challenges the previous belief that systemic vascular health determines the vascular component of cerebral neurovascular function, highlighting a specific link between circulating platelets and the neurovascular unit. Because altered cerebral neurovascular function marks the initial stages of neurodegenerative pathophysiology, understanding this novel association is now imperative, with the potential to lead to a significant advancement in our comprehension of early dementia pathophysiology. KEY POINTS: Haemostasis (platelet function) has been linked to the early stages of dementia, but the precise mechanisms are not well understood. This study considers whether a causal mechanism exists through atherothrombotic effects on the vasculature which can in turn affect brain health, or through platelet-specific interactions with brain physiology. Here, we show that elevated platelet reactivity is associated with blunted (delayed, shorter and smaller) cerebral blood flow responses to neuronal activation in healthy middle-aged and older adults. However, the association between platelet reactivity and cerebral neurovascular function was not mediated by systemic vascular reactivity. This finding challenges the previous belief that systemic vascular health determines the vascular component of cerebral neurovascular function, highlighting a specific link between circulating platelets and the neurovascular unit in early dementia pathophysiology.
Collapse
Affiliation(s)
- Gabriella M K Rossetti
- Centre for Integrative Neuroscience and Neurodynamics (CINN), School of Psychology and Clinical Language Sciences, University of Reading, Reading, UK
- Institute of Sport, Department of Sport and Exercise Sciences, Manchester Metropolitan University, Manchester, UK
| | - Joanne L Dunster
- Institute for Cardiovascular and Metabolic Research (ICMR), School of Biological Sciences, University of Reading, Reading, UK
| | - Aamir Sohail
- Centre for Integrative Neuroscience and Neurodynamics (CINN), School of Psychology and Clinical Language Sciences, University of Reading, Reading, UK
- Centre for Human Brain Health (CHBH), School of Psychology, University of Birmingham, Birmingham, UK
| | - Brendan Williams
- Centre for Integrative Neuroscience and Neurodynamics (CINN), School of Psychology and Clinical Language Sciences, University of Reading, Reading, UK
| | - Kiera M Cox
- Institute for Cardiovascular and Metabolic Research (ICMR), School of Biological Sciences, University of Reading, Reading, UK
| | - Suzannah Rawlings
- Institute for Cardiovascular and Metabolic Research (ICMR), School of Biological Sciences, University of Reading, Reading, UK
| | - Elysia Jewett
- Centre for Integrative Neuroscience and Neurodynamics (CINN), School of Psychology and Clinical Language Sciences, University of Reading, Reading, UK
- Institute for Cardiovascular and Metabolic Research (ICMR), School of Biological Sciences, University of Reading, Reading, UK
| | - Eleanor Benford
- Centre for Integrative Neuroscience and Neurodynamics (CINN), School of Psychology and Clinical Language Sciences, University of Reading, Reading, UK
- Institute for Cardiovascular and Metabolic Research (ICMR), School of Biological Sciences, University of Reading, Reading, UK
| | - Julie A Lovegrove
- Institute for Cardiovascular and Metabolic Research (ICMR), School of Biological Sciences, University of Reading, Reading, UK
- Institute of Food and Nutritional Health (IFNH), Department of Food and Nutritional Sciences, University of Reading, Reading, UK
| | - Jonathan M Gibbins
- Institute for Cardiovascular and Metabolic Research (ICMR), School of Biological Sciences, University of Reading, Reading, UK
| | - Anastasia Christakou
- Centre for Integrative Neuroscience and Neurodynamics (CINN), School of Psychology and Clinical Language Sciences, University of Reading, Reading, UK
| |
Collapse
|
18
|
Qi G, Tang H, Hu J, Kang S, Qin S. Potential role of tanycyte-derived neurogenesis in Alzheimer's disease. Neural Regen Res 2025; 20:1599-1612. [PMID: 38934388 PMCID: PMC11688558 DOI: 10.4103/nrr.nrr-d-23-01865] [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: 01/17/2024] [Revised: 02/19/2024] [Accepted: 04/17/2024] [Indexed: 06/28/2024] Open
Abstract
Tanycytes, specialized ependymal cells located in the hypothalamus, play a crucial role in the generation of new neurons that contribute to the neural circuits responsible for regulating the systemic energy balance. The precise coordination of the gene networks controlling neurogenesis in naive and mature tanycytes is essential for maintaining homeostasis in adulthood. However, our understanding of the molecular mechanisms and signaling pathways that govern the proliferation and differentiation of tanycytes into neurons remains limited. This article aims to review the recent advancements in research into the mechanisms and functions of tanycyte-derived neurogenesis. Studies employing lineage-tracing techniques have revealed that the neurogenesis specifically originating from tanycytes in the hypothalamus has a compensatory role in neuronal loss and helps maintain energy homeostasis during metabolic diseases. Intriguingly, metabolic disorders are considered early biomarkers of Alzheimer's disease. Furthermore, the neurogenic potential of tanycytes and the state of newborn neurons derived from tanycytes heavily depend on the maintenance of mild microenvironments, which may be disrupted in Alzheimer's disease due to the impaired blood-brain barrier function. However, the specific alterations and regulatory mechanisms governing tanycyte-derived neurogenesis in Alzheimer's disease remain unclear. Accumulating evidence suggests that tanycyte-derived neurogenesis might be impaired in Alzheimer's disease, exacerbating neurodegeneration. Confirming this hypothesis, however, poses a challenge because of the lack of long-term tracing and nucleus-specific analyses of newborn neurons in the hypothalamus of patients with Alzheimer's disease. Further research into the molecular mechanisms underlying tanycyte-derived neurogenesis holds promise for identifying small molecules capable of restoring tanycyte proliferation in neurodegenerative diseases. This line of investigation could provide valuable insights into potential therapeutic strategies for Alzheimer's disease and related conditions.
Collapse
Affiliation(s)
- Guibo Qi
- Department of Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Han Tang
- Department of Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jianian Hu
- Department of Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Siying Kang
- Department of Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Song Qin
- Department of Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
19
|
Kokkali M, Karali K, Thanou E, Papadopoulou MA, Zota I, Tsimpolis A, Efstathopoulos P, Calogeropoulou T, Li KW, Sidiropoulou K, Gravanis A, Charalampopoulos I. Multimodal beneficial effects of BNN27, a nerve growth factor synthetic mimetic, in the 5xFAD mouse model of Alzheimer's disease. Mol Psychiatry 2025; 30:2265-2283. [PMID: 39587294 PMCID: PMC12092300 DOI: 10.1038/s41380-024-02833-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 11/05/2024] [Accepted: 11/06/2024] [Indexed: 11/27/2024]
Abstract
Alzheimer's Disease (AD) is an incurable and debilitating progressive, neurodegenerative disorder which is the leading cause of dementia worldwide. Neuropathologically, AD is characterized by the accumulation of Aβ amyloid plaques in the microenvironment of brain cells and neurovascular walls, chronic neuroinflammation, resulting in neuronal and synaptic loss, myelin and axonal failure, as well as significant reduction in adult hippocampal neurogenesis. The hippocampal formation is particularly vulnerable to this degenerative process, due to early dysfunction of the cholinergic circuit. Neurotrophic factors consist major regulatory molecules and their decline in AD is considered as an important cause of disease onset and progression. Novel pharmacological approaches are targeting the downstream pathways controlled by neurotrophins, such as nerve growth factor (NGF) receptors, TrkA and p75NTR, which enhance hippocampal neurogenic capacity and neuroprotective mechanisms, and potentially counteract the neurotoxic effects of amyloid deposition. BNN27 is a non-toxic, newly developed 17-spiro-steroid analog, penetrating the blood-brain-barrier (BBB) and mimicking the neuroprotective effects of NGF, acting as selective activator of its receptors, both TrkA and p75NTR, thus promoting survival of various neuronal cell types. Our present research aims at determining whether and which aspects of the AD-related pathology, BNN27 is able to alleviate, exploring the cellular and molecular AD components and link these changes with improvements in the cognitive performance of an animal AD model, the 5xFAD mice. Our results clearly indicate that BNN27 administration significantly reduced amyloid-β load in whole brain of the animals, enhanced adult hippocampal neurogenesis, restored cholinergic function and synaptogenesis, reducing inflammatory activation and leading to significant restoration of cognitive functions. BNN27 may represent a new lead multimodal molecule with neuroprotective, neurogenic and anti-neuroinflammatory actions for developing druggable anti-Alzheimeric agents. Proteomics data are available via ProteomeXchange with the identifier PXD044699.
Collapse
Affiliation(s)
- Maria Kokkali
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, 71003, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, 71003, Greece
| | - Kanelina Karali
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, 71003, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, 71003, Greece
| | - Evangelia Thanou
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics & Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| | - Maria Anna Papadopoulou
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, 71003, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, 71003, Greece
| | - Ioanna Zota
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, 71003, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, 71003, Greece
| | - Alexandros Tsimpolis
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, 71003, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, 71003, Greece
| | | | | | - Ka Wan Li
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics & Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| | - Kyriaki Sidiropoulou
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, 71003, Greece
- Department of Biology, School of Sciences and Engineering, University of Crete, Heraklion, 71003, Greece
| | - Achille Gravanis
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, 71003, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, 71003, Greece
| | - Ioannis Charalampopoulos
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, 71003, Greece.
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, 71003, Greece.
| |
Collapse
|
20
|
Orellana AM, Port's NMS, de Sá Lima L, Leite JA, Andreotti DZ, Kinoshita PF, Cantanzaro AB, Neto JAM, Scavone C, Kawamoto EM. Ouabain increases neuronal differentiation of hippocampal neural precursor cells. CURRENT RESEARCH IN NEUROBIOLOGY 2025; 8:100147. [PMID: 40166632 PMCID: PMC11957680 DOI: 10.1016/j.crneur.2025.100147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 01/27/2025] [Accepted: 01/31/2025] [Indexed: 04/02/2025] Open
Abstract
Image 1.
