1
|
Maurya R, Sharma A, Naqvi S. Decoding NLRP3 Inflammasome Activation in Alzheimer's Disease: A Focus on Receptor Dynamics. Mol Neurobiol 2025:10.1007/s12035-025-04918-1. [PMID: 40232645 DOI: 10.1007/s12035-025-04918-1] [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: 12/02/2024] [Accepted: 04/03/2025] [Indexed: 04/16/2025]
Abstract
Alzheimer's disease (AD) is a leading neurodegenerative disorder marked by progressive cognitive decline and significant neuropsychiatric disturbances. Neuroinflammation, mediated by the NLRP3 inflammasome, is increasingly recognized as a critical factor in AD pathogenesis. The NLRP3 inflammasome, a crucial component of the innate immune system, is activated in response to both pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs). In AD, amyloid-beta (Aβ) plaques and tau aggregates act as DAMPs, triggering NLRP3 inflammasome activation in microglia and astrocytes. This activation leads to the production of pro-inflammatory cytokines IL-1β and IL-18, contributing to chronic neuroinflammation and neuronal death. This review explores the intricate mechanisms involved in NLRP3 activation, with a particular focus on TREM-2, Msn Kinase MINK, NF-κB, Toll-like receptors, and P2X7 receptors. Understanding these mechanisms offers insight into the multifaceted regulation of the NLRP3 inflammasome and its impact on AD pathology. By elucidating the roles of TREM-2, MINK1, NF-κB, TLRs, and P2X7 receptors, this review highlights potential therapeutic targets for modulating NLRP3 activity. Targeting these pathways could offer novel strategies for mitigating neuroinflammation and slowing the progression of AD. The interplay between these receptors and signaling pathways underscores the complexity of NLRP3 inflammasome regulation and its significance in AD, providing a foundation for future research aimed at developing effective therapeutic interventions.
Collapse
Affiliation(s)
- Ranika Maurya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER-R), Lucknow, UP, 226002, India
| | - Abha Sharma
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER-R), Lucknow, UP, 226002, India
| | - Saba Naqvi
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER-R), Lucknow, UP, 226002, India.
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER-R), Lucknow, UP, 226002, India.
| |
Collapse
|
2
|
Zhao Y, Zhao B. Protection of Green Tea Polyphenols against Neurodegenerative Diseases: Evidence and Possible Mechanisms. J Nutr 2025; 155:1077-1088. [PMID: 39956389 DOI: 10.1016/j.tjnut.2025.02.010] [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: 12/20/2024] [Revised: 02/07/2025] [Accepted: 02/11/2025] [Indexed: 02/18/2025] Open
Abstract
Aging is a major risk factor for neurodegenerative diseases. With aging of the global population, the prevalence of neurodegenerative diseases, such as Alzheimer's disease (AD) and Parkinson's disease (PD), has increased worldwide. Unfortunately, the available therapeutic options for these neurodegenerative diseases are limited, most of which only provide symptomatic relief and have potentially serious side effects. Epidemiological studies have shown that green tea consumption is associated with a lower prevalence of cognitive decline and decreased risk of AD and PD, providing an attractive preventive and therapeutic option. Polyphenols are major bioactive components in green tea, which contribute to the beneficial effects of green tea. Accumulating data suggest that green tea polyphenols (GTPs) have neuroprotective properties that inhibit the pathological development of neurodegenerative diseases; however, the underlying mechanisms are not yet completely understood. This paper reviews both in vitro and in vivo evidence that demonstrates the neuroprotective effects of GTPs against neurodegenerative diseases, with the main focus on AD and PD, and summarizes the possible molecular mechanisms by which GTPs impede the progression of neurodegeneration. In particular, this review highlights the modulation of GTPs on the common mechanisms involved in pathogenesis of neurodegenerative diseases, including oxidative stress-mediated neuronal toxicity, impaired proteostasis, and metal ion dyshomeostasis. The potential of using GTPs in the intervention of neurodegenerative diseases is also discussed, hopefully, providing useful insights into novel preventive and therapeutic strategies for these diseases.
