1
|
Yuasa-Kawada J, Kinoshita-Kawada M, Hiramoto M, Yamagishi S, Mishima T, Yasunaga S, Tsuboi Y, Hattori N, Wu JY. Neuronal guidance signaling in neurodegenerative diseases: Key regulators that function at neuron-glia and neuroimmune interfaces. Neural Regen Res 2026; 21:612-635. [PMID: 39995079 DOI: 10.4103/nrr.nrr-d-24-01330] [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: 10/31/2024] [Accepted: 01/27/2025] [Indexed: 02/26/2025] Open
Abstract
The nervous system processes a vast amount of information, performing computations that underlie perception, cognition, and behavior. During development, neuronal guidance genes, which encode extracellular cues, their receptors, and downstream signal transducers, organize neural wiring to generate the complex architecture of the nervous system. It is now evident that many of these neuroguidance cues and their receptors are active during development and are also expressed in the adult nervous system. This suggests that neuronal guidance pathways are critical not only for neural wiring but also for ongoing function and maintenance of the mature nervous system. Supporting this view, these pathways continue to regulate synaptic connectivity, plasticity, and remodeling, and overall brain homeostasis throughout adulthood. Genetic and transcriptomic analyses have further revealed many neuronal guidance genes to be associated with a wide range of neurodegenerative and neuropsychiatric disorders. Although the precise mechanisms by which aberrant neuronal guidance signaling drives the pathogenesis of these diseases remain to be clarified, emerging evidence points to several common themes, including dysfunction in neurons, microglia, astrocytes, and endothelial cells, along with dysregulation of neuron-microglia-astrocyte, neuroimmune, and neurovascular interactions. In this review, we explore recent advances in understanding the molecular and cellular mechanisms by which aberrant neuronal guidance signaling contributes to disease pathogenesis through altered cell-cell interactions. For instance, recent studies have unveiled two distinct semaphorin-plexin signaling pathways that affect microglial activation and neuroinflammation. We discuss the challenges ahead, along with the therapeutic potentials of targeting neuronal guidance pathways for treating neurodegenerative diseases. Particular focus is placed on how neuronal guidance mechanisms control neuron-glia and neuroimmune interactions and modulate microglial function under physiological and pathological conditions. Specifically, we examine the crosstalk between neuronal guidance signaling and TREM2, a master regulator of microglial function, in the context of pathogenic protein aggregates. It is well-established that age is a major risk factor for neurodegeneration. Future research should address how aging and neuronal guidance signaling interact to influence an individual's susceptibility to various late-onset neurological diseases and how the progression of these diseases could be therapeutically blocked by targeting neuronal guidance pathways.
Collapse
Affiliation(s)
| | | | | | - Satoru Yamagishi
- Department of Optical Neuroanatomy, Institute of Photonics Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takayasu Mishima
- Division of Neurology, Department of Internal Medicine, Sakura Medical Center, Toho University, Sakura, Japan
| | - Shin'ichiro Yasunaga
- Department of Biochemistry, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Yoshio Tsuboi
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Jane Y Wu
- Department of Neurology, Center for Genetic Medicine, Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
2
|
Orioli R, Sarno G, Seghetti F, Gobbi S, Belluti F, Feoli A, Massenzio F, Monti B, Spagnuolo R, Bartolini M, Castellano S, Bisi A. Exploiting acylaminopyrazole scaffold for polypharmacology approach in Alzheimer's disease. Eur J Med Chem 2025; 295:117799. [PMID: 40435831 DOI: 10.1016/j.ejmech.2025.117799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 05/21/2025] [Accepted: 05/22/2025] [Indexed: 06/11/2025]
Abstract
Neurodegenerative diseases currently represent one of the most serious health pitfalls for the world population. Considering their multifactorial nature, research has focused on the study of small molecules able to simultaneously tackle different targets involved in their onset and progression. In this paper, two sets of acylaminopyrazole-based compounds were designed to exploit the aminopyrazole core as a privileged structure properly decorated with an acyl moiety and a further amide function, connected with a proper spacer. Indeed, acylated aminopyrazoles could be able to establish the appropriate hydrogen bond pattern to both bind GSK-3β, responsible for tau hyperphosphorylation, prevent the formation of insoluble Aβ-protein aggregates and have the structural features to show chelating properties towards metals involved in neuroinflammation. The collection of compounds was tested in vitro for GSK-3β inhibition activity, antiaggregating and chelating properties. Selected compounds were able to inhibit GSK-3β in the low micromolar range with a reversible and competitive mechanism of action, as established by Microfluidic Mobility Shift Assay (MMSA) and showed metal chelating ability. Preliminary Structure Activity Relationships (SARs) to hit these distinct and interconnected targets for neuromodulation were established. Finally, selected compounds showed good apparent permeability values in parallel artificial membrane permeability assay (PAMPA) together with good cellular safety profile. The collected results validated acylaminopyrazole as promising scaffold for the development of multitarget-directed ligands. Compounds 1c and 4c emerged as promising prototypes, and deserve further optimization in the search for drug candidates for polypharmacological approach in neurodegenerative disease.
Collapse
Affiliation(s)
- Rebecca Orioli
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Giuliana Sarno
- Department of Pharmacy, Università degli Studi di Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano SA, Italy
| | - Francesca Seghetti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Silvia Gobbi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Federica Belluti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Alessandra Feoli
- Department of Pharmacy, Università degli Studi di Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano SA, Italy
| | - Francesca Massenzio
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Selmi 3, 40126, Bologna, Italy
| | - Barbara Monti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Selmi 3, 40126, Bologna, Italy; IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Rosaria Spagnuolo
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Manuela Bartolini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Sabrina Castellano
- Department of Pharmacy, Università degli Studi di Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano SA, Italy.
| | - Alessandra Bisi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy.
| |
Collapse
|
3
|
Lv B, Wang Z, Wang Q, Xu Z, Tang J, Pei Y, Bian Y, Sun H, Chen Y. Dual inhibitors of butyrylcholinesterase and histone deacetylase 6 for the treatment of Alzheimer's disease: design, synthesis, and biological evaluation. Bioorg Med Chem 2025; 127:118219. [PMID: 40347723 DOI: 10.1016/j.bmc.2025.118219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 04/18/2025] [Accepted: 04/29/2025] [Indexed: 05/14/2025]
Abstract
To address the multifactorial pathology of Alzheimer's disease (AD), eighteen butyrylcholinesterase (BChE) and histone deacetylase 6 (HDAC6) dual inhibitors were designed, synthesized, and biologically evaluated. Through structure-activity relationship studies, compound 17 emerged as the most potent candidate, with IC50 value of 0.3 nM for human BChE and 56.7 nM for HDAC6. This compound demonstrated favorable safety profiles, drug-like properties, and significant neuroprotective effects in vitro. In a mouse model of scopolamine-induced cognitive impairment, 17 (10 mg/kg) exhibited excellent safety and markedly improved cognitive deficits. These findings highlight compound 17 as a promising BChE/HDAC6 dual inhibitor, supporting its further development as a potential therapeutic agent for AD.
Collapse
Affiliation(s)
- Bingbing Lv
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, People's Republic of China
| | - Zhenqi Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, People's Republic of China
| | - Qinjie Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, People's Republic of China
| | - Zhaoxin Xu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, People's Republic of China
| | - Jixiong Tang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, People's Republic of China
| | - Yuqiong Pei
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, People's Republic of China
| | - Yaoyao Bian
- Jiangsu Provincial Engineering Center of TCM External Medication Researching and Industrializing, Nanjing University of Chinese Medicine, Nanjing 210023, People's Republic of China
| | - Haopeng Sun
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China.
| | - Yao Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, People's Republic of China.
| |
Collapse
|
4
|
Song R, Yin S, Wu J, Yan J. Neuronal regulated cell death in aging-related neurodegenerative diseases: key pathways and therapeutic potentials. Neural Regen Res 2025; 20:2245-2263. [PMID: 39104166 PMCID: PMC11759035 DOI: 10.4103/nrr.nrr-d-24-00025] [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: 01/08/2024] [Revised: 05/13/2024] [Accepted: 06/18/2024] [Indexed: 08/07/2024] Open
Abstract
Regulated cell death (such as apoptosis, necroptosis, pyroptosis, autophagy, cuproptosis, ferroptosis, disulfidptosis) involves complex signaling pathways and molecular effectors, and has been proven to be an important regulatory mechanism for regulating neuronal aging and death. However, excessive activation of regulated cell death may lead to the progression of aging-related diseases. This review summarizes recent advances in the understanding of seven forms of regulated cell death in age-related diseases. Notably, the newly identified ferroptosis and cuproptosis have been implicated in the risk of cognitive impairment and neurodegenerative diseases. These forms of cell death exacerbate disease progression by promoting inflammation, oxidative stress, and pathological protein aggregation. The review also provides an overview of key signaling pathways and crosstalk mechanisms among these regulated cell death forms, with a focus on ferroptosis, cuproptosis, and disulfidptosis. For instance, FDX1 directly induces cuproptosis by regulating copper ion valency and dihydrolipoamide S-acetyltransferase aggregation, while copper mediates glutathione peroxidase 4 degradation, enhancing ferroptosis sensitivity. Additionally, inhibiting the Xc- transport system to prevent ferroptosis can increase disulfide formation and shift the NADP + /NADPH ratio, transitioning ferroptosis to disulfidptosis. These insights help to uncover the potential connections among these novel regulated cell death forms and differentiate them from traditional regulated cell death mechanisms. In conclusion, identifying key targets and their crosstalk points among various regulated cell death pathways may aid in developing specific biomarkers to reverse the aging clock and treat age-related neurodegenerative conditions.
Collapse
Affiliation(s)
- Run Song
- Department of Neurology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
- Neuromolecular Biology Laboratory, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
| | - Shiyi Yin
- Department of Neurology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
- Neuromolecular Biology Laboratory, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
| | - Jiannan Wu
- Neuromolecular Biology Laboratory, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
| | - Junqiang Yan
- Department of Neurology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
- Neuromolecular Biology Laboratory, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
| |
Collapse
|
5
|
Hu Q, Wang S, Zhang W, Qu J, Liu GH. Unraveling brain aging through the lens of oral microbiota. Neural Regen Res 2025; 20:1930-1943. [PMID: 38993126 PMCID: PMC11691463 DOI: 10.4103/nrr.nrr-d-23-01761] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/22/2023] [Accepted: 05/31/2024] [Indexed: 07/13/2024] Open
Abstract
The oral cavity is a complex physiological community encompassing a wide range of microorganisms. Dysbiosis of oral microbiota can lead to various oral infectious diseases, such as periodontitis and tooth decay, and even affect systemic health, including brain aging and neurodegenerative diseases. Recent studies have highlighted how oral microbes might be involved in brain aging and neurodegeneration, indicating potential avenues for intervention strategies. In this review, we summarize clinical evidence demonstrating a link between oral microbes/oral infectious diseases and brain aging/neurodegenerative diseases, and dissect potential mechanisms by which oral microbes contribute to brain aging and neurodegeneration. We also highlight advances in therapeutic development grounded in the realm of oral microbes, with the goal of advancing brain health and promoting healthy aging.
Collapse
Affiliation(s)
- Qinchao Hu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Si Wang
- Advanced Innovation Center for Human Brain Protection and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Weiqi Zhang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Aging Biomarker Consortium, Beijing, China
| | - Jing Qu
- University of Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Aging Biomarker Consortium, Beijing, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Advanced Innovation Center for Human Brain Protection and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Aging Biomarker Consortium, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| |
Collapse
|
6
|
Socodato R, Relvas JB. Neuroinflammation revisited through the microglial lens. Neural Regen Res 2025; 20:1989-1990. [PMID: 39254552 PMCID: PMC11691469 DOI: 10.4103/nrr.nrr-d-24-00284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/22/2024] [Accepted: 06/03/2024] [Indexed: 09/11/2024] Open
Affiliation(s)
- Renato Socodato
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal
| | - João B. Relvas
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal
- Department of Biomedicine, Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal
| |
Collapse
|
7
|
Yang R, Deng MY, Yang LK, Wang GZ, Ma J, Wen Q, Gao N, Qiao HL. Identification of cytochrome P450 2E1 as a novel target in neuroinflammation and development of its inhibitor Q11 as a treatment strategy. Free Radic Biol Med 2025; 234:220-232. [PMID: 40122152 DOI: 10.1016/j.freeradbiomed.2025.03.032] [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: 01/27/2025] [Revised: 03/12/2025] [Accepted: 03/21/2025] [Indexed: 03/25/2025]
Abstract
Neuroinflammation is implicated in nearly all pathological processes of central nervous system (CNS) diseases. However, no medications specifically targeting neuroinflammation are clinically available, and conventional anti-inflammatory drugs exhibit limited efficacy. Here, we identified cytochrome P450 2E1 (CYP2E1) as a novel therapeutic target in neuroinflammation. Elevated CYP2E1 levels were observed in hippocampal tissues of mouse and rat neuroinflammation models, as well as in LPS-stimulated primary microglia. Genetic ablation of Cyp2e1 improved spatial learning and memory in neuroinflammatory rats and reduced pro-inflammatory cytokine levels in Cyp2e1-deficient microglia. Furthermore, Q11 (1-(4-methyl-5-thiazolyl) ethanone), a novel CYP2E1 inhibitor developed and synthesized in our laboratory, effectively ameliorated Alzheimer's disease-related spatial learning and memory functions and depression-related anxiety-like behaviors in mice/rats. Mechanistically, Q11 attenuated microglial activation, neuronal damage, oxidative stress, and neuroinflammation by suppressing the PI3K/Akt, STAT1/3, and NF-κB signaling pathways. These findings establish CYP2E1 as a druggable target for neuroinflammation and propose Q11 as a promising candidate for treating neuroinflammation-related diseases.
Collapse
Affiliation(s)
- Rui Yang
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Meng-Yan Deng
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Lu-Kui Yang
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Guan-Zhe Wang
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Jun Ma
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Qiang Wen
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Na Gao
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Hai-Ling Qiao
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China.
| |
Collapse
|
8
|
Wen J, Skampardoni I, Tian YE, Yang Z, Cui Y, Erus G, Hwang G, Varol E, Boquet-Pujadas A, Chand GB, Nasrallah IM, Satterthwaite TD, Shou H, Shen L, Toga AW, Zalesky A, Davatzikos C. Neuroimaging endophenotypes reveal underlying mechanisms and genetic factors contributing to progression and development of four brain disorders. Nat Biomed Eng 2025:10.1038/s41551-025-01412-w. [PMID: 40481237 DOI: 10.1038/s41551-025-01412-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 04/24/2025] [Indexed: 06/11/2025]
Abstract
Recent work leveraging artificial intelligence has offered promise to dissect disease heterogeneity by identifying complex intermediate brain phenotypes, called dimensional neuroimaging endophenotypes (DNEs). We advance the argument that these DNEs capture the degree of expression of respective neuroanatomical patterns measured, offering a dimensional neuroanatomical representation for studying disease heterogeneity and similarities of neurologic and neuropsychiatric diseases. We investigate the presence of nine DNEs derived from independent yet harmonized studies on Alzheimer's disease, autism spectrum disorder, late-life depression and schizophrenia in the UK Biobank study. Phenome-wide associations align with genome-wide associations, revealing 31 genomic loci (P < 5 × 10-8/9) associated with the nine DNEs. The nine DNEs, along with their polygenic risk scores, significantly enhanced the predictive accuracy for 14 systemic disease categories, particularly for conditions related to mental health and the central nervous system, as well as mortality outcomes. These findings underscore the potential of the nine DNEs to capture the expression of disease-related brain phenotypes in individuals of the general population and to relate such measures with genetics, lifestyle factors and chronic diseases.
