1
|
Jin J, Zhang H, Lu Q, Tian L, Yao S, Lai F, Liang Y, Liu C, Lu Y, Tian S, Zhao Y, Ren W. Nanocarrier-mediated siRNA delivery: a new approach for the treatment of traumatic brain injury-related Alzheimer's disease. Neural Regen Res 2025; 20:2538-2555. [PMID: 39314170 PMCID: PMC11801294 DOI: 10.4103/nrr.nrr-d-24-00303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 07/06/2024] [Accepted: 07/17/2024] [Indexed: 09/25/2024] Open
Abstract
Traumatic brain injury and Alzheimer's disease share pathological similarities, including neuronal loss, amyloid-β deposition, tau hyperphosphorylation, blood-brain barrier dysfunction, neuroinflammation, and cognitive deficits. Furthermore, traumatic brain injury can exacerbate Alzheimer's disease-like pathologies, potentially leading to the development of Alzheimer's disease. Nanocarriers offer a potential solution by facilitating the delivery of small interfering RNAs across the blood-brain barrier for the targeted silencing of key pathological genes implicated in traumatic brain injury and Alzheimer's disease. Unlike traditional approaches to neuroregeneration, this is a molecular-targeted strategy, thus avoiding non-specific drug actions. This review focuses on the use of nanocarrier systems for the efficient and precise delivery of siRNAs, discussing the advantages, challenges, and future directions. In principle, siRNAs have the potential to target all genes and non-targetable proteins, holding significant promise for treating various diseases. Among the various therapeutic approaches currently available for neurological diseases, siRNA gene silencing can precisely "turn off" the expression of any gene at the genetic level, thus radically inhibiting disease progression; however, a significant challenge lies in delivering siRNAs across the blood-brain barrier. Nanoparticles have received increasing attention as an innovative drug delivery tool for the treatment of brain diseases. They are considered a potential therapeutic strategy with the advantages of being able to cross the blood-brain barrier, targeted drug delivery, enhanced drug stability, and multifunctional therapy. The use of nanoparticles to deliver specific modified siRNAs to the injured brain is gradually being recognized as a feasible and effective approach. Although this strategy is still in the preclinical exploration stage, it is expected to achieve clinical translation in the future, creating a new field of molecular targeted therapy and precision medicine for the treatment of Alzheimer's disease associated with traumatic brain injury.
Collapse
Affiliation(s)
- Jie Jin
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Huajing Zhang
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Key Laboratory for Disaster Medicine Technology, Tianjin, China
| | - Qianying Lu
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Key Laboratory for Disaster Medicine Technology, Tianjin, China
| | - Linqiang Tian
- Henan Medical Key Laboratory for Research of Trauma and Orthopedics, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan Province, China
- Clinical Medical Center of Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang, Henan Province, China
| | - Sanqiao Yao
- Institutes of Health Central Plain, Xinxiang Medical University, Xinxiang, Henan Province, China
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, China
| | - Feng Lai
- Henan Medical Key Laboratory for Research of Trauma and Orthopedics, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan Province, China
| | - Yangfan Liang
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Chuanchuan Liu
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Yujia Lu
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Sijia Tian
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Yanmei Zhao
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Key Laboratory for Disaster Medicine Technology, Tianjin, China
| | - Wenjie Ren
- Henan Medical Key Laboratory for Research of Trauma and Orthopedics, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan Province, China
- Clinical Medical Center of Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang, Henan Province, China
- Institutes of Health Central Plain, Xinxiang Medical University, Xinxiang, Henan Province, China
| |
Collapse
|
2
|
Li Y, Xue J, Ma Y, Ye K, Zhao X, Ge F, Zheng F, Liu L, Gao X, Wang D, Xia Q. The complex roles of m 6 A modifications in neural stem cell proliferation, differentiation, and self-renewal and implications for memory and neurodegenerative diseases. Neural Regen Res 2025; 20:1582-1598. [PMID: 38845217 PMCID: PMC11688559 DOI: 10.4103/nrr.nrr-d-23-01872] [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: 11/13/2023] [Revised: 02/19/2024] [Accepted: 03/25/2024] [Indexed: 08/07/2024] Open
Abstract
N6-methyladenosine (m 6 A), the most prevalent and conserved RNA modification in eukaryotic cells, profoundly influences virtually all aspects of mRNA metabolism. mRNA plays crucial roles in neural stem cell genesis and neural regeneration, where it is highly concentrated and actively involved in these processes. Changes in m 6 A modification levels and the expression levels of related enzymatic proteins can lead to neurological dysfunction and contribute to the development of neurological diseases. Furthermore, the proliferation and differentiation of neural stem cells, as well as nerve regeneration, are intimately linked to memory function and neurodegenerative diseases. This paper presents a comprehensive review of the roles of m 6 A in neural stem cell proliferation, differentiation, and self-renewal, as well as its implications in memory and neurodegenerative diseases. m 6 A has demonstrated divergent effects on the proliferation and differentiation of neural stem cells. These observed contradictions may arise from the time-specific nature of m 6 A and its differential impact on neural stem cells across various stages of development. Similarly, the diverse effects of m 6 A on distinct types of memory could be attributed to the involvement of specific brain regions in memory formation and recall. Inconsistencies in m 6 A levels across different models of neurodegenerative disease, particularly Alzheimer's disease and Parkinson's disease, suggest that these disparities are linked to variations in the affected brain regions. Notably, the opposing changes in m 6 A levels observed in Parkinson's disease models exposed to manganese compared to normal Parkinson's disease models further underscore the complexity of m 6 A's role in neurodegenerative processes. The roles of m 6 A in neural stem cell proliferation, differentiation, and self-renewal, and its implications in memory and neurodegenerative diseases, appear contradictory. These inconsistencies may be attributed to the time-specific nature of m 6 A and its varying effects on distinct brain regions and in different environments.
Collapse
Affiliation(s)
- Yanxi Li
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Jing Xue
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yuejia Ma
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Ke Ye
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Xue Zhao
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Fangliang Ge
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Feifei Zheng
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Lulu Liu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Xu Gao
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- Basic Medical Institute, Heilongjiang Academy of Medical Sciences, Harbin, Heilongjiang Province, China
- Key Laboratory of Heilongjiang Province for Genetically Modified Animals, Harbin Medical University, Harbin, Heilongjiang Province, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, Heilongjiang Province, China
| | - Dayong Wang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, Heilongjiang Province, China
| | - Qing Xia
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
3
|
da Silva EA, Faber J, Penitente AR, Fernandes J, Bertolucci PHF, Longo BM, Arida RM. Effects of resistance exercise on behavioral and molecular changes in transgenic female mice for Alzheimer's disease in early and advanced stages. Exp Neurol 2025; 388:115217. [PMID: 40089002 DOI: 10.1016/j.expneurol.2025.115217] [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: 06/27/2024] [Revised: 02/26/2025] [Accepted: 03/11/2025] [Indexed: 03/17/2025]
Abstract
Alzheimer's disease (AD) is a neurodegenerative condition that affects memory and cognition, with a higher prevalence in women. Given the lack of effective treatment, physical activity stands out as a complementary approach to prevent or delay disease progression. While numerous studies on humans and animals indicate that aerobic exercise induces brain changes, the impact of resistance exercise (RE) on AD is not fully understood. OBJECTIVE This study aimed to investigate the behavioral and molecular changes induced by RE in female transgenic mice with AD at the early and advanced stages of the disease. MATERIALS AND METHODS Adult (initial phase - 7 to 8 months of age, n = 32) and adult/elderly (advanced phase - 22 to 23 months of age, n = 32) female mice (2xTg-AD) for the APPSWE/PS1dE9 mutation were subjected to a four-week RE protocol. Mobility, anxiety-like behavior, long-term memory (LTM), and depressive-like behavior were assessed. Beta-amyloid (βA) and cytokines were quantified using the ELISA technique. RESULTS There was a progressive increase in strength in both trained groups at different ages. RE reversed memory deficits only in adult AD animals and the anxiety-like behavior only in adult/elderly AD animals. RE reversed depressive-like behavior in adult and adult/elderly AD animals. RE reduced βA only in adult AD animals. RE modified the expression of several cytokines in animals in the early and advanced stage of AD. CONCLUSION RE can be a promising strategy to minimize the deleterious effects of AD; however, its effectiveness may be more limited to the early stages of the disease.
Collapse
Affiliation(s)
| | - Jean Faber
- Federal University of Sao Paulo - Neurology and Neurosurgery Department, Brazil
| | | | | | | | | | | |
Collapse
|
4
|
Nong W, Chen X, Chen Y, Feng X, Kong W, Chi R, Yan L, Wei Z. Natural polyphenol mangiferin delays neuronal cell senescence by inhibiting neuroinflammation mediated by microglial activation. IBRO Neurosci Rep 2025; 18:574-591. [PMID: 40271493 PMCID: PMC12017977 DOI: 10.1016/j.ibneur.2025.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 03/13/2025] [Accepted: 04/07/2025] [Indexed: 04/25/2025] Open
Abstract
Extracellular β-amyloid protein (Aβ) plaques are prominent pathological feature of Alzheimer's disease (AD). Aβ oligomers and plaques induce sustained microglial activation via the adenosine monophosphate-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR)/interferon regulatory factor 5 (IRF5) signaling pathway. This microglial activation-mediated neuroinflammation can accelerate neuronal cell senescence. Consequently, the regulation of the AMPK/mTOR/IRF5 pathway presents a potential therapeutic target for AD, as it may inhibit neuroinflammation and delay neuronal cell senescence. Mangiferin, a bioactive natural polyphenol extracted from the leaves of Mangifera indica Linn., has garnered significant attention for its anti-inflammatory properties. However, it remains unclear whether mangiferin can modulate the AMPK/mTOR/IRF5 pathway to inhibit microglial activation-mediated neuroinflammation and delay neuronal cell senescence. This study employed both cellular and animal models of neuronal cell senescence to explore the effects of mangiferin on the regulation of the AMPK/mTOR/IRF5 pathway, aiming to inhibit neuroinflammation and delay neuronal cell senescence in vitro and in vivo. Specifically, SH-SY5Y neuroblastoma cells were subjected to a neuroinflammatory microenvironment induced by Aβ1-42-mediated HMC3 microglial activation to induce neuronal cell senescence in vitro. Additionally, SAMP8 accelerated aging mice were utilized as an aging animal model. The results indicate that mangiferin significantly enhances AMPK phosphorylation in microglial cells, inhibits mTOR activation, and downregulates IRF5 expression. These effects collectively suppress microglial activation and markedly reduce the production of pro-inflammatory cytokines by activated microglia. Consequently, there is a decrease in the proportion of neurons arrested in the G0/G1 phase and a reduction in the number of senescence-associated β-galactosidase (SA-β-gal) positive neurons. Furthermore, mangiferin significantly decreases the expression of neuronal cell senescence markers P16Ink4a and P21Cip1. Collectively, these findings suggest that mangiferin effectively regulates the AMPK/mTOR/IRF5 pathway, inhibits neuroinflammation mediated by microglial activation, and delays neuronal cell senescence. This study underscores the potential of mangiferin for the treatment of neuroinflammation and neurodegenerative diseases.
Collapse
Affiliation(s)
- Wei Nong
- Guangxi Scientific Research Center of Traditional Chinese Medicine, Guangxi Traditional Chinese Medicine University, Nanning 530200, China
| | - Xiaoli Chen
- Laboratory of Zhuang Medicine Prescriptions Basis and Application Research, Guangxi Traditional Chinese Medicine University, Nanning 530001, China
| | - Yixin Chen
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi Traditional Chinese Medicine University, Nanning 530200, China
| | - Xueping Feng
- School of Basic Medical Sciences, Youjiang Medical University for Nationalities, Baise 533000, China
| | - Wen Kong
- Zhuang Medicine College, Guangxi Traditional Chinese Medicine University, Nanning 530001, China
| | - Rui Chi
- Zhuang Medicine College, Guangxi Traditional Chinese Medicine University, Nanning 530001, China
| | - Li Yan
- Laboratory of Zhuang Medicine Prescriptions Basis and Application Research, Guangxi Traditional Chinese Medicine University, Nanning 530001, China
| | - Zhiquan Wei
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi Traditional Chinese Medicine University, Nanning 530200, China
| |
Collapse
|
5
|
Wu H, Song Z, Chen Q, Yan R, Zhao H, Li H. Disrupting reconsolidation by systemic inhibition of Thioredoxin-1 attenuates cocaine and morphine relapse. Biomed Pharmacother 2025; 186:118037. [PMID: 40199134 DOI: 10.1016/j.biopha.2025.118037] [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: 03/30/2025] [Accepted: 04/03/2025] [Indexed: 04/10/2025] Open
Abstract
The enduring nature of drug-associated memories is an essential factor contributing to the relapse. Drug-related cues can activate drug memories, making them enter reconsolidation, during which interventions can effectively disrupt these memories. Interventions targeting memory reconsolidation present a promising therapeutic strategy for addressing substance use disorders (SUDs). Oxidative stress can disrupt neural function and impair memory. Thioredoxin-1 (Trx-1) effectively alleviates oxidative stress and reduces inflammation levels. However, few studies have connected Trx-1 to drug memory or explored its specific role in reconsolidation. This research employed the conditioned place preference (CPP) model to investigate the effects of Trx-1 inhibitors on the reconsolidation of morphine- and cocaine-related memories. Results show that immediate administration of PX-12, a Trx-1 inhibitor, after retrieval significantly attenuated the reinstatement of cocaine and morphine CPP induced by both cues and the drug itself, with the effect lasting for at least 14 days. In contrast, the inhibition of Trx-1, either 6 hours following retrieval or in the absence of retrieval, does not influence drug-seeking behaviors associated with cocaine or morphine. Furthermore, Trx-1 inhibitor itself did not produce any preferences. In summary, our results indicate that Trx-1 activity is crucial for cocaine- and morphine-related memories, and that the Trx-1 inhibitor may serve as a potential treatment for drug abuse.
Collapse
Affiliation(s)
- Hao Wu
- Department of Anesthesiology, Peking Union Medical College Hospital, Peking Union Medical College, and Chinese Academy of Medical Science, Beijing, China
| | - Zhihao Song
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China; Clinical Research Center for Skull Base Surgery and Neurooncology in Hunan Province, China; National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Qijun Chen
- Department of Clinical Laboratory, The Affiliated Guangzhou Twelfth People's Hospital, Guangzhou Medical University, Guangzhou 510620, China
| | - Ruyu Yan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China; Clinical Research Center for Skull Base Surgery and Neurooncology in Hunan Province, China; National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Haiting Zhao
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China; Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
| | - Haoyu Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China; Clinical Research Center for Skull Base Surgery and Neurooncology in Hunan Province, China; National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
6
|
Altahrawi AY, James AW, Shah ZA. The Role of Oxidative Stress and Inflammation in the Pathogenesis and Treatment of Vascular Dementia. Cells 2025; 14:609. [PMID: 40277934 DOI: 10.3390/cells14080609] [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: 03/11/2025] [Revised: 04/09/2025] [Accepted: 04/16/2025] [Indexed: 04/26/2025] Open
Abstract
Vascular dementia (VaD) is a heterogeneous group of brain disorders caused by cerebrovascular pathologies and the second most common cause of dementia, accounting for over 20% of cases and posing an important global health concern. VaD can be caused by cerebral infarction or injury in critical brain regions, including the speech area of the dominant hemisphere or arcuate fasciculus of the dominant hemisphere, leading to notable cognitive impairment. Although the exact causes of dementia remain multifactorial and complex, oxidative stress (reactive oxygen species), neuroinflammation (TNFα, IL-6, and IL-1β), and inflammasomes are considered central mechanisms in its pathology. These conditions contribute to neuronal damage, synaptic dysfunction, and cognitive decline. Thus, antioxidants and anti-inflammatory agents have emerged as potential therapeutic targets in dementia. Recent studies emphasize that cerebrovascular disease plays a dual role: first, as a primary cause of cognitive impairment and then as a contributor to the manifestation of dementia driven by other factors, such as Alzheimer's disease and other neurodegenerative conditions. This comprehensive review of VaD focuses on molecular mechanisms and their consequences. We provided up-to-date knowledge about epidemiology, pathophysiological mechanisms, and current therapeutic approaches for VaD.