Collapse
Affiliation(s)
- Yan Zhao
- Department of Bioengineering, Harbin Institute of Technology, Weihai, China
| | - Baolu Zhao
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
3
|
Sălcudean A, Bodo CR, Popovici RA, Cozma MM, Păcurar M, Crăciun RE, Crisan AI, Enatescu VR, Marinescu I, Cimpian DM, Nan AG, Sasu AB, Anculia RC, Strete EG. Neuroinflammation-A Crucial Factor in the Pathophysiology of Depression-A Comprehensive Review. Biomolecules 2025; 15:502. [PMID: 40305200 DOI: 10.3390/biom15040502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/26/2025] [Accepted: 03/27/2025] [Indexed: 05/02/2025] Open
Abstract
Depression is a multifactorial psychiatric condition with complex pathophysiology, increasingly linked to neuroinflammatory processes. The present review explores the role of neuroinflammation in depression, focusing on glial cell activation, cytokine signaling, blood-brain barrier dysfunction, and disruptions in neurotransmitter systems. The article highlights how inflammatory mediators influence brain regions implicated in mood regulation, such as the hippocampus, amygdala, and prefrontal cortex. The review further discusses the involvement of the hypothalamic-pituitary-adrenal (HPA) axis, oxidative stress, and the kynurenine pathway, providing mechanistic insights into how chronic inflammation may underlie emotional and cognitive symptoms of depression. The bidirectional relationship between inflammation and depressive symptoms is emphasized, along with the role of peripheral immune responses and systemic stress. By integrating molecular, cellular, and neuroendocrine perspectives, this review supports the growing field of immunopsychiatry and lays the foundation for novel diagnostic biomarkers and anti-inflammatory treatment approaches in depression. Further research in this field holds promise for developing more effective and personalized interventions for individuals suffering from depression.
Collapse
Affiliation(s)
- Andreea Sălcudean
- Department of Ethics and Social Sciences, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Târgu Mureș, Romania
| | - Cristina-Raluca Bodo
- Department of Ethics and Social Sciences, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Târgu Mureș, Romania
| | - Ramona-Amina Popovici
- Department of Management and Communication in Dental Medicine, Faculty of Dental Medicine, Victor Babes University of Medicine and Pharmacy of Timisoara, 9 Revolutiei 1989 Bv., 300070 Timisoara, Romania
| | - Maria-Melania Cozma
- Department of Ethics and Social Sciences, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Târgu Mureș, Romania
| | - Mariana Păcurar
- Orthodontic Department, Faculty of Dental Medicine, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Târgu Mures, Romania
| | | | - Andrada-Ioana Crisan
- Doctoral School, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Târgu Mureș, Romania
| | - Virgil-Radu Enatescu
- Department of Psychiatry, Faculty of Medicine, Victor Babes University of Medicine and Pharmacy of Timisoara, 300041 Timisoara, Romania
| | - Ileana Marinescu
- Discipline of Psychiatry, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Dora-Mihaela Cimpian
- Department of Ethics and Social Sciences, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Târgu Mureș, Romania
| | - Andreea-Georgiana Nan
- First Department of Psychiatry, Clinical County Hospital of Targu Mures, 540142 Târgu Mureș, Romania
| | - Andreea-Bianca Sasu
- First Department of Psychiatry, Clinical County Hospital of Targu Mures, 540142 Târgu Mureș, Romania
| | - Ramona-Camelia Anculia
- Discipline of Occupational Medicine, Faculty of Medicine, Victor Babes University of Medicine and Pharmacy of Timisoara, 300041 Timișoara, Romania
| | - Elena-Gabriela Strete
- Department of Psychiatry, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Târgu Mureș, Romania
| |
Collapse
|
4
|
Zhang B, Chen L, Kang M, Ai L, Tao Y. Gegen Qinlian Decoction improves Alzheimer's disease through TLR4/NF-κB/NLRP3 pathway. Tissue Cell 2025; 95:102818. [PMID: 40056656 DOI: 10.1016/j.tice.2025.102818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 02/12/2025] [Accepted: 02/24/2025] [Indexed: 03/10/2025]
Abstract
OBJECTIVE Alzheimer's disease (AD) is a neurodegenerative disease that leads to dementia, but effective treatments are lacking. This study aims to evaluate the therapeutic effects of Gegen Qinlian Decoction (GGQLD) on AD and investigate the underlying mechanisms. METHODS Using network pharmacology and bioinformatics, we identified 376 active ingredients of GGQLD and 427 drug targets. Among these, 7 potential targets (CASP1, MKI67, NFKB1, TLR4, NLRP3, IL1B, and AKT1) were identified as intersecting targets of both GGQLD and AD. Functional enrichment analysis revealed that GGQLD regulates pyroptosis-related pathways. In vivo, GGQLD was administered to AD rat models to assess its effects on spatial learning, memory, and brain tissue injury. RESULTS GGQLD significantly reduced latency time by 40 % and increased platform crossings by 60 % in AD rats, demonstrating improved spatial learning and memory abilities. It also reduced hippocampal tissue damage and abnormal Aβ deposition. Mechanistically, GGQLD downregulated pyroptosis-related targets (TLR4, NF-κB, NLRP3, IL-1β, and Caspase-1), which were significantly upregulated in AD. ROC analysis demonstrated strong diagnostic significance for these genes, with AUC values exceeding 0.70. Functional enrichment and KEGG analysis further indicated that GGQLD exerts its therapeutic effects through multiple pathways, particularly the NOD-like receptor pathway, Necroptosis, and NF-kappa B pathway. CONCLUSIONS This study demonstrates that GGQLD improves spatial learning, reduces brain tissue damage, and alleviates inflammation in AD through the regulation of pyroptosis-related pathways, providing evidence for its potential as a therapeutic agent for AD.