Collapse
Affiliation(s)
- Ana Maria Orellana
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, Brazil
- Laboratory of Molecular and Functional Neurobiology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, Brazil
| | - Natacha Medeiros S. Port's
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, Brazil
| | - Larissa de Sá Lima
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, Brazil
| | - Jacqueline Alves Leite
- Department of Pharmacology, Institute of Biological Sciences, Federal University of Goiás, 74045-155, Brazil
| | - Diana Zukas Andreotti
- Laboratory of Molecular and Functional Neurobiology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, Brazil
| | - Paula Fernanda Kinoshita
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, Brazil
| | - Arthur B. Cantanzaro
- Laboratory of Molecular and Functional Neurobiology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, Brazil
| | - João Agostinho M. Neto
- Laboratory of Cancer biology and Antineoplastic agents. Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, Brazil
| | - Cristoforo Scavone
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, Brazil
| | - Elisa M. Kawamoto
- Laboratory of Molecular and Functional Neurobiology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, Brazil
| |
Collapse
|
21
|
Peng XQ, Guo HS, Zhang X, Wu XY, Ruganzu JB, Wu SD, Zhao MT, Li L, Yang Y, Ji SF, Yang WN, Ren PY. TREM2 promotes hippocampal neurogenesis through regulating microglial M2 polarization in APP/PS1 mice. Exp Neurol 2025; 388:115205. [PMID: 40049315 DOI: 10.1016/j.expneurol.2025.115205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 02/25/2025] [Accepted: 03/03/2025] [Indexed: 03/10/2025]
Abstract
Triggering receptor expressed on myeloid cells-2 (TREM2) mainly expressed on microglia in the brain, and its mutations can increase the risk of Alzheimer's disease (AD). Upregulation or activation of TREM2 has been found to ameliorate several pathological features of AD, such as the reduction of amyloid beta (Aβ) plaques and tau hyperphosphorylation. However, the effects of TREM2 on neurogenesis are little understood. Here, we aimed to investigate the effects of TREM2 on hippocampal neurogenesis associated with microglial M2 polarization in APP/PS1 mice. Lentivirus vectors were used to interfere with the expression of TREM2 on microglia in the hippocampus of APP/PS1 mice and BV2 cells. The supernatant was collected from BV2 cells as a conditioned medium (CM) to culture neural stem cells (NSCs) in vitro. Upregulation of TREM2 partially salvaged the proliferation of NSCs and the decrease of the number of immature/mature neurons in the hippocampus of APP/PS1 mice, which was accompanied by an improvement in cognitive ability. Furthermore, upregulation of TREM2 increased the M2 microglia marker CD206, brain-derived neurotrophic factor (BDNF), and anti-inflammatory factors, while decreased the M1 microglia markers CD16/32 and CD86 and pro-inflammatory factors in vivo and in vitro. Importantly, the upregulation of TREM2 also led to a significant increase in the phosphorylation of PI3K and Akt. In vitro, treatment with LY294002, a PI3K inhibitor, abolished the beneficial effects of TREM2 on shifting microglia from M1 to M2 and the proliferation and differentiation of NSCs. Taken together, these findings indicated that upregulation of TREM2 activated the PI3K/Akt signaling pathway to promote microglial M2 polarization and led to the secretion of more BDNF, accompanied by an improved hippocampal neurogenesis and spatial cognitive function in APP/PS1 mice. Thus, TREM2 might be a promising target for the treatment of neurodegenerative disease.
Collapse
Affiliation(s)
- Xiao-Qian Peng
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Hong-Song Guo
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Xiao Zhang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Xiang-Yuan Wu
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - John Bosco Ruganzu
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Song-Di Wu
- Department of Neurology and Neuro-ophthalmology, The First Affiliated Hospital of Northwest University (The First Hospital of Xi'an), Xi'an 710002, China; Xi'an Key Laboratory for Innovation and Translation of Neuroimmunological Diseases, Xi'an 710002, China
| | - Ming-Tao Zhao
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China; Zonglian College, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Lei Li
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China; Zonglian College, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Yang Yang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China; Department of Traditional Chinese Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Sheng-Feng Ji
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Wei-Na Yang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China.
| | - Peng-Yu Ren
- Institute of Medical Artificial Intelligence, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China; Department of Neurosurgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China.
| |
Collapse
|
22
|
Shukla M, Narayan M. Proteostasis and Its Role in Disease Development. Cell Biochem Biophys 2025; 83:1725-1741. [PMID: 39422790 PMCID: PMC12123047 DOI: 10.1007/s12013-024-01581-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2024] [Indexed: 10/19/2024]
Abstract
Proteostasis (protein homeostasis) refers to the general biological process that maintains the proper balance between the synthesis of proteins, their folding, trafficking, and degradation. It ensures proteins are functional, locally distributed, and appropriately folded inside cells. Genetic information enclosed in mRNA is translated into proteins. To ensure newly synthesized proteins take on the exact three-dimensional conformation, molecular chaperones assist in proper folding. Misfolded proteins can be refolded or targeted for elimination to stop aggregation. Cells utilize different degradation pathways, for instance, the ubiquitin-proteasome system, the autophagy-lysosome pathway, and the unfolded protein response, to degrade unwanted or damaged proteins. Quality control systems of the cell monitor the folding of proteins. These checkpoint mechanisms are aimed at degrading or refolding misfolded or damaged proteins. Under stress response pathways, such as heat shock response and unfolded protein response, which are triggered under conditions that perturb proteostasis, the capacity for folding is increased, and degradation pathways are activated to help cells handle stressful conditions. The deregulation of proteostasis is implicated in a variety of illnesses, comprising cancer, metabolic diseases, cardiovascular diseases, and neurological disorders. Therapeutic strategies with a deeper insight into the mechanism of proteostasis are crucial for the treatment of illnesses linked with proteostasis and to support cellular health. Thus, proteostasis is required not only for the maintenance of cellular homeostasis and function but also for proper protein function and prevention of injurious protein aggregation. In this review, we have covered the concept of proteostasis, its mechanism, and how disruptions to it can result in a number of disorders.
Collapse
Affiliation(s)
- Manisha Shukla
- Department of Biotechnology, Pandit S.N. Shukla University, Shahdol, Madhya Pradesh, India
| | - Mahesh Narayan
- Department of Chemistry and Biochemistry, University of Texas, El Paso, TX, USA.
| |
Collapse
|
23
|
Dabbaghi MM, Fadaei MS, Goldoozian M, Fadaei MR, Baradaran Rahimi V, Askari VR. Promising impacts of Achillea spp., beyond A medicinal plant, against toxins, toxicities, and injuries: In vivo and in vitro mechanisms. Biochem Biophys Rep 2025; 42:102023. [PMID: 40330076 PMCID: PMC12051127 DOI: 10.1016/j.bbrep.2025.102023] [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: 02/04/2025] [Revised: 04/10/2025] [Accepted: 04/16/2025] [Indexed: 05/08/2025] Open
Abstract
Natural toxins produced by various living organisms pose significant risks to health, food security, and environmental balance through inhalation, ingestion, and other exposure routes. This review focuses on the ameliorative effects of different Achillea species, which comprise over 130 perennial herbs known for their therapeutic properties. A systematic examination of data from Scopus, PubMed, and Web of Science was conducted, encompassing various studies without date restrictions, ensuring a comprehensive selection of articles based on full-text availability. The results of this study indicate that Achillea millefolium exhibits anti-hyperglycemic and anti-hyperlipidemic properties, enhances collagen proliferation regulation, suppresses inflammatory responses, and displays significant antioxidant activity. Similarly, A. wilhelmsii has been shown to have hepatoprotective effects, as evidenced by reduced malondialdehyde levels and increased total thiol concentrations. A. fragrantissima has also been demonstrated to have cardioprotective effects, with a decrease in inflammatory markers and edema levels. The protective benefits of other species within the Achillea genus extend to various toxins. This comprehensive review underscores the potential of Achillea species as natural remedies for combating toxicities and promoting health.
Collapse
Affiliation(s)
- Mohammad Mahdi Dabbaghi
- Clinical Research Development Unit, Imam Reza Hospital, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Saleh Fadaei
- Clinical Research Development Unit, Imam Reza Hospital, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maral Goldoozian
- Clinical Research Development Unit, Imam Reza Hospital, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Reza Fadaei
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vafa Baradaran Rahimi
- Department of Cardiovascular Diseases, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vahid Reza Askari
- Clinical Research Development Unit, Imam Reza Hospital, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
24
|
Jiang M, Li Q, Chen J, Li R, Yao J, Hu Y, Zhang H, Cai L, Luo M, Sun Y, Zeng W. Microglial MS4A4A Protects against Epileptic Seizures in Alzheimer's Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2417733. [PMID: 40349168 PMCID: PMC12165070 DOI: 10.1002/advs.202417733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 04/16/2025] [Indexed: 05/14/2025]
Abstract
Alzheimer's disease (AD) is a predominant neurodegenerative disorder worldwide, with epileptic seizures being a common comorbidity that can exacerbate cognitive deterioration in affected individuals, thus highlighting the importance of early therapeutic intervention. It is determined that deletion of Ms4a4a, an AD-associated gene, exacerbates seizures in amyloid β (Aβ)-driven AD mouse model. MS4A4A is significantly upregulated in brain lesions in patients with epilepsy. Single-cell sequencing reveals that MS4A4A is highly expressed in microglia within these lesions, linked to enhanced phagocytic activity. Mechanistic investigation delineates that deletion of Ms4a4a impairs microglial phagocytosis, accompanied by diminished calcium influx and disruptions in mitochondrial metabolic fitness. The cytosolic fragment of Ms4a4a is anchored to the cytoskeletal components, supporting its critical role in mediating phagocytosis. Induction of Ms4a4a through central delivery of LNP-Il4 alleviates seizure conditions. Collectively, these findings identify Ms4a4a as a potential therapeutic target for managing seizures in AD treatment.