Collapse
Affiliation(s)
- Aseel Y Altahrawi
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, OH 43614, USA
| | - Antonisamy William James
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, OH 43614, USA
| | - Zahoor A Shah
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, OH 43614, USA
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, OH 43614, USA
| |
Collapse
|
7
|
Jasim MH, Saadoon Abbood R, Sanghvi G, Roopashree R, Uthirapathy S, Kashyap A, Sabarivani A, Ray S, Mustafa YF, Yasin HA. Flavonoids in the regulation of microglial-mediated neuroinflammation; focus on fisetin, rutin, and quercetin. Exp Cell Res 2025; 447:114537. [PMID: 40147710 DOI: 10.1016/j.yexcr.2025.114537] [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: 02/05/2025] [Revised: 03/22/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025]
Abstract
Neuroinflammation is a critical mechanism in central nervous system (CNS) inflammatory disorders, encompassing conditions such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), multiple sclerosis (MS), traumatic brain injury (TBI), encephalitis, spinal cord injury (SCI), and cerebral stroke. Neuroinflammation is characterized by increased blood vessel permeability, leukocyte infiltration, glial cell activation, and elevated production of inflammatory mediators, such as chemokines and cytokines. Microglia act as the resident macrophages of the central nervous system, serving as the principal defense mechanism in brain tissue. After CNS injury, microglia modify their morphology and downregulate genes that promote homeostatic functions. Despite comprehensive transcriptome analyses revealing specific gene modifications in "pathological" microglia, microglia's precise protective or harmful functions in neurological disorders remain insufficiently comprehended. Accumulating data suggests that the polarization of microglia into the M1 proinflammatory phenotype or the M2 antiinflammatory phenotype may serve as a sensible therapeutic strategy for neuroinflammation. Flavonoids, including rutin, fisetin, and quercetin, function as crucial chemical reservoirs with unique structures and diverse actions and are extensively used to modulate microglial polarization in treating neuroinflammation. This paper highlights the detrimental effects of neuroinflammation seen in neurological disorders such as stroke. Furthermore, we investigate their therapeutic benefits in alleviating neuroinflammation via the modulation of macrophage polarization.
Collapse
Affiliation(s)
- Mohannad Hamid Jasim
- Biology Department, College of Education, University of Fallujah, Fallujah, Iraq.
| | - Rosull Saadoon Abbood
- Medical Laboratory Techniques Department, College of Health and Medical Technology, University of Al-maarif, Anbar, Iraq.
| | - Gaurav Sanghvi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot, 360003, Gujarat, India.
| | - R Roopashree
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India.
| | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Erbil, Kurdistan Region, Iraq.
| | - Aditya Kashyap
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India.
| | - A Sabarivani
- Department of Biomedical, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India.
| | - Subhashree Ray
- Department of Biochemistry, IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, 751003, India.
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq.
| | - Hatif Abdulrazaq Yasin
- Department of Medical Laboratories Technology, Al-Nisour University College, Nisour Seq. Karkh, Baghdad, Iraq.
| |
Collapse
|
8
|
Müller L, Di Benedetto S. Neuroimmune crosstalk in chronic neuroinflammation: microglial interactions and immune modulation. Front Cell Neurosci 2025; 19:1575022. [PMID: 40260075 PMCID: PMC12009833 DOI: 10.3389/fncel.2025.1575022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 03/25/2025] [Indexed: 04/23/2025] Open
Abstract
Neuroinflammation is a fundamental feature of many chronic neurodegenerative diseases, where it contributes to disease onset, progression, and severity. This persistent inflammatory state arises from the activation of innate and adaptive immune responses within the central nervous system (CNS), orchestrated by a complex interplay of resident immune cells, infiltrating peripheral immune cells, and an array of molecular mediators such as cytokines, chemokines, and extracellular vesicles. Among CNS-resident cells, microglia play a central role, exhibiting a dynamic spectrum of phenotypes ranging from neuroprotective to neurotoxic. In chronic neurodegenerative diseases, sustained microglial activation often leads to the amplification of inflammatory cascades, reinforcing a pathogenic cycle of immune-mediated damage. Intercellular communication within the inflamed CNS is central to the persistence and progression of neuroinflammation. Microglia engage in extensive crosstalk with astrocytes, neurons, oligodendrocytes, and infiltrating immune cells, shaping both local and systemic inflammatory responses. These interactions influence key processes such as synaptic pruning, phagocytosis, blood-brain barrier integrity, and cytokine-mediated signaling. Understanding the mechanisms of cell-cell signaling in this context is critical for identifying therapeutic strategies to modulate the immune response and restore homeostasis. This review explores the key players in CNS neuroinflammation, with a focus on the role of microglia, the molecular pathways underlying intercellular communication, and potential therapeutic approaches to mitigate neuroinflammatory damage in chronic neurodegenerative diseases.
Collapse
Affiliation(s)
- Ludmila Müller
- Max Planck Institute for Human Development, Center for Lifespan Psychology, Berlin, Germany
| | | |
Collapse
|
9
|
Chen H, Li N, Liu N, Zhu H, Ma C, Ye Y, Shi X, Luo G, Dong X, Tan T, Wei X, Yin H. Photobiomodulation modulates mitochondrial energy metabolism and ameliorates neurological damage in an APP/PS1 mousmodel of Alzheimer's disease. Alzheimers Res Ther 2025; 17:72. [PMID: 40188044 PMCID: PMC11971757 DOI: 10.1186/s13195-025-01714-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: 11/28/2024] [Accepted: 03/10/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative disease. Amyloid β-protein (Aβ) is one of the key pathological features of AD, which is cytotoxic and can damage neurons, thereby causing cognitive dysfunction. Photobiomodulation (PBM) is a non-invasive physical therapy that induces changes in the intrinsic mechanisms of cells and tissues through low-power light exposure. Although PBM has been employed in the treatment of AD, the effect and precise mechanism of PBM on AD-induced neurological damage are still unclear. METHODS In vivo experiments, PBM (808 nm, 20 mW/cm2) was used to continuously interfere with APP/PS1 mice for 6 weeks, and then their cognitive function and AD pathological changes were evaluated. In vitro experiments, lipopolysaccharide (LPS) was used to induce microglia to model inflammation, and the effect of PBM treatment on microglia polarization status and phagocytic Aβ ability was evaluated. Hexokinase 2 (HK2) inhibitor 3-bromopyruvate (3BP) was used to study the effect of PBM treatment on mitochondrial energy metabolism in microglia. RESULTS PBM further ameliorates AD-induced cognitive impairment by alleviating neuroinflammation and neuronal apoptosis, thereby attenuating nerve damage. In addition, PBM can also reduce neuroinflammation by promoting microglial anti-inflammatory phenotypic polarization; Promotes Aβ clearance by enhancing the ability of microglia to engulf Aβ. Among them, PBM regulates microglial polarization and inhibits neuronal apoptosis, which may be related to its regulation of mitochondrial energy metabolism, promotion of oxidative phosphorylation, and inhibition of glycolysis. CONCLUSION PBM regulates neuroinflammatory response and inhibits neuronal apoptosis, thereby repairing Aβ-induced neuronal damage and cognitive dysfunction. Mitochondrial energy metabolism plays an important role in PBM in improving nerve injury in AD mice. This study provides theoretical support for the subsequent application of PBM in the treatment of AD.
Collapse
Affiliation(s)
- Hongli Chen
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Neuromodulation and Neurorepair, Integrative Regeneration Laboratory, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China.
- State Key Laboratory of Separation Membranes and Membrane Processes & Key Laboratory of Hollow Fiber Membrane Materials and Membrane Processes, Tianjin Key Laboratory of Optoelectronic Detection Technology and Systems, School of Life Sciences, Tiangong University, Tianjin, 300387, China.
- Cangzhou Institute of Tiangong University, Cangzhou, 061000, China.
| | - Na Li
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Neuromodulation and Neurorepair, Integrative Regeneration Laboratory, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
- State Key Laboratory of Separation Membranes and Membrane Processes & Key Laboratory of Hollow Fiber Membrane Materials and Membrane Processes, Tianjin Key Laboratory of Optoelectronic Detection Technology and Systems, School of Life Sciences, Tiangong University, Tianjin, 300387, China
| | - Na Liu
- State Key Laboratory of Separation Membranes and Membrane Processes & Key Laboratory of Hollow Fiber Membrane Materials and Membrane Processes, Tianjin Key Laboratory of Optoelectronic Detection Technology and Systems, School of Life Sciences, Tiangong University, Tianjin, 300387, China
- Cangzhou Institute of Tiangong University, Cangzhou, 061000, China
| | - Hongyu Zhu
- State Key Laboratory of Separation Membranes and Membrane Processes & Key Laboratory of Hollow Fiber Membrane Materials and Membrane Processes, Tianjin Key Laboratory of Optoelectronic Detection Technology and Systems, School of Life Sciences, Tiangong University, Tianjin, 300387, China
| | - Chunyan Ma
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Neuromodulation and Neurorepair, Integrative Regeneration Laboratory, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
- State Key Laboratory of Separation Membranes and Membrane Processes & Key Laboratory of Hollow Fiber Membrane Materials and Membrane Processes, Tianjin Key Laboratory of Optoelectronic Detection Technology and Systems, School of Life Sciences, Tiangong University, Tianjin, 300387, China
| | - Yutong Ye
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Neuromodulation and Neurorepair, Integrative Regeneration Laboratory, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
- State Key Laboratory of Separation Membranes and Membrane Processes & Key Laboratory of Hollow Fiber Membrane Materials and Membrane Processes, Tianjin Key Laboratory of Optoelectronic Detection Technology and Systems, School of Life Sciences, Tiangong University, Tianjin, 300387, China
| | - Xinyu Shi
- State Key Laboratory of Separation Membranes and Membrane Processes & Key Laboratory of Hollow Fiber Membrane Materials and Membrane Processes, Tianjin Key Laboratory of Optoelectronic Detection Technology and Systems, School of Life Sciences, Tiangong University, Tianjin, 300387, China
| | - Guoshuai Luo
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, 300222, China
| | - Xiaoxi Dong
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Neuromodulation and Neurorepair, Integrative Regeneration Laboratory, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| | - Tao Tan
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Zhejiang Provincial Clinical Research Center for Mental Disorders, The Affiliated Wenzhou Kangning Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. 325000, China
| | - Xunbin Wei
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Cancer Hospital & Institute, International Cancer Institute, Institute of Medical Technology, Peking University Health Science Center, Biomedical Engineering Department, Peking University, Beijing, 100142, China.
| | - Huijuan Yin
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Neuromodulation and Neurorepair, Integrative Regeneration Laboratory, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China.
| |
Collapse
|
10
|
Du Y, Zhang M, Chen M, Hu M, Zeng W, Cai X, Zhang W, Zhu J, Zhong M, Liu Y, Yang C. Adverse events of celecoxib associated with the central nervous system and cancer: a disproportionality analysis of the FDA adverse event reporting system. Expert Opin Drug Saf 2025:1-12. [PMID: 40159988 DOI: 10.1080/14740338.2025.2487140] [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: 10/10/2024] [Revised: 01/28/2025] [Accepted: 03/05/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND Celecoxib is now clinically recognized as a candidate for treating various neurological disorders and cancers. The recent emergence of some serious adverse reactions is concerning. RESEARCH DESIGN AND METHODS We carried out data mining on the FDA Adverse Event Reporting System (FAERS) for adverse events (AEs) with celecoxib as the main suspect drug and conducted a disproportionality analysis. RESULTS 111,155,092 AE reports were extracted from FAERS, and 32,841 AEs with celecoxib as the primary suspected drug were identified. Celecoxib AEs were predominantly reported in cardiac disorders (n = 9602) and nervous system disorders (n = 4045). Cerebrovascular accident (n = 3109, PRR = 3.24) ranked second in the number of reports and cerebrovascular disorder (n = 265, PRR = 5.06) ranked second in signal intensity, was described as rare in the instructions. Nine unexpected and serious AEs were discovered, such as Stevens-Johnson syndrome (n = 175, IC025 = 1.7), breast disease male (n = 4, IC025 = 1.54), and squamous cell carcinoma of the head and neck (n = 4, IC025 = 0.96). At 200 mg, celecoxib was more linked to musculoskeletal and connective AEs; At 400 mg, it was more linked to neurological and cardiovascular AEs. CONCLUSIONS Unexpected AEs of celecoxib in neurological diseases and cancer have been identified, offering valuable insights for monitoring and risk assessment in future clinical applications.
Collapse
Affiliation(s)
- Yikuan Du
- Central Laboratory, The Tenth Affiliated Hospital of Southern Medical University, Dongguan, China
| | - Mengting Zhang
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, China
| | - Mianhai Chen
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, China
| | - Mianda Hu
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, China
| | - Wenqi Zeng
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, China
| | - Xiaolin Cai
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, China
| | - Weichui Zhang
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, China
| | - Jinfeng Zhu
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, China
| | - Mingjun Zhong
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, China
| | - Yi Liu
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Chun Yang
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, China
| |
Collapse
|
11
|
Wang Y, Wu Z, Zheng Y, Wang H, Cheng B, Xia J. Unraveling the genetic underpinnings of mitochondrial traits and associated circulating inflammatory proteins in Alzheimer's disease: Mitochondrial HtrA2-T cell CD5 negative axis. J Alzheimers Dis 2025:13872877251329517. [PMID: 40170213 DOI: 10.1177/13872877251329517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2025]
Abstract
BackgroundPrevious studies with limited sample sizes have indicated a link between mitochondrial traits, inflammatory proteins, and Alzheimer's disease. The exact causality and their mediation relationships remain unclear.ObjectiveOur study aimed to delve into the genetic underpinnings of mitochondrial function and circulating inflammatory proteins in the pathogenesis of Alzheimer's disease.MethodsWe leveraged aggregated data from the largest genome-wide association study, including 69 mitochondrial traits, 91 circulating inflammatory proteins, and Alzheimer's disease. Bidirectional mendelian randomization (MR) analyses were performed to investigate their primary causal relationships. Thereafter a two-step MR mediation analysis was utilized to clarify the modulating effects of inflammatory proteins on mitochondria and Alzheimer's disease.ResultsOur study identified mitochondrial phenylalanine-tRNA ligase and 4-hydroxy-2-oxoglutarate aldolase as risk factors for Alzheimer's disease, and serine protease HtrA2 and carbonic anhydrase 5A as protective factors against Alzheimer's disease. Four inflammatory proteins (T-cell surface glycoprotein CD5, C-X-C motif chemokine 11, TGF-α, and TNF-related apoptosis-inducing ligand) played protective roles against Alzheimer's disease. Axin-1 and IL-6 increased the risk of Alzheimer's disease. Furthermore, T-cell surface glycoprotein CD5 was found to be a significant mediator between mitochondrial serine protease HTRA2 and Alzheimer's disease with the two-step MR method, accounting for 10.83% of the total effect.ConclusionsOur study emphasized mitochondrial HtrA2-T cell CD5 as a negative axis in Alzheimer's disease, offering novel perspectives on its etiology, pathogenesis, and treatment.
Collapse
Affiliation(s)
- Yixi Wang
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Zhuokai Wu
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Yiheng Zheng
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Haimeng Wang
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Bin Cheng
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Juan Xia
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
12
|
Radermacher J, Erhardt VKJ, Walzer O, Haas EC, Kuppler KN, Zügner JSR, Lauer AA, Hartmann T, Grimm HS, Grimm MOW. Influence of Ibuprofen on glycerophospholipids and sphingolipids in context of Alzheimer´s Disease. Biomed Pharmacother 2025; 185:117969. [PMID: 40073745 DOI: 10.1016/j.biopha.2025.117969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 02/21/2025] [Accepted: 03/05/2025] [Indexed: 03/14/2025] Open
Abstract
Alzheimer's disease (AD) is a multifactorial disorder associated with neuroinflammation, elevated oxidative stress, lipid alterations as well as amyloid-deposits and the formation of neurofibrillary tangles. Ibuprofen, a globally used analgesic, is discussed to influence disease progression due to its anti-inflammatory effect. However, changes in lipid-homeostasis induced by Ibuprofen have not yet been analyzed. Here we investigate the effect of Ibuprofen on lipid classes known to be associated with AD. Ibuprofen treatment leads to a significant increase in phosphatidylcholine, sphingomyelin and triacylglyceride (TAG) species whereas plasmalogens, which are highly susceptible for oxidation, were significantly decreased. The observed alterations in phosphatidylcholine and sphingomyelin levels in presence of Ibuprofen might counteract the reduced phosphatidylcholine- and sphingomyelin-levels found in AD brain tissue with potential positive aspects on synaptic plasticity and ceramide-induced apoptotic effects. On the other hand, Ibuprofen leads to elevated TAG-level resulting in the formation of lipid droplets which are associated with neuroinflammation. Reduction of plasmalogen-levels might accelerate decreased plasmalogen-levels found in AD brains. Treatment of Ibuprofen in terms of lipid-homeostasis reveals both potentially positive and negative changes relevant to AD. Therefore, understanding the influence of Ibuprofen on lipid-homeostasis may help to understand the heterogeneous results of studies treating AD with Ibuprofen.