Collapse
Affiliation(s)
- Bin Zhang
- Department of Traditional Chinese Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sun University, Guangzhou, Guangdong 510120, China
| | - Liudan Chen
- Department of Traditional Chinese Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sun University, Guangzhou, Guangdong 510120, China
| | - Mengru Kang
- Department of Traditional Chinese Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sun University, Guangzhou, Guangdong 510120, China
| | - Liang Ai
- Department of Traditional Chinese Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sun University, Guangzhou, Guangdong 510120, China
| | - Yangu Tao
- Department of Traditional Chinese Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sun University, Guangzhou, Guangdong 510120, China.
| |
Collapse
|
5
|
Akif A, My Nguyen TT, Liu L, Xu X, Kulkarni A, Jiang J, Zhang Y, Hao J. Targeting NLRP3 signaling with a novel sulfonylurea compound for the treatment of vascular cognitive impairment and dementia. RESEARCH SQUARE 2024:rs.3.rs-5611378. [PMID: 39764140 PMCID: PMC11702818 DOI: 10.21203/rs.3.rs-5611378/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2025]
Abstract
Background As a key inflammatory factor, the nucleotide-binding oligomerization domain (NOD)-like receptor protein 3 (NLRP3) inflammasome plays a crucial role in neuroinflammation and the progression of neurodegenerative diseases. Dysregulation of NLRP3 signaling can trigger various inflammatory responses in the brain, contributing to the development of neurodegenerative diseases such as ischemic stroke, vascular dementia (VaD), Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). Therefore, the NLRP3 signaling pathway is a promising therapeutic target for the treatment of neurodegenerative diseases, including VaD. Methods In this study, we investigated the therapeutic effects of a synthetic sulfonylurea NLRP3 inhibitor, AMS-17, in a VaD mouse model using bilateral common carotid artery stenosis (BCAS) and elucidated the underlying mechanisms. All mice were randomly divided into three groups: Sham, VaD + Vehicle, and VaD + AMS-17. Cognitive function was assessed using the Y-maze and Morris water maze (MWM) on the 50th day after BCAS. Brain sections and blood serum samples were collected for biomarker analysis and immunohistochemistry. Neurodegeneration, expressions of the molecules involved in the NLRP3 signaling pathways, tight junction proteins, and myelination were assessed using western blotting and immunofluorescence (IF). The levels of Interleukin-1 beta (IL-1β), Tumor Necrosis Factor-alpha (TNF-α) and Interleukin-4 (IL-4) in the blood were measured using ELISA. Results AMS-17 treatment improved cognitive function, enhanced blood-brain barrier (BBB) integrity, and promoted remyelination in VaD mice. Additionally, AMS-17 reduced neurodegeneration and decreased the expression of NLRP3 and its associated proteins, Apoptosis-associated speck-like protein (ASC), and cleaved caspase-1 in the brain. It also lowered pro-inflammatory TNF-α and IL-1β levels, while increasing the anti-inflammatory IL-4 level in the blood. Conclusions The findings of this study provide the first promising evidence for the use of AMS-17 in VaD treatment in mice. This study introduces AMS-17 as a novel chemical scaffold with NLRP3 inhibitory activity, which can be further developed for the treatment of VaD in humans.