Collapse
Affiliation(s)
- Junhao Wen
- Laboratory of AI and Biomedical Science (LABS), Columbia University, New York, NY, USA.
- Department of Radiology, Columbia University, New York, NY, USA.
- New York Genome Center (NYGC), New York, NY, USA.
- Department of Biomedical Engineering, Columbia University, New York, NY, USA.
- Data Science Institute (DSI), Columbia University, New York, NY, USA.
- Center for Innovation in Imaging Biomarkers and Integrated Diagnostics (CIMBID), Department of Radiology, Columbia University, New York, NY, USA.
- Zuckerman Institute, Columbia University, New York, NY, USA.
| | - Ioanna Skampardoni
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ye Ella Tian
- Systems Lab, Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Melbourne, Victoria, Australia
| | - Zhijian Yang
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yuhan Cui
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Guray Erus
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gyujoon Hwang
- Department of Psychiatry and Behavioral Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Erdem Varol
- Department of Computer Science and Engineering, New York University, New York, NY, USA
| | - Aleix Boquet-Pujadas
- Laboratory of AI and Biomedical Science (LABS), Columbia University, New York, NY, USA
| | - Ganesh B Chand
- Department of Radiology, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Ilya M Nasrallah
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Theodore D Satterthwaite
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Haochang Shou
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biostatistics, Epidemiology and Informatics University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Arthur W Toga
- Laboratory of Neuro Imaging (LONI), Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Andrew Zalesky
- Systems Lab, Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Melbourne, Victoria, Australia
| | - Christos Davatzikos
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
9
|
Liu Y, Aquili L, Wong KH, Lu Z, Lim LW. Past, present, and future of serotonin-targeting therapeutics for Alzheimer's disease: Perspectives from DNA methylation. Ageing Res Rev 2025; 108:102755. [PMID: 40239871 DOI: 10.1016/j.arr.2025.102755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 03/02/2025] [Accepted: 04/13/2025] [Indexed: 04/18/2025]
Abstract
With population aging, Alzheimer's disease (AD) is becoming increasingly prevalent, causing great health and economic burdens worldwide. Despite decades of research, there are still no effective disease-modifying treatments for AD, highlighting the urgent need for more in-depth understanding of the disease-causing mechanisms. The brain serotonin (5-HT) neurotransmission system undergoes structural and functional changes in aging and AD, which contributes to cognitive decline and comorbid mood disturbances. This review discusses the critical involvement of the brain 5-HT system in aging and AD. Existing findings on the changes in projection fiber innervation and receptor/transporter expression in AD are reviewed. Preclinical and clinical progress on the development of 5-HT-modulating drugs for AD and the obstacles faced by these development efforts are discussed. Epigenetic control of the brain 5-HT system and the potential of modulating 5-HT transmission via DNA methylation are also examined.
Collapse
Affiliation(s)
- Yanzhi Liu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong.
| | - Luca Aquili
- Department of Biosciences and Bioinformatics, and Suzhou Municipal Key Laboratory of Cancer Biology and Chronic Disease, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou 215123, China; School of Management, Ritsumeikan Asia Pacific University, Beppu, Oita, Japan.
| | - Kah Hui Wong
- Department of Anatomy, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia.
| | - Zhiliang Lu
- Department of Biosciences and Bioinformatics, and Suzhou Municipal Key Laboratory of Cancer Biology and Chronic Disease, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou 215123, China.
| | - Lee Wei Lim
- Department of Biosciences and Bioinformatics, and Suzhou Municipal Key Laboratory of Cancer Biology and Chronic Disease, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou 215123, China.
| |
Collapse
|
10
|
Ayyubova G, Madhu LN. Microglial NLRP3 Inflammasomes in Alzheimer's Disease Pathogenesis: From Interaction with Autophagy/Mitophagy to Therapeutics. Mol Neurobiol 2025; 62:7124-7143. [PMID: 39951189 DOI: 10.1007/s12035-025-04758-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 02/08/2025] [Indexed: 05/15/2025]
Abstract
The nucleotide-binding oligomerization domain-like receptor pyrin domain-containing 3 (NLRP3) inflammasome, discovered 20 years ago, is crucial in controlling innate immune reactions in Alzheimer's disease (AD). By initiating the release of inflammatory molecules (including caspases, IL-1β, and IL-18), the excessively activated inflammasome complex in microglia leads to chronic inflammation and neuronal death, resulting in the progression of cognitive deficiencies. Even though the involvement of NLRP3 has been implicated in neuroinflammation and widely explored in several studies, there are plenty of controversies regarding its precise roles and activation mechanisms in AD. Another prominent feature of AD is impairment in microglial autophagy, which can be either the cause or the consequence of NLRP3 activation and contributes to the aggregation of misfolded proteins and aberrant chronic inflammatory state seen in the disease course. Studies also demonstrate that intracellular buildup of dysfunctional and damaged mitochondria due to defective mitophagy enhances inflammasome activation, further suggesting that restoration of impaired autophagy and mitophagy can effectively suppress it, thereby reducing inflammation and protecting microglia and neurons. This review is primarily focused on the role of NLRP3 inflammasome in the etiopathology of AD, its interactions with microglial autophagy/mitophagy, and the latest developments in NLRP3 inflammasome-targeted therapeutic interventions being implicated for AD treatment.
Collapse
Affiliation(s)
- Gunel Ayyubova
- Department of Cytology, Embryology and Histology, Azerbaijan Medical University, Baku, Azerbaijan.
| | - Leelavathi N Madhu
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M Health Science Center School of Medicine, College Station, TX, USA
| |
Collapse
|
11
|
Huang J, Wu F, Cao W, Chen Y, Yao Q, Cen P, Wang J, Hong L, Zhang X, Zhou R, Jin C, Tian M, Zhang H, Zhong Y. Ultrasmall iron-gallic acid coordination polymer nanoparticles for scavenging ROS and suppressing inflammation in tauopathy-induced Alzheimer's disease. Biomaterials 2025; 317:123042. [PMID: 39805185 DOI: 10.1016/j.biomaterials.2024.123042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 12/19/2024] [Accepted: 12/21/2024] [Indexed: 01/16/2025]
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder globally, with no effective treatment available yet. A crucial pathological hallmark of AD is the accumulation of hyperphosphorylated tau protein, which is deteriorated by reactive oxygen species (ROS) and neuroinflammation in AD progression. Thus, alleviation of ROS and inflammation has become a potential therapeutic strategy in many studies. Herein, we reported ultrasmall coordination polymer nanoparticles formed by ferric ions and gallic acid (Fe-GA CPNs), which owned antioxidant and anti-inflammation properties for AD therapeutics. The facilely prepared Fe-GA CPNs exhibited remarkable superoxide dismutase-like, peroxidase-like enzyme activity, and ROS eliminating ability with great water solubility, compared with gallic acid. We demonstrated that Fe-GA CPNs effectively relieved oxidative stress, ameliorated inflammation by modulating microglial polarization towards anti-inflammation phenotype, and reduced hyperphosphorylated tau protein levels. Furthermore, Fe-GA CPNs treatment significantly improved cognitive function in tauopathy-induced AD rats, and achieved a neuroprotective effect against AD pathology. This study highlights the potential of coordination polymer nanoparticles as promising therapeutic candidates for AD and other tau-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Jiani Huang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China; Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Fei Wu
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China; Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Wenzhao Cao
- Human Phenome Institute, Fudan University, Shanghai, China
| | - Yuhan Chen
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China; Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Qiong Yao
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
| | - Peili Cen
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China; Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Jing Wang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China; Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Lu Hong
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China; Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Xiaohui Zhang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China; Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Rui Zhou
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China; Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Chentao Jin
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China; Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Mei Tian
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China; Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China; Human Phenome Institute, Fudan University, Shanghai, China.
| | - Hong Zhang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China; Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China; Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, China; College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, China.
| | - Yan Zhong
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China; Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China; Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, China.
| |
Collapse
|
12
|
Gaesser GA, Hall SE, Angadi SS, Poole DC, Racette SB. Increasing the health span: unique role for exercise. J Appl Physiol (1985) 2025; 138:1285-1308. [PMID: 40244910 DOI: 10.1152/japplphysiol.00049.2025] [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: 01/23/2025] [Revised: 02/21/2025] [Accepted: 04/02/2025] [Indexed: 04/19/2025] Open
Abstract
Health span, that period between birth and onset of major disease(s), when adequate physical and cognitive function permit those daily living activities essential to life quality, is lower in the United States than other developed countries. Physical inactivity and excessive calorie intake occupy dominant roles both in the problem, and by redressing them, in the solution. Consequently, this review focuses on evidence that appropriate exercise engagement and calorie restriction (CR) can improve physical and mental health with a view to extending the health span. Humanity, writ large, has grasped these underlying concepts for Millennia but has been largely intransigent to them. Thus, the final section proposes a novel Monty Python-esque approach that encompasses humanity's inimical sense of humor to increase physical fitness and mental health, restore energy balance, sustain better cognitive function, and extend the health span.
Collapse
Affiliation(s)
- Glenn A Gaesser
- College of Health Solutions, Arizona State University, Phoenix, Arizona, United States
| | - Stephanie E Hall
- Colleges of Veterinary Medicine and Health and Human Sciences, Kansas State University, Manhattan, Kansas, United States
| | - Siddhartha S Angadi
- School of Health Education and Human Development, University of Virginia, Charlottesville, Virginia, United States
| | - David C Poole
- Colleges of Veterinary Medicine and Health and Human Sciences, Kansas State University, Manhattan, Kansas, United States
| | - Susan B Racette
- College of Health Solutions, Arizona State University, Phoenix, Arizona, United States
| |
Collapse
|
13
|
Kathiresan DS, Balasubramani R, Marudhachalam K, Jaiswal P, Ramesh N, Sureshbabu SG, Puthamohan VM, Vijayan M. Role of Mitochondrial Dysfunctions in Neurodegenerative Disorders: Advances in Mitochondrial Biology. Mol Neurobiol 2025; 62:6827-6855. [PMID: 39269547 DOI: 10.1007/s12035-024-04469-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024]
Abstract
Mitochondria, essential organelles responsible for cellular energy production, emerge as a key factor in the pathogenesis of neurodegenerative disorders. This review explores advancements in mitochondrial biology studies that highlight the pivotal connection between mitochondrial dysfunctions and neurological conditions such as Alzheimer's, Parkinson's, Huntington's, ischemic stroke, and vascular dementia. Mitochondrial DNA mutations, impaired dynamics, and disruptions in the ETC contribute to compromised energy production and heightened oxidative stress. These factors, in turn, lead to neuronal damage and cell death. Recent research has unveiled potential therapeutic strategies targeting mitochondrial dysfunction, including mitochondria targeted therapies and antioxidants. Furthermore, the identification of reliable biomarkers for assessing mitochondrial dysfunction opens new avenues for early diagnosis and monitoring of disease progression. By delving into these advancements, this review underscores the significance of understanding mitochondrial biology in unraveling the mechanisms underlying neurodegenerative disorders. It lays the groundwork for developing targeted treatments to combat these devastating neurological conditions.
Collapse
Affiliation(s)
- Divya Sri Kathiresan
- Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Nadu, Tamil, 641046, India
| | - Rubadevi Balasubramani
- Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Nadu, Tamil, 641046, India
| | - Kamalesh Marudhachalam
- Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Nadu, Tamil, 641046, India
| | - Piyush Jaiswal
- Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Nadu, Tamil, 641046, India
| | - Nivedha Ramesh
- Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Nadu, Tamil, 641046, India
| | - Suruthi Gunna Sureshbabu
- Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Nadu, Tamil, 641046, India
| | - Vinayaga Moorthi Puthamohan
- Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Nadu, Tamil, 641046, India.
| | - Murali Vijayan
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA.
| |
Collapse
|
14
|
Li JY, Chen MB, Fu TT, Liu FF, Liu J, Xu CJ, Zhou J, Rao Y, Jiang ZP, Huang L. Discovery of structurally intriguing diterpenoids as anti-neuroinflammatory agents from mangrove plant Excoecaria agallocha L. via inhibiting macrophage polarization and inflammasome. PHYTOCHEMISTRY 2025; 234:114440. [PMID: 39952580 DOI: 10.1016/j.phytochem.2025.114440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 02/10/2025] [Accepted: 02/12/2025] [Indexed: 02/17/2025]
Abstract
Emerging evidence indicates the inhibition of neuroinflammation is an important and promising strategy for managing Alzheimer's disease. Anti-neuroinflammatory agents are potential drug leads. In this work, seventeen diterpenoids (1-17) including ten previously undescribed ones, named excoaglolides A-J (1-10) were isolated from the stems and twigs of the mangrove plant, Excoecaria agallocha L. The undescribed diterpenoids could be categorized into four subclasses, viz., ent-isopimaranes (1-4), ent-beyeranes (5-7), ent-atisanes (8-9), and ent-labdane (10). Their structures including absolute configurations were unambiguously characterized by spectroscopic methods, single crystal X-ray diffraction analyses, ECD calculations, and modified Mosher's method. Notably, compound 1 was a rare 3,4-seco-3,11-lactone-ent-isopimarane, whereas compound 2 was the first 2,3-seco-2,11-lactone-ent-isopimarane; compound 5 represents the first example of 2,3-seco-2-nor-1,3-lactone-ent-beyerane, compound 8 was the first member of 18(4 → 3)-abeo-ent-atisane forming through a Wagner-Meerwein rearrangement, compound 9 was an unusual rearranged 3,4-seco-ent-atisane possessing a cyclobutane ring, and compound 10 featuring a rare chlorine substitution at C-15. In addition, the stereochemistry of the known diterpenoid excoagallochaol D (11) with a unique 6/7/4-fused ring system was firstly determined by single crystal X-ray diffraction analyses in this study. Compounds 1, 12, 15, and 16 showed significant nitric oxide production inhibition in lipopolysaccharide-induced BV-2 microglial cells with IC50 values ranging from 2.0 to 13.5 μM. Further mechanistic investigations revealed that compound 1 inhibited macrophage polarization and decreased the expression levels of representative inflammation-related genes, including IL-1β, IL-6, TNF-α, COX-2, and iNOS. These were associated with suppressions of proteins related to inflammasome, including caspase 1 and NLRP3, and secretion of IL-1β. The present study revealed that the mangrove diterpenoid excoaglolide A (1) would be promising structure base for developing neuroinflammation-related neurodegeneration diseases.