Collapse
Affiliation(s)
| | | | - Oliver Walzer
- Experimental Neurology, Saarland University, Homburg, Saar 66424, Germany.
| | | | | | | | - Anna Andrea Lauer
- Experimental Neurology, Saarland University, Homburg, Saar 66424, Germany; Nutrition Therapy and Counseling, Campus Rheinland, SRH University of Applied Health Sciences, Leverkusen 51377, Germany.
| | - Tobias Hartmann
- Experimental Neurology, Saarland University, Homburg, Saar 66424, Germany; Deutsches Institut für Demenzprävention (DIDP), Saarland University, Homburg, Saar 66424, Germany.
| | - Heike Sabine Grimm
- Experimental Neurology, Saarland University, Homburg, Saar 66424, Germany; Nutrition Therapy and Counseling, Campus Rheinland, SRH University of Applied Health Sciences, Leverkusen 51377, Germany; Deutsches Institut für Demenzprävention (DIDP), Saarland University, Homburg, Saar 66424, Germany.
| | - Marcus Otto Walter Grimm
- Experimental Neurology, Saarland University, Homburg, Saar 66424, Germany; Nutrition Therapy and Counseling, Campus Rheinland, SRH University of Applied Health Sciences, Leverkusen 51377, Germany; Deutsches Institut für Demenzprävention (DIDP), Saarland University, Homburg, Saar 66424, Germany.
| |
Collapse
|
13
|
Yang C, Wu X, Jiang Z, Ru Y, Shen B, Li F, Cui J, Zhang C, Wang X, Yu W, Li Y, Huang Y, Kong A, Hao F, Xiao C, Wang Y, Gao Y. Evodiamine rescues lipopolysaccharide-induced cognitive impairment via C/EBP-β-COX2 axis-regulated neuroinflammation. Int J Biol Macromol 2025; 300:139597. [PMID: 39798734 DOI: 10.1016/j.ijbiomac.2025.139597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/02/2025] [Accepted: 01/06/2025] [Indexed: 01/15/2025]
Abstract
Neuroinflammation is a key driver of neurological disorders. Evodiamine (EVO), an alkaloid from the traditional Chinese herb Evodia rutaecarpa, possesses potent biological activities, notably anticancer and anti-inflammatory effects. This study investigates EVO's potential to attenuate LPS-induced neuroinflammation, focusing on identifying its therapeutic targets and mechanisms of action. EVO treatment significantly improved mitochondrial function and reduced oxidative stress in LPS-stimulated BV2 cells, while also lowering levels of pro-inflammatory factors (IL-6, NO, IL-1β) in brain organoids. In mice, EVO treatment alleviated behavioral abnormalities, especially in cognition and memory, and lowered hippocampal inflammation marker levels. To elucidate the critical mechanisms by which EVO exerts its anti-inflammatory effects, we analyzed LPS-induced inflammatory injury in BV2 cells and used transcriptomics to investigate whether EVO modulates the C/EBP-β signaling pathway. Further validation using si-C/EBP-β confirmed EVO's regulatory effect on the C/EBP-β-COX2 axis, showing that knockdown significantly reduced pro-inflammatory factor expression, thereby providing neuroprotection. Moreover, molecular docking and dynamics simulations confirmed a stable interaction between EVO and C/EBP-β, supporting its role in attenuating LPS-induced neuroinflammation. In summary, these findings suggest that EVO regulates inflammation-related pathways by targeting the C/EBP-β-COX2 axis, offering neuroprotective benefits and mitigating neuroinflammatory responses.
Collapse
Affiliation(s)
- Chunqi Yang
- College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China; Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Xiangjun Wu
- School of Pharmacy, Henan University, Kaifeng 475000, China; Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Ziyu Jiang
- Beijing Institute of Radiation Medicine, Beijing 100850, China; School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yi Ru
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Baoying Shen
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Fangyang Li
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Jialu Cui
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Cheng Zhang
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Xiaoqiang Wang
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Wenrun Yu
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Yina Li
- Beijing Institute of Radiation Medicine, Beijing 100850, China; School of Life Sciences, Hebei University, Baoding, Hebei 071002, China
| | - Ying Huang
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Ao Kong
- School of Pharmacy, Henan University, Kaifeng 475000, China; Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Feiran Hao
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Chengrong Xiao
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Yuguang Wang
- School of Pharmacy, Henan University, Kaifeng 475000, China; Beijing Institute of Radiation Medicine, Beijing 100850, China; School of Life Sciences, Hebei University, Baoding, Hebei 071002, China.
| | - Yue Gao
- College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China; Beijing Institute of Radiation Medicine, Beijing 100850, China; School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
14
|
Wang J, Wang DZ, Li BH, Yang S, Guo FQ, Zheng B, Wang JH. Elevated circulating cathepsin S levels are associated with cognitive decline and neurodegeneration in a cohort of patients reporting memory complaints. J Alzheimers Dis 2025; 104:1147-1153. [PMID: 40095668 DOI: 10.1177/13872877251322809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
BackgroundAs a member of the cysteine protease family, cathepsin S has been implicated in the pathogenesis of various diseases, including Alzheimer's disease (AD), primarily by promoting inflammation.ObjectiveCurrent evidence regarding the role of cathepsin S primarily comes from animal studies. This study aims to explore the clinical relevance of cathepsin S in AD.MethodsIn a cohort of older adults aged 60 or above with memory complaints, we examined baseline plasma levels of cathepsin S and assessed their association with cognitive decline and biomarkers of neurodegeneration during a 36-month follow-up.ResultsPlasma levels of cathepsin S were significantly higher in individuals experiencing longitudinal cognitive decline compared to those without cognitive decline. Furthermore, plasma levels of cathepsin S were associated with declines in Mini-Mental State Examination (MMSE) scores and increases in neurofilament light and pTau181 levels. Higher plasma cathepsin S levels were linked to an increased risk of longitudinal cognitive decline (decrease in MMSE scores of 3 or more), adjusting for age, sex, education, APOE genotype, alcohol consumption, smoking, and comorbidities.ConclusionsThis study provides additional evidence supporting the potential role of cathepsin S in the pathogenesis of AD from a clinical perspective.
Collapse
Affiliation(s)
- Jian Wang
- Department of Neurology, Ya'an People's Hospital, Ya'an, Sichuan Province, China
| | - Duo-Zi Wang
- Department of Neurology, the Affiliated Hospital of University of Electronic Science and Technology, Sichuan Provincial People's Hospital, Chengdu, Sichuan Province, China
| | - Bing-Hu Li
- Department of Neurology, the Affiliated Hospital of University of Electronic Science and Technology, Sichuan Provincial People's Hospital, Chengdu, Sichuan Province, China
| | - Shu Yang
- Department of Neurology, the Affiliated Hospital of University of Electronic Science and Technology, Sichuan Provincial People's Hospital, Chengdu, Sichuan Province, China
| | - Fu-Qiang Guo
- Department of Neurology, the Affiliated Hospital of University of Electronic Science and Technology, Sichuan Provincial People's Hospital, Chengdu, Sichuan Province, China
| | - Bo Zheng
- Department of Neurology, Ya'an People's Hospital, Ya'an, Sichuan Province, China
| | - Jian-Hong Wang
- Department of Neurology, the Affiliated Hospital of University of Electronic Science and Technology, Sichuan Provincial People's Hospital, Chengdu, Sichuan Province, China
| |
Collapse
|
15
|
Ors H, Alimogullari E, Aslan Erdem S, Elmazoglu Z, Ceylan AF. Rosmarinus officinalis Ethanolic Extracts Rescues BV-2 Cells via Modulating Inflammation and Redox Balance: Comparative Study With Carnosol and Carnosic Acid. Cell Biochem Funct 2025; 43:e70073. [PMID: 40219627 PMCID: PMC11992469 DOI: 10.1002/cbf.70073] [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: 06/06/2024] [Revised: 03/03/2025] [Accepted: 04/02/2025] [Indexed: 04/14/2025]
Abstract
Neuroinflammation generally refers to an inflammatory response within the central nervous system caused by various pathological insults, including infection, trauma, ischemia, and toxins. As the brain's sentinel immune cell, microglia are tasked as the first responders to infection or tissue injury and initiating an inflammatory response. The perennial shrub plant Rosmarinus officinalis L. was reported to possess anti-inflammatory, anticancer, anti-nociceptive, antidiabetic, neuroprotective, and antioxidative properties. The present study aimed to investigate the effects of Rosmarinus officinalis ethanolic extracts on the lipopolysaccharide (LPS)-induced neuroinflammation model of BV-2 cells in comparison to carnosol and carnosic acid, phenolic diterpenes of the plant. Ultrasound-assisted extraction was used to have ethanolic extract of the plant. LPS was used to induce inflammation in BV-2 cells. Tumor necrosis alpha (TNF-α), interleukin 1 beta (IL-1β) secretion, reactive oxygen species (ROS) production, GSH/GSSG ratio, protein carbonyl level, and caspase-3 activity were evaluated. Inflammation induced by LPS was reduced by the ethanolic extract. Both carnosol and carnosic acid decreased the TNF-α and IL-1β levels as well. The ethanolic extract reduced ROS production and protein carbonylation, and increased GSH/GSSG ratio more effectively compared to the effects of carnosol and carnosic acid. Results depicted that caspase-3 activity was reduced by the ethanolic extract and this effect was more pronounced compared to carnosol and carnosic acid. The present study indicates the ethanolic extract of Rosmarinus officinalis rescues BV-2 cells from apoptosis via alleviating inflammation and oxidative stress.
Collapse
Affiliation(s)
- Hatice Ors
- Faculty of Medicine, Department of Medical PharmacologyAnkara Yildirim Beyazit UniversityAnkaraTurkey
| | - Ebru Alimogullari
- Faculty of Medicine, Department of Histology and EmbryologyAnkara Yildirim Beyazit UniversityAnkaraTurkey
| | - Sinem Aslan Erdem
- Faculty of Pharmacy, Department of PharmacognosyAnkara UniversityAnkaraTurkey
| | - Zubeyir Elmazoglu
- Faculty of Pharmacy, Department of PharmacologyAnkara Medipol UniversityAnkaraTurkey
| | - Asli F. Ceylan
- Faculty of Medicine, Department of Medical PharmacologyAnkara Yildirim Beyazit UniversityAnkaraTurkey
| |
Collapse
|
16
|
de Souza AA, Goularte KCM, Piccoli RC, Custódio SV, de Mello JE, Victor MG, Domingues WB, de Souza LP, Dos Santos Gonçalves L, Campos VF, Cunico W, Oses JP, Stefanello FM, de Aguiar MSS, Spanevello RM. 3-(3-(diethylamino)propyl)-2-(4-(methylthio)phenyl)thiazolidin-4-one Attenuates Scopolamine-induced Cognitive Impairment in Rats: Insights Into Neuroprotective Effects. Mol Neurobiol 2025:10.1007/s12035-025-04887-5. [PMID: 40164887 DOI: 10.1007/s12035-025-04887-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 03/24/2025] [Indexed: 04/02/2025]
Abstract
Alzheimer's Disease (AD) is characterized by memory decline, dysregulation in cholinergic and purinergic signaling, neuroinflammation, and oxidative stress. Current treatments are limited, highlighting the need for new compounds to prevent or delay AD progression. Thiazolidinones have emerged as promising candidates due to their antioxidant, anti-inflammatory, and anticholinesterase properties. The aim of this study was to evaluate the effects of 3-(3-(diethylamino)propyl)-2-(4-(methylthio)phenyl)thiazolidin-4-one (DS27) in a rat model of scopolamine-induced memory deficits. Male rats were divided into groups: I - Control, II - Scopolamine (SCO) (1 mg/kg), III - SCO and DS27 (5 mg/kg), IV - SCO and DS27 (10 mg/kg), V - SCO and donepezil (5 mg/kg). The animals were treated orally with DS27 or donepezil for seven days. On the 8th day, they underwent the open field test and inhibitory avoidance training, followed by intraperitoneal administration SCO. Twenty-four hours later, an inhibitory avoidance test was conducted. Acetylcholinesterase (AChE) activity, oxidative stress, and inflammatory and purinergic parameters were analyzed in the cerebral cortex, hippocampus, cerebrospinal fluid, serum, lymphocytes, and liver. DS27 prevented memory deficits, alterations in AChE activity, and oxidative damage induced by SCO in brain structures. Additionally, DS27 prevented SCO-induced decrease in IL-10 levels, and increase in IL-6, and TNF-α expression in the cerebral cortex, and normalized ATP and ADP hydrolysis in cerebrospinal fluid and lymphocytes. DS27 did not induce oxidative stress in the liver or alter serum biochemical parameters. These findings suggest that DS27 has significant neuroprotective properties and could be a promising alternative for treating neurodegenerative diseases like AD.
Collapse
Affiliation(s)
- Anita Avila de Souza
- Graduate Program in Biochemistry and Bioprospection - Laboratory of Neurochemistry, Inflammation, and Cancer - Center the Chemical, Pharmaceutical, and Food Sciences, Federal University of Pelotas, Capão Do Leão Campus S/N, Pelotas-RS, CEP 96010‑900, Brazil
| | - Kelen Cristiane Machado Goularte
- Graduate Program in Biochemistry and Bioprospection - Laboratory of Neurochemistry, Inflammation, and Cancer - Center the Chemical, Pharmaceutical, and Food Sciences, Federal University of Pelotas, Capão Do Leão Campus S/N, Pelotas-RS, CEP 96010‑900, Brazil
| | - Raphaela Cassol Piccoli
- Graduate Program in Biochemistry and Bioprospection - Laboratory of Neurochemistry, Inflammation, and Cancer - Center the Chemical, Pharmaceutical, and Food Sciences, Federal University of Pelotas, Capão Do Leão Campus S/N, Pelotas-RS, CEP 96010‑900, Brazil
| | - Solange Vega Custódio
- Graduate Program in Biochemistry and Bioprospection - Laboratory of Neurochemistry, Inflammation, and Cancer - Center the Chemical, Pharmaceutical, and Food Sciences, Federal University of Pelotas, Capão Do Leão Campus S/N, Pelotas-RS, CEP 96010‑900, Brazil
| | - Julia Eisenhardt de Mello
- Graduate Program in Biochemistry and Bioprospection - Laboratory of Neurochemistry, Inflammation, and Cancer - Center the Chemical, Pharmaceutical, and Food Sciences, Federal University of Pelotas, Capão Do Leão Campus S/N, Pelotas-RS, CEP 96010‑900, Brazil
| | - Melinda Gomes Victor
- Graduate Program in Biochemistry and Bioprospecting - Laboratory of Chemistry Applied to Bioactive - Center the Chemical, Pharmaceutical and Food Sciences, Federal University of Pelotas, Capão Do Leão Campus S/N, Pelotas-RS, CEP 96010‑900, Brazil
| | - William Borges Domingues
- Graduate Program in Biotechnology - Laboratory of Structural Genomics - Technological Development Center, Federal University of Pelotas, Capão Do Leão Campus, S/N, Pelotas-RS, RS, CEP 96010‑900, Brazil
| | - Lucas Petitemberte de Souza
- Graduate Program in Biotechnology - Laboratory of Structural Genomics - Technological Development Center, Federal University of Pelotas, Capão Do Leão Campus, S/N, Pelotas-RS, RS, CEP 96010‑900, Brazil
| | - Laís Dos Santos Gonçalves
- Graduate Program in Biotechnology - Laboratory of Structural Genomics - Technological Development Center, Federal University of Pelotas, Capão Do Leão Campus, S/N, Pelotas-RS, RS, CEP 96010‑900, Brazil
| | - Vinicius Farias Campos
- Graduate Program in Biochemistry and Bioprospection - Laboratory of Neurochemistry, Inflammation, and Cancer - Center the Chemical, Pharmaceutical, and Food Sciences, Federal University of Pelotas, Capão Do Leão Campus S/N, Pelotas-RS, CEP 96010‑900, Brazil
- Graduate Program in Biotechnology - Laboratory of Structural Genomics - Technological Development Center, Federal University of Pelotas, Capão Do Leão Campus, S/N, Pelotas-RS, RS, CEP 96010‑900, Brazil
| | - Wilson Cunico
- Graduate Program in Biochemistry and Bioprospecting - Laboratory of Chemistry Applied to Bioactive - Center the Chemical, Pharmaceutical and Food Sciences, Federal University of Pelotas, Capão Do Leão Campus S/N, Pelotas-RS, CEP 96010‑900, Brazil
| | - Jean Pierre Oses
- Graduate Program in Biochemistry and Bioprospection - Laboratory of Neurochemistry, Inflammation, and Cancer - Center the Chemical, Pharmaceutical, and Food Sciences, Federal University of Pelotas, Capão Do Leão Campus S/N, Pelotas-RS, CEP 96010‑900, Brazil
| | - Francieli Moro Stefanello
- Graduate Program in Biochemistry and Bioprospection - Laboratory of Neurochemistry, Inflammation, and Cancer - Center the Chemical, Pharmaceutical, and Food Sciences, Federal University of Pelotas, Capão Do Leão Campus S/N, Pelotas-RS, CEP 96010‑900, Brazil
| | - Mayara Sandrielly Soares de Aguiar
- Department of Clinical Medicine, Faculty of Medicine - Laboratory of Neuroscience and Behavior - Drug Research and Development Center, Federal University of Ceará, Fortaleza-CE, CEP 60020-181, Brazil
| | - Roselia Maria Spanevello
- Graduate Program in Biochemistry and Bioprospection - Laboratory of Neurochemistry, Inflammation, and Cancer - Center the Chemical, Pharmaceutical, and Food Sciences, Federal University of Pelotas, Capão Do Leão Campus S/N, Pelotas-RS, CEP 96010‑900, Brazil.