Collapse
Affiliation(s)
| | | | - Langni Liu
- The University of Texas Health Science Center at Houston
| | - Xiaotian Xu
- The Affiliated Hospital of Yangzhou University
| | | | | | | | | |
Collapse
|
6
|
Liang K, Ma S, Luo K, Wang R, Xiao C, Zhang X, Gao Y, Li M. Salidroside: An Overview of Its Promising Potential and Diverse Applications. Pharmaceuticals (Basel) 2024; 17:1703. [PMID: 39770545 PMCID: PMC11678419 DOI: 10.3390/ph17121703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/05/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
Salidroside, a phenolic compound isolated from various Rhodiola plants, is the principal active constituent of Traditional Chinese Medicine known for its adaptogenic properties. Due to the challenging environment of Rhodiola species, such as high altitude, high radiation, drought, and hypoxia, the source of salidroside is scarce. However, numerous studies have shown that salidroside has a range of biological activities, including cardiovascular and central nervous system activity, and anti-hypoxia, anti-inflammatory, and anti-aging activities. Although previous studies have partially summarized the pharmacological effects of salidroside, the overall pharmacological effects have not been analyzed. Hence, this review will systematically summarize the isolation, purification, synthesis, derivatization, pharmacological activity, pharmacokinetics, clinical application, and safety of salidroside. It is expected to provide new insights for the further research and pharmaceutical development of salidroside.
Collapse
Affiliation(s)
- Keke Liang
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China; (K.L.); (S.M.); (K.L.); (R.W.)
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (C.X.); (X.Z.)
| | - Shuhe Ma
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China; (K.L.); (S.M.); (K.L.); (R.W.)
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (C.X.); (X.Z.)
| | - Kai Luo
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China; (K.L.); (S.M.); (K.L.); (R.W.)
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (C.X.); (X.Z.)
| | - Renjie Wang
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China; (K.L.); (S.M.); (K.L.); (R.W.)
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (C.X.); (X.Z.)
| | - Chenrong Xiao
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (C.X.); (X.Z.)
| | - Xianxie Zhang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (C.X.); (X.Z.)
| | - Yue Gao
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (C.X.); (X.Z.)
- National Key Laboratory of Kidney Diseases, Beijing 100850, China
| | - Maoxing Li
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China; (K.L.); (S.M.); (K.L.); (R.W.)
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (C.X.); (X.Z.)
- National Key Laboratory of Kidney Diseases, Beijing 100850, China
| |
Collapse
|
7
|
Dou RX, Zhang YM, Hu XJ, Gao FL, Zhang LL, Liang YH, Zhang YY, Yao YP, Yin L, Zhang Y, Gu C. Aβ 1-42 promotes microglial activation and apoptosis in the progression of AD by binding to TLR4. Redox Biol 2024; 78:103428. [PMID: 39550828 PMCID: PMC11615585 DOI: 10.1016/j.redox.2024.103428] [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: 10/08/2024] [Revised: 11/12/2024] [Accepted: 11/12/2024] [Indexed: 11/19/2024] Open
Abstract
Alzheimer's disease (AD) is one of the most common age-related neurodegenerative diseases and the most devastating form of senile dementia. It has a complex mechanism and no effective treatment. Exploring the pathogenesis of AD and providing ideas for treatment can effectively improve the prognosis of AD. Microglia were incubated with β-amyloid protein 1-42 (Aβ1-42) to construct an AD cell model. After microglia were activated, cell morphology changed, the expression level of inflammatory factors increased, cell apoptosis was promoted, and the expression of microtubule-associated protein (Tau protein) and related proteins increased. By up-regulating and down-regulating Toll-like receptor 4 (TLR4), the cells were divided into TLR4 knockdown negative control group(Lv-NC group), TLR4 knockdown group(Lv-TLR4 group), TLR4 overexpression negative control group(Sh-NC group), and TLR4 overexpression group(Sh-TLR4 group). The expression of inflammatory factors was detected again. It was found that compared with the Lv-NC group, the expression of various inflammatory factors in the Lv-TLR4 group decreased, cell apoptosis was inhibited, and the expression of Tau protein and related proteins decreased. Compared with the Sh-NC group, the expression of inflammatory factors in the Sh-TLR4 group increased, cell apoptosis was promoted, and the expression of Tau protein and related proteins increased. These results indicate that Aβ1-42 may promote microglial activation and apoptosis by binding to TLR4. Reducing the expression of TLR4 can reduce the occurrence of inflammatory response in AD cells and slow down cell apoptosis. Therefore, TLR4 is expected to become a new target for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Rui-Xia Dou
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China; Department of Neurology, Gansu Provincial People's Hospital, Lanzhou, 730000, Gansu, China.