Collapse
Affiliation(s)
- Jun-Yi Li
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, 570200, China
| | - Ming-Bin Chen
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, 570200, China
| | - Ting-Ting Fu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, 570200, China
| | - Fei-Fei Liu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, 570200, China
| | - Jin Liu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, 570200, China
| | - Cong-Jun Xu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, 570200, China
| | - Jing Zhou
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, 570200, China
| | - Yong Rao
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, 570200, China.
| | - Zhong-Ping Jiang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, 570200, China.
| | - Ling Huang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, 570200, China.
| |
Collapse
|
15
|
Li J, Li J, Liu Y, Hu C, Xu H, Cao D, Zhang R, Zhang K. Nrf2 Ameliorates Sevoflurane-Induced Cognitive Deficits in Aged Mice by Inhibiting Neuroinflammation in the Hippocampus. Mol Neurobiol 2025; 62:8048-8064. [PMID: 39969679 DOI: 10.1007/s12035-025-04777-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 02/11/2025] [Indexed: 02/20/2025]
Abstract
Perioperative neurocognitive disorders (PND), common complications that occur after anesthetized surgery in elderly patients, are major challenges to our rapidly growing aging population. The transcription factor known as nuclear factor erythroid-2-related factor 2 (Nrf2) is an essential component of the cellular antioxidant response, purportedly contributing to the preservation of cognitive functions such as learning and memory. Nevertheless, the function and intracellular processes involving Nrf2 in PND remain largely unknown. Therefore, we evaluate the influence and fundamental mechanism of Nrf2 on PND in aged mice. To establish the postoperative neurocognitive dysfunction (PND) model, aged mice were subjected to anesthesia via inhalation of 3% sevoflurane for a duration of 2 h. The role of Nrf2 in PND was investigated by administering microinjections of either the adeno-associated virus (AAV)-Nrf2 vector or a null virus vector into the hippocampal CA1 region of aged mice 28 days before exposure to sevoflurane. Various assays including enzyme-linked immunosorbent assay (ELISA), immunofluorescence staining, and western blotting were employed to assess levels of pro-inflammatory cytokines, microglial activation, and the oxidative stress response. Furthermore, synaptic plasticity was evaluated through long-term potentiation (LTP) recording and Golgi staining techniques. Elevated expression of Nrf2 within the hippocampal CA1 region ameliorated sevoflurane-induced cognitive deficits, synaptic plasticity anomalies, and the oxidative stress reaction in aged mice. Furthermore, the activation of microglia and the release of pro-inflammatory cytokines (including IL-6, TNF-α, and IL-1β) within the hippocampus post-sevoflurane exposure were modulated in an Nrf2-dependent fashion. Based on the findings from present research, we conclude that Nrf2 ameliorates sevoflurane-induced cognitive dysfunction by inhibiting hippocampal neuroinflammation, thereby proposing a potential therapeutic target for PND.
Collapse
Affiliation(s)
- Junhua Li
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yanjiang West Road, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Jinfeng Li
- Department of Anesthesiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (the Second Clinical Medical College of Guangzhou University of Chinese Medicine), Guangzhou, China
| | - Yafang Liu
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yanjiang West Road, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Chuwen Hu
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yanjiang West Road, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Hui Xu
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yanjiang West Road, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Dong Cao
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yanjiang West Road, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Rong Zhang
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yanjiang West Road, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China.
| | - Kun Zhang
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yanjiang West Road, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China.
| |
Collapse
|
16
|
Marino C, Malotaux V, Giudicessi A, Aguillon D, Sepulveda-Falla D, Lopera F, Quiroz YT. Protective genetic variants against Alzheimer's disease. Lancet Neurol 2025; 24:524-534. [PMID: 40409316 DOI: 10.1016/s1474-4422(25)00116-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 01/24/2025] [Accepted: 03/31/2025] [Indexed: 05/25/2025]
Abstract
Genetic studies can offer powerful insights for the development of disease-modifying therapies for Alzheimer's disease. Protective genetic variants that delay the onset of cognitive impairment have been found in people with sporadic Alzheimer's disease and in carriers of mutations that usually cause autosomal-dominant Alzheimer's disease in mid-life. The study of families who carry autosomal dominant mutations provides a unique opportunity to uncover genetic modifiers of disease progression, including rare variants in genes such as APOE and RELN. Understanding how these variants confer protection can help identify the biological pathways that contribute to cognitive resilience, such as the heparan-sulphate proteoglycan-APOE receptor pathway, the TREM-2-driven signalling pathways in the microglia, and phagocytosis. Therapies able to replicate the beneficial effects of these natural defences could provide novel strategies for slowing or preventing the progression of Alzheimer's disease.
Collapse
Affiliation(s)
- Claudia Marino
- The Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA, USA; Department of Neurology, Sealy Institute for Drug Discovery and Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, Galveston, TX, USA
| | - Vincent Malotaux
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Averi Giudicessi
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Boston University, Department of Psychological and Brain Sciences, Boston, MA, USA
| | - David Aguillon
- The Neuroscience Group of Antioquia, School of Medicine, University of Antioquia, Medellín, Colombia
| | - Diego Sepulveda-Falla
- The Institute of Neuropathology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Francisco Lopera
- The Neuroscience Group of Antioquia, School of Medicine, University of Antioquia, Medellín, Colombia
| | - Yakeel T Quiroz
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Boston University, Department of Psychological and Brain Sciences, Boston, MA, USA; The Neuroscience Group of Antioquia, School of Medicine, University of Antioquia, Medellín, Colombia; Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
17
|
Zhang H, Zhang J, Huang K, Cai C, Jiang J, Su Z, Gu H, Duan Z, Shao S, Zhou M, Du Q, He F. Novel p-terphenyls with anti-neuroinflammatory activity from fruiting bodies of the Chinese edible mushroom Thelephora ganbajun Zang. Bioorg Chem 2025; 159:108414. [PMID: 40174532 DOI: 10.1016/j.bioorg.2025.108414] [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: 12/31/2024] [Revised: 03/24/2025] [Accepted: 03/25/2025] [Indexed: 04/04/2025]
Abstract
The detailed mycochemical exploration of the EtOAc extract of a famous edible mushroom Thelephora ganbajun, resulted in the isolation of six new p-terphenyl derivatives, named theleganbanins A - F (1-6), together with five known ones, namely atromentin (7), fendleryl B (8), 2-O-methylatromentin (9), vialinin B (10), and ganbajunin B (11). Their structures were precisely determined through comprehensive spectroscopic analyses, especially 1D and 2D NMR data and HRMS measurement. Single crystal X-ray diffraction and comparison of calculated and experimental ECD spectra were conducted to further confirm the absolute configurations of compounds 1-6. Theleganbanins A (1) and B (2) featuring a rare α, β-unsaturated-γ-butyrolactone core were proposed to be biosynthesized through aldol condensation for the first time in naturally occurring p-terphenyl derivatives. Theleganbanin C (3) was identified as a pair of p-terphenyl enantiomers with a novel 1', 6'-dyhydro-2', 5'-pyridinedione ring. Theleganbanin D (4) was the first example of p-terphenyl derivatives with a hemiacetal furanone moiety. The anti-neuroinflammatory activities of compounds 1-2 and 4-10 were screened. As a result, these compounds showed inhibitory activity on the production of pro-inflammatory cytokines TNF-α, IL-6 and IL-1β in lipopolysaccharide (LPS)-induced BV-2 microglial cells. Further investigation showed that compound 2 could inhibit the phosphorylation of JAK2/STAT3 signaling pathway. These finding indicated that p-terphenyl derivatives from edible mushroom Thelephora ganbajun Zang would be promising drug candidates in treatment of neuroinflammatory related diseases.
Collapse
Affiliation(s)
- Hang Zhang
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Jingyi Zhang
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Keyin Huang
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Cheng Cai
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Jinyan Jiang
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Science, The University of Tokyo, Tokyo 113-8657, Japan
| | - Zijie Su
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Haixin Gu
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Zidan Duan
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Shijie Shao
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Min Zhou
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Qingfeng Du
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China; Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou 510515, China
| | - Fei He
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China; Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou 510515, China.
| |
Collapse
|
18
|
Jasim SA, Altalbawy FMA, Abohassan M, Oghenemaro EF, Bishoyi AK, Singh RP, Kaur P, Sivaprasad GV, Mohammed JS, Hulail HM. Histone Deacetylases (HDACs) Roles in Inflammation-mediated Diseases; Current Knowledge. Cell Biochem Biophys 2025; 83:1375-1386. [PMID: 39419931 DOI: 10.1007/s12013-024-01587-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2024] [Indexed: 10/19/2024]
Abstract
The histone acetyl transferases (HATs) and histone deacetylases (HDACs), which are mostly recognized for their involvement in regulating chromatin remodeling via histone acetylation/deacetylation, have been shown to also change several non-histone proteins to regulate other cellular processes. Acetylation affects the activity or function of cytokine receptors, nuclear hormone receptors, intracellular signaling molecules, and transcription factors in connection to inflammation. Some small-molecule HDAC inhibitors are utilized as anticancer medications in clinical settings due to their capability to regulate cellular growth arrest, differentiation, and death. Here, we summarize our present knowledge of the innate and adaptive immunological pathways that classical HDAC enzymes control. The aim is to justify the targeted (or non-targeted) use of inhibitors against certain HDAC enzymes in inflammatory diseases such as arthritis, inflammatory bowel diseases (IBD), airways inflammation and neurological diseases.
Collapse
Affiliation(s)
- Saade Abdalkareem Jasim
- Medical Laboratory Techniques department, College of Health and Medical Technology, University of Al-maarif, Anbar, Iraq
| | - Farag M A Altalbawy
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia.
| | - Mohammad Abohassan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Enwa Felix Oghenemaro
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Delta State University, Abraka, Delta State, Nigeria
| | - Ashok Kumar Bishoyi
- Department of Microbiology, Faculty of Science, Marwadi University Research Center, Marwadi University, Rajkot, 360003, Gujarat, India
| | - Ravindra Pal Singh
- Department of Pharmaceutics, NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Parjinder Kaur
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - G V Sivaprasad
- Department of Basic Science & Humanities, Raghu Engineering College, Visakhapatnam, India
| | | | - Hanen Mahmod Hulail
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| |
Collapse
|
19
|
Targas ABA, Victoriano PHM, Garcia MBB, Alexandre-Silva V, Cominetti MR. Exploring the connection between dementia and cardiovascular risk with a focus on ADAM10. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167825. [PMID: 40174790 DOI: 10.1016/j.bbadis.2025.167825] [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: 03/26/2025] [Accepted: 03/27/2025] [Indexed: 04/04/2025]
Abstract
Alzheimer's disease (AD) represents a leading cause of dementia, characterized by progressive cognitive and functional decline. Although extensive research has unraveled critical aspects of AD pathology, its etiology remains incompletely understood, urging further exploration into potential risk factors. Growing evidence underscores a significant link between cardiovascular disease (CVD) risk factors and AD, with modifiable lifestyle elements - such as physical inactivity, high low-density lipoprotein (LDL) levels, obesity, hypertension, atherosclerosis, and diabetes - emerging as contributors to cerebrovascular damage and neurodegeneration. ADAM10, a disintegrin and metalloproteinase involved in the non-amyloidogenic processing of amyloid precursor protein (APP), has garnered interest for its dual role in cardiovascular and neurodegenerative processes. ADAM10's regulation of neuroinflammation, endothelial function, and proteolytic cleavage of APP potentially moderates amyloid-β (Aβ) peptide formation, thus influencing both cardiovascular and brain health. Given these interconnected roles, this narrative review investigates whether ADAM10-driven vascular dysfunction accelerates neurodegeneration, how lipid metabolism influences ADAM10 activity in CVD and AD, and whether targeting ADAM10 could offer a dual-benefit therapeutic strategy to mitigate disease burden. By exploring epidemiological data, clinical studies, and molecular pathways, we aim to clarify ADAM10's bridging function between AD and cardiovascular risk, offering a new perspective into therapeutic opportunities to alleviate the dual burden of these interrelated conditions.
Collapse
Affiliation(s)
| | | | | | | | - Marcia Regina Cominetti
- Department of Gerontology, Federal University of São Carlos, São Carlos, SP, Brazil; Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
20
|
Zhang B, Wang J, Chen J, Pan X. Arsenic exposure activates microglia, inducing neuroinflammation and promoting the occurrence and development of Alzheimer's disease-like neurodegeneration in mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 297:118251. [PMID: 40294499 DOI: 10.1016/j.ecoenv.2025.118251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/27/2025] [Accepted: 04/24/2025] [Indexed: 04/30/2025]
Abstract
Health damage caused by environmental arsenic pollution has attracted widespread attention, with Alzheimer's disease (AD) thought to be one of the adverse effects of long-term arsenic exposure. In this study, we sought to investigate the relationship between arsenic exposure and AD-like neurodegeneration and to determine the role of microglia in the process of AD-like neurodegeneration induced by arsenic exposure. The relationship between arsenic exposure and AD-like neurodegeneration was investigated using cognitive assessments and biological experiments. Arsenic exposure induced memory impairment in C57BL/6 mice and resulted in a significant increase in the number of Aβ+ and pTau+ cells in the entorhinal cortex and hippocampus with neuronal granular vacuolar degeneration and necroptosis, accompanied by upregulated expression of related proteins, which showed dose- and time-response relationships. Arsenic exposure intensified memory decline in FAD4T mice with accelerated AD-like neurodegeneration. Correlation analysis showed a negative correlation between memory impairment and neurodegeneration in the entorhinal cortex and hippocampus in mice. Arsenic exposure also activated microglia in the entorhinal cortex and hippocampus, with enlargement of the cytosol, shortened and thickened cell protrusions, hypertrophic changes, and abnormal proliferation, as well as upregulated expression of the pro-inflammatory cytokines TNF-α and IL-1β. Arsenic exposure induced overactivation of microglia in the entorhinal cortex and hippocampus of FAD4T mice, resulting in de-branching or bulbous protrusions and fragmented cytoplasm. Our findings suggest that arsenic exposure promotes the occurrence and development of AD-like neurodegeneration via the activation of microglia, which induces neuroinflammation in mice.
Collapse
Affiliation(s)
- Bo Zhang
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 561113, China; Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guizhou Medical University, Guiyang 561113, China.
| | - Jiaojiao Wang
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 561113, China; Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guizhou Medical University, Guiyang 561113, China
| | - Junhong Chen
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 561113, China; Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guizhou Medical University, Guiyang 561113, China
| | - Xueli Pan
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 561113, China; Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guizhou Medical University, Guiyang 561113, China.
| |
Collapse
|
21
|
Festa LK, Jordan-Sciutto KL, Grinspan JB. Neuroinflammation: An Oligodendrocentric View. Glia 2025; 73:1113-1129. [PMID: 40059542 PMCID: PMC12014387 DOI: 10.1002/glia.70007] [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: 12/17/2024] [Revised: 02/18/2025] [Accepted: 02/20/2025] [Indexed: 03/16/2025]
Abstract
Chronic neuroinflammation, driven by central nervous system (CNS)-resident astrocytes and microglia, as well as infiltration of the peripheral immune system, is an important pathologic mechanism across a range of neurologic diseases. For decades, research focused almost exclusively on how neuroinflammation impacted neuronal function; however, there is accumulating evidence that injury to the oligodendrocyte lineage is an important component for both pathologic and clinical outcomes. While oligodendrocytes are able to undergo an endogenous repair process known as remyelination, this process becomes inefficient and usually fails in the presence of sustained inflammation. The present review focuses on our current knowledge regarding activation of the innate and adaptive immune systems in the chronic demyelinating disease, multiple sclerosis, and provides evidence that sustained neuroinflammation in other neurologic conditions, such as perinatal white matter injury, traumatic brain injury, and viral infections, converges on oligodendrocyte injury. Lastly, the therapeutic potential of targeting the impact of inflammation on the oligodendrocyte lineage in these diseases is discussed.