Collapse
Affiliation(s)
- Meng Jiang
- Institute for Immunology and School of Basic Medical Sciences and Beijing Key Laboratory of Immunological Research of Allergy (LIRA)Tsinghua UniversityBeijing100084China
| | - Qingqing Li
- Institute for Immunology and School of Basic Medical Sciences and Beijing Key Laboratory of Immunological Research of Allergy (LIRA)Tsinghua UniversityBeijing100084China
| | - Jianhui Chen
- Institute for Immunology and School of Basic Medical Sciences and Beijing Key Laboratory of Immunological Research of Allergy (LIRA)Tsinghua UniversityBeijing100084China
| | - Ruochong Li
- School of Life SciencesTsinghua UniversityBeijing100084China
| | - Jun Yao
- ENO Bio mRNA Innovation InstituteShenzhen Rhegen Biotechnology Co. LtdShenzhen518000China
| | - Yong Hu
- ENO Bio mRNA Innovation InstituteShenzhen Rhegen Biotechnology Co. LtdShenzhen518000China
| | - Haizheng Zhang
- School of Life SciencesTsinghua UniversityBeijing100084China
| | - Lixin Cai
- Pediatric Epilepsy CenterPeking University First HospitalBeijing100034China
| | - Maoguo Luo
- School of Life SciencesTsinghua UniversityBeijing100084China
| | - Yu Sun
- Pediatric Epilepsy CenterPeking University First HospitalBeijing100034China
| | - Wenwen Zeng
- Institute for Immunology and School of Basic Medical Sciences and Beijing Key Laboratory of Immunological Research of Allergy (LIRA)Tsinghua UniversityBeijing100084China
- SXMU‐Tsinghua Collaborative Innovation Center for Frontier MedicineTaiyuan030001China
- Tsinghua‐Peking Center for Life SciencesBeijing100084China
| |
Collapse
|
25
|
Kim E, Tanzi RE, Choi SH. Therapeutic potential of exercise-hormone irisin in Alzheimer's disease. Neural Regen Res 2025; 20:1555-1564. [PMID: 38993140 PMCID: PMC11688551 DOI: 10.4103/nrr.nrr-d-24-00098] [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: 01/24/2024] [Revised: 05/20/2024] [Accepted: 06/04/2024] [Indexed: 07/13/2024] Open
Abstract
Irisin is a myokine that is generated by cleavage of the membrane protein fibronectin type III domain-containing protein 5 (FNDC5) in response to physical exercise. Studies reveal that irisin/FNDC5 has neuroprotective functions against Alzheimer's disease, the most common form of dementia in the elderly, by improving cognitive function and reducing amyloid-β and tau pathologies as well as neuroinflammation in cell culture or animal models of Alzheimer's disease. Although current and ongoing studies on irisin/FNDC5 show promising results, further mechanistic studies are required to clarify its potential as a meaningful therapeutic target for alleviating Alzheimer's disease. We recently found that irisin treatment reduces amyloid-β pathology by increasing the activity/levels of amyloid-β-degrading enzyme neprilysin secreted from astrocytes. Herein, we present an overview of irisin/FNDC5's protective roles and mechanisms against Alzheimer's disease.
Collapse
Affiliation(s)
- Eunhee Kim
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
| | - Rudolph E. Tanzi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
| | - Se Hoon Choi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
26
|
Martins YA, Cardinali CAEF, Torrão AS. Age-related differences in long-term memory performance and astrocyte morphology in rat hippocampus. Neurobiol Aging 2025; 150:19-43. [PMID: 40043468 DOI: 10.1016/j.neurobiolaging.2025.02.006] [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: 02/10/2025] [Accepted: 02/24/2025] [Indexed: 04/10/2025]
Abstract
Astrocytes are neuromodulator cells. Their complex and dynamic morphology regulates neuronal signaling, synaptic plasticity, and neurogenesis. The impact of aging on astrocyte morphology is still under ongoing debate. Therefore, this study aimed to characterize astrocyte morphology in the hippocampus of older rats. 2-, 18-, and 20-month-old male Wistar rats were submitted to the object recognition test to assess their short- and long-term memories. CA1, CA2, CA3, and the dentate gyrus were collected for immunohistochemistry analysis and glial fibrillary acid protein (GFAP) immunostaining. Our results indicate that 20-month-old rats did not recognize or discriminate the novel object in the long-term memory test. Also, GFAP staining was greater in the oldest group for all analyzed areas. Morphometric and fractal analysis indicated shorter branch lengths and smaller sizes for astrocytes of 20-month-old rats. Overall, our results suggest that 20-month-old rats have long-term memory impairment, increased GFAP staining, and astrocyte dystrophy. These age-related alterations in astrocyte morphology are a resource for future studies exploring the role of astrocytes in age-related cognitive decline and age-related diseases.
Collapse
Affiliation(s)
- Yandara A Martins
- Departamento de Fisiologia e Biofisica, Universidade de São Paulo, Sao Paulo, Brazil.
| | | | - Andréa S Torrão
- Departamento de Fisiologia e Biofisica, Universidade de São Paulo, Sao Paulo, Brazil.
| |
Collapse
|
27
|
Aggarwal R, Kumar P, Kumar S, Tiwari S, Chaturvedi RK. Synthesis and biological evaluation of novel Trifluoromethylated Arylidene-hydrazinyl-thiazoles as neuroprotective agents. Bioorg Chem 2025; 159:108390. [PMID: 40139118 DOI: 10.1016/j.bioorg.2025.108390] [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: 01/22/2025] [Revised: 03/05/2025] [Accepted: 03/14/2025] [Indexed: 03/29/2025]
Abstract
Neurodegenerative diseases, a substantial global health challenge affecting millions, underscore the pressing need for novel and effective pharmacotherapeutic drugs to address these disorders. In this concern, a library of novel trifluoromethylated arylidene-hydrazinyl-thiazoles has been synthesized and assessed for their anti-neurodegenerative potential. Multicomponent regioselective chemical transformation has been carried out utilizing thiosemicarbazide, trifluoromethylated 1,3-diketones and heteroaryl aldehydes in the presence of N-bromosuccinimide (NBS) in refluxing ethanol. The regioisomeric structure of the synthesized products was unambiguously characterized by employing heteronuclear 2D NMR spectroscopic studies. All the synthesized derivatives were evaluated for their anti-neurodegenerative properties on rat brain hippocampus-derived Neural Stem Cells (NSCs), examining their impact on survival, proliferation and neuronal differentiation in vitro. Among the tested thiazole derivatives, compounds 4a, 4b, 4c, 4f, 4 g, 4b' and 4i' demonstrated a remarkable increase in the number of neuronal cells as compared to the control group within the NSC culture and also exhibited the ability to promote NSC differentiation towards the neuronal lineage. Additionally, the selected compounds showed protection against amyloid beta (Aβ)-induced neurotoxicity in NSCs culture. Incorporating the trifluoromethyl group into the thiazole scaffold is a pivotal factor in augmenting biopotency, resulting in a marked increase in the count of neuronal cells compared to their non-fluorinated thiazole counterparts.
Collapse
Affiliation(s)
- Ranjana Aggarwal
- Department of Chemistry, Kurukshetra University, Kurukshetra 136119, Haryana, India; Council of Scientific and Industrial Research-National Institute of Science Communication and Policy Research, New Delhi 110012, India.
| | - Prince Kumar
- Department of Chemistry, Kurukshetra University, Kurukshetra 136119, Haryana, India
| | - Suresh Kumar
- Department of Chemistry, Kurukshetra University, Kurukshetra 136119, Haryana, India
| | - Saurabh Tiwari
- Molecular Neurotoxicology and Cell Integrity Laboratory, Systems Toxicology and Health Risk Assessment Group, FEST Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh (U.P.), India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Rajnish Kumar Chaturvedi
- Molecular Neurotoxicology and Cell Integrity Laboratory, Systems Toxicology and Health Risk Assessment Group, FEST Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh (U.P.), India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
28
|
Cerqueni G, Terenzi V, Preziuso A, Serfilippi T, Piccirillo S, Di Vincenzo M, Ambrogini P, Amoroso S, Orciani M, Lariccia V, Magi S. Identification of glutamate-related disease-dependent alterations in subventricular NSCs of the 3xTg Alzheimer's disease model, could they be involved in attempting damage repair? Cell Tissue Res 2025; 400:241-253. [PMID: 39960548 DOI: 10.1007/s00441-025-03954-6] [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: 11/12/2024] [Accepted: 01/30/2025] [Indexed: 06/01/2025]
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disorder characterised by several factors, such as impaired glutamate neurotransmission affecting crucial functions. Neural stem cells (NSCs) are present in the adult brains of all mammalian species and contribute to the continuous generation of neural cells throughout life. The disruption of glutamate levels during the development of AD could impact NSCs' functionality, influencing their response to the microenvironment. In this work, we isolated adult neural stem cells from both triple transgenic (3xTg)-AD mice and age-matched wild type (WT) mice in order to gather information on any differences between them, particularly concerning the potential mechanisms involved in the internalisation of glutamate and its utilisation for energy production. The 3xTg model offers the ability to recapitulate human pathology with both plaque and tangle hallmarks that are involved in the process of glutamate release. In vitro culture 3xTg NSCs showed a slight morphological difference compared to WT cells and a massive reduction of proliferation and viability. Furthermore, 3xTg NSCs displayed an increase in the expression of glutamate transporters and glutamine synthetase, while glutamate dehydrogenase did not show any reduction, which is typical in AD brains. Data obtained from this basic research study suggest a possible involvement of glutamate in the cellular energy balance, indicating an attempted response of NSCs to the cytotoxic microenvironment in the early stage of AD pathology. This finding is of great interest, as it corroborates the hypothesis that targeting the glutamatergic system could be an extremely promising strategy for new therapeutics in AD.
Collapse
Affiliation(s)
- Giorgia Cerqueni
- Department of Biomedical Sciences and Public Health, School of Medicine, Marche Polytechnic University, Via Tronto 10/A, 60126, Ancona, AN, Italy.