| |
Collapse
|
17
|
Fan X, Diao W, Wang H, Yin X, Qian W. Interferon Regulatory Factors as a Potential Therapeutic Target for Neuroinflammation: A Focus on Alzheimer's Disease. Int J Mol Sci 2025; 26:2906. [PMID: 40243463 PMCID: PMC11988619 DOI: 10.3390/ijms26072906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 03/19/2025] [Accepted: 03/21/2025] [Indexed: 04/18/2025] Open
Abstract
Interferon Regulatory Factors (IRFs) are critical modulators of immune and inflammatory responses, yet their roles in Alzheimer's disease (AD) and other neurodegenerative disorders remain incompletely understood. While IRFs are recognized for their regulatory functions in neuroinflammation, microglial activation, and neuronal survival, their dual roles as both drivers of pathological inflammation and mediators of neuroprotective pathways underscore a sophisticated regulatory paradox in neurodegenerative disorders. This review aims to synthesize current evidence on IRF-mediated neuroinflammation in AD and related diseases, focusing on the multifaceted functions of key IRF family members, including IRF1, IRF3, and IRF7. We critically evaluate their divergent roles: IRF1 and IRF3, for instance, exacerbate neuroinflammatory cascades and amyloid-beta (Aβ) pathology in AD, whereas IRF7 may paradoxically suppress inflammation under specific conditions. Additionally, we explore IRF dysregulation in Parkinson's disease, multiple sclerosis, amyotrophic lateral sclerosis, and Huntington's disease, emphasizing shared and distinct mechanisms across neurodegenerative disorders. Restoring IRF balance through genetic manipulation, small-molecule inhibitors, or microbiome-derived modulators could attenuate neuroinflammation, enhance Aβ clearance, and protect neuronal integrity. Ultimately, this work provides a framework for future research to harness IRF signaling pathways in the development of precision therapies for AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
| | | | | | | | - Wei Qian
- Department of Biochemistry and Molecular Biology, School of Medicine, Nantong University, Nantong 226001, China; (X.F.); (W.D.); (H.W.); (X.Y.)
| |
Collapse
|
18
|
Dhapola R, Kumari S, Sharma P, Vellingiri B, HariKrishnaReddy D. Advancements in autophagy perturbations in Alzheimer's disease: Molecular aspects and therapeutics. Brain Res 2025; 1851:149494. [PMID: 39922409 DOI: 10.1016/j.brainres.2025.149494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/11/2024] [Accepted: 02/02/2025] [Indexed: 02/10/2025]
Abstract
Emerging evidences suggest that autophagy, a key cellular process responsible for degrading and recycling damaged organelles and proteins, plays a crucial role in maintaining neuronal health. Dysfunctional autophagy has been linked to the pathogenesis of Alzheimer's disease (AD), contributing to the accumulation of misfolded proteins and cellular debris. Molecular mechanisms underlying autophagy dysfunction in AD involve amyloid-beta (Aβ) and tau accumulation, neuroinflammation, mitochondrial dysfunction, oxidative stress and endoplasmic reticulum stress. Disrupted signaling pathways such as TRIB3, Nmnat and BAG3 that regulate key processes like autophagosome initiation, lysosome function, and protein homeostasis also play a crucial role in the pathogenesis. Restoration of autophagy by modulating these molecular and signaling pathways may be an effective therapeutic strategy for AD. Studies have found few drugs targeting autophagy dysregulation in AD. These drugs include metformin that has been found to modulate the expression of TRIB3 for autophagy regulation. Another drug, resveratrol has been reported to augment the activity of Nmnat thus, increases autophagy flux. BACE1 and mTOR inhibitors like arctigenin, nilvadipine and dapagliflozin were also found to restore autophagy. This study elaborates recent advances in signaling and molecular pathways and discusses current and emerging therapeutic interventions targeting autophagy dysfunction in AD.
Collapse
Affiliation(s)
- Rishika Dhapola
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda 151401 Punjab, India
| | - Sneha Kumari
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda 151401 Punjab, India
| | - Prajjwal Sharma
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda 151401 Punjab, India
| | - Balachandar Vellingiri
- Human Cytogenetics and Stem Cell Laboratory, Department of Zoology, School of Basic Sciences, Central University of Punjab 151401 Bathinda, Punjab, India.
| | - Dibbanti HariKrishnaReddy
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda 151401 Punjab, India.
| |
Collapse
|
19
|
Beltran-Velasco AI, Clemente-Suárez VJ. Impact of Peripheral Inflammation on Blood-Brain Barrier Dysfunction and Its Role in Neurodegenerative Diseases. Int J Mol Sci 2025; 26:2440. [PMID: 40141084 PMCID: PMC11942216 DOI: 10.3390/ijms26062440] [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: 01/27/2025] [Revised: 02/26/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
The blood-brain barrier (BBB) is essential for maintaining brain homeostasis by regulating molecular exchange between the systemic circulation and the central nervous system. However, its dysfunction, often driven by peripheral inflammatory processes, has been increasingly linked to the development and progression of neurodegenerative diseases such as Alzheimer's and Parkinson's. Emerging evidence suggests that the gut-brain axis plays a key role in BBB integrity, with intestinal dysbiosis and chronic inflammation contributing to barrier disruption through immune and metabolic pathways. Furthermore, the selective vulnerability of specific brain regions to BBB dysfunction appears to be influenced by regional differences in vascularization, metabolic activity, and permeability, making certain areas more susceptible to neurodegenerative processes. This review explored the molecular mechanisms linking peripheral inflammation, gut microbiota, and BBB dysfunction, emphasizing their role in neurodegeneration. A comprehensive literature review was conducted using Web of Science, PubMed, Scopus, Wiley, ScienceDirect, and Medline, covering publications from 2015 to 2025. The findings highlight a complex interplay between gut microbiota-derived metabolites, immune signaling, and BBB permeability, underscoring the need for targeted interventions such as microbiome modulation, anti-inflammatory therapies, and advanced drug delivery systems. The heterogeneity of the BBB across different brain regions necessitates the development of region-specific therapeutic strategies. Despite advancements, critical knowledge gaps persist regarding the precise mechanisms underlying BBB dysfunction. Future research should leverage cutting-edge methodologies such as single-cell transcriptomics and organ-on-chip models to translate preclinical findings into effective clinical applications. Addressing these challenges will be crucial for developing personalized therapeutic approaches to mitigate the impact of BBB dysfunction in neurodegenerative diseases.
Collapse
Affiliation(s)
- Ana Isabel Beltran-Velasco
- NBC Group, Psychology Department, School of Life and Nature Sciences, Nebrija University, 28248 Madrid, Spain;
| | - Vicente Javier Clemente-Suárez
- Faculty of Medicine, Health and Sports, Universidad Europea de Madrid, Villaviciosa de Odón, 28670 Madrid, Spain
- Grupo de Investigación en Cultura, Educación y Sociedad, Universidad de la Costa, Barranquilla 080002, Colombia
| |
Collapse
|
20
|
Jia X, Sun X, Wang K, Li M. DRGCL: Drug Repositioning via Semantic-Enriched Graph Contrastive Learning. IEEE J Biomed Health Inform 2025; 29:1656-1667. [PMID: 38437145 DOI: 10.1109/jbhi.2024.3372527] [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: 03/06/2024]
Abstract
Drug repositioning greatly reduces drug development costs and time by discovering new indications for existing drugs. With the development of technology and large-scale biological databases, computational drug repositioning has increasingly attracted remarkable attention, which can narrow down repositioning candidates. Recently, graph neural networks (GNNs) have been widely used and achieved promising results in drug repositioning. However, the existing GNNs based methods usually focus on modeling the complex drug-disease association graph, but ignore the semantic information on the graph, which may lead to a lack of consistency of global topology information and local semantic information for the learned features. To alleviate the above challenge, we propose a novel drug repositioning model based on graph contrastive learning, termed DRGCL. First, we treat the known drug-disease associations as the topology graph. Second, we select the top- similar neighbor from drug/disease similarity information to construct the semantic graph rather than use the traditional data augmentation strategy, thereby maximally retaining rich semantic information. Finally, we pull closer to embedding consistency of the different embedding spaces by graph contrastive learning to enhance the topology and semantic feature on the graph. We have evaluated DRGCL on four benchmark datasets and the experiment results show that the proposed DRGCL is superior to the state-of-the-art methods. Especially, the average result of DRGCL is 11.92% higher than that of the second-best method in terms of AUPRC. The case studies further demonstrate the reliability of DRGCL.
Collapse
|
21
|
Guan J, Wu P, Liu M, Jiang C, Meng X, Wu X, Lu M, Fan Y, Gan L. Egln3 expression in microglia enhances the neuroinflammatory responses in Alzheimer's disease. Brain Behav Immun 2025; 125:21-32. [PMID: 39701332 DOI: 10.1016/j.bbi.2024.12.022] [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: 08/06/2024] [Revised: 11/27/2024] [Accepted: 12/14/2024] [Indexed: 12/21/2024] Open
Abstract
Alzheimer's disease (AD), characterized by cognitive and behavioral abnormalities, is the most prevalent neurodegenerative disease worldwide. Neuroinflammation, which is induced by microglial activation, resulting in the expression of a multitude of inflammatory factors, is one of the principal characteristics of AD. Herein, we found that Egln3 is differentially expressed in microglia in the brains of AD mice. Egln3 is a member of the Egln family of proline hydroxylases, which regulates a variety of biological processes, including transcription, the cell cycle, and apoptosis, through hydroxylation, ubiquitylation, and participation in glycolysis. To further observe the effects of Egln3 on cognitive function, we utilized APP/PS1 mice as a pathological model of AD to conduct behavioral experiments and assess the expression levels of Aβ and inflammatory factors. The specific mechanisms by which Egln3 affects microglial activation were analyzed using in vitro experiments and transcriptome sequencing. The results of these analyses demonstrated that Egln3 is highly expressed in microglia in AD. Inhibition of Egln3 expression in the brains of APP/PS1 mice improves neuroinflammatory responses and cognitive function, indicating that a high expression of Egln3 promotes AD progression. Furthermore, our findings indicate that Egln3 could activate the MAPK pathway, which in turn contributes to the aggravation of neuroinflammation. Inhibition of the MAPK pathway results in attenuation of the pro-inflammatory state of microglia. Consequently, Egln3 may exacerbate neuroinflammation and promote AD progression via the MAPK pathway in microglia, making it a promising target for AD-related therapies.
Collapse
Affiliation(s)
- Jiaxin Guan
- Department of Geriatrics, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, Heilongjiang 150081, China
| | - Pengfei Wu
- Department of Neurosurgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China; Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu Medical University, Bengbu 233030, China
| | - Meiling Liu
- Department of Geriatrics, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, Heilongjiang 150081, China
| | - Chuanlu Jiang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Xiangqi Meng
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Xiaowei Wu
- Department of Geriatrics, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, Heilongjiang 150081, China
| | - Meijiao Lu
- Department of Geriatrics, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, Heilongjiang 150081, China
| | - Ying Fan
- Department of Geriatrics, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, Heilongjiang 150081, China
| | - Lu Gan
- Department of Geriatrics, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, Heilongjiang 150081, China.
| |
Collapse
|
22
|
Chen L, Zhao X, Sheng R, Lazarovici P, Zheng W. Artemisinin alleviates astrocyte overactivation and neuroinflammation by modulating the IRE1/NF-κB signaling pathway in in vitro and in vivo Alzheimer's disease models. Free Radic Biol Med 2025; 229:96-110. [PMID: 39826816 DOI: 10.1016/j.freeradbiomed.2025.01.027] [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: 08/28/2024] [Revised: 12/25/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
Recent studies have shown that neuroinflammation and heightened glial activity, particularly astrocyte overactivation, are associated with Alzheimer's disease (AD). Abnormal accumulation of amyloid-beta (Aβ) induces endoplasmic reticulum (ER) stress and activates astrocytes. Artemisinin (ART), a frontline anti-malarial drug, has been found to have neuroprotective properties. However, its impact on astrocytes remains unclear. In this study, we used Aβ1-42 induced astrocyte cultures and 3 × Tg-AD mice as in vitro and in vivo models, respectively, to investigate the effects of ART on AD related astrocyte overactivation and its underlying mechanisms. ART attenuated Aβ1-42-induced astrocyte activation, ER stress, and inflammatory responses in astrocyte cultures by inhibiting IRE1 phosphorylation and the NF-κB pathway, as evidenced by the overexpression of IRE1 WT and IRE1-K599A (kinase activity invalidated), along with application of activators and inhibitors related to ER stress. Furthermore, ART alleviated the detrimental effects and restored neurotrophic function of astrocytes on co-cultured neurons, preventing neuronal apoptosis during Aβ1-42 treatment. In 3 × Tg-AD mice, ART treatment improved cognitive function and reduced astrocyte overactivation, neuroinflammation, ER stress, and neuronal apoptosis. Moreover, ART attenuated the upregulation of IRE1/NF-κB pathway activity in AD mice. Astrocyte-specific overexpression of IRE1 via adeno-associated virus in AD mice reversed the ameliorating effects of ART. Our findings suggest that ART inhibits astrocyte overactivation and neuroinflammation in both in vitro and in vivo AD models by modulating the IRE1/NF-κB signaling pathway, thereby enhancing neuronal functions. This study underscores the therapeutic potential of ART in AD and highlights the significance of modulating the ER stress-inflammatory cycle and normalizing astrocyte-neuron communication.
Collapse
Affiliation(s)
- Lei Chen
- Pharmaceutical Science, Faculty of Health Sciences, University of Macau, Macau, Taipa, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, China
| | - Xia Zhao
- Pharmaceutical Science, Faculty of Health Sciences, University of Macau, Macau, Taipa, China
| | - Rui Sheng
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, China.
| | - Philip Lazarovici
- School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 9112002, Israel
| | - Wenhua Zheng
- Pharmaceutical Science, Faculty of Health Sciences, University of Macau, Macau, Taipa, China.
| |
Collapse
|
23
|
Redhwan A, Adnan M, Bakhsh HR, Alshammari N, Surti M, Parashar M, Patel M, Patel M, Manjegowda DS, Sharma S. Computational Identification and Functional Analysis of Potentially Pathogenic nsSNPs in the NLRP3 Gene Linked to Alzheimer's Disease. Cell Biochem Biophys 2025; 83:357-375. [PMID: 39167281 DOI: 10.1007/s12013-024-01465-9] [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: 07/25/2024] [Indexed: 08/23/2024]
Abstract
Single Nucleotide Polymorphisms (SNPs) are key in understanding complex diseases. Nonsynonymous single-nucleotide polymorphisms (nsSNPs) occur in protein-coding regions, potentially altering amino acid sequences, protein structure and function. Computational methods are vital for distinguishing deleterious nsSNPs from neutral ones. We investigated the role of NLRP3 gene in neuroinflammation associated with Alzheimer's disease (AD) pathogenesis. A total of 893 missense (nsSNPs) were obtained from the dbSNP database and subjected to rigorous filtering using bioinformatics tools like SIFT, Align GVGD, PolyPhen-2, and PANTHER to identify potentially damaging variants. Of these, 18 nsSNPs were consistently predicted to have deleterious effects across all tools. Notably, 16 of these variants exhibited reduced protein stability, while only 4 were predicted to be buried within the protein structure. Among the identified nsSNPs, rs180177442 (R262L and R262P), rs201875324 (T659I), and rs139814109 (T897M) were classified as high-risk variants due to their significant deleterious impact, probable damaging effects, and association with decreased protein stability. Molecular docking and simulation analyses were conducted utilizing Memantine, a standard drug utilized in AD treatment, to investigate potential interactions with the altered protein structures. Additional clinical and genetic investigations are necessary to elucidate the underlying mechanisms that link NLRP3 polymorphisms with the initiation of AD.