Collapse
Affiliation(s)
- Lindsay K Festa
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Neurology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Kelly L Jordan-Sciutto
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Judith B Grinspan
- Department of Neurology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| |
Collapse
|
22
|
Almutary AG, Begum MY, Siddiqua A, Gupta S, Chauhan P, Wadhwa K, Singh G, Iqbal D, Padmapriya G, Kumar S, Kedia N, Verma R, Kumar R, Sinha A, Dheepak B, Abomughaid MM, Jha NK. Unlocking the Neuroprotective Potential of Silymarin: A Promising Ally in Safeguarding the Brain from Alzheimer's Disease and Other Neurological Disorders. Mol Neurobiol 2025; 62:7975-7997. [PMID: 39956886 DOI: 10.1007/s12035-024-04654-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 12/02/2024] [Indexed: 02/18/2025]
Abstract
Medicinal plants and their phytochemicals have been extensively employed worldwide for centuries to address a diverse range of ailments, boasting a history that spans several decades. These plants are considered the source of numerous medicinal compounds. For instance, silymarin is a polyphenolic flavonoid extract obtained from the milk thistle plant or Silybum marianum which has been shown to have significant neuroprotective effects and great therapeutic benefits. Neurodegenerative diseases (NDs) are a class of neurological diseases that have become more prevalent in recent years, and although treatment is available, there is no complete cure developed yet. Silymarin utilizes a range of molecular mechanisms, including modulation of MAPK, AMPK, NF-κB, mTOR, and PI3K/Akt pathways, along with various receptors, enzymes, and growth factors. These mechanisms collectively contribute to its protective effects against NDs such as Alzheimer's disease, Parkinson's disease, and depression. Despite its safety and efficacy, silymarin faces challenges related to bioavailability and aqueous solubility, hindering its development as a clinical drug. This review highlights the molecular mechanisms underlying silymarin's neuroprotective effects, suggesting its potential as a promising therapeutic strategy for NDs.
Collapse
Affiliation(s)
- Abdulmajeed G Almutary
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, P.O. Box 59911, Abu Dhabi, United Arab Emirates
| | - M Yasmin Begum
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Ayesha Siddiqua
- Department of Clinical Pharmacy, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Saurabh Gupta
- Deparment of Biotechnology, GLA University, Mathura, India
| | - Payal Chauhan
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| | - Karan Wadhwa
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| | - Govind Singh
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| | - Danish Iqbal
- Department of Health Information Management, College of Applied Medical Sciences, Buraydah Private Colleges, 51418, Buraydah, Saudi Arabia
| | - Gopalakrishnan Padmapriya
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Sanjay Kumar
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan, 303012, India
| | - Navin Kedia
- NIMS School of Civil Engineering, NIMS University Rajasthan, Jaipur, India
| | - Rajni Verma
- Department of Applied Sciences, Chandigarh Engineering College, Chandigarh Group of Colleges Jhanjeri, Mohali, 140307, Punjab, India
| | - Ravi Kumar
- Department of Basic Science & Humanities, Raghu Engineering College, Visakhapatnam, India
| | - Aashna Sinha
- School of Applied and Life Sciences, Department of Research and Innovation, Uttaranchal University, Dehradun, Uttarakhand, India
| | - B Dheepak
- Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Mosleh Mohammad Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, 61922, Bisha, Saudi Arabia
| | - Niraj Kumar Jha
- Department of Biotechnology & Bioengineering, School of Biosciences & Technology, Galgotias University, Greater Noida, Uttar Pradesh, 203201, India.
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India.
- School of Bioengineering & Biosciences, Lovely Professional University, Phagwara, 144411, India.
| |
Collapse
|
23
|
Yan ZJ, Ye M, Li J, Zhang DF, Yao YG. Early transcriptional and cellular abnormalities in choroid plexus of a mouse model of Alzheimer's disease. Mol Neurodegener 2025; 20:62. [PMID: 40450296 DOI: 10.1186/s13024-025-00853-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 05/20/2025] [Indexed: 06/03/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by the accumulation of amyloid-β plaques, tau hyperphosphorylation, and neuroinflammation. The choroid plexus (ChP), serving as the blood-cerebrospinal fluid-brain barrier, plays essential roles in immune response to stress and brain homeostasis. However, the cellular and molecular contributions of the ChP to AD progression remain inadequately understood. METHODS To elucidate the molecular abnormalities during the early stages of AD, we acquired single-cell transcription profiling of ChP from APP/PS1 mice with early-stage of Aβ pathology and litter-mate controls. The transcriptional alterations that occurred in each cell type were identified by differentially expressed genes, cell-cell communications and pseudotemporal trajectory analysis. The findings were subsequently validated by a series of in situ and in vitro assays. RESULTS We constructed a comprehensive atlas of ChP at single-cell resolution and identified six major cell types and immune subclusters in male mice. The majority of dysregulated genes were found in the epithelial cells of APP/PS1 mice in comparison to wild-type (WT) mice, and most of these genes belonged to down-regulated module involved in mitochondrial respirasome assembly, cilium organization, and barrier integrity. The disruption of the epithelial barrier resulted in the downregulation of macrophage migration inhibitory factor (MIF) secretion in APP/PS1 mice, leading to macrophage activation and increased phagocytosis of Aβ. Concurrently, ligands (e.g., APOE) secreted by macrophages and other ChP cells facilitated the entry of lipids into ependymal cells, leading to lipid accumulation and the activation of microglia in the brain parenchyma in APP/PS1 mice compared to WT controls. CONCLUSIONS Taken together, these data profiled early transcriptional and cellular abnormalities of ChP within an AD mouse model, providing novel insights of cerebral vasculature into the pathobiology of AD.
Collapse
Affiliation(s)
- Zhong-Jiang Yan
- State Key Laboratory of Genetic Evolution & Animal Models, Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, and KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Maosen Ye
- State Key Laboratory of Genetic Evolution & Animal Models, Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, and KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Jiexi Li
- State Key Laboratory of Genetic Evolution & Animal Models, Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, and KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Deng-Feng Zhang
- State Key Laboratory of Genetic Evolution & Animal Models, Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, and KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650204, China.
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China.
- National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), National Resource Center for Non-Human Primates, Yunnan Engineering Center on Brain Disease Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650107, China.
| | - Yong-Gang Yao
- State Key Laboratory of Genetic Evolution & Animal Models, Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, and KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650204, China.
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China.
- National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), National Resource Center for Non-Human Primates, Yunnan Engineering Center on Brain Disease Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650107, China.
| |
Collapse
|
24
|
Qu Y, Ding M, Zhang M, Zheng L, Hu B, An H. Iridoid glycosides in kidney-tonifying Chinese medicinal herbs: Mechanisms and implications for Alzheimer's disease therapy. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119870. [PMID: 40288663 DOI: 10.1016/j.jep.2025.119870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 03/22/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Alzheimer's disease (AD) is an incurable and irreversible type of dementia. Existing drugs cannot meet clinical needs; thus, developing new treatments is necessary. Traditional Chinese medicine (TCM) has been used in the prevention and treatment of AD. TCM holds the theory that "the kidney support brain function" and believes that dementia can be addressed from a kidney-based perspective. Kidney-tonifying herbs are a class of medicines that have the effect of tonifying the kidney and benefiting the brain. Some of these herbs have been shown to have anti-AD effects. Iridoid glycosides (IGs), which are important components of kidney-tonifying herbs, may have the potential to prevent and treat AD. However, their effects on AD have not yet been reviewed. AIM OF THE REVIEW This literature review provides a comprehensive summary of the potential of IGs in the prevention and treatment of AD. It also sets the foundation for future studies that will make the use of such drugs in clinical practice possible. MATERIAL AND METHODS Kidney-tonifying Chinese herbs were selected with reference to the Chinese Pharmacopoeia (2020 edition) and the textbook of Chinese Materia Medica (5th edition). Literature survey was conducted using PubMed, Web of Science, Google Scholar, and CNKI, with "Alzheimer's disease," "kidney-tonifying Chinese medicinal herbs," and "Iridoid Glycosides" as the primary keywords. RESULTS Kidney-tonifying herbal IGs include loganin, morroniside, verbenalin, cornuside, catalpol, rehmannioside A, geniposidic acid, and aucubin. These IGs have shown multiple pharmacological effects, including anti-AD effects. The effective mechanisms of IGs for AD treatment include anti-oxidative stress, inhibiting neuronal apoptosis, antagonizing amyloid neurotoxicity and tau protein hyperphosphorylation, regulating immune function, anti-inflammation, normalizing the function of the cholinergic nervous system, recuperating neurobiochemical, and regulating AD-related genes. Consequently, IGs can combat AD by modulating multiple targets and pathways. CONCLUSION Kidney-tonifying herbal IGs have great potential to combat AD.
Collapse
Affiliation(s)
- Yanjie Qu
- Department of Traditional Chinese Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Minrui Ding
- Department of Neurology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Mengxue Zhang
- Department of Neurology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Lan Zheng
- Department of Traditional Chinese Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Bing Hu
- Cancer Institute, Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Hongmei An
- Department of Science & Technology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| |
Collapse
|
25
|
Dahlén AD, Roshanbin S, Aguilar X, Bucher NM, Lopes van den Broek S, Sehlin D, Syvänen S. PET imaging of TREM2 in amyloid-beta induced neuroinflammation. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07358-0. [PMID: 40434494 DOI: 10.1007/s00259-025-07358-0] [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: 02/05/2025] [Accepted: 05/14/2025] [Indexed: 05/29/2025]
Abstract
PURPOSE The triggering receptor expressed on myeloid cells 2 (TREM2) has become a promising target for biologics in both monitoring and treating neuroinflammation in Alzheimer's disease (AD). This study aimed to develop and compare bispecific anti-TREM2 antibodies featuring different transferrin receptor (TfR) binders to enhance brain delivery, identifying the most suitable format for in vivo PET imaging of TREM2 in transgenic AD mice. METHODS Three bispecific TREM2-antibody formats were produced and evaluated for their ability to cross the blood-brain barrier (BBB) via TfR-mediated transcytosis and bind TREM2. Blood concentration profiles up to 72 h post-injection (p.i.), and ex vivo brain uptake of iodine-125-labeled antibody constructs were quantified in AppNL-G-F and age-matched wild type (WT) mice using a γ-counter. The best-performing bispecific TREM2-antibody was radiolabeled with iodine-124 and used for in vivo PET imaging of brain TREM2 levels in AppNL-G-F mice at 72 h p.i. Brain TREM2 concentrations were subsequently quantified using ELISA. RESULTS The antibody format carrying two scFv8D3 TfR-binders (IgG-scFv2), demonstrated the highest brain concentrations of all tested bispecific constructs. This antibody also exhibited significantly higher brain concentrations in AppNL-G-F mice compared to WT mice at both 48 and 72 h p.i. This difference was further visualized and quantified through in vivo PET imaging. Moreover, brain concentrations of the antibody ligand correlated with elevated TREM2 levels in brain homogenates. CONCLUSION These findings highlight IgG-scFv2 as a promising radioligand for in vivo PET imaging of TREM2, advancing non-invasive neuroinflammation studies and supporting drug development for AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Amelia D Dahlén
- Department of Public Health and Caring Sciences, Section of Molecular Geriatrics, Uppsala University, Uppsala, Sweden
| | - Sahar Roshanbin
- Department of Public Health and Caring Sciences, Section of Molecular Geriatrics, Uppsala University, Uppsala, Sweden
| | - Ximena Aguilar
- Department of Public Health and Caring Sciences, Section of Molecular Geriatrics, Uppsala University, Uppsala, Sweden
| | - Nadja M Bucher
- Department of Public Health and Caring Sciences, Section of Molecular Geriatrics, Uppsala University, Uppsala, Sweden
| | - Sara Lopes van den Broek
- Department of Public Health and Caring Sciences, Section of Molecular Geriatrics, Uppsala University, Uppsala, Sweden
| | - Dag Sehlin
- Department of Public Health and Caring Sciences, Section of Molecular Geriatrics, Uppsala University, Uppsala, Sweden
| | - Stina Syvänen
- Department of Public Health and Caring Sciences, Section of Molecular Geriatrics, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
26
|
Chen W, Liu X, Muñoz VR, Kahn CR. Loss of insulin signaling in microglia impairs cellular uptake of Aβ and neuroinflammatory response exacerbating AD-like neuropathology. Proc Natl Acad Sci U S A 2025; 122:e2501527122. [PMID: 40388612 DOI: 10.1073/pnas.2501527122] [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: 01/21/2025] [Accepted: 04/07/2025] [Indexed: 05/21/2025] Open
Abstract
Insulin receptors are present on cells throughout the body, including the brain. Dysregulation of insulin signaling in neurons and astrocytes has been implicated in altered mood, cognition, and the pathogenesis of Alzheimer's disease (AD). To define the role of insulin signaling in microglia, the primary phagocytes in the brain critical for maintenance and damage repair, we created mice with an inducible microglia-specific insulin receptor knockout (MG-IRKO). RiboTag profiling of microglial mRNAs revealed that loss of insulin signaling results in alterations of gene expression in pathways related to innate immunity and cellular metabolism. In vitro, loss of insulin signaling in microglia results in metabolic reprogramming with an increase in glycolysis and impaired uptake of Aβ. In vivo, MG-IRKO mice exhibit alterations in mood and social behavior, and when crossed with the 5xFAD mouse model of AD, the resultant mice exhibit increased levels of Aβ plaque and elevated neuroinflammation. Thus, insulin signaling in microglia plays a key role in microglial cellular metabolism and the ability of the cells to take up Aβ, such that reduced insulin signaling in microglia alters mood and social behavior and accelerates AD pathogenesis. Together, these data indicate key roles of insulin action in microglia and the potential of targeting insulin signaling in microglia in treatment of AD.
Collapse
Affiliation(s)
- Wenqiang Chen
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, MA 02215
- Clinical and Translational Research, Steno Diabetes Center Copenhagen, Herlev 2730, Denmark
| | - Xiangyu Liu
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, MA 02215
| | - Vitor Rosetto Muñoz
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, MA 02215
- Laboratory of Molecular Biology of Exercise, University of Campinas, Limeira, São Paulo 13484-350, Brazil
| | - C Ronald Kahn
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, MA 02215
| |
Collapse
|
27
|
Yan F, Qiao Y, Pan S, Kang A, Chen H, Bai Y. RIPK1: A Promising Target for Intervention Neuroinflammation. J Neuroimmune Pharmacol 2025; 20:59. [PMID: 40418439 DOI: 10.1007/s11481-025-10208-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 04/16/2025] [Indexed: 05/27/2025]
Abstract
Necroptosis is a novel mode of cell death that differs from traditional apoptosis, characterized by distinct molecular mechanisms and physiopathological features. Recent research has increasingly underscored the pivotal role of necroptosis in various neurological diseases, including stroke, Alzheimer's disease and multiple sclerosis. A defining hallmark of these conditions is neuroinflammation, a complex inflammatory response that critically influences neuronal survival. This review provides a comprehensive analysis of the mechanistic underpinnings of necroptosis and its intricate interplay with neuroinflammation, exploring the interrelationship between the two processes and their impact on neurological disorders. In addition, we discuss potential therapeutic strategies that target the intervention of necroptosis and neuroinflammation, offering novel avenues for intervention. By deepening our understanding of these interconnected processes, the development of more effective treatments approaches holds significant promise for improving patient outcomes in neurological disorders.