- Department of Life Science, Health, and Health Professions, Link Campus University, Via del Casale Di San Pio V, 00165, Rome, RM, Italy.
| | - Valentina Terenzi
- Department of Biomedical Sciences and Public Health, School of Medicine, Marche Polytechnic University, Via Tronto 10/A, 60126, Ancona, AN, Italy
| | - Alessandra Preziuso
- Department of Biomedical Sciences and Public Health, School of Medicine, Marche Polytechnic University, Via Tronto 10/A, 60126, Ancona, AN, Italy
| | - Tiziano Serfilippi
- Department of Biomedical Sciences and Public Health, School of Medicine, Marche Polytechnic University, Via Tronto 10/A, 60126, Ancona, AN, Italy
| | - Silvia Piccirillo
- Department of Biomedical Sciences and Public Health, School of Medicine, Marche Polytechnic University, Via Tronto 10/A, 60126, Ancona, AN, Italy
| | - Mariangela Di Vincenzo
- Department of Clinical and Molecular Sciences, School of Medicine, Marche Polytechnic University, Via Tronto 10/A, 60126, Ancona, AN, Italy
| | - Patrizia Ambrogini
- Department of Biomolecular Sciences, University of Urbino "Carlo Bo", Via Ca' Le Suore 2-4, 61029, Urbino, PU, Italy
| | - Salvatore Amoroso
- Department of Biomedical Sciences and Public Health, School of Medicine, Marche Polytechnic University, Via Tronto 10/A, 60126, Ancona, AN, Italy
| | - Monia Orciani
- Department of Clinical and Molecular Sciences, School of Medicine, Marche Polytechnic University, Via Tronto 10/A, 60126, Ancona, AN, Italy.
| | - Vincenzo Lariccia
- Department of Biomedical Sciences and Public Health, School of Medicine, Marche Polytechnic University, Via Tronto 10/A, 60126, Ancona, AN, Italy
| | - Simona Magi
- Department of Biomedical Sciences and Public Health, School of Medicine, Marche Polytechnic University, Via Tronto 10/A, 60126, Ancona, AN, Italy
| |
Collapse
|
29
|
Zhang X, Xu H, Yin S, Gozal D, Khalyfa A. Obstructive sleep apnea and memory impairments: Clinical characterization, treatment strategies, and mechanisms. Sleep Med Rev 2025; 81:102092. [PMID: 40286536 DOI: 10.1016/j.smrv.2025.102092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 03/31/2025] [Accepted: 04/07/2025] [Indexed: 04/29/2025]
Abstract
Obstructive sleep apnea (OSA), is associated with dysfunction in the cardiovascular, metabolic and neurological systems. However, the relationship between OSA and memory impairment, intervention effects, and underlying pathways are not well understood. This review summarizes recent advances in the clinical characterization, treatment strategies, and mechanisms of OSA-induced memory impairments. OSA patients may exhibit significant memory declines, including impairments in working memory from visual and verbal sources. The underlying mechanisms behind OSA-related memory impairment are complex and multifactorial with poorly understood aspects that require further investigation. Neuroinflammation, oxidative stress, neuronal damage, synaptic plasticity, and blood-brain barrier dysfunction, as observed under exposures to intermittent hypoxia and sleep fragmentation are likely contributors to learning and memory dysfunction. Continuous positive airway pressure treatment can provide remarkable relief from memory impairment in OSA patients. Other treatments are emerging but need to be rigorously evaluated for cognitive improvement. Clinically, reliable and objective diagnostic tools are necessary for accurate diagnosis and clinical characterization of cognitive impairments in OSA patients. The complex links between gut-brain axis, epigenetic landscape, genetic susceptibility, and OSA-induced memory impairments suggest new directions for research. Characterization of clinical phenotypic clusters can facilitate advances in precision medicine to predict and treat OSA-related memory deficits.
Collapse
Affiliation(s)
- Xiaoman Zhang
- Department of Otolaryngology Head and Neck Surgery & Shanghai Key Laboratory of Sleep Disordered Breathing & Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Huajun Xu
- Department of Otolaryngology Head and Neck Surgery & Shanghai Key Laboratory of Sleep Disordered Breathing & Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Shankai Yin
- Department of Otolaryngology Head and Neck Surgery & Shanghai Key Laboratory of Sleep Disordered Breathing & Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - David Gozal
- Department of Pediatrics and Office of the Dean, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, 25755, USA
| | - Abdelnaby Khalyfa
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, 25755, USA.
| |
Collapse
|
30
|
Micheli L, Caruso M, D’Andrea G, Volpe D, Ceccarelli M, Tirone F. Survey of transcriptome analyses of hippocampal neurogenesis with focus on adult dentate gyrus stem cells. Front Cell Dev Biol 2025; 13:1605116. [PMID: 40519263 PMCID: PMC12162651 DOI: 10.3389/fcell.2025.1605116] [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: 04/02/2025] [Accepted: 05/05/2025] [Indexed: 06/18/2025] Open
Abstract
Adult mammalian brains generate new neurons throughout life in two main niches, the dentate gyrus of the hippocampus and the subventricular zone, starting from neural stem cells (NSCs). Adult hippocampal neurogenesis is crucial for learning and memory and decreases during aging. As defined in mouse models, NSCs, which are prevalently quiescent, develop into proliferating progenitor cells, neuroblasts, and immature and mature neurons. Two visions for NSC self-renewal in the dentate gyrus have been proposed, one postulating persistent self-renewal, with cycles of rest and reactivation even in old age, and the other proposing a short-lived NSC model. Single-cell RNA sequencing and clonal studies, discussed in this review, have shed light on the developmental steps of neurogenic cells and the modality of self-renewal, revealing the presence in the adult dentate gyrus of NSC heterogeneous populations, one long-lived and another rapidly depleted at an early age. Another relevant question is whether adult neurogenesis occurs in humans. A few single-cell RNA-seq studies show that new neurons, with prolonged neuronal maturation, are continuously generated at low frequency from stem/progenitor cells, which results in the accumulation of immature granule cell neurons. This suggests an important role of these cells in human neurogenesis and hence interspecies differences in the neurogenic process dynamics. This review is focused on transcriptomic studies that have faced these and other NSC issues by analyzing developmental trajectories of neural cells and NSCs gene expression profiles in specific experimental settings of hippocampal neurogenesis, and also in mouse models with deletion or overexpression of specific genes to reproduce neural pathologies.
Collapse
Affiliation(s)
- Laura Micheli
- Institute of Biochemistry and Cell Biology, National Research Council, Rome, Italy
| | - Maurizia Caruso
- Institute of Biochemistry and Cell Biology, National Research Council, Rome, Italy
| | - Giorgio D’Andrea
- Institute of Biochemistry and Cell Biology, National Research Council, Rome, Italy
| | - Daniel Volpe
- Institute of Biochemistry and Cell Biology, National Research Council, Rome, Italy
| | - Manuela Ceccarelli
- Onco-Hematology, Cell Therapy, Gene Therapies and Hemopoietic Transplant, Bambino Gesù Children’s Hospital IRCCS, Rome, Italy
| | - Felice Tirone
- Institute of Biochemistry and Cell Biology, National Research Council, Rome, Italy
| |
Collapse
|
31
|
Dolgacheva LP, Zinchenko VP, Nadeev AD, Goncharov NV. Serotonergic Regulation in Alzheimer's Disease. Int J Mol Sci 2025; 26:5218. [PMID: 40508026 PMCID: PMC12154332 DOI: 10.3390/ijms26115218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 05/18/2025] [Accepted: 05/27/2025] [Indexed: 06/16/2025] Open
Abstract
Serotonin (5-HT) is a neurotransmitter that also plays an important role in the regulation of vascular tone and angiogenesis. This review focuses on the involvement of the 5-HT system in pathological processes leading to the development of Alzheimer's disease (AD). There is evidence that damage or dysfunction of the 5-HT system contributes to the development of AD, and different subtypes of 5-HT receptors are a potential target for the treatment of AD. A link has been established between AD, depression, stress, and 5-HT deficiency in the brain. There are new data on the role of circadian rhythms in modulating stress, depression, and the 5-HT system; amyloid β (Aβ) plaque clearance; and AD progression. Circadian disruption inhibits Aβ plaque clearance and modulates AD progression. The properties and functions of 5-HT, its receptors, and serotonergic neurons are presented. Special attention is paid to the central role of 5-HT in brain development, including neurite outgrowth, regulation of somatic morphology, motility, synaptogenesis, control of dendritic spine shape and density, neuronal plasticity determining its role in network regeneration, and changes in innervation after brain damage. The results of different studies indicate that the interaction of amyloid β oligomers (AβO) with mitochondria is a sufficient trigger for AD-related neurodegeneration. The action of 5-HT leads to an improvement in mitochondrial quality and the restoration of brain areas after traumatic brain injury, chronic stress, or developmental disorders in AD. The role of a healthy lifestyle and drugs acting on serotonin receptors in the prevention and treatment of AD is discussed.
Collapse
Affiliation(s)
- Lyudmila P. Dolgacheva
- Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, Institute of Cell Biophysics of the Russian Academy of Sciences, Pushchino 142290, Russia (V.P.Z.)
| | - Valery P. Zinchenko
- Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, Institute of Cell Biophysics of the Russian Academy of Sciences, Pushchino 142290, Russia (V.P.Z.)
| | - Alexander D. Nadeev
- Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, Institute of Cell Biophysics of the Russian Academy of Sciences, Pushchino 142290, Russia (V.P.Z.)
| | - Nikolay V. Goncharov
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, Saint Petersburg 194223, Russia
| |
Collapse
|
32
|
Ristori S, Bertoni G, Bientinesi E, Monti D. The Role of Nutraceuticals and Functional Foods in Mitigating Cellular Senescence and Its Related Aspects: A Key Strategy for Delaying or Preventing Aging and Neurodegenerative Disorders. Nutrients 2025; 17:1837. [PMID: 40507106 PMCID: PMC12157746 DOI: 10.3390/nu17111837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2025] [Revised: 05/13/2025] [Accepted: 05/22/2025] [Indexed: 06/16/2025] Open
Abstract
As life expectancy continues to increase, it becomes increasingly important to extend healthspan by targeting mechanisms associated with aging. Cellular senescence is recognized as a significant contributor to aging and neurodegenerative disorders. This review examines the emerging role of nutraceuticals and functional foods as potential modulators of cellular senescence, which may, in turn, influence the development of neurodegenerative diseases. An analysis of experimental studies indicates that bioactive compounds, including polyphenols, vitamins, and spices, possess substantial antioxidants, anti-inflammatory and epigenetic properties. These nutritional senotherapeutic agents effectively scavenge reactive oxygen species, modulate gene expression, and decrease the secretion of senescence-associated secretory phenotype factors, minimizing cellular damage. Nutraceuticals can enhance mitochondrial function, reduce oxidative stress, and regulate inflammation, key factors in aging and diseases like Alzheimer's and Parkinson's. Furthermore, studies reveal that specific bioactive compounds can reduce senescence markers in cellular models, while others exhibit senostatic and senolytic properties, both directly and indirectly. Diets enriched with these nutraceuticals, such as the Mediterranean diet, have been correlated with improved brain health and the deceleration of aging. Despite these promising outcomes, direct evidence linking these compounds to reducing senescent cell numbers remains limited, highlighting the necessity for further inquiry. This review presents compelling arguments for the potential of nutraceuticals and functional foods to promote longevity and counteract neurodegeneration by exploring their molecular mechanisms. The emerging relationship between dietary bioactive compounds and cellular senescence sets the stage for future research to develop effective preventive and therapeutic strategies for age-related diseases.