Collapse
Affiliation(s)
- Alya Redhwan
- Department of Health, College of Health and Rehabilitation Sciences, Princess Nourah bint Abdulrahman University, Riyadh, 11671, Saudi Arabia
| | - Mohd Adnan
- Department of Biology, College of Science, University of Ha'il, Ha'il, Saudi Arabia
| | - Hadeel R Bakhsh
- Department of Rehabilitation Sciences, College of Health and Rehabilitation Sciences, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Nawaf Alshammari
- Department of Health, College of Health and Rehabilitation Sciences, Princess Nourah bint Abdulrahman University, Riyadh, 11671, Saudi Arabia
| | - Malvi Surti
- 4Research and Development Cell, Department of Biotechnology, Parul Institute of Applied Sciences, Parul University, Vadodara, Gujarat, India
| | - Mansi Parashar
- 4Research and Development Cell, Department of Biotechnology, Parul Institute of Applied Sciences, Parul University, Vadodara, Gujarat, India
| | - Mirav Patel
- 4Research and Development Cell, Department of Biotechnology, Parul Institute of Applied Sciences, Parul University, Vadodara, Gujarat, India
| | - Mitesh Patel
- 4Research and Development Cell, Department of Biotechnology, Parul Institute of Applied Sciences, Parul University, Vadodara, Gujarat, India
| | - Dinesh Sosalagere Manjegowda
- 4Department of Human Genetics, School of Basic and Applied Sciences, Dayananda Sagar University, Bangalore, 560078, India
| | - Sameer Sharma
- Department of Bioinformatics, BioNome, Bangalore, India.
| |
Collapse
|
24
|
Zhang X, Wang SJ, Wan SC, Li X, Chen G. Ozone: complicated effects in central nervous system diseases. Med Gas Res 2025; 15:44-57. [PMID: 39436168 PMCID: PMC11515058 DOI: 10.4103/mgr.medgasres-d-24-00005] [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: 01/15/2024] [Revised: 06/20/2024] [Accepted: 08/25/2024] [Indexed: 10/23/2024] Open
Abstract
Oxidative stress is closely related to various diseases. Ozone can produce redox reactions through its unique response. As a source of the oxidative stress response, the strong oxidizing nature of ozone can cause severe damage to the body. On the other hand, low ozone concentrations can activate various mechanisms to combat oxidative stress and achieve therapeutic effects. Some animal experiments and clinical studies have revealed the potential medical value of ozone, indicating that ozone is not just a toxic gas. By reviewing the mechanism of ozone and its therapeutic value in treating central nervous system diseases (especially ischemic stroke and Alzheimer's disease) and the toxic effects of ozone, we find that ozone inhalation and a lack of antioxidants or excessive exposure lead to harmful impacts. However, with adequate antioxidants, ozone can transmit oxidative stress signals, reduce inflammation, reduce amyloid β peptide levels, and improve tissue oxygenation. Similar mechanisms to those of possible new drugs for treating ischemic stroke and Alzheimer's disease indicate the potential of ozone. Nevertheless, limited research has restricted the application of ozone. More studies are needed to reveal the exact dose-effect relationship and healing effect of ozone.
Collapse
Affiliation(s)
- Xu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Shi-Jun Wang
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Si-Cen Wan
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Xiang Li
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Gang Chen
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
25
|
Huang Z, Hao M, Shi N, Wang X, Yuan L, Yuan H, Wang X. Porphyromonas gingivalis: a potential trigger of neurodegenerative disease. Front Immunol 2025; 16:1482033. [PMID: 40028317 PMCID: PMC11867964 DOI: 10.3389/fimmu.2025.1482033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 01/27/2025] [Indexed: 03/05/2025] Open
Abstract
Porphyromonas gingivalis (P. gingivalis) is a gram-negative bacterium and the main causative agent of periodontitis, a disease closely associated with the development of periodontal disease. The progression of periodontitis, a chronic infectious disease, is intricately linked to the inflammatory immune response. Inflammatory cytokines act on periodontal tissues via immunomodulation, resulting in the destruction of the periodontal tissue. Recent studies have established connections between periodontitis and various systemic diseases, including cardiovascular diseases, tumors, and neurodegenerative diseases. Neurodegenerative diseases are neurological disorders caused by immune system dysfunction, including Alzheimer's and Parkinson's diseases. One of the main characteristics of neurodegenerative diseases is an impaired inflammatory response, which mediates neuroinflammation through microglial activation. Some studies have shown an association between periodontitis and neurodegenerative diseases, with P. gingivalis as the primary culprit. P. gingivalis can cross the blood-brain barrier (BBB) or mediate neuroinflammation and injury through a variety of pathways, including the gut-brain axis, thereby affecting neuronal growth and survival and participating in the onset and progression of neurodegenerative diseases. However, comprehensive and systematic summaries of studies on the infectious origin of neurodegenerative diseases are lacking. This article reviews and summarizes the relationship between P. gingivalis and neurodegenerative diseases and its possible regulatory mechanisms. This review offers new perspectives into the understanding of neurodegenerative disease development and highlights innovative approaches for investigating and developing tailored medications for treating neurodegenerative conditions, particularly from the viewpoint of their association with P. gingivalis.
Collapse
Affiliation(s)
- Ziyan Huang
- Department of Stomatology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Miao Hao
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Naixu Shi
- Department of Stomatology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Xinyu Wang
- Department of Stomatology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Lin Yuan
- Department of Stomatology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Haotian Yuan
- Department of Stomatology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Xiaofeng Wang
- Department of Stomatology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
26
|
Mangoni AA, Zinellu A. A systematic review and meta-analysis of pteridines in mild cognitive impairment and Alzheimer's disease. BMC Geriatr 2025; 25:94. [PMID: 39948480 PMCID: PMC11823259 DOI: 10.1186/s12877-025-05760-9] [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: 09/23/2024] [Accepted: 02/05/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Alterations in specific pteridine metabolites, particularly neopterin, biopterin, and tetrahydrobiopterin have been reported in mild cognitive impairment, Alzheimer's disease, and other types of dementia. However, the available evidence regarding such alterations has not been comprehensively and critically appraised. METHODS We systematically reviewed studies reporting the concentrations of biopterin, tetrahydrobiopterin, and neopterin in different biological fluids in patients with mild cognitive impairment, Alzheimer's disease or other types of dementia, and healthy controls. Electronic databases were searched from inception to 29 February 2024. RESULTS Overall, there were no significant differences in plasma/serum concentrations of neopterin between patients with mild cognitive impairment, Alzheimer's disease, or other types of dementia, when grouped together, and healthy controls after adjusting for publication bias (11 studies, standard mean difference, SMD = 0.20, 95% CI -0.02 to 0.41, p = 0.076). In meta-regression and subgroup analysis, the effect size was significantly associated with age, number of participants, study continent, presence of mild cognitive impairment, presence of Alzheimer's disease, analytical method, and assessment of serum vs. plasma. One study reported higher urine neopterin in patients with Alzheimer's disease vs. controls whereas another study reported non-significant between-group differences in cerebrospinal neopterin. The cerebrospinal fluid concentrations of biopterin were significantly lower in patients with Alzheimer's disease vs. controls (two studies, SMD = -0.75, 95% CI -1.23 to -0.27, p = 0.002; I2 = 0.0%, p = 0.46). One study showed non-significant between-group differences in plasma biopterin whereas another study showed higher concentrations of urine biopterin in patients with Alzheimer's disease. Our search did not identify studies investigating tetrahydrobiopterin. CONCLUSION Our study showed no significant differences in circulating neopterin between patients with mild cognitive impairment, Alzheimer's disease, or other types of dementia, when grouped together, and healthy controls. The significant associations observed between the effect size and mild cognitive impairment and Alzheimer's disease in subgroup analysis warrant further investigation. (PROSPERO registration number: CRD42024523478).
Collapse
Affiliation(s)
- Arduino A Mangoni
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, Australia.
- Department of Clinical Pharmacology, Flinders Medical Centre, Southern Adelaide Local Health Network, Bedford ParkAdelaide, SA, 5042, Australia.
| | - Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| |
Collapse
|
27
|
de Lima BRF, de Siqueira Patriota LL, de Oliveira Marinho A, da Costa JA, Ribeiro BG, de Souza Santos VB, Napoleão DC, Cavalcanti JVFL, Vieira LD, Pereira MC, de Melo Rego MJB, Pitta MGDR, Napoleão TH, Paiva PMG, da Rosa MM. Subacute symptoms of depression and anxiety in stress-exposed mice: Role of Schinus terebinthifolia Raddi leaf lectin (SteLL). JOURNAL OF ETHNOPHARMACOLOGY 2025; 341:119343. [PMID: 39805478 DOI: 10.1016/j.jep.2025.119343] [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: 10/02/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 01/16/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Anxiety and depression are leading causes of disability worldwide, often exacerbated by chronic stress. Schinus terebinthifolia Raddi. has been used in traditional medicine for several purposes. Among these, the use of bark-and-leaf tea and leaf decoction to treat depression has been reported. Previous studies showed that the S. terebinthifolia leaf lectin (SteLL) can ameliorate anxiety and depression symptoms in mice. AIM OF THE STUDY To investigate SteLL as a compound from S. terebinthifolia leaf able to alleviate symptoms of depression and anxiety in an unpredictable chronic mild stress (UCMS) animal model. METHODS Mice were subjected to four-week UCMS and then treated with SteLL at 2 and 4 mg/kg (i.p.) or with fluoxetine at 10 mg/kg i.p. (positive control) for 21 days. Behavioral assessments were conducted using the open field test, elevated plus maze, tail suspension test, and sucrose preference test. Serum corticosterone and inflammatory markers (cytokines) levels were determined. The levels of cytokine, oxidative stress indicators and monoamines in brain homogenates were also measured to understand the biochemical changes induced by SteLL treatment. RESULTS SteLL treatment at both doses significantly (p < 0.05) alleviated the stress-induced behavior in mice, reducing the anxiety and depression signals in all tests. SteLL administration increased the brain levels of monoamines noradrenaline and serotonin in comparison with UCMS control mice that received only vehicle. SteLL reduced superoxide production, lipid peroxidation and improved reduced glutathione (GSH) levels in the brain. The lectin also increased serum and brain levels of anti-inflammatory cytokine IL-4, while reducing levels of pro-inflammatory cytokines. Serum corticosterone levels were not decreased by lectin treatment. CONCLUSION Our findings highlight SteLL as a neuromodulatory agent from S. terebinthifolia leaves effective in subacute and stress-induced anxiety and depression through modulation of monoaminergic, oxidative stress, and inflammatory pathways. The data shows the potential of this lectin as a therapeutic agent for stress-related neuropsychological disorders.
Collapse
Affiliation(s)
| | | | | | - Jainaldo Alves da Costa
- Biochemistry Department, Center of Biosciences, Universidade Federal de Pernambuco, Recife, Brazil.
| | - Beatriz Galdino Ribeiro
- Department of Chemical Engineering, Center for Technology and Geosciences, Universidade Federal de Pernambuco, Recife, Brazil.
| | | | - Daniella Carla Napoleão
- Department of Chemical Engineering, Center for Technology and Geosciences, Universidade Federal de Pernambuco, Recife, Brazil.
| | | | - Leucio Duarte Vieira
- Department of Physiology and Pharmacology, Center for Biosciences, Universidade Federal de Pernambuco, Recife, Brazil.
| | - Michelly Cristiny Pereira
- Department of Physiology and Pharmacology, Center for Biosciences, Universidade Federal de Pernambuco, Recife, Brazil; Center for Therapeutic Innovation Suely Galdino (NUPIT-SG), Universidade Federal de Pernambuco, Recife, Brazil.
| | - Moacyr Jesus Barreto de Melo Rego
- Biochemistry Department, Center of Biosciences, Universidade Federal de Pernambuco, Recife, Brazil; Center for Therapeutic Innovation Suely Galdino (NUPIT-SG), Universidade Federal de Pernambuco, Recife, Brazil.
| | - Maira Galdino da Rocha Pitta
- Biochemistry Department, Center of Biosciences, Universidade Federal de Pernambuco, Recife, Brazil; Center for Therapeutic Innovation Suely Galdino (NUPIT-SG), Universidade Federal de Pernambuco, Recife, Brazil.
| | - Thiago Henrique Napoleão
- Biochemistry Department, Center of Biosciences, Universidade Federal de Pernambuco, Recife, Brazil.
| | | | - Michelle Melgarejo da Rosa
- Biochemistry Department, Center of Biosciences, Universidade Federal de Pernambuco, Recife, Brazil; Center for Therapeutic Innovation Suely Galdino (NUPIT-SG), Universidade Federal de Pernambuco, Recife, Brazil.
| |
Collapse
|
28
|
Kaspute G, Ramanavicius A, Prentice U. Natural drug delivery systems for the treatment of neurodegenerative diseases. Mol Biol Rep 2025; 52:217. [PMID: 39928236 DOI: 10.1007/s11033-025-10286-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 01/22/2025] [Indexed: 02/11/2025]
Abstract
Today, herbal drugs are prominent in the pharmaceutical industry due to their well-known therapeutic and side effects. Plant-based compounds often face limitations such as poor solubility, low bioavailability, and instability in physiological environments, restricting their therapeutic efficacy and delivery. Nanotechnology-based solutions, including nanoparticle formulations and advanced delivery systems like liposomes and transfersomes, address these issues by enhancing solubility, stability, bioavailability, and targeted delivery, thereby optimizing the therapeutic potential of phytoactive compounds. Neuroinflammation can be a cause of neurodegenerative disorders such as Alzheimer's and Parkinson's diseases, or amyotrophic lateral sclerosis. Consequently, there is a need for the optimal delivery of a pharmacological anti-inflammatory agents to the CNS. Thus, the non-invasive administration of a stable compound at a therapeutic concentration is needed to assure molecule crossing through the blood-brain barrier. Natural resources have more structural diversity and novelty than synthetic compounds, e.g. plant-derived drug products have higher molecular weights, incorporate more oxygen atoms, and are more complex. As a result, plant-derived products have unique features which can be used to effectively modulate neuroinflammation. Therefore, this review aims to identify herbal molecules capable of targeting neuroinflammation and present novel strategies for their efficient delivery.
Collapse
Affiliation(s)
- Greta Kaspute
- Department of Nanotechnology, State Research Institute Center for Physical Sciences and Technology (FTMC), Sauletekio Av. 3, 10257, Vilnius, Lithuania
| | - Arunas Ramanavicius
- Department of Nanotechnology, State Research Institute Center for Physical Sciences and Technology (FTMC), Sauletekio Av. 3, 10257, Vilnius, Lithuania.
- Department of Physical Chemistry, Faculty of Chemistry and Geosciences, Institute of Chemistry, Vilnius University, Naugarduko St. 24, 03225, Vilnius, Lithuania.
| | - Urte Prentice
- Department of Nanotechnology, State Research Institute Center for Physical Sciences and Technology (FTMC), Sauletekio Av. 3, 10257, Vilnius, Lithuania.
- Department of Physical Chemistry, Faculty of Chemistry and Geosciences, Institute of Chemistry, Vilnius University, Naugarduko St. 24, 03225, Vilnius, Lithuania.
| |
Collapse
|
29
|
Meng T, Zhang Y, Ye Y, Li H, He Y. Bioinformatics insights into mitochondrial and immune gene regulation in Alzheimer's disease. Eur J Med Res 2025; 30:89. [PMID: 39920860 PMCID: PMC11806906 DOI: 10.1186/s40001-025-02297-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: 03/27/2024] [Accepted: 01/13/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND There is growing evidence that the pathogenesis of Alzheimer's disease is closely linked to the resident innate immune cells of the central nervous system, including microglia and astrocytes. Mitochondrial dysfunction in microglia has also been reported to play an essential role in the pathogenesis of AD and other neurological diseases. Therefore, finding the mitochondrial and immune-related gene (MIRG) signatures in AD can be significant in diagnosing and treating AD. METHODS In this study, the intersection of the differentially expressed genes (DEGs) from the GSE109887 cohort, immune-related genes (IRGs) obtained from WGCNA analysis, and mitochondria-related genes (MRGs) was taken to identify mitochondria-immune-related genes (MIRGs). Then, using machine learning algorithms, biomarkers with good diagnostic value were selected, and a nomogram was constructed. Subsequently, we further analyzed the signaling pathways and potential biological mechanisms of the biomarkers through gene set enrichment analysis, prediction of transcription factors (TFs), miRNAs, and drug prediction. RESULTS Using machine learning algorithms, five biomarkers (TSPO, HIGD1A, NDUFAB1, NT5DC3, and MRPS30) were successfully identified, and a nomogram model with strong diagnostic ability and accuracy (AUC > 0.9) was constructed. In addition, single-gene enrichment analysis revealed that NDUFAB1 was significantly enriched in pathways associated with diseases, such as Alzheimer's and Parkinson's, providing valuable insights for future clinical research on Alzheimer's in the context of mitochondrial-immune interactions. Interestingly, brain tissue pathology showed neuronal atrophy and demyelination in AD mice, along with a reduction in Nissl bodies. Furthermore, the escape latency of AD mice was significantly longer than that of the control group. After platform removal, there was a notable increase in the path complexity and time required to reach the target quadrant, suggesting a reduction in spatial memory capacity in AD mice. Moreover, qRT-PCR validation confirmed that the mRNA expression of the five biomarkers was consistent with bioinformatics results. In AD mice, TSPO expression was increased, while HIGD1A, NDUFAB1, NT5DC3, and MRPS30 expressions were decreased. However, peripheral blood samples did not show expression of HIGD1A or MRPS30. These findings provide new insights for research on Alzheimer's disease in the context of mitochondrial-immune interactions, further exploring the pathogenesis of Alzheimer's disease and offering new perspectives for the clinical development of novel drugs. CONCLUSIONS Five mitochondrial and immune biomarkers, i.e., TSPO, HIGD1A, NDUFAB1, NT5DC3, and MRPS30, with diagnostic value in Alzheimer's disease, were screened by machine-learning algorithmic models, which will be a guide for future clinical research of Alzheimer's disease in the mitochondria-immunity-related direction.