Collapse
Affiliation(s)
- Feixing Yan
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730000, China
| | - Yujun Qiao
- Department of Pharmacy, Lanzhou University Second Hospital, Lanzhou, 730000, China
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730000, China
| | - Shunli Pan
- Department of Pharmacy, Lanzhou University Second Hospital, Lanzhou, 730000, China
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730000, China
| | - Anjuan Kang
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730000, China
| | - Haile Chen
- Department of Pharmacy, Lanzhou University Second Hospital, Lanzhou, 730000, China
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730000, China
| | - Yinliang Bai
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China.
- Department of Pharmacy, Lanzhou University Second Hospital, Lanzhou, 730000, China.
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730000, China.
| |
Collapse
|
28
|
Maynes MA, Owens CA, Anani-Wolf DM, Tritz ZP, Jin F, Hansen MJ, Seady M, Johnson AJ. Heightened effector immune cell infiltration of the hippocampus concurrently with brain ventricular volume expansion in aged APP/PS1 mice. J Neuroimmunol 2025; 406:578646. [PMID: 40449343 DOI: 10.1016/j.jneuroim.2025.578646] [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: 01/10/2025] [Revised: 04/16/2025] [Accepted: 05/19/2025] [Indexed: 06/03/2025]
Abstract
Alzheimer's disease (AD) is the most common form of dementia for which the role of neuroinflammation is becoming more realized. Recent studies have shown that immune cells infiltrate the hippocampus and the cortex of AD patients as well as mouse models of the disease. In this study, we employed T2-weighted magnetic resonance imaging (MRI) to view changes in ventricular volume in addition to spectral flow cytometric assessment of the hippocampus infiltrating immune profile in the aged APP/PS1 mice. Aged APP/PS1 mice present with increased size of lateral, dorsal, and ventral ventricles plus increased numbers of hippocampus infiltrating effector immune cell subsets, including CD8 T cells expressing IFNγ, granzyme B, and perforin along with other cell types such as γδ T cells, neutrophils, and NK cells. The concurrent increase in effector cell types with ventricular enlargement expands the putative mechanism of brain atrophy in the APP/PS1 mouse model to include cytolytic functions of these aforementioned immune cell subsets.
Collapse
Affiliation(s)
- Mark A Maynes
- Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, USA
| | - Carley A Owens
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
| | | | | | - Fang Jin
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
| | | | - Marina Seady
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
| | - Aaron J Johnson
- Department of Immunology, Mayo Clinic, Rochester, MN, USA; Department of Neurology, Mayo Clinic, Rochester, MN, USA; Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
29
|
Choi J, Strickland A, Loo HQ, Dong W, Barbar L, Bloom AJ, Sasaki Y, Jin SC, DiAntonio A, Milbrandt J. Diverse cell types establish a pathogenic immune environment in peripheral neuropathy. J Neuroinflammation 2025; 22:138. [PMID: 40410792 PMCID: PMC12100903 DOI: 10.1186/s12974-025-03459-7] [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: 02/24/2025] [Accepted: 05/01/2025] [Indexed: 05/25/2025] Open
Abstract
Neuroinflammation plays a complex and context-dependent role in many neurodegenerative diseases. We identified a key pathogenic function of macrophages in a mouse model of a rare human congenital neuropathy in which SARM1, the central executioner of axon degeneration, is activated by hypomorphic mutations in the axon survival factor NMNAT2. Macrophage depletion blocked and reversed neuropathic phenotypes in this sarmopathy model, revealing SARM1-dependent neuroimmune mechanisms as key drivers of disease pathogenesis. In this study, we investigated the impact of chronic subacute SARM1 activation on the peripheral nerve milieu using single cell/nucleus RNA-sequencing (sc/snRNA-seq). Our analyses reveal an expansion of immune cells (macrophages and T lymphocytes) and repair Schwann cells, as well as significant transcriptional alterations to a wide range of nerve-resident cell types. Notably, endoneurial fibroblasts show increased expression of chemokines (Ccl9, Cxcl5) and complement components (C3, C4b, C6) in response to chronic SARM1 activation, indicating enhanced immune cell recruitment and immune response regulation by non-immune nerve-resident cells. Analysis of CD45+ immune cells in sciatic nerves revealed an expansion of an Il1b+ macrophage subpopulation with increased expression of markers associated with phagocytosis and T cell activation/proliferation. We also found a significant increase in T cells in sarmopathic nerves. Remarkably, T cell depletion rescued motor phenotypes in the sarmopathy model. These findings delineate the significant changes chronic SARM1 activation induces in peripheral nerves and highlights the potential of immunomodulatory therapies for SARM1-dependent peripheral neurodegenerative disease.
Collapse
Affiliation(s)
- Julie Choi
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Amy Strickland
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Hui Qi Loo
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Wendy Dong
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Lilianne Barbar
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - A Joseph Bloom
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Needleman Center for Neurometabolism and Axonal Therapeutics, St. Louis, MO, 63110, USA
| | - Yo Sasaki
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Sheng Chih Jin
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Aaron DiAntonio
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Needleman Center for Neurometabolism and Axonal Therapeutics, St. Louis, MO, 63110, USA
| | - Jeffrey Milbrandt
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA.
- Needleman Center for Neurometabolism and Axonal Therapeutics, St. Louis, MO, 63110, USA.
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
30
|
Cao Q, Shen M, Li R, Liu Y, Zeng Z, Zhou J, Niu D, Zhang Q, Wang R, Yao J, Zhang G. Elucidating the specific mechanisms of the gut-brain axis: the short-chain fatty acids-microglia pathway. J Neuroinflammation 2025; 22:133. [PMID: 40400035 PMCID: PMC12093714 DOI: 10.1186/s12974-025-03454-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Accepted: 04/22/2025] [Indexed: 05/23/2025] Open
Abstract
In recent years, the gut microbiota has been increasingly recognized for its influence on various central nervous system diseases mediated by microglia, yet the underlying mechanisms remain unclear. As key metabolites of the gut microbiota, short-chain fatty acids (SCFAs) have emerged as a focal point in understanding microglia-related interactions. In this review, we further refine the connection between the gut microbiota and microglia by introducing the concept of the "SCFAs-microglia" pathway. We summarize current knowledge on this pathway, recent discoveries regarding its role in neurological diseases, and potential pharmacological strategies targeting it. Finally, we outlined the current challenges and limitations in this field of research. We hope this review provides new insights into the role of the gut microbiota in neuroimmune regulation.
Collapse
Affiliation(s)
- Qingyu Cao
- College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd, Linyi, 276005, China
| | - Mengmeng Shen
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd, Linyi, 276005, China
| | - Ruoqiu Li
- Key Laboratory of Marine Drugs, School of Medicine and Pharmacy, Ministry of Education, Ocean University of China, Qingdao, 266003, China
| | - Yan Liu
- School of Pharmacy, Qingdao University, Qingdao, 266071, China
| | - Zhen Zeng
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd, Linyi, 276005, China
| | - Jidong Zhou
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd, Linyi, 276005, China
| | - Dejun Niu
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd, Linyi, 276005, China
| | - Quancai Zhang
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd, Linyi, 276005, China
| | - Rongrong Wang
- Key Laboratory of Marine Drugs, School of Medicine and Pharmacy, Ministry of Education, Ocean University of China, Qingdao, 266003, China
| | - Jingchun Yao
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd, Linyi, 276005, China.
| | - Guimin Zhang
- College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China.
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd, Linyi, 276005, China.
| |
Collapse
|
31
|
Lin Z, Sun M. Phytochemical regulation of CaMKII in Alzheimer's disease: A review of molecular mechanisms and therapeutic potential. Pharmacol Res 2025; 216:107790. [PMID: 40409522 DOI: 10.1016/j.phrs.2025.107790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 04/08/2025] [Accepted: 05/19/2025] [Indexed: 05/25/2025]
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disorder that leads to cognitive decline. CaMKII is a calcium-regulated kinase that is crucial for synaptic plasticity and memory. Phytochemicals with diverse origins, safety, and biological activity have attracted considerable attention in AD research. This systematic analysis of phytochemicals targeting CaMKII reveals their neuroprotective mechanisms against AD pathogenesis, highlighting CaMKII as a promising therapeutic target that warrants further preclinical investigation and drug development. We conducted a comprehensive review of the literature of phytochemicals that target CaMKII as a protective mechanism against AD. The search was conducted across multiple databases, including PubMed, Web of Science, China National Knowledge Internet, and Google Scholar, and covered the period from January 2000 to October 2024. A total of 301 articles were retrieved, of which 22 articles were included. The results showed that flavonoid, glycoside, terpene, and polyphenol analogs positively regulated CaMKII expression, whereas alkaloid analogs negatively regulated CaMKII expression. Different components of traditional Chinese medicine played different roles in CaMKII expression. Flavonoid compounds upregulated the expression of SYN, PSD-95, MAP2, and GluR1 to exert neuroprotective effects. Alkaloid and glycoside analogs inhibited Aβ deposition and tau hyperphosphorylation. Terpene analogs upregulated the SYN, PSD-95, NMDAR, BDNF, and PI3K/Akt signaling pathways to exert neuroprotection. Polyphenol analogs upregulated PSD-95, Munc18-1, SNAP25, SYN, and BDNF to exert neuroprotective effects. Emerging evidence demonstrates that select phytochemicals and traditional Chinese medicine compounds exert neuroprotective effects in AD by modulating CaMKII activity, thereby reducing Aβ accumulation, attenuating tau hyperphosphorylation, and enhancing synaptic plasticity, suggesting promising therapeutic potential.
Collapse
Affiliation(s)
- Zhongying Lin
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China.
| | - Miao Sun
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China.
| |
Collapse
|
32
|
Ke S, Liu Z, Wan Y. Let-7 Family as a Mediator of Exercise on Alzheimer's Disease. Cell Mol Neurobiol 2025; 45:43. [PMID: 40389769 PMCID: PMC12089606 DOI: 10.1007/s10571-025-01559-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Accepted: 04/28/2025] [Indexed: 05/21/2025]
Abstract
Memory loss, and behavioral impairments. Hallmark pathological features include amyloid-beta (Aβ) plaques, tau neurofibrillary tangles, chronic inflammation, and impaired neuronal signaling. Physical exercise is increasingly recognized as a non-pharmacological intervention to attenuate Alzheimer's disease (AD) risk and progression by enhancing neuroplasticity, improving mitochondrial function, and modulating immune responses. The let-7 family of microRNAs is critically involved in AD pathology. Elevated levels of let-7b and let-7e have been reported in the cerebrospinal fluid of AD patients, with let-7b levels correlating positively with total tau and phosphorylated tau concentrations. Overexpression of let-7a enhances Aβ-induced neurotoxicity, increases neuronal apoptosis by up to 45%, and alters autophagy-related signaling via the PI3K/Akt/mTOR pathway, as shown by 1.8-fold increases in LC3-II/I ratios and 2.2-fold upregulation of Beclin-1 expression. Exercise modulates let-7 expression in a tissue-specific and context-dependent manner. Aerobic training reduces skeletal muscle expression of let-7b-5p by 30-35%, while increasing its suppressor Lin28a by 40%, thereby improving mitochondrial respiration. Overall, modulation of let-7 by exercise influences neuronal survival, autophagy, and inflammation, offering a potential mechanism through which physical activity exerts neuroprotective effects in AD. Quantitative characterization of let-7 expression patterns may support its use as a diagnostic and therapeutic biomarker, though further research is needed to establish optimal modulation strategies.
Collapse
Affiliation(s)
- Shanbin Ke
- College of Education, Jiangxi Institute of Applied Science and Technology, Nanchang, 330100, China
| | - Zhengqiong Liu
- College of Education, Jiangxi Institute of Applied Science and Technology, Nanchang, 330100, China
| | - Yuwen Wan
- College of Education, Jiangxi Institute of Applied Science and Technology, Nanchang, 330100, China.
| |
Collapse
|
33
|
Ebrahimi R, Shahrokhi Nejad S, Falah Tafti M, Karimi Z, Sadr SR, Ramadhan Hussein D, Talebian N, Esmaeilpour K. Microglial activation as a hallmark of neuroinflammation in Alzheimer's disease. Metab Brain Dis 2025; 40:207. [PMID: 40381069 DOI: 10.1007/s11011-025-01631-9] [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: 03/24/2025] [Accepted: 05/08/2025] [Indexed: 05/19/2025]
Abstract
Microglial activation has emerged as a hallmark of neuroinflammation in Alzheimer's disease (AD). Central to this process is the formation and accumulation of amyloid beta (Aβ) peptide and neurofibrillary tangles, both of which contribute to synaptic dysfunction and neuronal cell death. Aβ oligomers trigger microglial activation, leading to the release of pro-inflammatory cytokines, which further exacerbates neuroinflammation and neuronal damage. Importantly, the presence of activated microglia surrounding amyloid plaques is correlated with heightened production of cytokines such as interleukin (IL)-1β and tumor necrosis factor-alpha (TNF-α), creating a vicious cycle of inflammation. While microglia play a protective role by clearing Aβ plaques during the early stages of AD, their chronic activation can lead to detrimental outcomes, including enhanced tau pathology and neuronal apoptosis. Recent studies have highlighted the dualistic nature of microglial activation, showcasing both inflammatory (M1) and anti-inflammatory (M2) phenotypes that fluctuate based on the surrounding microenvironment. Disruption in microglial function and regulation can lead to neurovascular dysfunction, further contributing to the cognitive decline seen in AD. Moreover, emerging biomarkers and imaging techniques are unveiling the complexity of microglial responses in AD, providing avenues for targeted therapeutics aimed at modulating these cells. Understanding the intricate interplay between microglia, Aβ, and tau pathology is vital for developing potential interventions to mitigate neuroinflammation and its impact on cognitive decline in AD. This review synthesizes current findings regarding microglial activation and its implications for AD pathogenesis, offering insights into future therapeutic strategies.