| | - Ya-Min Zhang
- Department of Neurology, Gansu Provincial People's Hospital, Lanzhou, 730000, Gansu, China
| | - Xiao-Juan Hu
- Department of Neurology, Gansu Provincial People's Hospital, Lanzhou, 730000, Gansu, China
| | - Fu-Lin Gao
- Department of Neurology, Gansu Provincial People's Hospital, Lanzhou, 730000, Gansu, China
| | - Lu-Lu Zhang
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China; Department of Neurology, Gansu Provincial People's Hospital, Lanzhou, 730000, Gansu, China
| | - Yun-Hua Liang
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China; Department of Neurology, Gansu Provincial People's Hospital, Lanzhou, 730000, Gansu, China
| | - Yin-Ying Zhang
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China; Department of Neurology, Gansu Provincial People's Hospital, Lanzhou, 730000, Gansu, China
| | - Yu-Ping Yao
- Department of Neurology, Gansu Provincial People's Hospital, Lanzhou, 730000, Gansu, China
| | - Li Yin
- Western Medical District of Chinese PLA General Hospital, Haidian District, 100097, Beijing, China
| | - Yi Zhang
- Department of Neurology, Gansu Provincial People's Hospital, Lanzhou, 730000, Gansu, China
| | - Cheng Gu
- Department of Neurology, Gansu Provincial People's Hospital, Lanzhou, 730000, Gansu, China.
| |
Collapse
|
8
|
Shi C, Gao T, Lyu W, Qiang B, Chen Y, Chen Q, Zhang L, Liu Z. Deep-Learning-Driven Discovery of SN3-1, a Potent NLRP3 Inhibitor with Therapeutic Potential for Inflammatory Diseases. J Med Chem 2024; 67:17833-17854. [PMID: 39302813 DOI: 10.1021/acs.jmedchem.4c01857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
The NLRP3 inflammasome plays a central role in the pathogenesis of various intractable human diseases, making it an urgent target for therapeutic intervention. Here, we report the development of SN3-1, a novel orally potent NLRP3 inhibitor, designed through a lead compound strategy centered on deep-learning-based molecular generative models. Our strategy enables rapid fragment enumeration and takes into account the synthetic accessibility of the compounds, thereby significantly enhancing the optimization of lead compounds and facilitating the discovery of potent inhibitors. X-ray crystallography provided insights into the SN3-1 inhibitory mechanism. SN3-1 has shown a favorable safety profile in both acute and chronic toxicity assessments and exhibits robust pharmacokinetic properties. Furthermore, SN3-1 demonstrated significant therapeutic efficacy in various disease models characterized by NLRP3 activation. This study introduces a potent candidate for developing NLRP3 inhibitors and significantly expands the repertoire of tools available for the discovery of novel inhibitors.
Collapse
Affiliation(s)
- Cheng Shi
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Tongfei Gao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Weiping Lyu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Bo Qiang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yanming Chen
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Qixuan Chen
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Liangren Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Zhenming Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|
9
|
AmeliMojarad M, AmeliMojarad M. The neuroinflammatory role of microglia in Alzheimer's disease and their associated therapeutic targets. CNS Neurosci Ther 2024; 30:e14856. [PMID: 39031970 PMCID: PMC11259573 DOI: 10.1111/cns.14856] [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: 04/30/2024] [Revised: 06/17/2024] [Accepted: 07/02/2024] [Indexed: 07/22/2024] Open
Abstract
INTRODUCTION Alzheimer's disease (AD), the main cause of dementia, is characterized by synaptic loss and neurodegeneration. Amyloid-β (Aβ) accumulation, hyperphosphorylation of tau protein, and neurofibrillary tangles (NFTs) in the brain are considered to be the initiating factors of AD. However, this hypothesis falls short of explaining many aspects of AD pathogenesis. Recently, there has been mounting evidence that neuroinflammation plays a key role in the pathophysiology of AD and causes neurodegeneration by over-activating microglia and releasing inflammatory mediators. METHODS PubMed, Web of Science, EMBASE, and MEDLINE were used for searching and summarizing all the recent publications related to inflammation and its association with Alzheimer's disease. RESULTS Our review shows how inflammatory dysregulation influences AD pathology as well as the roles of microglia in neuroinflammation, the possible microglia-associated therapeutic targets, top neuroinflammatory biomarkers, and anti-inflammatory drugs that combat inflammation. CONCLUSION In conclusion, microglial inflammatory reactions are important factors in AD pathogenesis and need to be discussed in more detail for promising therapeutic strategies.