Collapse
Affiliation(s)
| | | | | | - Daniela Monti
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy; (S.R.); (G.B.); (E.B.)
| |
Collapse
|
33
|
Surya K, Rathinam A, Abubakkar MN, Jayachandran KS, Kandasamy M, Anusuyadevi M. Resveratrol mitigates activated astrocytes and microglia preventing Alzheimer's Disease (AD) progression and facilitates neuronal communication in Amyloid-β25-35 induced rat model for AD: A special emphasis on non-neuronal involvement in AD pathophysiology. Psychopharmacology (Berl) 2025:10.1007/s00213-025-06814-x. [PMID: 40423784 DOI: 10.1007/s00213-025-06814-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 05/06/2025] [Indexed: 05/28/2025]
Abstract
RATIONALE Amyloid deposits initiate neuroinflammation by activating astrocytes and microglia in the hippocampus, increasing neuronal vulnerability and loss. Astrocytes, while essential for cerebral function, can contribute to neuronal dysfunction by retracting neuronal synapses, that forms a consequence of neuroinflammation, leading to cognitive deficits in Alzheimer's disease (AD). Upon Amyloid-β (Aβ) deposition, astrocytes become reactive as part of a repair mechanism, however this process can impair neurogenesis resulting in AD progression. OBJECTIVE The current study hypothesizes that resveratrol (RSV) can address inflammation and promote neural regeneration, mitigating cognitive decline. Our previous research highlights RSV's homeostatic effect through SIRT1 normalization, which is crucial in preventing AD progression. However, its neurogenic potential in AD remains underexplored. METHODS In this study, Aβ25-35-induced AD rat model was used to study the anti-inflammatory, neurogenic and cellular homeostatic effect of RSV (30 mg/kg) for four weeks. RESULTS Results showed increased Doublecortin expressing cells, indicating favorable neurogenesis in hippocampus. Immunofluorescence of microglia and astrocytes in the hippocampus revealed that RSV counteracted their activation by reducing the formation of engulfing microglia and elongated astrocytes. Behavioral assessments using the Morris water maze and cued radial arm maze demonstrated significant improvements in spatial and learning memory. These cognitive improvements were supported by increased choline acetyltransferase and SIRT1 levels. CONCLUSION These findings suggest that RSV effectively reduces neuroinflammation, promotes neurogenesis in the sub granular zone of the hippocampus, and improves learning and memory in both control and AD conditions via SIRT1. This study highlights RSV's potential as a suitable therapeutic agent for AD.
Collapse
Affiliation(s)
- Kumar Surya
- Molecular Neurogerontology Laboratory, Department of Biochemistry, School of Life Sciences, Bharathidasan University, Tiruchirappalli, 620024, Tamil Nadu, India
| | - Anitha Rathinam
- Molecular Neurogerontology Laboratory, Department of Biochemistry, School of Life Sciences, Bharathidasan University, Tiruchirappalli, 620024, Tamil Nadu, India
| | - Meher Nisha Abubakkar
- Molecular Neurogerontology Laboratory, Department of Biochemistry, School of Life Sciences, Bharathidasan University, Tiruchirappalli, 620024, Tamil Nadu, India
| | - Kesavan Swaminathan Jayachandran
- Molecular Cardiology and Drug Discovery Laboratory, Department of Bioinformatics, School of Life Sciences, Bharathidasan University, Tiruchirappalli, 620024, Tamil Nadu, India
| | - Mahesh Kandasamy
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, 620024, Tamil Nadu, India
- University Grants Commission-Faculty Recharge Programme, (UGC-FRP), New Delhi- 110002, India
| | - Muthuswamy Anusuyadevi
- Molecular Neurogerontology Laboratory, Department of Biochemistry, School of Life Sciences, Bharathidasan University, Tiruchirappalli, 620024, Tamil Nadu, India.
| |
Collapse
|
34
|
Keeling EG, Bergamino M, Ott LR, McElvogue MM, Stokes AM. Repeatability and reliability of cerebrovascular reactivity in young adults using multi-echo, multi-contrast MRI. J Cereb Blood Flow Metab 2025:271678X251345292. [PMID: 40415374 PMCID: PMC12106380 DOI: 10.1177/0271678x251345292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 05/05/2025] [Accepted: 05/08/2025] [Indexed: 05/27/2025]
Abstract
Cerebrovascular reactivity (CVR) shows promise as a biomarker of vascular integrity and may benefit from a repeatable, reliable, and microvasculature-sensitive acquisition. A combined spin- and gradient-echo (SAGE) functional MRI (fMRI) acquisition may improve repeatability and reliability compared to single spin- (SE) and gradient-echo (GRE) fMRI and provide a microvascular-weighted analysis. The most repeatable and reliable MRI acquisition CVR maps were compared across three CVR paradigms: a breath-hold task, a breath modulation task, and a resting state acquisition. SAGE-fMRI data was acquired in fifteen young adults at two timepoints. Mean gray matter (GM) within-subject coefficient of variation (wCV) and intraclass correlation coefficient (ICC) were compared within the quantitative and weighted SAGE-fMRI CVR maps and single GRE- and SE-fMRI CVR. Total and microvascular MRI inputs with lowest wCV and highest ICC were used to compare three CVR paradigms. Total and microvascular weighted SAGE-fMRI CVR had the lowest wCV and highest ICC across paradigms. The breath-hold paradigm produced significantly higher GM CVR estimates. SAGE repeatably and reliably measures CVR and offers a simultaneous, complementary analysis on total and microvascular scales. The breath-hold paradigm showed significantly higher CVR estimates, but less compliance-dependent protocols may be ideal for applications in patient populations.
Collapse
Affiliation(s)
- Elizabeth G Keeling
- Barrow Neuroimaging Innovation Center, Barrow Neurological Institute, Phoenix, AZ, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Maurizio Bergamino
- Barrow Neuroimaging Innovation Center, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Lauren R Ott
- Barrow Neuroimaging Innovation Center, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Molly M McElvogue
- Barrow Neuroimaging Innovation Center, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Ashley M Stokes
- Barrow Neuroimaging Innovation Center, Barrow Neurological Institute, Phoenix, AZ, USA
| |
Collapse
|
35
|
Li W, Ma S, Li M. Intervention Role of APOE in CNS Diseases: APOE Actions and APOE Neurogenesis Capability. Mol Neurobiol 2025:10.1007/s12035-025-05028-8. [PMID: 40402407 DOI: 10.1007/s12035-025-05028-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 05/02/2025] [Indexed: 05/23/2025]
Abstract
Neurogenesis is a biological process in which new neurons are generated from neural stem cells (NSCs) in specific neural niches in the brain. Impaired neurogenesis, characterized by the progressive loss of neurons, leads to cognitive and motor disabilities and is a hallmark of central nervous system (CNS) diseases. Conversely, enhancing neurogenesis has been shown to alleviate the symptoms of CNS diseases. Apolipoprotein E (APOE) is a protein that plays various biological roles in CNS diseases. Emerging research indicates that APOE is involved in adult neurogenesis, which is crucial for maintaining the neural progenitor pool in the dentate gyrus (DG) and synaptic activity. Therefore, APOE could be a therapeutic target for promoting neurogenesis in the treatment and intervention of CNS diseases. In this context, we present a comprehensive overview of the clinical evidence supporting the role of APOE in CNS diseases on the basis of a meta-analysis. We also discuss the neurogenic potential of APOE, which has significant implications not only for understanding the biological underpinnings of neurological diseases but also for developing novel treatment strategies for CNS diseases.
Collapse
Affiliation(s)
- Wenhua Li
- State Key Laboratory of Component-Based Chinese Medicine, Ministry of Education Key Laboratory of Pharmacologyof, Traditional Chinese Medicine Formulae, Institute of Traditional Chinese Medicine , Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Suya Ma
- Guang'anmen Hospital, China, Academy of Chinese Medicine Sciences , Beijing, 100053, China
| | - Min Li
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China
| |
Collapse
|
36
|
Ferrero-Sereno P, Palomo-López P, Mendoza-Muñoz M, Luna-Castaño P, Caballero-De la Calle R, Muñoz-Bermejo L. Digital Health Literacy and Physical Activity Programme for Improvement of Quality of Life in Caregivers of People with Dementia (CAREFIT): Study Protocol. Healthcare (Basel) 2025; 13:1219. [PMID: 40508835 PMCID: PMC12154072 DOI: 10.3390/healthcare13111219] [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: 04/12/2025] [Revised: 05/15/2025] [Accepted: 05/20/2025] [Indexed: 06/16/2025] Open
Abstract
Background/Objectives: Dementia involves progressive cognitive and functional deterioration that leads to dependence and overload on family caregivers. This overload has a negative impact on the physical, mental, emotional, and occupational health of caregivers, leading to occupational imbalance and problems arising from an inadequate distribution of time devoted to caregiving. This project aims to evaluate the effects of the technology-based CAREFIT programme, structured around physical activity interventions, education, and psychoemotional and social support, on the health-related quality of life and emotional well-being of informal caregivers. Methods: The experimental group will develop the intervention programme, which will last 8 weeks and combine educational activities, physical activities, and psychoemotional and social support. Before beginning the intervention, the entire experimental group will receive a training session and educational materials on how to access and use the platform. The CAREFIT platform will consist of two educational sessions and two weekly physical sessions, combined with psychoemotional and social support activities that participants must complete. Initial, final, and follow-up evaluations will be conducted. The HRQoL and psychoemotional health (stress, anxiety, depression, and perceived social support and burden) of caregivers of people with dementia will be the main outcome measures. The effects of the intervention on the study variables will be assessed using a repeated-measures analysis of variance (ANOVA). Conclusions: The proposed protocol for the CAREFIT programme represents an innovative and multidisciplinary initiative that leverages a digital platform to promote the well-being of informal caregivers of people with dementia. This approach combines health literacy and strengthened psychoemotional and social support. Through this integration, the goal is to reduce the levels of burden, stress, anxiety, and depression among primary caregivers, while strengthening their self-care capabilities and social support networks.