Collapse
Affiliation(s)
- Rasoul Ebrahimi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Mahdi Falah Tafti
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Karimi
- Ross and Carol Nese College of Nursing, Pennsylvania State University, University Park, PA, USA
| | - Seyyedeh Reyhaneh Sadr
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Niki Talebian
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Preventative Gynecology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Khadijeh Esmaeilpour
- Department of Psychology, University of Toronto Mississagua, Mississauga, ON, Canada.
| |
Collapse
|
34
|
Bartra C, Vuraić K, Yuan Y, Codony S, Valdés-Quiroz H, Casal C, Slevin M, Máquez-Kisinousky L, Planas AM, Griñán-Ferré C, Pallàs M, Morisseau C, Hammock BD, Vázquez S, Suñol C, Sanfeliu C. Microglial pro-inflammatory mechanisms induced by monomeric C-reactive protein are counteracted by soluble epoxide hydrolase inhibitors. Int Immunopharmacol 2025; 155:114644. [PMID: 40215773 PMCID: PMC12147942 DOI: 10.1016/j.intimp.2025.114644] [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] [Received: 09/20/2024] [Revised: 04/01/2025] [Accepted: 04/07/2025] [Indexed: 04/29/2025]
Abstract
Monomeric C-reactive protein (mCRP) is a pro-inflammatory molecule generated by the dissociation of native CRP. Clinical and experimental studies suggest that mCRP deposition in the brain induces Alzheimer's disease (AD) pathology and cognitive loss. Pathological neuroinflammation is increasingly suggested as relevant in AD. Innovative therapies against neuroinflammation are desperately needed, and inhibitors of the enzyme soluble epoxide hydrolase (sEH) are a promising new generation of anti-inflammatory drugs. Mouse primary microglia and BV2 cell line cultures were exposed to mCRP to analyze its pro-inflammatory mechanisms. sEH inhibitors, both newly synthesized UB-SCG-55 and UB-SCG-65, and the reference agent TPPU, were tested for their anti-inflammatory action against mCRP. Phenotypic changes were analyzed through cell imaging techniques, as well as molecular analysis of inflammatory mediators and gene activation pathways. Results show that mCRP triggers a pro-inflammatory response through three main inflammatory pathways: iNOS, NLRP3, and COX-2, followed by increased cytokine generation. Polarization of microglia toward a M1-like phenotype was confirmed by morphological analysis. Also, mCRP can bind to and cross the cell membrane, providing further insight into its mechanisms of action. sEH inhibitors were effective against mCRP induction of a reactive microglial phenotype. The first-line compound UB-SCG-55 emerged as the most potent anti-inflammatory against mCRP injury. Therefore, the direct activation of microglia by mCRP provides evidence of its role in triggering and exacerbating neurodegenerative diseases with a neuroinflammatory component, such as AD. Furthermore, the protection given by inhibitors of sEH confirms its potential as innovative drugs against deleterious effects of neuroinflammation.
Collapse
Affiliation(s)
- Clara Bartra
- Department of Neuroscience and Experimental Therapeutics, Institute of Biomedical Research of Barcelona (IIBB), CSIC, 08036 Barcelona, Spain; PhD Program in Biotechnology, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, 08034 Barcelona, Spain.
| | - Kristijan Vuraić
- Department of Neuroscience and Experimental Therapeutics, Institute of Biomedical Research of Barcelona (IIBB), CSIC, 08036 Barcelona, Spain
| | - Yi Yuan
- Department of Neuroscience and Experimental Therapeutics, Institute of Biomedical Research of Barcelona (IIBB), CSIC, 08036 Barcelona, Spain
| | - Sandra Codony
- Laboratory of Medicinal Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, Universitat de Barcelona, Barcelona, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Haydeé Valdés-Quiroz
- Department of Neuroscience and Experimental Therapeutics, Institute of Biomedical Research of Barcelona (IIBB), CSIC, 08036 Barcelona, Spain
| | - Carme Casal
- Microscopy Service, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain
| | - Mark Slevin
- CCAMF, George Emil Palade Universitatea de Medicina, Farmacie, Stiinte se Technologie, "George Emil Palade" din Targu-Mures, 540142, Tirgu Mures, Romania
| | - Leonardo Máquez-Kisinousky
- Department of Neuroscience and Experimental Therapeutics, Institute of Biomedical Research of Barcelona (IIBB), CSIC, 08036 Barcelona, Spain
| | - Anna M Planas
- Department of Neuroscience and Experimental Therapeutics, Institute of Biomedical Research of Barcelona (IIBB), CSIC, 08036 Barcelona, Spain
| | - Christian Griñán-Ferré
- Department of Pharmacology and Therapeutic Chemistry, Institut de Neurociències-Universitat de Barcelona, Barcelona, Spain; Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Mercè Pallàs
- Department of Pharmacology and Therapeutic Chemistry, Institut de Neurociències-Universitat de Barcelona, Barcelona, Spain; Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Christophe Morisseau
- Department of Entomology and Nematology and Comprehensive Cancer Center, University of California, Davis, CA 95616, United States
| | - Bruce D Hammock
- Department of Entomology and Nematology and Comprehensive Cancer Center, University of California, Davis, CA 95616, United States
| | - Santiago Vázquez
- Laboratory of Medicinal Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, Universitat de Barcelona, Barcelona, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Cristina Suñol
- Department of Neuroscience and Experimental Therapeutics, Institute of Biomedical Research of Barcelona (IIBB), CSIC, 08036 Barcelona, Spain
| | - Coral Sanfeliu
- Department of Neuroscience and Experimental Therapeutics, Institute of Biomedical Research of Barcelona (IIBB), CSIC, 08036 Barcelona, Spain.
| |
Collapse
|
35
|
Zhang H, Song L, Zhou L, Li X, Xuan M, Liu C, Zhao H. α -Lipoic acid alleviates Parkinson's disease by suppressing S100A9-mediated pyroptosis. Int Immunopharmacol 2025; 155:114539. [PMID: 40233449 DOI: 10.1016/j.intimp.2025.114539] [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: 01/08/2025] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 04/17/2025]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease, and inflammation is a key factor in the progression of PD. S100A9 mediates pyroptosis and implicates in various diseases including PD. Pyroptosis, an emerging form of programmed cell death, usually causes cell rupture and death via an inflammatory response. α-Lipoic acid (α-ALA), a cellular coenzyme, participates in anti-inflammatory and antioxidant processes. Although its role in PD has been confirmed, but the exact mechanism of its anti-inflammatory effect remains unclear. In our research, we examined the potential mechanisms of pyroptosis mediated by S100A9 in PD and the neuroprotective effects of α-ALA. We used 6-hydroxydopamine (6-OHDA) to induce SH-SY5Y cells in vitro and in C57BL/6 mice in vivo. The cell viability of SH-SY5Y cells confirmed the neuroprotective effect of α-ALA. Proteomics analysis indicated that S100A9 was involved in 6-OHDA-mediated neuronal injury, while α-ALA could inhibit. We found that α-ALA ameliorated PD symptoms induced by 6-OHDA and decreased the levels of NLRP3 inflammasome, Gasdermin D, and IL-1β, which are major hallmarks of pyroptosis. Furthermore, our research demonstrated that α-ALA mitigated cell injury by suppressing NLRP3-dependent pyroptosis mediated by S100A9. In brief, pyroptosis is pivotal in PD, while α-ALA protects dopaminergic neurons by suppressing pyroptosis mediated through the NLRP3 inflammasome, directly reducing S100A9, and subsequently inhibiting the NLRP3/Gasdermin D signaling pathways. Our results collectively suggest that suppressing S100A9-mediated pyroptosis and administering α-ALA may represent a novel approach in treating of PD.
Collapse
Affiliation(s)
- Hongxu Zhang
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Ling Song
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Lin Zhou
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Xiaoyuan Li
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Mingwen Xuan
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Chang Liu
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China.
| | - Hong Zhao
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China.
| |
Collapse
|
36
|
Zeng X, Yuan Y, Li Y, Hu Z, Hu S. Deciphering the NLRP3 inflammasome in diabetic encephalopathy: Molecular insights and emerging therapeutic targets. Exp Neurol 2025; 391:115304. [PMID: 40383363 DOI: 10.1016/j.expneurol.2025.115304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 05/01/2025] [Accepted: 05/11/2025] [Indexed: 05/20/2025]
Abstract
Diabetic encephalopathy (DE) is a neurological complication characterized by neuroinflammation, cognitive impairment, and memory decline, with its pathogenesis closely linked to the activation of the NLRP3 inflammasome. As a central regulator of the innate immune system, the NLRP3 inflammasome plays a pivotal role in DE progression by mediating neuroinflammation, pyroptosis, mitochondrial dysfunction, oxidative stress, endoplasmic reticulum (ER) stress, and microglial polarization. This review systematically explores the molecular mechanisms by which the NLRP3 inflammasome contributes to DE, focusing on its role in neuroinflammatory cascades and neuronal damage, as well as the diabetes-associated physiological changes that exacerbate DE pathogenesis. Furthermore, we summarize emerging therapeutic strategies targeting the NLRP3 inflammasome, including small-molecule inhibitors and bioactive compounds derived from traditional herbal medicine, highlighting their potential for DE treatment. These findings not only advance our understanding of DE but also provide a foundation for developing NLRP3-targeted pharmacological interventions.
Collapse
Affiliation(s)
- Xinyi Zeng
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330031, China; The First Clinical Medical College of Nanchang University, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330031, China
| | - Yi Yuan
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330031, China; School of Huankui Academy, Nanchang University, Nanchang, Jiangxi 330031, China
| | - Yujia Li
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330031, China; The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330031, China
| | - Ziyan Hu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330031, China; The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330031, China
| | - Shan Hu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330031, China.
| |
Collapse
|
37
|
Sun C, Gao X, Sha S, Wang S, Shan Y, Li L, Xing C, Guan H, Du H. Berberine alleviates Alzheimer's disease by activating autophagy and inhibiting ferroptosis through the JNK-p38MAPK signaling pathway. Int Immunopharmacol 2025; 155:114550. [PMID: 40215776 DOI: 10.1016/j.intimp.2025.114550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 03/19/2025] [Accepted: 03/22/2025] [Indexed: 04/29/2025]
Abstract
INTRODUCTION Alzheimer's disease (AD) is a neurodegenerative disease characterized by amyloid beta (Aβ) deposition, phosphorylated Tau protein aggregation, inflammation, and neuronal damage. Neuronal autophagy plays an important role in ameliorating central nervous system diseases such as AD. As an emerging form of iron-dependent cell death, ferroptosis has attracted great attention in the field of neurodegenerative diseases. Berberine (BBR), a natural alkaloid, has demonstrated excellent in inflammation reduction, inhibition of Aβ production, and neuroprotection, making it a potential candidate for AD treatment. However, the mechanisms of autophagy and ferroptosis in BBR treatment of AD have not been elucidated. OBJECTIVES This study aimed to investigate the potential of BBR in alleviating AD and evaluate its molecular mechanism through a combination of network pharmacology and biological experiments. METHODS We assessed alterations in Aβ plaques, neurons, neuroinflammation, and autophagy-related markers in the mice brain using immunofluorescence staining. Network pharmacology and molecular docking were used to analyze the potential targets and signaling pathways of BBR in the treatment of AD. Morris Water Maze (MWM) and new object recognition (NOR) experiments were used to test the spatial memory ability of mice. In addition, we validated the relationship between JNK-P38MAPK, autophagy, ferroptosis, and BBR treatment in 5xFAD mice and A β-induced SH-SY5Y cell models. RESULTS The results of immunofluorescence staining showed that BBR effectively mitigated Aβ plaque deposition, ameliorated neuronal damage and neuroinflammation. The autophagy-related markers Beclin1 and LC3B were upregulated and P62 was downregulated after BBR treatment. The expression levels of ROS and lipid peroxide MDA decreased significantly after BBR treatment. qPCR results showed that the expression levels of ferroptosis-related genes TFR1, ASCL4, DMT1, and IREB2 were decreased, while the expression levels of FTH1 and SLC7A11 increased after BBR treatment. Behavioral experiments showed that BBR treatment enhanced spatial memory impairment in 5xFAD mice. Network pharmacological and in vitro analyses demonstrated that BBR activated autophagy and inhibited ferroptosis by inhibiting the JNK-P38MAPK signaling pathway. Following treatment with an autophagy inhibitor on SH-SY5Y cells, autophagy was markedly suppressed, and ferroptosis was induced. CONCLUSION In summary, we found that BBR alleviates AD by inhibiting the JNK-P38MAPK pathway to enhance autophagy and inhibit ferroptosis, further reducing Aβ plaque deposition, inhibiting inflammatory response, and improving neuronal damage.
Collapse
Affiliation(s)
- Chunbin Sun
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Xiaoyu Gao
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Shuang Sha
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Si Wang
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Yubang Shan
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Luping Li
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Cencan Xing
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China; Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083,China.
| | - Hongyan Guan
- China Testing & Certification International Group Co., Ltd., Beijing 100024, China.
| | - Hongwu Du
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China; Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083,China.
| |
Collapse
|
38
|
Yang W, Yu Q, Wang N, Lam KK, Lin ZX, Xian YF. Far-Infrared Radiation Ameliorates the Cognitive Dysfunction in an Alzheimer's Disease Transgenic Mouse via Modulating Jak-2/Stat3 and Nrf-2/HO-1 Pathways. Neuromolecular Med 2025; 27:34. [PMID: 40374872 DOI: 10.1007/s12017-025-08860-2] [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: 04/02/2025] [Accepted: 05/02/2025] [Indexed: 05/18/2025]
Abstract
Alzheimer's disease (AD) is the primary cause of dementia in the elderly. However, effective therapies that modify the disease process in AD remain elusive. Far-infrared radiation (FIR) is commonly utilized as a complementary treatment a range of disease, for example insomnia and rheumatoid arthritis. In this research, we explored how FIR light impacts the cognitive functions of TgCRND8 AD mice and elucidated its underlying molecular mechanism. The cognitive capabilities of TgCRND8 mice assessed by employing the Morris water maze. The concentrations of IL-1β, TNF-α, IL-4, Aβ40, and Aβ42 protein were assessed by enzyme-linked immunosorbent assay. Immunostaining was conducted to assess the Aβ deposits and microglial presence in the brains of TgCRND8 mice. Western blot was applied to detect the protein expressions of tau phosphorylation, amyloid-β (Aβ) production, Jak-2/Stat3, and Nrf-2/HO-1 pathways. The results indicated that FIR light notably ameliorated the cognitive impairments of the AD mice, reduced both Aβ deposition and tau protein hyperphosphorylation at sites of Thr205, Ser369, Ser404, and Thr181, suppressed the release of TNF-α and IL-1β, attenuated the ratios of p-Jak-2/Jak-2 and p-Stat3/Stat3, while increased the protein levels of IL-4, Nrf-2, and HO-1 in the brains of TgCRND8 mice. These findings amply demonstrated that FIR light ameliorated cognitive deficits of TgCRND8 mice via reducing both Aβ burden and tau protein hyperphosphorylation, suppressing the neuroinflammation, and restoring the levels of the oxidative-related proteins through modulating Jak-2/Stat3 and Nrf-2/HO-1 pathways. These experimental findings indicate that FIR light treatment is a promising treatment approach for AD.
Collapse
Affiliation(s)
- Wen Yang
- School of Chinese Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China
| | - Qiuxia Yu
- School of Chinese Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China
| | - Nick Wang
- Nick Wang Technology Limited, TML Tower, 3 Hoi Shing Road, Tsuen Wan, Kowloon, Hong Kong SAR, People's Republic of China
| | - Koon Kit Lam
- School of Chinese Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China
| | - Zhi-Xiu Lin
- School of Chinese Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China.
- Hong Kong Institute of Integrative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, People's Republic of China.
| | - Yan-Fang Xian
- School of Chinese Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China.
| |
Collapse
|
39
|
You Z, Liu Y, Li Z, Liu J, Li J. Application of Upper Limb Multimodal Tasks Combined With fNIRS Technology in the Assessment of Mild Cognitive Impairment. JOURNAL OF BIOPHOTONICS 2025:e202500020. [PMID: 40364662 DOI: 10.1002/jbio.202500020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/16/2025] [Accepted: 04/17/2025] [Indexed: 05/15/2025]
Abstract
Mild cognitive impairment (MCI) is primarily characterized by a gradual decline in cognitive function, where early detection and intervention are crucial to preventing Alzheimer's disease progression. This study integrates upper limb multimodal tasks (ULMTs) with functional near-infrared spectroscopy (fNIRS) to assess cognitive and motor functions in MCI patients. Thirty-seven elderly participants were categorized into healthy control (HC) and MCI groups. The experiment consisted of resting state, numerical cognitive task (NCT), motor task (MT), and ULMT phases. fNIRS measured hemodynamic responses in the prefrontal and motor cortices, while an upper limb trainer recorded motor data. Results showed weaker cortical responses in the MCI group during rest and reduced motor cortex activation during NCT. Both groups displayed increased cortical activity during ULMT compared to NCT but reduced motor performance compared to MT. These findings demonstrate the potential of ULMTs combined with fNIRS for early MCI assessment and intervention.