Collapse
Affiliation(s)
- Tian Meng
- Yunnan Yunke Institute of Biotechnology, No. 871 Longquan Rd, Kunming, 650500, China
| | - Yazhou Zhang
- Department of Geriatrics, The Second People's Hospital of Kunming, No. 338Guangming Rd, Kunming, 650233, Yunnan, China
| | - Yuan Ye
- Department of Geriatrics, The Second People's Hospital of Kunming, No. 338Guangming Rd, Kunming, 650233, Yunnan, China
| | - Hui Li
- Yunnan Labreal Biotechnology Co., LTD, No. 871 Longquan Rd, Kunming, 650500, China
| | - Yongsheng He
- Yunnan Yunke Institute of Biotechnology, No. 871 Longquan Rd, Kunming, 650500, China.
- Yunnan Labreal Biotechnology Co., LTD, No. 871 Longquan Rd, Kunming, 650500, China.
| |
Collapse
|
30
|
Linzhu, Zhang J, Fan W, Su C, Jin Z. Influence of immune cells and inflammatory factors on Alzheimer's disease axis: evidence from mediation Mendelian randomization study. BMC Neurol 2025; 25:49. [PMID: 39910474 PMCID: PMC11796147 DOI: 10.1186/s12883-025-04057-z] [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: 03/27/2024] [Accepted: 01/28/2025] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is one of the most common forms of dementia in the elderly, characterized by progressive neurodegeneration. While the exact etiology of AD remains unclear, immune inflammation is known to play a significant role in the disease. METHODS This study utilized a two-sample Mendelian randomization (MR) approach to assess the causal relationship between different types of immune cells and AD, while considering inflammatory factors as intermediate variables. Data were collected from three sources: immune cell data (731 phenotypes), inflammatory factors (48 cytokines from 8,293 individuals), and AD data (35,274 cases, 59,163 controls). Multiple MR methods were employed to minimize bias, and detailed descriptions of instrumental variable selection and statistical methods were provided. RESULTS The study findings suggest potential causal relationships between six different types of immune cells and AD, as well as causal relationships between 13 immune cells and inflammatory factors. Additionally, two statistically significant inflammatory factors were found to have potential causal relationships with AD. Specifically, immune cells CD33-HLA DR + and CD45 on CD33-HLA DR + may further influence AD by regulating Interleukin-2 levels. CONCLUSION This study provides valuable insights into the immunoinflammatory pathogenesis of AD and offers partial guidance for the development of relevant interventions, thereby contributing beneficial information for the prevention and treatment of related diseases.
Collapse
Affiliation(s)
- Linzhu
- Department of Traditional Chinese Medicine, the Second Hospital of Shandong University, Jinan, 250014, China
| | - Jianxin Zhang
- Department of Traditional Chinese Medicine, the Second Hospital of Shandong University, Jinan, 250014, China
| | - Wenhui Fan
- Department of Traditional Chinese Medicine, the Second Hospital of Shandong University, Jinan, 250014, China
| | - Chen Su
- Department of Traditional Chinese Medicine, the Second Hospital of Shandong University, Jinan, 250014, China
| | - Zhi Jin
- Department of Traditional Chinese Medicine, the Second Hospital of Shandong University, Jinan, 250014, China.
| |
Collapse
|
31
|
Yang X, Wang J, Jia X, Yang Y, Fang Y, Ying X, Li H, Zhang M, Wei J, Pan Y. Microglial polarization in Alzheimer's disease: Mechanisms, implications, and therapeutic opportunities. J Alzheimers Dis 2025:13872877241313223. [PMID: 39894910 DOI: 10.1177/13872877241313223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by the accumulation of amyloid-β plaques, neurofibrillary tangles, and chronic neuroinflammation. Microglial cells, the resident immune cells in the central nervous system, play a crucial role in the pathogenesis of AD. Microglia can undergo polarization, shifting between pro-inflammatory (M1) and anti-inflammatory (M2) phenotypes in response to different stimuli. Dysregulation of microglial polarization towards the pro-inflammatory phenotype leads to the release of inflammatory cytokines, oxidative stress, and synaptic dysfunction. These processes contribute to neuronal damage and cognitive decline in AD. However, several challenges remain in this field. The complex molecular mechanisms governing microglial polarization in AD need to be further elucidated. In this review, we discuss the mechanisms underlying microglial polarization in AD and its implications in disease progression.
Collapse
Affiliation(s)
- Xinmao Yang
- Department of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Jie Wang
- Department of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Xiaotao Jia
- Department of Neurology, The Affifiliated Xi'an Central Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi, PR China
| | - Yaqian Yang
- Department of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Yan Fang
- Department of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Xiaoping Ying
- Department of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Hong Li
- Department of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Meiqian Zhang
- Department of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Jing Wei
- Department of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Yanfang Pan
- Department of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
| |
Collapse
|
32
|
Liu Y, Wu L, Peng W, Mao X. Glial polarization in neurological diseases: Molecular mechanisms and therapeutic opportunities. Ageing Res Rev 2025; 104:102638. [PMID: 39672208 DOI: 10.1016/j.arr.2024.102638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 12/05/2024] [Accepted: 12/07/2024] [Indexed: 12/15/2024]
Abstract
Glial cell polarization plays a pivotal role in various neurological disorders. In response to distinct stimuli, glial cells undergo polarization to either mitigate neurotoxicity or facilitate neural repair following injury, underscoring the importance of glial phenotypic polarization in modulating central nervous system function. This review presents an overview of glial cell polarization, focusing on astrocytes and microglia. It explores the involvement of glial polarization in neurological diseases such as Alzheimer's disease, Parkinson's disease, stroke, epilepsy, traumatic brain injury, amyotrophic lateral sclerosis, multiple sclerosis and meningoencephalitis. Specifically, it emphasizes the role of glial cell polarization in disease pathogenesis through mechanisms including neuroinflammation, neurodegeneration, calcium signaling dysregulation, synaptic dysfunction and immune response. Additionally, it summarizes various therapeutic strategies including pharmacological treatments, dietary supplements and cell-based therapies, aimed at modulating glial cell polarization to ameliorate brain dysfunction. Future research focused on the spatio-temporal manipulation of glial polarization holds promise for advancing precision diagnosis and treatment of neurological diseases.
Collapse
Affiliation(s)
- Yuqing Liu
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China; National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Lei Wu
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha 410008, China; Institute of Clinical Pharmacology and Engineering Research Center of Applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha 410078, China
| | - Weijun Peng
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China; National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha 410011, China.
| | - Xiaoyuan Mao
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha 410008, China; Institute of Clinical Pharmacology and Engineering Research Center of Applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha 410078, China.
| |
Collapse
|
33
|
Wang P, Dong YL, Li SS, Jin Y, Sun WL, Zhao BS, Li QB, Chen X. Integrating network pharmacology and component analysis to investigate the potential mechanisms of Sheng-Hui-Yi-Zhi decoction in the treatment of Alzheimer's disease. J Alzheimers Dis 2025; 103:666-678. [PMID: 39772836 DOI: 10.1177/13872877241305744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by cognitive impairment. OBJECTIVE To elucidate the potential mechanisms of Sheng-Hui-Yi-Zhi (SHYZ) for the treatment of AD and explore the effective substances of SHYZ. METHODS Liquid chromatography-mass spectrometry (LC-MS) was used to identify the active components of SHYZ. Network pharmacology was employed to predict the potential targets and pathways of SHYZ in the treatment of AD. SAMP8 mice were used as a model for AD and were treated with SHYZ. The Morris water maze was utilized to assess the learning and memory capabilities of mice. Additionally, the levels of TNF-α, IL-1β, and IL-6 in the brain hippocampus of mice were quantified using ELISA. The protein expression of PI3 K/p-PI3 K, AKT/p-AKT, MAPK38/p-MAPK38, and NFκB p65/p-NFκB p65 in the hippocampus was analyzed using Western blotting. Additionally, qRT-PCR was employed to assess the gene expressions of TNF-α, IL-1β, and IL-6 in the hippocampus. RESULT The network pharmacological prediction results showed that the treatment of AD with SHYZ was closely related to the inhibition of inflammatory response. Behavioral experiments revealed that SHYZ significantly reduced the time taken to escape, increased the number of times the platform was crossed, and prolonged the residence time in the target quadrant. Meanwhile, SHYZ treatment suppressed the expression of Aβ1-42 protein and inflammatory factors. SHYZ significantly inhibited the expression of proteins of PI3 K, AKT, MAPK p38, and NF-κB p65. CONCLUSIONS SHYZ has been shown to effectively ameliorate learning and memory impairment in SAMP8 AD mice by inhibiting the expression of Aβ1-42 and reducing the increase of inflammatory factors.
Collapse
Affiliation(s)
- Peng Wang
- The Second Department of Healthcare, China-Japan Friendship Hospital, Beijing, China
| | - Yuan-Li Dong
- Department of Traditional Chinese Medicine, Peking University People's Hospital, Beijing, China
| | - Shan-Shan Li
- Department of Education, China-Japan Friendship Hospital, Beijing, China
| | - Yi Jin
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
| | - Wei-Liang Sun
- Clinical Medical Research Institute, China-Japan Friendship Hospital, Beijing, China
| | - Bao-Sheng Zhao
- Beijing Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Qiu-Bing Li
- The Second Department of Healthcare, China-Japan Friendship Hospital, Beijing, China
| | - Xin Chen
- The Second Department of Healthcare, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
34
|
Bhardwaj V, Kumari S, Dhapola R, Sharma P, Beura SK, Singh SK, Vellingiri B, HariKrishnaReddy D. Shedding light on microglial dysregulation in Alzheimer's disease: exploring molecular mechanisms and therapeutic avenues. Inflammopharmacology 2025; 33:679-702. [PMID: 39609333 DOI: 10.1007/s10787-024-01598-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 10/26/2024] [Indexed: 11/30/2024]
Abstract
Alzheimer's disease (AD) stands out as the foremost prevalent neurodegenerative disorder, characterized by a complex etiology. Various mechanisms have been proposed to elucidate its onset, encompassing amyloid-beta (Aβ) toxicity, tau hyperphosphorylation, oxidative stress and reactive gliosis. The hallmark of AD comprises Aβ and tau aggregation. These misfolded protein aggregates trigger the activation of glial cells, primarily microglia. Microglial cells serve as a major source of inflammatory mediators and their cytotoxic activation has been implicated in various aspects of AD pathology. Activated microglia can adopt M1 or M2 phenotypes, where M1 promotes inflammation by increasing pro-inflammatory cytokines and M2 suppresses inflammation by boosting anti-inflammatory factors. Overexpressed pro-inflammatory cytokines include interleukin (IL)-1β, IL-6 and tumor necrosis factor-α (TNF-α) in adjacent brain regions. Furthermore, microglial signaling pathways dysregulated in AD are myeloid differentiation primary-response protein 88 (Myd 88), colony-stimulating factor-1 receptor (CSF1R) and dedicator of cytokinesis 2 (DOCK2), which alter the physiology. Despite numerous findings, the causative role of microglia-mediated neuroinflammation in AD remains elusive. This review concisely explores cellular and molecular mechanisms of activated microglia and their correlation with AD pathogenesis. Additionally, it highlights promising therapeutics targeting microglia modulation, currently undergoing preclinical and clinical studies, for developing effective treatment for AD.
Collapse
Affiliation(s)
- Vanshu Bhardwaj
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Sneha Kumari
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Rishika Dhapola
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Prajjwal Sharma
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Samir Kumar Beura
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India
| | - Sunil Kumar Singh
- Department of Bio-Chemistry, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India
| | - Balachandar Vellingiri
- Human Cytogenetics and Stem Cell Laboratory, Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Dibbanti HariKrishnaReddy
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India.
| |
Collapse
|
35
|
Udagatti VD, Kumar RD, Rao AG, Sowmya S. Pathological Sequelae of Chronic Vestibular Migraine and Cerebrovascular Insufficiency Leading to Cognitive Decline and Dementia. Indian J Otolaryngol Head Neck Surg 2025; 77:975-982. [PMID: 40070764 PMCID: PMC11890893 DOI: 10.1007/s12070-024-05200-x] [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: 09/28/2024] [Accepted: 11/05/2024] [Indexed: 03/14/2025] Open
Abstract
Chronic vestibular migraine (CVM) and cerebrovascular insufficiency are recognized contributors to cognitive impairment. Vestibular dysfunction affects spatial orientation and balance, while compromised cerebral perfusion impacts neuronal health. This study explores the combined pathological effects of these conditions and their role in cognitive decline and dementia. To evaluate the association between chronic vestibular migraine, cerebrovascular insufficiency, and their potential role in accelerating cognitive decline and the onset of dementia. A patient was evaluated from the ages of 48 to 74 years, initially diagnosed with chronic vestibular migraine and later developing cerebrovascular insufficiency. Cognitive function was assessed using the Mini-Mental State Examination (MMSE) over a 5-year follow-up. Neuroimaging, including MRI and CT angiography, was employed to assess cerebral perfusion and white matter changes. The incidence of dementia was evaluated based on the Diagnostic and Statistical Manual of Mental Disorders (DSM-5) criteria. The patient with coexisting CVM and cerebrovascular insufficiency demonstrated rapid cognitive decline, with MMSE scores decreasing by an average of 5 points over the follow-up period. Neuroimaging revealed microvascular ischemic changes and white matter lesions. Frequent vestibular migraines were associated with the progression to dementia over years of regular assessment and follow-up. Chronic vestibular migraine and cerebrovascular insufficiency are significantly associated with accelerated cognitive decline and an increased risk of dementia. The presence of white matter lesions and ischemic changes appears to exacerbate neurodegeneration. Early diagnosis and management of vestibular dysfunction and cerebrovascular risk factors may help mitigate cognitive decline and reduce the incidence of dementia in affected patients.
Collapse
Affiliation(s)
| | - Rajendran Dinesh Kumar
- Dr.M.G.R. Educational and Research Institute, Rajarajeswari Medical College and Hospital, Department of ENT and Head-Neck Surgery, Bengaluru, Karnataka 560074 India
| | - Ashish Gurudutt Rao
- Dr.M.G.R. Educational and Research Institute, Rajarajeswari Medical College and Hospital, Department of ENT and Head-Neck Surgery, Bengaluru, Karnataka 560074 India
| | - S. Sowmya
- Dr.M.G.R. Educational and Research Institute, Rajarajeswari Medical College and Hospital, Department of ENT and Head-Neck Surgery, Bengaluru, Karnataka 560074 India
| |
Collapse
|
36
|
Ni W, Ding J, Gong P, Tan X, Li J. Inhibition of Kv1.1 channels ameliorates Cu(II)-induced microglial activation and cognitive impairment in mice. Neurochem Int 2025; 183:105936. [PMID: 39814318 DOI: 10.1016/j.neuint.2025.105936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 12/31/2024] [Accepted: 01/13/2025] [Indexed: 01/18/2025]
Abstract
Microglia-mediated neuroinflammation plays a critical role in neuronal damage in neurodegenerative disorders such as Alzheimer's disease. Evidence shows that voltage-gated potassium (Kv) channels regulate microglial activation. We previously reported that copper dyshomeostasis causes neuronal injury via activating microglia. This study was designed to explore the role of Kv1.1 channels in copper-evoked microglial neuroinflammation. BV-2 microglial cells were treated with Cu(II). DiBAC4(3) was used to measure membrane potential. Microglial activation and neuronal loss were detected by enzyme-linked immunosorbent assay, Western blotting, and immunostaining. Learning and memory function was assessed with Morris water maze task. Cu(II) caused a hyperpolarized membrane potential in microglial cells, an effect abolished by functional Kv1.1 blockade. Blockade of Kv1.1 and knock-down of Kv1.1 with small interfering RNA repressed Cu(II)-induced microglial production of pro-inflammatory mediators. Also, Kv1.1 inhibition attenuated activation of PI3K/Akt-ERK1/2 signaling pathway and production of mitochondrial reactive oxidative species as well as nuclear factor-κB activation in Cu(II)-stimulated microglia. Moreover, the Cu(II)-caused, microglia-mediated neurotoxicity (indicated by reduced neuronal survival and increased dendritic loss) was attenuated by Kv1.1 knock-down. In an in vivo mouse model, hippocampal injection of Cu(II) caused elevated Kv1.1 mRNA (but not other Kv1 channels) expression and enhanced microglial Kv1.1 immunoreactivity in the hippocampus. Furthermore, blockade of Kv1.1 attenuated Cu(II)-induced microglial activation and neuronal dendritic loss in the hippocampus and learning and memory dysfunction. These findings suggest that inhibition of Kv1.1 ameliorates Cu(II)-induced microglial activation and cognitive impairment. Thus, it might represent a potential molecular target for anti-inflammatory therapy of neurodegenerative disorders.