Collapse
Affiliation(s)
- Patricia Ferrero-Sereno
- Social Impact and Innovation in Health (INHealth), University Centre of Merida, University of Extremadura, 06800 Mérida, Spain
- Research Group Nurse-In, University Alfonso X el Sabio, Villanueva de la Cañada, Comunidad de, 28691 Madrid, Spain; (P.L.-C.); (R.C.-D.l.C.)
| | | | - María Mendoza-Muñoz
- Research Group on Physical and Heath Literacy and Health-Related Quality of Life (PHYQOL), Faculty of Sports Sciences, University of Extremadura, 10003 Cáceres, Spain;
| | - Patricia Luna-Castaño
- Research Group Nurse-In, University Alfonso X el Sabio, Villanueva de la Cañada, Comunidad de, 28691 Madrid, Spain; (P.L.-C.); (R.C.-D.l.C.)
| | - Raquel Caballero-De la Calle
- Research Group Nurse-In, University Alfonso X el Sabio, Villanueva de la Cañada, Comunidad de, 28691 Madrid, Spain; (P.L.-C.); (R.C.-D.l.C.)
| | - Laura Muñoz-Bermejo
- Social Impact and Innovation in Health (INHealth), University Centre of Merida, University of Extremadura, 06800 Mérida, Spain
| |
Collapse
|
37
|
Wang J, Yao J, Wang Z. Identification of shared mechanisms between Alzheimer's disease and atherosclerosis by integrated bioinformatics analysis. Eur J Med Res 2025; 30:408. [PMID: 40405226 PMCID: PMC12096694 DOI: 10.1186/s40001-025-02642-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 04/27/2025] [Indexed: 05/24/2025] Open
Abstract
Alzheimer's disease (AD) and atherosclerosis (AS) are two interacting diseases mostly affecting aged adults. AD is characterized by the deposition of neuritic plaques mainly consisting of Aβ, and AS is defined by the formation of atheromatous plaque along the vascular wall. The shared mechanisms underlying the pathogenesis of AD and AS remain unclear. Here we applied several bioinformatic analyses of bulk sequencing data sets of AD brain tissues and atherosclerotic plaques to seek relevant genes between AD and AS. WIPF3, was identified as the most affected gene in both diseases using weighted gene co-expression network analysis, machine-learning-based Lasso Cox regression analysis and random forest analysis. Furthermore, immune cell infiltration analysis of AS data sets and cell portion of single-cell RNA sequencing data from AD patients revealed an essential role of inflammation in the co-occurrence of AD and AS. Taken together, WIPF3 deficiency and inflammation may simultaneously mediate both AD and AS and could be potential targets for the prevention and therapy of these two closely related diseases.
Collapse
Affiliation(s)
- Jukun Wang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jing Yao
- Cerebrovascular Disease Department, Neurological Disease Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Zhe Wang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China.
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
38
|
Sun L, Wu Y, Yang J, Liang J, Li P, Yu X, Meng N, Sun T, Wang M, Chen C. The brain-white adipose tissue axis may play a crucial role in diabetes mellitus: a metabolic network analysis using total-body PET/CT imaging. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07337-5. [PMID: 40392299 DOI: 10.1007/s00259-025-07337-5] [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: 12/19/2024] [Accepted: 05/05/2025] [Indexed: 05/22/2025]
Abstract
PURPOSE This study aims to construct an individualized glucose metabolism network using total-body 18F-FDG PET imaging, which provides a comprehensive view of glucose metabolism across various organs, to explore the role of inter-organ interactions in Diabetes Mellitus (DM). METHODS In this study, we constructed covariance metabolic networks using static total-body PET images, normalized by lean body mass, from 36 patients with DM (DM group) and 36 age- and sex-matched healthy controls (HC group). Differences in network properties between the DM and HC groups were evaluated at both group and individual levels. In addition, correlation analysis was performed to explore the relationship between network properties and baseline clinical data in the DM subjects. RESULTS We observed that the same edges in the first three edges with the largest values were brain-subcutaneous adipose tissue (SAT) and brain-visceral adipose tissue (VAT) at both group and individual levels. There was a positive correlation between the brain-VAT and BMI and there was a negative correlation between the brain-SAT and age. The most perturbed organ was the brain at both group and individual levels, and there was a positive correlation between the strength of abnormality of brain and age. CONCLUSION This study successfully used static total-body PET imaging to construct individualized glucose metabolism networks for patients with DM, identifying the brain-VAT and brain-SAT as the most significantly altered edge and the brain as the most affected organ. These findings provide novel insights into the role of the brain-white adipose tissue axis in glucose metabolism in DM.
Collapse
Affiliation(s)
- Lubing Sun
- Department of Medical Imaging, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, Zhengzhou, China
- Clinical Bioinformatics Experimental Center, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, Zhengzhou, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Yaping Wu
- Department of Medical Imaging, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, Zhengzhou, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Junpeng Yang
- Department of Endocrinology, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, Zhengzhou, China
| | - Junting Liang
- Clinical Bioinformatics Experimental Center, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, Zhengzhou, China
| | - Panlong Li
- Department of Medical Imaging, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, Zhengzhou, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Xuan Yu
- Department of Medical Imaging, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, Zhengzhou, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Nan Meng
- Department of Medical Imaging, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, Zhengzhou, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Tao Sun
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| | - Meiyun Wang
- Department of Medical Imaging, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, Zhengzhou, China.
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
| | - Chuanliang Chen
- Department of Medical Imaging, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, Zhengzhou, China.
- Clinical Bioinformatics Experimental Center, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, Zhengzhou, China.
| |
Collapse
|
39
|
Campbell FC. Diet, dementia, and the hippocampus. Stem Cells Transl Med 2025; 14:szaf007. [PMID: 40387785 PMCID: PMC12087336 DOI: 10.1093/stcltm/szaf007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Accepted: 01/19/2025] [Indexed: 05/20/2025] Open
|
40
|
Ong J, Wu Q, Sasaki K, Isoda H, Szele FG. Nutraceuticals: using food to enhance brain health by modulating postnatal neurogenesis in animal models and patient populations. Stem Cells Transl Med 2025; 14:szaf006. [PMID: 40387786 PMCID: PMC12087346 DOI: 10.1093/stcltm/szaf006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 11/30/2024] [Indexed: 05/20/2025] Open
Abstract
Adult hippocampal neurogenesis, while occurring throughout life, decreases with age and in some neurodegenerative diseases. As decreased hippocampal neurogenesis is correlated with cognitive decline, efforts have been made to increase levels of neurogenesis, either through natural compounds, environmental interventions or novel pharmacological compounds. Nutraceuticals are food products with medical benefits such as antioxidation, anti-inflammation or neuroprotection. There has been increasing interest in these "functional foods" and their active compounds in recent years, providing natural alternatives to de novo pharmaceuticals. This review highlights key nutraceuticals that promote neurogenesis and/or improve cognitive outcomes. By outlining the effects of these compounds in the animal models employed and in clinical populations, we also suggest further investigations. We examine common targets and pathways through which these nutraceuticals are believed to exert pro-neurogenic effects. Most nutraceutical preparations contain multiple components, any of which may exert effects on neurogenesis. Identifying key active compounds in nutraceuticals may enable researchers to better understand their effects and standardize doses across studies. The less stringent regulatory requirements for nutraceuticals can be a double-edged sword. While allowing easier access to the beneficial effects, higher doses of these compounds may have detrimental effects. Hence, research in this field should not only aim to identify the benefits of these compounds but also to identify efficacious and safe dosages for them. Our aims are to provide understanding of nutraceuticals, provide evidence for their benefits on neurogenesis and neurogenesis-related behaviors and finally to summarize potential mechanisms and help guide future work.
Collapse
Affiliation(s)
- Jun Ong
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, United Kingdom
| | - Qingqing Wu
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, United Kingdom
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba, Ibaraki 305-8572, Japan
| | - Kazunori Sasaki
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba, Ibaraki 305-8572, Japan
- Open Innovation Laboratory for Food and Medicinal Resource Engineering, National Institute of Advanced Industrial Science and Technology (AIST) and University of Tsukuba, Tsukuba, Ibaraki 305-8572, Japan
- Faculty of Pure and Applied Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8571, Japan
| | - Hiroko Isoda
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba, Ibaraki 305-8572, Japan
- Open Innovation Laboratory for Food and Medicinal Resource Engineering, National Institute of Advanced Industrial Science and Technology (AIST) and University of Tsukuba, Tsukuba, Ibaraki 305-8572, Japan
- Faculty of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8572, Japan
| | - Francis G Szele
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, United Kingdom
| |
Collapse
|
41
|
Cho J, Bae S, Jeon J, Transfeld J, Lee C, Nott A, Gao F, Seo J. Enhanced differentiation of neural progenitor cells in Alzheimer's disease into vulnerable immature neurons. iScience 2025; 28:112446. [PMID: 40384927 PMCID: PMC12084003 DOI: 10.1016/j.isci.2025.112446] [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: 11/11/2024] [Revised: 02/26/2025] [Accepted: 04/11/2025] [Indexed: 05/20/2025] Open
Abstract
Focusing on the early stages of Alzheimer's disease (AD) holds great promise. However, the specific events in neural cells preceding AD onset remain elusive. To address this, we utilized human-induced pluripotent stem cells carrying APPswe mutation to explore the initial changes associated with AD progression. We observed enhanced neural activity and early neuronal differentiation in APPswe cerebral organoids cultured for one month. This phenomenon was also evident when neural progenitor cells (NPCs) were differentiated into neurons. Furthermore, transcriptomic analyses of NPCs and neurons confirmed altered expression of neurogenesis-related genes in APPswe NPCs. We also found that the upregulation of reactive oxygen species (ROS) is crucial for early neuronal differentiation in these cells. In addition, APPswe neurons remained immature after initial differentiation with increased susceptibility to toxicity, providing valuable insights into the premature exit from the neural progenitor state and the increased vulnerability of neural cells in AD.