Collapse
Affiliation(s)
- Zhenda You
- School of Mechanical Engineering, Hebei University of Technology, Tianjin, China
| | - Ying Liu
- Laboratory of Robotics Mechanism and Cross Innovation, School of Intelligent Engineering and Automation, Beijing University of Posts and Telecommunications, Beijing, China
- Beijing Key Laboratory of Rehabilitation Technical Aids for Old-Age Disability, National Research Center for Rehabilitation Technical Aids, Beijing, China
- Key Laboratory of Neuro-Functional Information and Rehabilitation Engineering of the Ministry of Civil Affairs, Beijing, P. R. China
| | - Zengyong Li
- Beijing Key Laboratory of Rehabilitation Technical Aids for Old-Age Disability, National Research Center for Rehabilitation Technical Aids, Beijing, China
- Key Laboratory of Neuro-Functional Information and Rehabilitation Engineering of the Ministry of Civil Affairs, Beijing, P. R. China
| | - Jixiao Liu
- School of Mechanical Engineering, Hebei University of Technology, Tianjin, China
| | - Jian Li
- Laboratory of Robotics Mechanism and Cross Innovation, School of Intelligent Engineering and Automation, Beijing University of Posts and Telecommunications, Beijing, China
| |
Collapse
|
40
|
Wang K, Yang R, Li J, Wang H, Wan L, He J. Nanocarrier-based targeted drug delivery for Alzheimer's disease: addressing neuroinflammation and enhancing clinical translation. Front Pharmacol 2025; 16:1591438. [PMID: 40438598 PMCID: PMC12116324 DOI: 10.3389/fphar.2025.1591438] [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: 03/11/2025] [Accepted: 05/05/2025] [Indexed: 06/01/2025] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by cognitive decline, amyloid-beta (Aβ) aggregation, tau pathology, and chronic neuroinflammation. Among these, neuroinflammation plays a crucial role in exacerbating disease progression, making it an attractive therapeutic target. However, the presence of the blood-brain barrier (BBB) significantly limits the effective delivery of therapeutic agents to the brain, necessitating novel drug delivery strategies. Nanocarrier-based delivery systems have emerged as a promising solution to these challenges, offering targeted drug transport, enhanced BBB penetration, and improved bioavailability while minimizing systemic toxicity. This review explores the current advancements in nanocarrier-mediated drug delivery for AD, focusing on the mechanisms of neuroinflammation, the role of nanocarriers in overcoming the BBB, and their ability to modulate inflammatory pathways. Furthermore, the review discusses preclinical validation strategies and key challenges, including safety concerns, large-scale production limitations, and regulatory hurdles that must be addressed to enable clinical translation. Future perspectives emphasize the integration of nanotechnology with precision medicine, gene therapy, and artificial intelligence to optimize nanocarrier design for individualized AD treatment. By overcoming these obstacles, nanocarriers hold the potential to revolutionize therapeutic approaches for AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Kang Wang
- Acupuncture and Moxibustion Department, Beijing Massage Hospital, Beijing, China
| | - Rongying Yang
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| | - Jing Li
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Haitao Wang
- The school of Clinical Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Li Wan
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jiale He
- Department of Rheumatology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
41
|
Chen YH, Wang ZB, Liu XP, Mao ZQ. Cerebrospinal Fluid CCL25 as a Biomarker for Alzheimer's Disease: Associations with Pathology, Neurodegeneration, and Cognitive Decline. Mol Neurobiol 2025:10.1007/s12035-025-05007-z. [PMID: 40366557 DOI: 10.1007/s12035-025-05007-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 04/29/2025] [Indexed: 05/15/2025]
Abstract
Neuroinflammation plays a crucial role in Alzheimer's disease (AD) pathogenesis. We investigated the relationship between cerebrospinal fluid (CSF) C-C chemokine ligand 25 (CCL25), an inflammatory regulator, and AD pathology and progression. We analyzed data on CSF CCL25, AD biomarkers (CSF β-amyloid [Aβ]42, phosphorylated tau [pTau]181, amyloid positron emission tomography [PET]), postmortem neuropathology, magnetic resonance imaging-based neurodegeneration, and cognitive function from 703 participants in the Alzheimer's Disease Neuroimaging Initiative cohort. We found that elevated CSF CCL25 levels were associated with cognitive impairment, abnormal Aβ and tau pathology, greater brain atrophy, and worse cognitive performance (all P < 0.05). Notably, CSF CCL25 exhibited nonlinear relationships with Aβ and tau pathology, reaching a plateau as AD pathology increased. CSF CCL25 showed acceptable diagnostic accuracy in distinguishing amyloid-positive/negative (A ±) and tau-positive/negative (T ±) participants (area under the curve [AUC] = 0.71-0.77) and autopsy-confirmed AD cases (AUC = 0.77), with optimal performance in differentiating A + T + from A-T- participants (AUC = 0.82-0.85 with age and sex adjustment). Longitudinally, higher baseline CSF CCL25 predicted accelerated amyloid accumulation, hippocampal atrophy, and cognitive decline. Mediation analyses revealed that CCL25 partially mediated associations between Aβ pathology and tau pathology (mediating effect: 54.5%), neurodegeneration (18.2%), and cognitive decline (7.4%). Among 37 CSF CCL and CXCL chemokines examined, 28 were associated with at least one AD-related outcome, with CCL25 demonstrating the strongest associations overall. These findings suggest that CSF CCL25 is involved in early AD pathological progression and may serve as an inflammatory biomarker for diagnosis and monitoring of disease progression in AD.
Collapse
Affiliation(s)
- Yu-Han Chen
- Department of Human Anatomy, Neuroscience Research Center, Hebei Medical University, Shijiazhuang, 050017, China
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang, 050017, China
- The First Clinical Medical School, Hebei North University, Zhangjiakou, 075000, China
| | - Zhi-Bo Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing, 100053, China
| | - Xi-Peng Liu
- Department of Neurosurgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China
| | - Zhi-Qi Mao
- Department of Neurosurgery, the First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
42
|
Hermine O, Gros L, Tran TA, Loussaief L, Flosseau K, Moussy A, Mansfield CD, Vermersch P. Tyrosine kinase inhibitor, masitinib, limits neuronal damage, as measured by serum neurofilament light chain concentration in a model of neuroimmune-driven neurodegenerative disease. PLoS One 2025; 20:e0322199. [PMID: 40367050 PMCID: PMC12077730 DOI: 10.1371/journal.pone.0322199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 03/17/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND Masitinib is an orally administered tyrosine kinase inhibitor that targets activated cells of the innate neuroimmune system. We have studied the neuroprotective action of masitinib on the manifestations of experimental autoimmune encephalitis (EAE) induced axonal and neuronal damage. EAE is a model of neuroimmune-driven chronic neuroinflammation and therefore highly relevant to masitinib's mechanism of action in neurodegenerative diseases. Importantly, neuronal damage, or prevention thereof, can be rapidly assessed by measuring serum neurofilament light chain (NfL) concentration in EAE-induced mice. METHODS EAE induction was performed in healthy female C57BL/6 mice via active MOG 35-55 peptide immunization. Treatments were initiated 14 days post EAE induction. On day-0, 39 mice with established EAE symptoms were randomly assigned to 3 treatment groups (n = 13): EAE control, masitinib 50 mg/kg/day (M50), and masitinib 100 mg/kg/day (M100). The treatment started on day-1 and ended on day-15. Blood samples were collected on day-1 and day-8, via tail vein sampling, and on day-15, via intracardiac puncture. Assessments included quantification of serum NfL levels along the disease duration, cytokine quantification at day-15, and clinical assessments. RESULTS Masitinib treatment significantly (p < 0.0001) limited NfL production with respect to control; specifically, relative change in serum NfL concentration at day-8 was 43% and 60% lower for the M50 and M100 groups, respectively. Likewise, for the assessment of absolute serum NfL at day-8 and day-15, there was a significantly lower NfL concentration for masitinib treatment as compared with control. Furthermore, EAE mice treated with masitinib showed significantly lower concentrations of several well-established pro-inflammatory cytokines relative to control at day-15. A beneficial effect of masitinib on functional performance was also observed, with both M50 and M100 groups showing significantly less relative deterioration in grip strength at day-15 as compared with control (p < 0.001). CONCLUSION This study is the first demonstration that masitinib, a drug that targets the innate as opposed to the adaptive neuroimmune system, can lower serum NfL levels, and by extension therefore, neuronal damage, in a neuroimmune-driven neurodegenerative disease model. Overall, findings indicate that masitinib has a neuroprotective effect under conditions of chronic neuroinflammation and therefore plausible disease-modifying activity across a broad range of neurodegenerative diseases.
Collapse
MESH Headings
- Animals
- Female
- Mice
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/blood
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Neurofilament Proteins/blood
- Benzamides
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Mice, Inbred C57BL
- Disease Models, Animal
- Neurons/drug effects
- Neurons/pathology
- Thiazoles/pharmacology
- Thiazoles/therapeutic use
- Neurodegenerative Diseases/drug therapy
- Neurodegenerative Diseases/blood
- Neurodegenerative Diseases/pathology
- Pyridines
- Neuroprotective Agents/pharmacology
- Neuroprotective Agents/therapeutic use
- Tyrosine Kinase Inhibitors
- Piperidines
Collapse
Affiliation(s)
- Olivier Hermine
- Imagine Institute, INSERM UMR 1163, University of Paris, Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutic Implication, Hôpital Necker, Paris, France
- Department of Hematology, Necker Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | | | | | | | | | | | | | - Patrick Vermersch
- Univ. Lille, UMR Inserm U1172, CHU Lille, FHU Precise, Lille, France
| |
Collapse
|
43
|
Wang Y, Li Y, Zhou L, Yuan Y, Liu C, Zeng Z, Chen Y, He Q, Wu Z. Identification and validation of pyroptosis-related genes in Alzheimer's disease based on multi-transcriptome and machine learning. Front Aging Neurosci 2025; 17:1568337. [PMID: 40438507 PMCID: PMC12116433 DOI: 10.3389/fnagi.2025.1568337] [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: 01/29/2025] [Accepted: 04/29/2025] [Indexed: 06/01/2025] Open
Abstract
Background Alzheimer's disease (AD) progression is characterized by persistent neuroinflammation, where pyroptosis-an inflammatory programmed cell death mechanism-has emerged as a key pathological contributor. However, the molecular mechanisms through which pyroptosis-related genes (PRGs) drive AD pathogenesis remain incompletely elucidated. Methods We integrated multiple transcriptomes of AD patients from the GEO database and analyzed the expression of PRGs in combined datasets. Machine learning algorithms and comprehensive bioinformatics analysis (including immune infiltration and receiver operating characteristic (ROC)) were applied to identify the hub genes. Additionally, we validated the expression patterns of these key genes using the expression data from AD mice and constructed potential regulatory networks through time series and correlation analysis. Results We identified 91 PRGs in AD using the weighted gene co-expression network analysis (WGCNA) and differentially expressed genes analysis. By application of the protein-protein interaction and machine learning algorithms, seven pyroptosis feature genes (CHMP2A, EGFR, FOXP3, HSP90B1, MDH1, METTL3, and PKN2) were identified. Crucially, MDH1 and PKN2 demonstrated superior performance in terms of immune cell infiltration, ROC curves, and experimental validation. Furthermore, we constructed the long non-coding RNA and mRNA (lncRNA-mRNA) regulatory network of these characteristic genes using the gene expression profiles from AD mice at varying ages, revealing the potential regulatory mechanism in AD. Conclusion This study provides the first comprehensive characterization of pyroptosis-related molecular signatures in AD. Seven hub genes were identified, with particular emphasis on MDH1 and PKN2. Their superior performances were validated through comprehensive bioinformatic analysis in both patient and mouse transcriptomes, as well as the experimental data. Our findings establish foundational insights into pyroptosis mechanisms in AD that may inform novel treatment strategies targeting neuroinflammatory pathways.
Collapse
Affiliation(s)
- Yuntai Wang
- Institute of Basic Medicine, North Sichuan Medical College, Nanchong, China
- School of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Yilin Li
- School of Integrated Traditional Chinese and Western Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Lu Zhou
- School of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Yihuan Yuan
- School of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Chuanfei Liu
- School of Medical Imaging, North Sichuan Medical College, Nanchong, China
| | - Zimeng Zeng
- Institute of Basic Medicine, North Sichuan Medical College, Nanchong, China
| | - Yuanqi Chen
- School of Nursing, North Sichuan Medical College, Nanchong, China
| | - Qi He
- Institute of Basic Medicine, North Sichuan Medical College, Nanchong, China
| | - Zhuoze Wu
- Institute of Basic Medicine, North Sichuan Medical College, Nanchong, China
| |
Collapse
|
44
|
Aathira NS, Kaur A, Kumar A, Dar GM, Nimisha, Sharma AK, Bera P, Mahajan B, Chatterjee A, Saluja SS. The genetic risk factors, molecular pathways, microRNAs, and the gut microbiome in Alzheimer's disease. Neuroscience 2025; 577:217-227. [PMID: 40374065 DOI: 10.1016/j.neuroscience.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: 11/23/2024] [Revised: 04/25/2025] [Accepted: 05/12/2025] [Indexed: 05/17/2025]
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia worldwide. It is a multifaceted condition resulting from interplay of genetic mutations (e.g., APP, PSEN1, PSEN2) that account for less than 5% of cases, several genetic risk variants such as APOE4, TREM2, CD33, CLU, SORL1, and CR1 contribute to disease susceptibility and epigenetic factors, which may mediate the influence of environmental and lifestyle factors over time. Other critical contributors such as aging, protein misfolding and aggregation (amyloid-β and tau), molecular and transcriptomic dysregulation affecting neuronal function, and modifiable lifestyle factors like diet, physical activity, and environmental exposures presents challenges in accurate diagnosis and management. Research has predominantly focused on the diverse molecular pathways in the pathogenesis of AD, with particular attention given to the amyloidogenic pathways, tau pathology, calcium signalling, endolysosomal pathways, and others, whether they are directly or indirectly involved. Apart from these known molecular pathways, miRNAs are gaining attention as important regulators, which have been implicated in moderating the expression of mRNA targets involved in various processes associated with the clearance of pathogenic β-amyloid proteins. A mounting body of research suggests the possible role of gut microbiota in AD which regulates inflammation, neurotransmitters, and the blood-brain barrier. Gut dysbiosis can trigger neuroinflammation and amyloid-beta aggregation, making microbiome composition a potential early AD biomarker. This review aims to explore briefly the diverse risk encompassing genetic polymorphisms, altered molecular pathways implicated in AD pathogenesis, miRNA regulatory mechanisms, and the potential impact of gut microbiota on AD risk.