Collapse
Affiliation(s)
- Wenwen Ni
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, PR China
| | - Jiani Ding
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, PR China
| | - Ping Gong
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, PR China
| | - Xiaofang Tan
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, PR China
| | - Juan Li
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, PR China.
| |
Collapse
|
37
|
Shan X, Li D, Yin H, Tao W, Zhou L, Gao Y, Xing C, Zhang C. Recent Insights on the Role of Nuclear Receptors in Alzheimer's Disease: Mechanisms and Therapeutic Application. Int J Mol Sci 2025; 26:1207. [PMID: 39940973 PMCID: PMC11818835 DOI: 10.3390/ijms26031207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/13/2025] [Accepted: 01/23/2025] [Indexed: 02/16/2025] Open
Abstract
Nuclear receptors (NRs) are ligand-activated transcription factors that regulate a broad array of biological processes, including inflammation, lipid metabolism, cell proliferation, and apoptosis. Among the diverse family of NRs, peroxisome proliferator-activated receptors (PPARs), estrogen receptor (ER), liver X receptor (LXR), farnesoid X receptor (FXR), retinoid X receptor (RXR), and aryl hydrocarbon receptor (AhR) have garnered significant attention for their roles in neurodegenerative diseases, particularly Alzheimer's disease (AD). NRs influence the pathophysiology of AD through mechanisms such as modulation of amyloid-beta (Aβ) deposition, regulation of inflammatory pathways, and improvement of neuronal function. However, the dual role of NRs in AD progression, where some receptors may exacerbate the disease while others offer therapeutic potential, presents a critical challenge for their application in AD treatment. This review explores the functional diversity of NRs, highlighting their involvement in AD-related processes and discussing the therapeutic prospects of NR-targeting strategies. Furthermore, the key challenges, including the necessity for the precise identification of beneficial NRs, detailed structural analysis through molecular dynamics simulations, and further investigation of NR mechanisms in AD, such as tau pathology and autophagy, are also discussed. Collectively, continued research is essential to clarify the role of NRs in AD, ultimately facilitating their potential use in the diagnosis, prevention, and treatment of AD.
Collapse
Affiliation(s)
- Xiaoxiao Shan
- Anhui Academy of Chinese Medicine, Anhui University of Chinese Medicine, Hefei 230012, China; (X.S.); (D.L.); (H.Y.); (W.T.); (L.Z.); (Y.G.); (C.X.)
- Center for Xin’an Medicine and Modernization of Traditional Chinese Medicine of IHM, Grand Health Research Institute of Hefei Comprehensive National Science Center, Anhui University of Chinese Medicine, Hefei 230012, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei 230012, China
- School of Pharmacy, Institute of Pharmacokinetics, Anhui University of Chinese Medicine, Hefei 230012, China
- Anhui Genuine Chinese Medicinal Materials Quality Improvement Collaborative Innovation Center, Hefei 230012, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Dawei Li
- Anhui Academy of Chinese Medicine, Anhui University of Chinese Medicine, Hefei 230012, China; (X.S.); (D.L.); (H.Y.); (W.T.); (L.Z.); (Y.G.); (C.X.)
- Center for Xin’an Medicine and Modernization of Traditional Chinese Medicine of IHM, Grand Health Research Institute of Hefei Comprehensive National Science Center, Anhui University of Chinese Medicine, Hefei 230012, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei 230012, China
- School of Pharmacy, Institute of Pharmacokinetics, Anhui University of Chinese Medicine, Hefei 230012, China
- Anhui Genuine Chinese Medicinal Materials Quality Improvement Collaborative Innovation Center, Hefei 230012, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Huihui Yin
- Anhui Academy of Chinese Medicine, Anhui University of Chinese Medicine, Hefei 230012, China; (X.S.); (D.L.); (H.Y.); (W.T.); (L.Z.); (Y.G.); (C.X.)
- Center for Xin’an Medicine and Modernization of Traditional Chinese Medicine of IHM, Grand Health Research Institute of Hefei Comprehensive National Science Center, Anhui University of Chinese Medicine, Hefei 230012, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei 230012, China
- School of Pharmacy, Institute of Pharmacokinetics, Anhui University of Chinese Medicine, Hefei 230012, China
- Anhui Genuine Chinese Medicinal Materials Quality Improvement Collaborative Innovation Center, Hefei 230012, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Wenwen Tao
- Anhui Academy of Chinese Medicine, Anhui University of Chinese Medicine, Hefei 230012, China; (X.S.); (D.L.); (H.Y.); (W.T.); (L.Z.); (Y.G.); (C.X.)
- Center for Xin’an Medicine and Modernization of Traditional Chinese Medicine of IHM, Grand Health Research Institute of Hefei Comprehensive National Science Center, Anhui University of Chinese Medicine, Hefei 230012, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei 230012, China
- School of Pharmacy, Institute of Pharmacokinetics, Anhui University of Chinese Medicine, Hefei 230012, China
- Anhui Genuine Chinese Medicinal Materials Quality Improvement Collaborative Innovation Center, Hefei 230012, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Lele Zhou
- Anhui Academy of Chinese Medicine, Anhui University of Chinese Medicine, Hefei 230012, China; (X.S.); (D.L.); (H.Y.); (W.T.); (L.Z.); (Y.G.); (C.X.)
- Center for Xin’an Medicine and Modernization of Traditional Chinese Medicine of IHM, Grand Health Research Institute of Hefei Comprehensive National Science Center, Anhui University of Chinese Medicine, Hefei 230012, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei 230012, China
- School of Pharmacy, Institute of Pharmacokinetics, Anhui University of Chinese Medicine, Hefei 230012, China
- Anhui Genuine Chinese Medicinal Materials Quality Improvement Collaborative Innovation Center, Hefei 230012, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Yu Gao
- Anhui Academy of Chinese Medicine, Anhui University of Chinese Medicine, Hefei 230012, China; (X.S.); (D.L.); (H.Y.); (W.T.); (L.Z.); (Y.G.); (C.X.)
- Center for Xin’an Medicine and Modernization of Traditional Chinese Medicine of IHM, Grand Health Research Institute of Hefei Comprehensive National Science Center, Anhui University of Chinese Medicine, Hefei 230012, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei 230012, China
- School of Pharmacy, Institute of Pharmacokinetics, Anhui University of Chinese Medicine, Hefei 230012, China
- Anhui Genuine Chinese Medicinal Materials Quality Improvement Collaborative Innovation Center, Hefei 230012, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Chengjie Xing
- Anhui Academy of Chinese Medicine, Anhui University of Chinese Medicine, Hefei 230012, China; (X.S.); (D.L.); (H.Y.); (W.T.); (L.Z.); (Y.G.); (C.X.)
- Center for Xin’an Medicine and Modernization of Traditional Chinese Medicine of IHM, Grand Health Research Institute of Hefei Comprehensive National Science Center, Anhui University of Chinese Medicine, Hefei 230012, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei 230012, China
- School of Pharmacy, Institute of Pharmacokinetics, Anhui University of Chinese Medicine, Hefei 230012, China
- Anhui Genuine Chinese Medicinal Materials Quality Improvement Collaborative Innovation Center, Hefei 230012, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Caiyun Zhang
- Anhui Academy of Chinese Medicine, Anhui University of Chinese Medicine, Hefei 230012, China; (X.S.); (D.L.); (H.Y.); (W.T.); (L.Z.); (Y.G.); (C.X.)
- Center for Xin’an Medicine and Modernization of Traditional Chinese Medicine of IHM, Grand Health Research Institute of Hefei Comprehensive National Science Center, Anhui University of Chinese Medicine, Hefei 230012, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei 230012, China
- School of Pharmacy, Institute of Pharmacokinetics, Anhui University of Chinese Medicine, Hefei 230012, China
- Anhui Genuine Chinese Medicinal Materials Quality Improvement Collaborative Innovation Center, Hefei 230012, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Anhui University of Chinese Medicine, Hefei 230012, China
| |
Collapse
|
38
|
Yoo J, Lee J, Ahn B, Han J, Lim MH. Multi-target-directed therapeutic strategies for Alzheimer's disease: controlling amyloid-β aggregation, metal ion homeostasis, and enzyme inhibition. Chem Sci 2025; 16:2105-2135. [PMID: 39810997 PMCID: PMC11726323 DOI: 10.1039/d4sc06762b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative dementia, marked by progressive cognitive decline and memory impairment. Despite advances in therapeutic research, single-target-directed treatments often fall short in addressing the complex, multifactorial nature of AD. This arises from various pathological features, including amyloid-β (Aβ) aggregate deposition, metal ion dysregulation, oxidative stress, impaired neurotransmission, neuroinflammation, mitochondrial dysfunction, and neuronal cell death. This review illustrates their interrelationships, with a particular emphasis on the interplay among Aβ, metal ions, and AD-related enzymes, such as β-site amyloid precursor protein cleaving enzyme 1 (BACE1), matrix metalloproteinase 9 (MMP9), lysyl oxidase-like 2 (LOXL2), acetylcholinesterase (AChE), and monoamine oxidase B (MAOB). We further underscore the potential of therapeutic strategies that simultaneously inhibit Aβ aggregation and address other pathogenic mechanisms. These approaches offer a more comprehensive and effective method for combating AD, overcoming the limitations of conventional therapies.
Collapse
Affiliation(s)
- Jeasang Yoo
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Jimin Lee
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Byeongha Ahn
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Jiyeon Han
- Department of Applied Chemistry, University of Seoul Seoul 02504 Republic of Korea
| | - Mi Hee Lim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| |
Collapse
|
39
|
Kunwar OK, Singh S. Neuroinflammation and neurodegeneration in Huntington's disease: genetic hallmarks, role of metals and organophosphates. Neurogenetics 2025; 26:21. [PMID: 39820855 DOI: 10.1007/s10048-025-00801-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 01/07/2025] [Indexed: 01/19/2025]
Abstract
Huntington's disease (HDs) is a fatal, autosomal dominant, and hereditary neurodegenerative disorder characterized by progressive motor dysfunction, cognitive decline, and psychiatric disturbances. HD is well linked to mutation in the HTT gene, which leads to an abnormal expansion of trinucleotide CAG repeats, resulting in the production of the mHTT protein and responsible for abnormally long poly-Q tract. These abnormal proteins disrupt cellular processes, including neuroinflammation, endoplasmic reticulum (ER) stress, and mitochondrial dysfunction, ultimately leading to selective neuronal loss in the brain. Epidemiological studies reveal significant regional variability in HDs prevalence, with the highest rates observed in North America and the lowest in Africa. In addition to genetic factors, environmental influences such as exposure to metals, and chemicals, and lifestyle factors like alcohol and tobacco use may exacerbate disease progression. This review explores the molecular mechanisms underlying HDs and emphasize the role of neuroinflammatory mediators and environmental factors, in HD research. Understanding these complex interactions is crucial for developing targeted interventions that can slow or halt the progression of this devastating disease.
Collapse
Affiliation(s)
- Omkar Kumar Kunwar
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Shamsher Singh
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India.
| |
Collapse
|
40
|
Guo Y, Zhao J, Liu X, Lu P, Liang F, Wang X, Wu J, Hai Y. Ghrelin Induces Ferroptosis Resistance and M2 Polarization of Microglia to Alleviate Neuroinflammation and Cognitive Impairment in Alzheimer's Disease. J Neuroimmune Pharmacol 2025; 20:6. [PMID: 39797928 DOI: 10.1007/s11481-024-10165-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 11/29/2024] [Indexed: 01/13/2025]
Abstract
Microglial polarization and ferroptosis are important pathological features in Alzheimer's disease (AD). Ghrelin, a brain-gut hormone, has potential neuroprotective effects in AD. This study aimed to explore the potential mechanisms by which ghrelin regulates the progression of AD, as well as the crosstalk between microglial polarization and ferroptosis. Mouse BV2 microglial cells and male mice were treated with beta-amyloid (Aβ) (1-42) to simulate the AD environment. Microglia ferroptosis was measured by detecting levels of ferroptosis-related proteins (SLC7A11, GPX4, FTL1, and FTH1), metabolic markers (ROS, MDA, GSH, SOD), and observing mitochondrial morphological changes. Microglial polarization was evaluated by measuring levels of inflammatory markers and surface markers. The impact of ghrelin on Aβ1-42-exposed microglia was assessed by coupling with the ferroptosis activator Erastin. Cognitive impairment in AD mice was evaluated through behavioral tests. Tissue staining was applied to determine neuronal damage. In Aβ1-42-exposed microglia, ghrelin upregulated the protein expression of SLC7A11, GPX4, FTL1 and FTH1, reduced ROS and MDA levels, and elevated GSH and SOD levels through the BMP6/SMAD1 pathway. Ghrelin alleviated mitochondrial structural damage. Additionally, ghrelin reduced levels of pro-inflammatory factors and CD86, while increasing levels of anti-inflammatory factors and CD206. Erastin reversed the effects of ghrelin on ferroptosis and phenotypic polarization in Aβ1-42-exposed microglia. In AD mice, ghrelin ameliorated abnormal behavior, neuroinflammation, and plaque deposition. Ghrelin attenuated iNOS/IBA1-positive expression and enhanced Arg-1/IBA1-positive expression in the hippocampus. Ghrelin induces microglial M2 polarization by inhibiting microglia ferroptosis, thereby alleviating neuroinflammation. Our results indicate that ghrelin may serve as a promising potential agent for treating cognitive impairment in AD.
Collapse
Affiliation(s)
- Yaoxue Guo
- Department of Clinical Pharmacy, Baotou Central Hospital, Baotou, 014040, Inner Mongolia, China
| | - Junli Zhao
- Pharmacy Department, Baotou Central Hospital, Baotou, 014040, Inner Mongolia, China
| | - Xing Liu
- Pharmacy Department, Baotou Central Hospital, Baotou, 014040, Inner Mongolia, China
| | - Pu Lu
- Oncology Department, Baotou Central Hospital, Baotou, 014040, Inner Mongolia, China
| | - Furu Liang
- Department of Neurology, Baotou Central Hospital, Baotou, 014040, Inner Mongolia, China
| | - Xueyan Wang
- Oncology Department, Baotou Central Hospital, Baotou, 014040, Inner Mongolia, China
| | - Jing Wu
- Pharmacy Department, Baotou Central Hospital, Baotou, 014040, Inner Mongolia, China
| | - Yan Hai
- Pharmacy Department, Baotou Central Hospital, Baotou, 014040, Inner Mongolia, China.
- Baotou Central Hospital, 61 Huancheng Road, Donghe District, Baotou, 014040, Inner Mongolia, China.
| |
Collapse
|
41
|
Elsworthy RJ, Pearce A, Masoudzadeh F, Koska K, Lodhiya H, Meher G, Adjej J, Brookes KJ. OAS1: A Protective Mechanism for Alzheimer's Disease? An Exploration of Data and Possible Mechanisms. Int J Mol Sci 2025; 26:524. [PMID: 39859237 PMCID: PMC11765370 DOI: 10.3390/ijms26020524] [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: 12/03/2024] [Revised: 01/06/2025] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
The immune system and neuroinflammation are now well established in the aetiology of neurodegeneration. Previous studies of transcriptomic and gene association studies have highlighted the potential of the 2'-5' oligoadenylate synthetase 1 (OAS1) to play a role in Alzheimer's disease. OAS1 is a viral response gene, interferon-induced, dsRNA activated enzyme, which binds RNase L to degrade dsRNA, and has been associated with COVID-19 response. This study explores whether a viral defence gene could play a vital role in neurodegeneration pathology. The genotyping of five SNPs across the OAS1 locus was conducted in the Brains for Dementia Research (BDR) Cohort for association with AD. RNA-sequencing data were explored for differences in OAS1 gene expression between phenotypes and genotypes. Finally, levels of dsRNA were measured in control cell lines, prior to and after exposure to amyloid oligomers and in cells harbouring a dementia-relevant mutation. No association of any of the OAS1 SNPs investigated were associated with the AD phenotype in the BDR cohort. However, gene expression data supported the previous observation that the minor allele haplotype was associated with higher levels of the OAS1 gene expression and the presence of an alternative transcript. Further to this, the presence of endogenous dsRNA was found to increase with exposure to amyloid oligomers and in the cell line with a dementia-relevant mutation. The data presented here suggest further exploration of the OAS1 gene in relation to dementia is warranted. Investigations of whether carriers of the protective OAS1 haplotype lower dsRNA presence and in turn lower inflammation and cell death are required to support the role of the gene as a moderator of neurodegeneration.