Collapse
Affiliation(s)
- Joonho Cho
- Department of Brain Sciences, DGIST, Daegu 42988, South Korea
| | - Simsung Bae
- Department of Brain Sciences, DGIST, Daegu 42988, South Korea
| | - Juyeong Jeon
- Department of Brain Sciences, DGIST, Daegu 42988, South Korea
| | - Janis Transfeld
- UK Dementia Research Institute at Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Changyeob Lee
- Department of Brain Sciences, DGIST, Daegu 42988, South Korea
| | - Alexi Nott
- UK Dementia Research Institute at Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Fan Gao
- Bioinformatics Resource Center, Beckman Institute of Caltech, Pasadena, CA 91125, USA
| | - Jinsoo Seo
- Department of Brain Sciences, DGIST, Daegu 42988, South Korea
- Center for Synapse Diversity and Specificity, DGIST, Daegu 42988, South Korea
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, South Korea
| |
Collapse
|
42
|
Pan W, Xu LF, Wang YX, Wang YJ, Wang JQ, Qian X, Zhou CZ, Wang H, Fan XH, Wang J. FOXG1 Improves Cognitive Function in Alzheimer's Disease by Promoting Endogenous Neurogenesis. FASEB J 2025; 39:e70582. [PMID: 40297942 DOI: 10.1096/fj.202403299rr] [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: 12/19/2024] [Revised: 04/09/2025] [Accepted: 04/18/2025] [Indexed: 04/30/2025]
Abstract
Strategies aimed at enhancing the capacity of neural stem cells (NSCs) to generate multipotential, proliferative, and migratory cell populations capable of efficient neuronal differentiation are crucial for structural repair following neurodegenerative damage. The role of Forkhead-box gene 1 (FOXG1) in pattern formation, cell proliferation, and specification has been established. However, its involvement in Alzheimer's disease (AD) remains largely unknown. Here, we investigated the association between Foxg1 gene variants and AD-like behavioral deficits, amyloid-β (Aβ) aggregate formation, as well as p21 expression. Furthermore, we explored whether targeting the FOXG1-regulated cell cycle contributes to the promotion of adult neurogenesis in the context of AD. In this study, we successfully induced overexpression of FOXG1 in the hippocampus of AD brains through adeno-associated virus-Foxg1 infusion. Activation of FOXG1 rescued spatial learning disabilities, short-term memory deficits, and sensorimotor gating impairments observed in AD transgenic animals. By inhibiting p21 WAF1/cyclin-dependent kinase interacting protein 1 (p21cip1/waf1)-mediated cell cycle arrest, FOXG1 facilitates the activation and proliferation of NSCs. Additionally, the Foxg1 gene promotes an increase in precursor population size and enhances neuroblast differentiation. These combined effects on proliferation and differentiation lead to the generation of postmitotic neurons within the hippocampus in AD animals. Together, these findings demonstrate the importance of cooperation between FOXG1 and p21 for maintaining NSC self-renewal while facilitating neuronal lineage progression and contributing to endogenous neurogenesis during AD. Elevating levels of FOXG1 either pharmacologically or through alternative means could potentially serve as a therapeutic strategy for treating AD.
Collapse
Affiliation(s)
- Wen Pan
- The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | - Long-Fei Xu
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | - Yu-Xin Wang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | - Yi-Jie Wang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | - Jia-Qing Wang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | - Xin Qian
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | - Cheng-Zhi Zhou
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | - Hua Wang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
- Zhenjiang Jieshengrui Biotechnology Co., Ltd, Zhenjiang, Jiangsu, People's Republic of China
| | - Xiao-Hua Fan
- The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | - Jia Wang
- The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
- Zhenjiang Jieshengrui Biotechnology Co., Ltd, Zhenjiang, Jiangsu, People's Republic of China
| |
Collapse
|
43
|
Zhang Q, Zheng J, Sun H, Zheng J, Ma Y, Ji Q, Chen D, Tang Z, Zhang J, He Y, Song T. The Notch Signaling Pathway: A Potential Target for Mental Disorders. Mol Neurobiol 2025:10.1007/s12035-025-05034-w. [PMID: 40372672 DOI: 10.1007/s12035-025-05034-w] [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: 08/04/2024] [Accepted: 05/02/2025] [Indexed: 05/16/2025]
Abstract
The highly conserved Notch signaling pathway plays a critical role in cell fate determination during metazoan development through cell-to-cell communication. The classical pathway consists of Notch receptors, ligands, intracellular effectors, DNA-binding proteins, and other regulatory molecules. Recent research has highlighted its involvement in the pathogenesis of several diseases. In autism, bipolar disorder, and schizophrenia, the Notch signaling pathway is implicated in key processes such as neuronal development and synaptic plasticity. Furthermore, it has been shown to play significant roles in other mental health conditions, including anxiety, depression, post-traumatic stress disorder, and neurocognitive disorders. However, the precise mechanisms underlying the contribution of Notch to these conditions remain poorly understood. This review examines the current understanding of the Notch signaling pathway in mental disorders, highlighting its role in their pathophysiology and summarizing therapeutic strategies aimed at modulating this pathway to improve mental health outcomes.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Immunology, Zunyi Medical University, Zunyi, China
| | - Jingxuan Zheng
- The Eighth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Hongqin Sun
- Department of Immunology, Zunyi Medical University, Zunyi, China
| | - Jishan Zheng
- Department of Immunology, Zunyi Medical University, Zunyi, China
| | - Yunyan Ma
- Department of Immunology, Zunyi Medical University, Zunyi, China
| | - Qinglu Ji
- School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Dengwang Chen
- Department of Immunology, Zunyi Medical University, Zunyi, China
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Zhengzhen Tang
- Department of Pediatrics, Third Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jidong Zhang
- Department of Immunology, Zunyi Medical University, Zunyi, China
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi, China
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Yuqi He
- School of Pharmacy, Zunyi Medical University, Zunyi, China
- Engineering Research Center of Key Technologies for Industrial Development of Dendrobium in Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Tao Song
- Department of Immunology, Zunyi Medical University, Zunyi, China.
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi, China.
| |
Collapse
|
44
|
Zhang Y, Ma T, Lu X, Hua H, Wu L, Chen Z. Mechanical mechanics-reclaiming a new battlefield for chronic liver disease. J Adv Res 2025:S2090-1232(25)00346-7. [PMID: 40379238 DOI: 10.1016/j.jare.2025.05.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 04/17/2025] [Accepted: 05/12/2025] [Indexed: 05/19/2025] Open
Abstract
BACKGROUND In the 21st century, significant breakthroughs have been made in the research of chronic liver disease. New biochemical markers of pathogenicity and corresponding drugs continue to emerge. However, current treatment strategies remain unsatisfactory due to complex pathological changes in the liver, including vascular dysfunction, myofibroblast-like transition, and local tissue necrosis in liver sinusoids. These challenges have created an urgent need for innovative, synergistic treatments. Mechanical mechanics is a growing field, with increasing evidence suggesting that mechanical signals play a role similar to that of biochemical markers. These signals influence response speed, conduction intensity, and functional diversity in regulating cell activities. AIM OF REVIEW This review summarizes the three main mechanical characteristics involved in the progression of "liver fibrosis-cirrhosis-hepatocellular carcinoma" and provides an in-depth interpretation of several mechanically-related targets. Finally, current and cutting-edge therapeutic strategies are proposed from a cellular perspective. Despite the many challenges that remain, this review is both relevant and significant.
Collapse
Affiliation(s)
- Yiheng Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Tianle Ma
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - XingXing Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Haibing Hua
- Department of Gastroenterology, Jiangyin Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Jiangyin 214400, China.
| | - Li Wu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Zhipeng Chen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
45
|
Wang X, Wang L, Bu Q, Xiao Y, Zhao Y, Jiang L, Dai Y, Li H, Liu H, Chen Y, Flores AD, Zhao Y, Cen X. LUZP1 Regulates Dendritic Spine Maturation and Synaptic Plasticity in the Hippocampal Dentate Gyrus of Mice. J Neurosci 2025; 45:e1867242025. [PMID: 40180573 PMCID: PMC12079723 DOI: 10.1523/jneurosci.1867-24.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 02/16/2025] [Accepted: 03/18/2025] [Indexed: 04/05/2025] Open
Abstract
Leucine zipper protein 1 (LUZP1) functions in the maintenance and dynamics of the cytoskeleton by interacting with actin and microtubules. Deficiency or mutation of LUZP1 is associated with brain developmental disorders; however, its precise role in brain function remains unclear. We showed that LUZP1 localizes to actin and is highly expressed in CaMKIIα-expressing neurons within the mouse hippocampal dentate gyrus. Depletion of LUZP1 impedes dendritic spine maturation, which is characterized by excess immature filopodia and loss of mature mushroom spines both in vitro and in vivo. LUZP1 knockdown reduces spontaneous electrical activity and synaptic plasticity in hippocampal neurons. Conditional deletion of LUZP1 in CaMKIIα-expressing neurons causes impaired learning and memory behavior in mice of both sexes. Mechanistically, LUZP1 control dendritic maturation by directly interacting with filamin A and modulating the Rac1-PAK1 signaling pathway. These findings shed light on the role of LUZP1 in regulating synaptic plasticity and brain function.
Collapse
Affiliation(s)
- Xiaojie Wang
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Neuroscience, City University of Hong Kong, Hong Kong 999077, China
| | - Liang Wang
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qian Bu
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuzhou Xiao
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yue Zhao
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Linhong Jiang
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yanping Dai
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hongchun Li
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Haxiaoyu Liu
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yaxing Chen
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Angelo D Flores
- Department of Neuroscience, City University of Hong Kong, Hong Kong 999077, China
| | - Yinglan Zhao
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaobo Cen
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
46
|
Butovsky O, Rosenzweig N. Alzheimer's disease and age-related macular degeneration: Shared and distinct immune mechanisms. Immunity 2025; 58:1120-1139. [PMID: 40324382 DOI: 10.1016/j.immuni.2025.04.013] [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: 01/22/2025] [Revised: 04/09/2025] [Accepted: 04/10/2025] [Indexed: 05/07/2025]
Abstract
Alzheimer's disease (AD) and age-related macular degeneration (AMD) represent the leading causes of dementia and vision impairment in the elderly, respectively. The retina is an extension of the brain, yet these two central nervous system (CNS) compartments are often studied separately. Despite affecting cognition vs. vision, AD and AMD share neuroinflammatory pathways. By comparing these diseases, we can identify converging immune mechanisms and potential cross-applicable therapies. Here, we review immune mechanisms highlighting the shared and distinct aspects of these two age-related neurodegenerative conditions, focusing on responses to hallmark disease manifestations, the opposite role of overlapping immune risk loci, and potential unified therapeutic approaches. We also discuss unique tissue requirements that may dictate different outcomes of conserved immune mechanisms and how we can reciprocally utilize lessons from AD therapeutics to AMD. Looking forward, we suggest promising directions for research, including the exploration of regenerative medicine, gene therapies, and innovative diagnostics.