Collapse
Affiliation(s)
- N S Aathira
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Amanpreet Kaur
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Arun Kumar
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Ghulam Mehdi Dar
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Nimisha
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Abhay Kumar Sharma
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Pinki Bera
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Bhawna Mahajan
- Department of Biochemistry, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Atri Chatterjee
- Department of Neurology, VMMC and Safdarjung Hospital, New Delhi, India.
| | - Sundeep Singh Saluja
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India; Department of GI Surgery, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India.
| |
Collapse
|
45
|
Dimkovski A, Dobričić V, Simić MR, Jurhar Pavlova M, Mihajloska E, Sterjev Z, Poceva Panovska A. Synthesis, Biological Evaluation, and Molecular Docking Studies of Novel Coumarin-Triazole-Isatin Hybrids as Selective Butyrylcholinesterase Inhibitors. Molecules 2025; 30:2121. [PMID: 40430294 PMCID: PMC12114297 DOI: 10.3390/molecules30102121] [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: 04/17/2025] [Revised: 05/02/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
A series of 21 novel coumarin-triazole-isatin hybrids was synthesized and evaluated for their potential as multitarget agents in Alzheimer's disease (AD). The compounds featured variations in alkyl linker length that connects coumarin and triazole and substitution at the 5-position of the isatin ring. Several derivatives showed potent butyrylcholinesterase (BChE) inhibition with selectivity over acetylcholinesterase (AChE). The lead compound, 6c1, exhibited strong BChE inhibition (IC50 = 1.74 μM), surpassing donepezil. Enzyme kinetics revealed a mixed-type mechanism, while molecular docking studies confirmed dual binding at catalytic and peripheral sites. Structure-activity relationship (SAR) analysis highlighted the influence of linker flexibility and steric/electronic effects of substituents. The observed BChE selectivity, combined with favorable in vitro profiles, identifies these hybrids as promising leads for AD drug development.
Collapse
Affiliation(s)
- Aleksandar Dimkovski
- Institute for Pharmaceutical Chemistry, Faculty of Pharmacy, Ss Cyril and Methodius University in Skopje, 1000 Skopje, North Macedonia; (E.M.); (Z.S.)
| | - Vladimir Dobričić
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, 11221 Belgrade, Serbia
| | - Milena R. Simić
- Department of Organic Chemistry, Faculty of Pharmacy, University of Belgrade, 11221 Belgrade, Serbia;
| | - Maja Jurhar Pavlova
- Institute for Microbiology and Parasitology, Faculty of Medicine, Ss Cyril and Methodius University in Skopje, 1000 Skopje, North Macedonia;
| | - Evgenija Mihajloska
- Institute for Pharmaceutical Chemistry, Faculty of Pharmacy, Ss Cyril and Methodius University in Skopje, 1000 Skopje, North Macedonia; (E.M.); (Z.S.)
| | - Zoran Sterjev
- Institute for Pharmaceutical Chemistry, Faculty of Pharmacy, Ss Cyril and Methodius University in Skopje, 1000 Skopje, North Macedonia; (E.M.); (Z.S.)
| | - Ana Poceva Panovska
- Institute for Applied Chemistry and Pharmaceutical Analysis, Faculty of Pharmacy, Ss Cyril and Methodius University in Skopje, 1000 Skopje, North Macedonia;
| |
Collapse
|
46
|
McCrea LT, Batorsky RE, Bowen JJ, Yeh H, Thanos JM, Fu T, Perlis RH, Sheridan SD. Identifying brain-penetrant small-molecule modulators of human microglia using a cellular model of synaptic pruning. Neuropsychopharmacology 2025:10.1038/s41386-025-02123-1. [PMID: 40346178 DOI: 10.1038/s41386-025-02123-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 04/03/2025] [Accepted: 04/30/2025] [Indexed: 05/11/2025]
Abstract
Microglia dysregulation is implicated across a range of neurodevelopmental and neurodegenerative disorders, making their modulation a promising therapeutic target. Using PBMC-derived induced microglia-like cells (piMGLCs) in a scalable assay, we screened 489 CNS-penetrant compounds for modulation of microglial phagocytosis of human synaptosomes in a validated assay for microglia-mediated synaptic pruning. Compounds from the library that reduced phagocytosis by ≥2 standard deviations across the library without cytotoxicity were validated in secondary screens, with 28 of them further confirmed to reduce phagocytosis by 50% or more. These compounds comprise a wide range of therapeutic classes with different mechanisms of action, including immunosuppressants, kinase inhibitors, antipsychotics, and epigenetic modulators. Image-based morphological measurements were calculated to measure the degree of ramified vs. ameboid morphotypes as an indicator of activation state. Additionally, transcriptomic profiling indicated divergent effects on cell signaling, metabolism, activation, and actin dynamics across confirmed compounds. In particular, multiple CNS-penetrant small molecules with prior FDA approval or demonstration of safety in vivo demonstrated modulatory effects on microglia. For example, identified drugs such as the tyrosine kinase inhibitors lapatinib, alectinib, and lazertinib and the epigenetic modulator vorinostat have been approved for various cancer treatments and are being investigated for other indications; however, they have not been extensively studied in patients for neurodevelopmental and neurodegenerative disorders. These potential disease-modifying agents represent high-priority candidates for repositioning studies in neurodevelopmental, neuroinflammatory, or neurodegenerative disorders.
Collapse
Affiliation(s)
- Liam T McCrea
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Rebecca E Batorsky
- Tufts Institute for Artificial Intelligence, Tufts University, Medford, MA, USA
| | - Joshua J Bowen
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Hana Yeh
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Jessica M Thanos
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Ting Fu
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Roy H Perlis
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA.
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA.
| | - Steven D Sheridan
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA.
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
47
|
Guo TY, Zhang M, Lv YL, Qiu NZ, Chen RM, Zhang FF, Chen W, Zhang F, Gao YF, Wang XD, Zhang XH, Chen MH, Zhang HT, Wang H. Cognitive improvement effects of PF-04957325, a phosphodiesterase-8 inhibitor, in mouse models of Alzheimer's disease via modulating neuroinflammation. Int J Neuropsychopharmacol 2025; 28:pyaf028. [PMID: 40312965 PMCID: PMC12116884 DOI: 10.1093/ijnp/pyaf028] [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: 01/16/2025] [Accepted: 04/24/2025] [Indexed: 05/03/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative disease characterized by memory deficit and has emerged as a growing global health concern. Phosphodiesterase-8 (PDE8) is a cyclic adenosine monophosphate (cAMP)-specific hydrolase and its correlation with AD pathogenesis remains underexplored. Here, the effects and mechanisms of PF-04957325 (denoted as PF), a PDE8 inhibitor, were investigated in reversing AD both in vitro and in vivo. METHODS Briefly, BV2 cells were incubated with amyloid-β oligomers (AβO) to construct an AD cell model. Then, 2-month-old male C57BL/6J mice injected with AβO into the hippocampus and 10-month-old male amyloid precursor protein/presenilin-1 (APP/PS1) mice were used to construct AD animal models. Cells and mice were treated with PF to observe the effects of PDE8 on behavior and pathology related to AD. The Y-maze, novel object recognition (NOR), and Morris water maze (MWM) were performed to investigate cognitive function in mice. Western blot and immunofluorescence staining were used to identify the microglial activation state. Lastly, Western blot and ELISA were conducted to determine the levels of inflammatory factors and the proteins of PDE8/cAMP/CREB signaling. RESULTS PF-04957325 pretreatment reversed the conversation of proinflammatory microglia in BV2 cells induced by AβO, while also suppressing the levels of inflammatory factors, including interleukin-1β, interleukin-6, tumor necrosis factor-α, inducible nitric oxide synthase , and cyclooxygenase-2. In addition, AβO incubation upregulated the expression of PDE8 and concurrently downregulated that of brain-derived neurotrophic factor (BDNF), cAMP, p-PKA/PKA, and p-CREB/CREB in BV2 cells, all of which were reversed by PF. In vivo experiments evidenced impaired performance in the Y-maze, NOR, and MWM; these effects were reversed by PF. Similarly, PF treatment significantly attenuated microglia activation and the release of the inflammatory factors, and reversed the changes in the expression of BDNF and PDE8/cAMP/CREB signaling in AD mice. Finally, PF reduced the generation of Aβ1-42 by suppressing the expression of APP and PS1 in APP/PS1 mice. CONCLUSIONS PF alleviated AD-like changes in behavior and pathology through various mechanisms, including attenuating microglia-mediated neuroinflammation, upregulating the expression of BDNF, restoring synaptic dysfunction, and inhibiting Aβ generation, which appear to be involved by PDE8/cAMP/CREB signaling. These results highlight the therapeutic potential of targeting PDE8 inhibition for AD treatment.
Collapse
Affiliation(s)
- Tian-yang Guo
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai’an, Shandong, China
| | - Meng Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai, China
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Shandong Luye Pharmaceutical Co., Ltd., Yantai, Shandong, China
| | - Yu-li Lv
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai’an, Shandong, China
| | - Nian-zhuang Qiu
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai’an, Shandong, China
| | - Rui-min Chen
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai’an, Shandong, China
| | - Fang-fang Zhang
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai’an, Shandong, China
| | - Wei Chen
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai’an, Shandong, China
| | - Feng Zhang
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai’an, Shandong, China
| | - Yong-feng Gao
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai’an, Shandong, China
| | - Xiao-dan Wang
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai’an, Shandong, China
| | - Xue-hui Zhang
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai’an, Shandong, China
| | - Mei-hua Chen
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai’an, Shandong, China
| | - Han-ting Zhang
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai’an, Shandong, China
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, Shandong, China
| | - Hao Wang
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai’an, Shandong, China
| |
Collapse
|
48
|
Hu X, Ma YN, Peng J, Wang Z, Liang Y, Xia Y. Exosomes derived from olfactory mucosa mesenchymal stem cells attenuate cognitive impairment in a mouse model of Alzheimer's disease. Biosci Trends 2025; 19:189-201. [PMID: 40101983 DOI: 10.5582/bst.2025.01065] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by cognitive decline, neuroinflammation, and endoplasmic reticulum (ER) stress. In recent years, exosomes have garnered significant attention as a potential therapeutic tool for neurodegenerative diseases. This study, for the first time, investigates the neuroprotective effects of exosomes derived from olfactory mucosa mesenchymal stem cells (OM-MSCs-Exos) in AD and further explore the potential role of low-density lipoprotein receptor-related protein 1 (LRP1) in this process. Using an Aβ1-42-induced AD mouse model, we observed that OM-MSCs-Exos significantly improved cognitive function in behavioral tests, reduced neuroinflammatory responses, alleviated ER stress, and decreased neuronal apoptosis. Further analysis revealed that OM-MSCs-Exos exert neuroprotective effects by modulating the activation of microglia and astrocytes and influencing the ER stress response, a process that may involve LRP1. Although these findings support the potential neuroprotective effects of OM-MSCs-Exos, further studies are required to explore their long-term stability, dose dependency, and immunogenicity to assess their feasibility for clinical applications.
Collapse
Affiliation(s)
- Xiqi Hu
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
| | - Ya-Nan Ma
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
| | - Jun Peng
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
| | - Zijie Wang
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
| | - Yuchang Liang
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
| | - Ying Xia
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
| |
Collapse
|
49
|
Casquero-Veiga M, Ceron C, Cortes-Canteli M. Alzheimer's disease and vascular biology - A focus on the procoagulant state. Curr Opin Cell Biol 2025; 95:102528. [PMID: 40347710 DOI: 10.1016/j.ceb.2025.102528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/25/2025] [Accepted: 04/10/2025] [Indexed: 05/14/2025]
Abstract
Alzheimer's disease (AD) is characterized by a multifactorial pathophysiology. Beyond its classical hallmarks, growing evidence highlights vascular contributions, including hemostatic dysregulation and a prothrombotic state in AD. This review focuses on recent findings concerning two key blood clot components-fibrin(ogen) and platelets-and their roles in AD pathology, including fibrinogen's abnormal accumulation in the AD brain, its interaction with amyloid-β, together with the associated impacts on clot stability, vascular occlusion, and neuroinflammation; and the potential switch of platelets along the AD continuum from protective to deleterious. This review provides an update on the interplay between vascular dysfunction and AD, underscoring the need for comprehensive integrative research to address AD's complexity and advocating for personalized approaches to tackle this multifaceted disorder.
Collapse
Affiliation(s)
- Marta Casquero-Veiga
- Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain; Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Carlos Ceron
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Marta Cortes-Canteli
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Centro Internacional de Neurociencia Cajal - Consejo Superior de Investigaciones Científicas (CINC - CSIC), Madrid, Spain.
| |
Collapse
|
50
|
Han X, Cao X, Ju Q, Ge C, Lin Y, Shi J, Zhang X, Sun C, Li H. Microglial TAK1 promotes neurotoxic astrocytes and cognitive impairment in LPS-induced hippocampal neuroinflammation. J Biol Chem 2025:110225. [PMID: 40349778 DOI: 10.1016/j.jbc.2025.110225] [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: 11/25/2024] [Revised: 04/27/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025] Open
Abstract
The peripheral immune system has a strong effect on the central nervous system (CNS). Systemic lipopolysaccharides (LPS) administration triggers robust microglial activation and induces significant inflammatory responses in the hippocampus. This study investigates the role of Transforming Growth Factor-β-Activated Kinase 1 (TAK1) in mediating LPS-induced hippocampal neuroinflammation and cognitive impairment. Our findings reveal that LPS induces activation of microglial TAK1, which in turn actives downstream effector NF-κB/p65 to release pro-inflammatory cytokines. The activated microglia also promote astrocytes to polarize into a neurotoxic phenotype (A1-like phenotype), and cause the loss of newborn neurons in the hippocampal dentate gyrus (DG). However, TAK1 reduction inhibits microglial responses, limits neurotoxic astrocytes, rescues newborn neurons, and subsequently improves LPS-induced cognitive deficits, suggesting that targeting TAK1 may be an effective strategy for alleviating neuroinflammation. The interaction between TAK1 activation, microglial responses, and the transition of neurotoxic astrocytes enhances our understanding of the cellular dynamics driving LPS-induced neuroinflammation, suggesting that TAK1 may be a therapeutic target for treating cognitive impairment.
Collapse
Affiliation(s)
- Xiao Han
- Department of Human Anatomy, Medical School of Nantong University, Nantong, 226001, Jiangsu, China; Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu, China
| | - Xin Cao
- Department of Human Anatomy, Medical School of Nantong University, Nantong, 226001, Jiangsu, China
| | - Qianqian Ju
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu, China
| | - Chengxin Ge
- Department of Human Anatomy, Medical School of Nantong University, Nantong, 226001, Jiangsu, China
| | - Yongqi Lin
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu, China
| | - Jinhong Shi
- Department of Human Anatomy, Medical School of Nantong University, Nantong, 226001, Jiangsu, China
| | - Xinhua Zhang
- Department of Human Anatomy, Medical School of Nantong University, Nantong, 226001, Jiangsu, China; Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu, China.
| | - Cheng Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu, China.
| | - Haoming Li
- Department of Human Anatomy, Medical School of Nantong University, Nantong, 226001, Jiangsu, China.
| |
Collapse
|