Collapse
Affiliation(s)
- Melika AmeliMojarad
- Department of Bioprocess Engineering, Institute of Industrial and Environmental BiotechnologyNational Institute of Genetic Engineering and BiotechnologyTehranIran
| | - Mandana AmeliMojarad
- Department of Bioprocess Engineering, Institute of Industrial and Environmental BiotechnologyNational Institute of Genetic Engineering and BiotechnologyTehranIran
| |
Collapse
|
10
|
Zeng L, Hu P, Zhang Y, Li M, Zhao Y, Li S, Luo A. Macrophage migration inhibitor factor (MIF): Potential role in cognitive impairment disorders. Cytokine Growth Factor Rev 2024; 77:67-75. [PMID: 38548489 DOI: 10.1016/j.cytogfr.2024.03.003] [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: 01/07/2024] [Revised: 03/11/2024] [Accepted: 03/15/2024] [Indexed: 06/22/2024]
Abstract
Macrophage migration inhibitory factor (MIF) is a cytokine in the immune system, participated in both innate and adaptive immune responses. Except from immune cells, MIF is also secreted by a variety of non-immune cells, including hematopoietic cells, endothelial cells (ECs), and neurons. MIF plays a crucial role in various diseases, such as sepsis, rheumatoid arthritis, acute kidney injury, and neurodegenerative diseases. The role of MIF in the neuropathogenesis of cognitive impairment disorders is emphasized, as it recruits multiple inflammatory mediators, leading to activating microglia or astrocyte-derived neuroinflammation. Furthermore, it contributes to the cell death of neurons and ECs with the binding of apoptosis-inducing factor (AIF) through parthanatos-associated apoptosis-inducing factor nuclease (PAAN) / MIF pathway. This review comprehensively delves into the relationship between MIF and the neuropathogenesis of cognitive impairment disorders, providing a series of emerging MIF-targeted pharmaceuticals as potential treatments for cognitive impairment disorders.
Collapse
Affiliation(s)
- Lian Zeng
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Pengchao Hu
- Hubei Provincial Clinical Research Center for Parkinson's Disease, Central Laboratory, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang 44100, China; Hubei Key Laboratory of Precision Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yu Zhang
- Hubei Provincial Clinical Research Center for Parkinson's Disease, Central Laboratory, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang 44100, China
| | - Mingyue Li
- Hubei Provincial Clinical Research Center for Parkinson's Disease, Central Laboratory, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang 44100, China
| | - Yilin Zhao
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shiyong Li
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Ailin Luo
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
11
|
Kargbo RB. Advancements in Predictive Medicine: NLRP3 Inflammasome Inhibitors and AI-Driven Predictive Health Analytics. ACS Med Chem Lett 2024; 15:331-333. [PMID: 38505847 PMCID: PMC10945530 DOI: 10.1021/acsmedchemlett.4c00056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Indexed: 03/21/2024] Open
Abstract
Recent advancements in predictive medicine are significantly reshaping the field, primarily through developing novel NLRP3 inflammasome inhibitors and applying AI-driven predictive health analytics. NLRP3 inflammasome inhibitors offer new therapeutic strategies for treating inflammatory and neurodegenerative diseases. Concurrently, AI's role in predictive health analytics marks a transformative shift in disease management and personalized healthcare. By analyzing complex biomarker data, AI provides crucial insights into individual health trajectories, enabling early interventions and customized treatment plans. This convergence of cutting-edge therapies and AI technology heralds a new era in precision medicine and personalized healthcare strategies.