Collapse
Affiliation(s)
- Oleg Butovsky
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Neta Rosenzweig
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
47
|
Song KW, Lim M, Monje M. Complex neural-immune interactions shape glioma immunotherapy. Immunity 2025; 58:1140-1160. [PMID: 40324379 DOI: 10.1016/j.immuni.2025.04.017] [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: 02/20/2025] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 05/07/2025]
Abstract
Rich neural-immune interactions in the central nervous system (CNS) shape its function and create a unique immunological microenvironment for immunotherapy in CNS malignancies. Far from the now-debunked concept of CNS "immune privilege," it is now understood that unique immunological niches and constant immune surveillance of the brain contribute in multifaceted ways to brain health and robustly influence immunotherapy approaches for CNS cancers. Challenges include immune-suppressive and neurotoxicity-promoting crosstalk between brain, immune, and tumor cells. Developing effective immunotherapies for cancers of the nervous system will require a deeper understanding of these neural-immune-malignant cell interactions. Here, we review progress and challenges in immunotherapy for gliomas of the brain and spinal cord in light of these unique neural-immune interactions and highlight future work needed to optimize promising immunotherapies for gliomas.
Collapse
Affiliation(s)
- Kun-Wei Song
- Department of Neurology and Neurological Sciences, Stanford University, Palo Alto, CA, USA
| | - Michael Lim
- Department of Neurosurgery, Stanford University, Palo Alto, CA, USA
| | - Michelle Monje
- Department of Neurology and Neurological Sciences, Stanford University, Palo Alto, CA, USA; Department of Neurosurgery, Stanford University, Palo Alto, CA, USA; Howard Hughes Medical Institute, Stanford University, Palo Alto, CA, USA.
| |
Collapse
|
48
|
Yalçin T, Kaya S. Effect of thymoquinone on hippocampal spexin levels in cisplatin-induced rats. Neurol Res 2025:1-9. [PMID: 40340641 DOI: 10.1080/01616412.2025.2504158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 05/04/2025] [Indexed: 05/10/2025]
Abstract
Neurotoxicity is a known side effect of the chemotherapeutic drug cisplatin (CIS). Thymoquinone (THQ) is a natural compound with strong neuroprotective, antioxidant, and anti-inflammatory effects. The objective of this study is to ascertain the impact of CIS on histopathological, biochemical, and spexin (SPX) immunoreactivity in the hippocampus, and to determine whether THQ has a protective role against these effects.Twenty-eight male Sprague - Dawley rats (8-10 weeks old,200 ± 20 g) were used in the study and randomly divided into four groups (n = 7): control (no administration), CIS (7 mg/kg on the first day), CIS+THQ (7 mg/kg CIS on the first day + 10 mg/kg/day THQ), and THQ (10 mg/kg/day THQ). On the 15th day, the rats were sacrificed. Hippocampus tissue samples were used for biochemical, histological, and immunohistochemical analyses. CISadministration significantly increased interleukin-6 (IL-6), malondialdehyde(MDA), histopathological changes, and SPX immunoreactivity in the hippocampus.THQ treatment was found to significantly reduce the adverse effects of.THQ treatment demonstrated neuroprotective effects againstCIS-induced damage in the hippocampus by modulating antioxidant activity, inflammatory response, and SPX immunoreactivity. We suggest that SPX, whose role and mechanism of action in cognitive, physiological, and pathological processes remains unclear, plays an active role in hippocampus-related functions. Further and more comprehensive studies on SPX are warranted.
Collapse
Affiliation(s)
- Tuba Yalçin
- Vocational Higher School of Healthcare Studies, Batman University, Batman, Turkey
| | - Sercan Kaya
- Vocational Higher School of Healthcare Studies, Batman University, Batman, Turkey
| |
Collapse
|
49
|
Huang D, Lv J, Gong W, Tian J, Gao X, Qin X, Du G, Zhou Y. Combining Metabolomics and Quantitative Analysis to Investigate Purine Metabolism Disorders in Depression and the Therapeutic Effect of Chaigui Granules. ACS Chem Neurosci 2025; 16:1749-1766. [PMID: 40209102 DOI: 10.1021/acschemneuro.4c00804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2025] Open
Abstract
Depression is a complex mental disorder. Studies have shown that purine metabolism disorders in depression and regulation of purine metabolites and related purinergic receptors may be an effective way to alleviate depression. Chaigui granules (CG) are a Chinese medicine prescription with antidepressant effects. Its antidepressant effect has been shown to be related to the improvement of purine metabolism disorders in depression. In this study, exogenous purine metabolite adenosine supplementation and adenosine A1 receptor antagonist (DPCPX) were employed to investigate the potential of Chaigui granules to exert an antidepressant effect by examining the behavioral indices of CUMS rats. The aim of this study was to determine whether the antidepressant effect of Chaigui granules is mediated by A1R receptors using DPCPX, an A1R receptor antagonist. Nontargeted metabolomic analysis was employed to compare and analyze the alterations in the metabolic profile of plasma and peripheral blood mononuclear cells (PBMCs) in each experimental group. Subsequently, combining the results from the metabolomics profile, targeted metabolomics was employed to identify key metabolites for purine metabolism. The objective was to investigate the effects of Chaigui granules, exogenous adenosine supplementation, and DPCPX on purine metabolism in depressed rats. Finally, the relevant signal pathways were validated by molecular biological means. The results of the depression-like behavior indicate that the antidepressant efficacy of Chaigui granules was associated with the modulation of adenosine and adenosine A1 receptor. Metabolomic analysis demonstrated that the Chaigui granule and adenosine exerted a pronounced regulatory effect on purine metabolism, and the regulatory effect on peripheral blood mononuclear cells (PBMCs) was markedly superior to that observed in plasma. In addition, targeted quantitative analysis showed that all eight purine metabolites were reversed after the administration of Chaigui granules and adenosine. Concurrently, the administration of an adenosine A1 receptor antagonist may serve to mitigate the regulatory impact of Chaigui granules on purine metabolites. Finally, the molecular biological results indicate that the antidepressant effect of Chaigui granules may be mediated by the A1R receptor, and it can play an antidepressant role by regulating the CAMP-PKA-CREB-BDNF pathway.
Collapse
Affiliation(s)
- Dehua Huang
- Modern Research Center for Traditional Chinese Medicine, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Road, Taiyuan, Shanxi 030006, P. R. China
- Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Shanxi University, No. 92, Wucheng Road, Taiyuan, Shanxi 030006, P. R. China
| | - Jiale Lv
- Modern Research Center for Traditional Chinese Medicine, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Road, Taiyuan, Shanxi 030006, P. R. China
- Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Shanxi University, No. 92, Wucheng Road, Taiyuan, Shanxi 030006, P. R. China
| | - Wenxia Gong
- Modern Research Center for Traditional Chinese Medicine, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Road, Taiyuan, Shanxi 030006, P. R. China
- Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Shanxi University, No. 92, Wucheng Road, Taiyuan, Shanxi 030006, P. R. China
| | - Junsheng Tian
- Modern Research Center for Traditional Chinese Medicine, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Road, Taiyuan, Shanxi 030006, P. R. China
- Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Shanxi University, No. 92, Wucheng Road, Taiyuan, Shanxi 030006, P. R. China
| | - Xiaoxia Gao
- Modern Research Center for Traditional Chinese Medicine, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Road, Taiyuan, Shanxi 030006, P. R. China
- Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Shanxi University, No. 92, Wucheng Road, Taiyuan, Shanxi 030006, P. R. China
| | - Xuemei Qin
- Modern Research Center for Traditional Chinese Medicine, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Road, Taiyuan, Shanxi 030006, P. R. China
- Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Shanxi University, No. 92, Wucheng Road, Taiyuan, Shanxi 030006, P. R. China
| | - Guanhua Du
- Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, P. R. China
| | - Yuzhi Zhou
- Modern Research Center for Traditional Chinese Medicine, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Road, Taiyuan, Shanxi 030006, P. R. China
- Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Shanxi University, No. 92, Wucheng Road, Taiyuan, Shanxi 030006, P. R. China
| |
Collapse
|
50
|
Zhang J, Liu T, Wu H, Wei J, Qu Q. Identification of NDUFV2, NDUFS7, OPA1, and NDUFA1 as biomarkers for Alzheimer's disease: Insights from oxidative stress and mitochondrial dysfunction in the hippocampus. J Alzheimers Dis 2025:13872877251339771. [PMID: 40329774 DOI: 10.1177/13872877251339771] [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: 05/08/2025]
Abstract
BackgroundAlzheimer's disease (AD) is characterized by amyloid-β deposits, neurofibrillary tangles, and hippocampal neurodegeneration, with oxidative stress and mitochondrial dysfunction playing critical roles in its pathogenesis. Identifying hub genes associated with these processes could advance biomarker discovery and therapeutic strategies.ObjectiveThis study aimed to identify key oxidative stress- and mitochondrial dysfunction-related genes in the AD hippocampus, evaluate their diagnostic potential, and explore therapeutic agents targeting these genes.MethodsWe analyzed datasets GSE48350 and GSE5281, encompassing 56 controls and 29 AD patients. Weighted gene co-expression network analysis (WGCNA) selected genes with significance (adjusted p-value < 0.05 and |logFC| ≥ 0.5). Further studies involved immune cell infiltration, Gene set enrichment analysis (GSEA), and intersecting differentially expressed genes (DEGs) with oxidative stress-related genes (ORGs) and mitochondrial dysfunction-related genes (MDRGs). Functional enrichment and Protein-protein interaction (PPI) analyses were conducted. Experimental validation was done in AD mouse models, and diagnostic potential was tested using datasets GSE28146 and GSE29652. Therapeutic drugs were predicted based on hub genes.ResultsAD showed altered immune cell expression. GSEA linked DEGs to nervous system processes and neurotransmitters. 194 oxidative stress-related DEGs were enriched in neuronal death and mitochondrial processes. PPI analysis identified 24 DEGs related to both oxidative stress and mitochondrial dysfunction (DEO-MDRGs), with diagnostic potential (AUC > 0.5). LASSO regression selected four DEO-MDRGs: NDUFV2, NDUFS7, OPA1, and NDUFA1. Their protein levels were reduced in AD mice with decreased mitochondrial function. These genes showed good diagnostic performance. Potential drugs, like ME-344 and metformin hydrochloride, may be useful in AD treatment.ConclusionsNDUFV2, NDUFS7, OPA1, and NDUFA1 can serve as biomarkers for AD diagnosis.
Collapse
Affiliation(s)
- Junshi Zhang
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Neurology, Huaihe Hospital of Henan University, Kaifeng, China
| | - Tingting Liu
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, China
| | - Haojie Wu
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, China
| | - Jianshe Wei
- Department of Neurology, Huaihe Hospital of Henan University, Kaifeng, China
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, China
| | - Qiumin Qu
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|