Collapse
|
12
|
Pan Y, Li J, Lin P, Wan L, Qu Y, Cao L, Wang L. A review of the mechanisms of abnormal ceramide metabolism in type 2 diabetes mellitus, Alzheimer's disease, and their co-morbidities. Front Pharmacol 2024; 15:1348410. [PMID: 38379904 PMCID: PMC10877008 DOI: 10.3389/fphar.2024.1348410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 01/18/2024] [Indexed: 02/22/2024] Open
Abstract
The global prevalence of type 2 diabetes mellitus (T2DM) and Alzheimer's disease (AD) is rapidly increasing, revealing a strong association between these two diseases. Currently, there are no curative medication available for the comorbidity of T2DM and AD. Ceramides are structural components of cell membrane lipids and act as signal molecules regulating cell homeostasis. Their synthesis and degradation play crucial roles in maintaining metabolic balance in vivo, serving as important mediators in the development of neurodegenerative and metabolic disorders. Abnormal ceramide metabolism disrupts intracellular signaling, induces oxidative stress, activates inflammatory factors, and impacts glucose and lipid homeostasis in metabolism-related tissues like the liver, skeletal muscle, and adipose tissue, driving the occurrence and progression of T2DM. The connection between changes in ceramide levels in the brain, amyloid β accumulation, and tau hyper-phosphorylation is evident. Additionally, ceramide regulates cell survival and apoptosis through related signaling pathways, actively participating in the occurrence and progression of AD. Regulatory enzymes, their metabolites, and signaling pathways impact core pathological molecular mechanisms shared by T2DM and AD, such as insulin resistance and inflammatory response. Consequently, regulating ceramide metabolism may become a potential therapeutic target and intervention for the comorbidity of T2DM and AD. The paper comprehensively summarizes and discusses the role of ceramide and its metabolites in the pathogenesis of T2DM and AD, as well as the latest progress in the treatment of T2DM with AD.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Lei Wang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
13
|
Almeaqli MT, Alaidaa Y, Alnajjar FM, Al Shararh AS, Alharbi DS, Almslmani YI, Alotibi YA, Alrashidi HS, Alshehri WA, Hassan HM, Al-Gayyar MMH. Therapeutic Effects of Arctiin on Alzheimer's Disease-like Model in Rats by Reducing Oxidative Stress, Inflammasomes and Fibrosis. Curr Alzheimer Res 2024; 21:276-288. [PMID: 39136502 DOI: 10.2174/0115672050333388240801043509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 06/28/2024] [Accepted: 07/19/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) affects approximately 50 million people globally and is expected to triple by 2050. Arctiin is a lignan found in the Arctium lappa L. plant. Arctiin possesses anti-proliferative, antioxidative and anti-adipogenic. OBJECTIVES We aimed to explore the potential therapeutic effects of Arctiin on rats with AD by evaluating the expression of TLR4, NLRP3, STAT3, TGF-β, cyclin D1, and CDK2. METHODS AD was induced in rats by administering 70 mg/kg of aluminum chloride through intraperitoneal injection daily for six weeks. After inducing AD, some rats were treated with 25 mg/kg of Arctiin daily for three weeks through oral gavage. Furthermore, to examine the brain tissue structure, hippocampal sections were stained with hematoxylin/eosin and anti-TLR4 antibodies. The collected samples were analyzed for gene expression and protein levels of TLR4, NLRP3, STAT3, TGF-β, cyclin D1, and CDK2. RESULTS In behavioral tests, rats showed a significant improvement in their behavior when treated with Arctiin. Microimages stained with hematoxylin/eosin showed that Arctiin helped to improve the structure and cohesion of the hippocampus, which was previously impaired by AD. Furthermore, Arctiin reduced the expression of TLR4, NLRP3, STAT3, TGF-β, cyclin D1, and CDK2. CONCLUSION Arctiin can enhance rats' behavior and structure of the hippocampus in AD rats. This is achieved through its ability to reduce the expression of both TLR4 and NLRP3, hence inhibiting the inflammasome pathway. Furthermore, Arctiin can improve tissue fibrosis by regulating STAT3 and TGF-β. Lastly, it can block the cell cycle proteins cyclin D1 and CDK2.
Collapse
Affiliation(s)
- Mohamed T Almeaqli
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia
| | - Yazeed Alaidaa
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia
| | - Faisal M Alnajjar
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia
| | - Abdullah S Al Shararh
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia
| | - Danah S Alharbi
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia
| | - Yazeed I Almslmani
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia
| | - Yousef A Alotibi
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia
| | - Hani S Alrashidi
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia
| | - Wael A Alshehri
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia
| | - Hanan M Hassan
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa City, Egypt
| | - Mohammed M H Al-Gayyar
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia
| |
Collapse
|