Collapse
Affiliation(s)
- Richard J. Elsworthy
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham B15 2TT, UK;
| | - Alex Pearce
- Department of Biosciences, School of Science & Technology, Nottingham Trent University, Nottingham NG11 8NF, UK
| | - Farnoush Masoudzadeh
- Department of Biosciences, School of Science & Technology, Nottingham Trent University, Nottingham NG11 8NF, UK
| | - Klaudia Koska
- Department of Biosciences, School of Science & Technology, Nottingham Trent University, Nottingham NG11 8NF, UK
| | - Honey Lodhiya
- Department of Biosciences, School of Science & Technology, Nottingham Trent University, Nottingham NG11 8NF, UK
| | - Gargi Meher
- Department of Biosciences, School of Science & Technology, Nottingham Trent University, Nottingham NG11 8NF, UK
| | - Jodelle Adjej
- Department of Biosciences, School of Science & Technology, Nottingham Trent University, Nottingham NG11 8NF, UK
| | - Keeley J. Brookes
- Department of Biosciences, School of Science & Technology, Nottingham Trent University, Nottingham NG11 8NF, UK
| |
Collapse
|
42
|
Cummings JL, Atri A, Feldman HH, Hansson O, Sano M, Knop FK, Johannsen P, León T, Scheltens P. evoke and evoke+: design of two large-scale, double-blind, placebo-controlled, phase 3 studies evaluating efficacy, safety, and tolerability of semaglutide in early-stage symptomatic Alzheimer's disease. Alzheimers Res Ther 2025; 17:14. [PMID: 39780249 PMCID: PMC11708093 DOI: 10.1186/s13195-024-01666-7] [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: 09/06/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND Disease-modifying therapies targeting the diverse pathophysiology of Alzheimer's disease (AD), including neuroinflammation, represent potentially important and novel approaches. The glucagon-like peptide-1 receptor agonist semaglutide is approved for the treatment of type 2 diabetes and obesity and has an established safety profile. Semaglutide may have a disease-modifying, neuroprotective effect in AD through multimodal mechanisms including neuroinflammatory, vascular, and other AD-related processes. Large randomized controlled trials are needed to assess the efficacy and safety of semaglutide in early-stage symptomatic AD. METHODS evoke and evoke+ are randomized, double-blind, placebo-controlled phase 3 trials investigating the efficacy, safety, and tolerability of once-daily oral semaglutide versus placebo in early-stage symptomatic AD. Eligible participants were men or women aged 55-85 years with mild cognitive impairment or mild dementia due to AD with confirmed amyloid abnormalities (assessed by positron emission tomography or cerebrospinal fluid [CSF] analysis). After a maximum 12-week screening phase, an anticipated 1840 patients in each trial are randomized (1:1) to semaglutide or placebo for 156 weeks (104-week main treatment phase and 52-week extension). Randomized participants follow an 8-week dose escalation regimen (3 mg [weeks 0-4], 7 mg [weeks 4-8], and 14 mg [weeks 8-156]). The primary endpoint is the semaglutide-placebo difference on change from baseline to week 104 in the Clinical Dementia Rating - Sum of Boxes score. Analyses of plasma biomarkers, collected from all participants, and a CSF sub-study (planned n = 210) will explore semaglutide effects on AD biomarkers and neuroinflammation. RESULTS Enrollment was undertaken between May 18, 2021, and September 8, 2023. Completion of the trials' main phase is expected in September 2025, and the 52-week extension (in which participants and investigators remain blinded to treatment assignment) will continue to October 2026. CONCLUSION evoke and evoke+ are the first large-scale trials to investigate the disease-modifying potential of semaglutide in participants with early-stage symptomatic AD, including exploration of effects on AD biomarkers and neuroinflammation. The trials will provide data on the potential disease-modifying effects of semaglutide and will be important in evaluating its utility in the treatment of early-stage symptomatic AD. TRIAL REGISTRATION Clinicaltrials.gov, NCT04777396 and NCT04777409. Date: 02/03/2021.
Collapse
Affiliation(s)
- Jeffrey L Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, Kirk Kerkorian School of Medicine, University of Nevada, Las Vegas, NV, USA.
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences, University of Nevada, Las Vegas, NV, USA.
| | - Alireza Atri
- Banner Sun Health Research Institute, Sun City, AZ, USA
- Banner Alzheimer's Institute, Phoenix, AZ, USA
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Howard H Feldman
- Department of Neurosciences, University of California San Diego, La Jolla, San Diego, CA, USA
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Mary Sano
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Filip K Knop
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Steno Diabetes Center Copenhagen, Herlev, Denmark
- Novo Nordisk A/S, Søborg, Denmark
| | | | | | - Philip Scheltens
- Alzheimer Center Amsterdam, Department of Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- EQT Life Sciences, Amsterdam, The Netherlands
| |
Collapse
|
43
|
Long C, Fritts A, Broadway J, Brawman-Mintzer O, Mintzer J. Neuroinflammation: A Driving Force in the Onset and Progression of Alzheimer's Disease. J Clin Med 2025; 14:331. [PMID: 39860337 PMCID: PMC11766252 DOI: 10.3390/jcm14020331] [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: 12/19/2024] [Revised: 01/03/2025] [Accepted: 01/05/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: The goal of this commentary is to highlight several key components of the inflammatory process as it relates to amyloid toxicity in Alzheimer's disease (AD), including the role of neuroinflammatory factors and peripheral inflammatory events. Methods: Google Scholar and PubMed were used to find articles with the following keywords: Alzheimer's disease, amyloids, neuroinflammation, peripheral inflammation, microglia, cytokines, and treatments. Sources that were case reports, not peer-reviewed, or older than 30 years were excluded. Abstracts were reviewed first for their relevance before the full text was considered. Methods sections were reviewed to ensure the interventional papers included were randomized controlled trials, meta-analyses, or systematic reviews; however, several literature reviews were also included due to the relevance of their background information. Results: Based on the literature review, we chose to concentrate on microglia, cytokine signaling, and peripheral inflammation markers. We found that microglia activation and subsequent microglia-driven inflammation play a pivotal role in the pathomechanism of AD. Additionally, cytokines such as interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-a) appear to contribute to amyloid accumulation and cell damage. Finally, the increased permeability of the blood-brain barrier (BBB) allows for the peripheral inflammatory process to contribute to the inflammation of the central nervous system (CNS) and amyloid-beta (Aβ) accumulation. Conclusions: Current evidence suggests that the immune system plays a pivotal role in the pathogenesis of AD, both in the CNS and the periphery.
Collapse
Affiliation(s)
- Campbell Long
- Ralph H. Johnson VA Health Care System, 109 Bee St, Charleston, SC 29401, USA
| | - Arianne Fritts
- College of Health Professions, Department of Health Sciences and Research, Medical University of South Carolina, 171 Ashley Avenue, Charleston, SC 29425, USA
| | - Jessica Broadway
- Ralph H. Johnson VA Health Care System, 109 Bee St, Charleston, SC 29401, USA
- College of Medicine, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, 171 Ashley Avenue, Charleston, SC 29425, USA
| | - Olga Brawman-Mintzer
- Ralph H. Johnson VA Health Care System, 109 Bee St, Charleston, SC 29401, USA
- College of Medicine, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, 171 Ashley Avenue, Charleston, SC 29425, USA
| | - Jacobo Mintzer
- Ralph H. Johnson VA Health Care System, 109 Bee St, Charleston, SC 29401, USA
- College of Health Professions, Department of Health Studies, Medical University of South Carolina, 171 Ashley Avenue, Charleston, SC 29425, USA
| |
Collapse
|
44
|
Mahwish, Imran M, Naeem H, Hussain M, Alsagaby SA, Al Abdulmonem W, Mujtaba A, Abdelgawad MA, Ghoneim MM, El‐Ghorab AH, Selim S, Al Jaouni SK, Mostafa EM, Yehuala TF. Antioxidative and Anticancer Potential of Luteolin: A Comprehensive Approach Against Wide Range of Human Malignancies. Food Sci Nutr 2025; 13:e4682. [PMID: 39830909 PMCID: PMC11742186 DOI: 10.1002/fsn3.4682] [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: 08/01/2024] [Revised: 10/25/2024] [Accepted: 11/30/2024] [Indexed: 01/22/2025] Open
Abstract
Luteolin is widely distributed phytochemical, a flavonoid, in kingdom plantae. Luteolin with potential antioxidant activity prevent ROS-induced damages and reduce oxidative stress which is mainly responsible in pathogenesis of many diseases. Several chemo preventive activities and therapeutic benefits are associated with luteolin. Luteolin prevents cancer via modulation of numerous pathways, that is, by inactivating proteins; such as procaspase-9, CDC2 and cyclin B or upregulation of caspase-9 and caspase-3, cytochrome C, cyclin A, CDK2, and APAF-1, in turn inducing cell cycle arrest as well as apoptosis. It also enhances phosphorylation of p53 and expression level of p53-targeted downstream gene. By Increasing BAX protein expression; decreasing VEGF and Bcl-2 expression it can initiate cell cycle arrest and apoptosis. Luteolin can stimulate mitochondrial-modulated functions to cause cellular death. It can also reduce expression levels of p-Akt, p-EGFR, p-Erk1/2, and p-STAT3. Luteolin plays positive role against cardiovascular disorders by improving cardiac function, decreasing the release of inflammatory cytokines and cardiac enzymes, prevention of cardiac fibrosis and hypertrophy; enhances level of CTGF, TGFβ1, ANP, Nox2, Nox4 gene expressions. Meanwhile suppresses TGFβ1 expression and phosphorylation of JNK. Luteolin helps fight diabetes via inhibition of alpha-glucosidase and ChE activity. It can reduce activity levels of catalase, superoxide dismutase, and GS4. It can improve blood glucose, insulin, HOMA-IR, and HbA1c levels. This review is an attempt to elaborate molecular targets of luteolin and its role in modulating irregularities in cellular pathways to overcome severe outcomes during diseases including cancer, cardiovascular disorders, diabetes, obesity, inflammation, Alzheimer's disease, Parkinson's disease, hepatic disorders, renal disorders, brain injury, and asthma. As luteolin has enormous therapeutic benefits, it could be a potential candidate in future drug development strategies.
Collapse
Affiliation(s)
- Mahwish
- Institute of Food Science and NutritionUniversity of SargodhaSargodhaPakistan
| | - Muhammad Imran
- Department of Food Science and TechnologyUniversity of NarowalNarowalPakistan
| | - Hammad Naeem
- Department of Food Science and TechnologyMuhammad Nawaz Shareef University of AgricultureMultanPakistan
| | - Muzzamal Hussain
- Department of Food SciencesGovernment College University FaisalabadFaisalabadPakistan
| | - Suliman A. Alsagaby
- Department of Medical Laboratory Sciences, College of Applied Medical SciencesMajmaah UniversityAL‐MajmaahSaudi Arabia
| | - Waleed Al Abdulmonem
- Department of Pathology, College of MedicineQassim UniversityBuraidahSaudi Arabia
| | - Ahmed Mujtaba
- Department of Food Sciences and Technology, Faculty of Engineering and TechnologyHamdard University Islamabad campusIslamabadPakistan
| | - Mohamed A. Abdelgawad
- Department of Pharmaceutical Chemistry, College of PharmacyJouf UniversityAljoufSaudi Arabia
| | - Mohammed M. Ghoneim
- Department of Pharmacy Practice, College of PharmacyAlMaarefa UniversityRiyadhSaudi Arabia
| | - Ahmed H. El‐Ghorab
- Department of Chemistry, College of ScienceJouf UniversitySakakaSaudi Arabia
| | - Samy Selim
- Department of Clinical Laboratory Sciences, College of Applied Medical SciencesJouf UniversitySakakaSaudi Arabia
| | - Soad K. Al Jaouni
- Department of Hematology/Oncology, Yousef Abdulatif Jameel Scientific Chair of Prophetic Medicine Application, Faculty of MedicineKing Abdulaziz UniversityJeddahSaudi Arabia
| | - Ehab M. Mostafa
- Department of Pharmacognosy, College of PharmacyJouf UniversitySakakaSaudi Arabia
- Pharmacognosy and Medicinal Plants Department, Faculty of Pharmacy (Boys)Al‐Azhar UniversityCairoEgypt
| | - Tadesse Fenta Yehuala
- Faculty of Chemical and Food Engineering, Bahir Dar Institute of TechnologyBahir Dar UniversityBahir DarEthiopia
| |
Collapse
|
45
|
Duan ZM, Wu LF. Role of oral-gut-brain axis in psychiatric and neurological disorders. Shijie Huaren Xiaohua Zazhi 2024; 32:878-886. [DOI: 10.11569/wcjd.v32.i12.878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/06/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024] Open
Abstract
The oral cavity and gut are two important microbial habitats in the human body, harboring the most ecologically rich and taxonomically diverse microbial communities in humans, which play a key role in microbiome related diseases. In recent years, the emerging concept of the oral-gut-brain axis has attracted widespread attention in the fields of neuroscience, digestive science, and microbiology. It is not only an anatomical description, but also a comprehensive concept that covers multiple physiological functions and pathological mechanisms. Simply put, the oral-gut-brain axis refers to the complex network that connects the mouth, gut, and brain tightly together through neural connections and hormonal and immune pathways. With the deepening of research on the oral-gut-brain axis theory, more and more evidence shows that it plays an important role in depression, Parkinson's disease, and other neurodegenerative diseases. This article reviews the recent progress in research of the oral-gut-brain axis in psychiatric and neurological diseases.
Collapse
Affiliation(s)
- Zhi-Min Duan
- Department of Gastroenterology, The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Ling-Fei Wu
- Department of Gastroenterology, The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
| |
Collapse
|
46
|
Yang F, Gao W, Wang J, Li X, Li H. Progress of Chinese Medicine in Regulating Microglial Polarization against Alzheimer's Disease. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:2255-2275. [PMID: 39721955 DOI: 10.1142/s0192415x24500873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2024]
Abstract
Alzheimer's disease (AD), the predominant form of dementia, is a neurodegenerative disorder of the central nervous system (CNS) characterized by a subtle onset and a spectrum of cognitive and functional declines. The clinical manifestation of AD encompasses memory deficits, cognitive deterioration, and behavioral disturbances, culminating in a severe impairment of daily living skills. Despite its high prevalence, accounting for 60-70% of all dementia cases, there remains an absence of curative therapeutics. Microglia (MG), the resident immune cells of the CNS, exhibit a bifurcated role in AD pathogenesis. Functioning in a neuroprotective capacity, MGs express scavenger receptors, facilitating the clearance of [Formula: see text]-amyloid protein (A[Formula: see text]) and cellular debris. Conversely, aberrant activation of MGs can lead to the secretion of pro-inflammatory cytokines, thereby propagating neuroinflammatory responses that are detrimental to neuronal integrity. The dynamics of MG activation and the ensuing neuroinflammation are pivotal in the evolution of AD. Chinese medicine (CM), a treasure trove of traditional Chinese cultural practices, has demonstrated significant potential in the therapeutic management of AD. Over the past triennium, CM has garnered considerable research attention for its multifaceted approaches to AD, including the regulation of MG polarization. This review synthesizes current knowledge on the origins, polarization dynamics, and mechanistic interplay of MG with AD pathology. It further explores the nexus between MG polarization and cardinal pathological hallmarks of AD, such as A[Formula: see text] plaque deposition, hyperphosphorylation of tau, synaptic plasticity impairments, neuroinflammation, and brain-gut-axis dysregulation. The review also encapsulates the therapeutic strategies of CM, which encompass monomers, formulae, and acupuncture. These strategies modulate MG polarization in the context of AD treatment, thereby providing a robust theoretical framework in which to conduct future investigative endeavors in both the clinical and preclinical realms.
Collapse
Affiliation(s)
- Fengge Yang
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P. R. China
| | - Wei Gao
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P. R. China
- Jiangsu College of Nursing Jiangsu, Huaian, Huaiyin 223001, P. R. China
| | - Junting Wang
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P. R. China
- The Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150001, P. R. China
| | - Xue Li
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P. R. China
| | - Honglin Li
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P. R. China
- The Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150001, P. R. China
| |
Collapse
|
47
|
Nie RZ, Luo HM, Liu YP, Wang SS, Hou YJ, Chen C, Wang H, Lv HL, Tao XY, Jing ZH, Zhang HK, Li PF. Food Functional Factors in Alzheimer's Disease Intervention: Current Research Progress. Nutrients 2024; 16:3998. [PMID: 39683391 DOI: 10.3390/nu16233998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Alzheimer's disease (AD) is a complex multifactorial neurodegenerative disease. With the escalating aging of the global population, the societal burden of this disease is increasing. Although drugs are available for the treatment of AD, their efficacy is limited and there remains no effective cure. Therefore, the identification of safe and effective prevention and treatment strategies is urgently needed. Functional factors in foods encompass a variety of natural and safe bioactive substances that show potential in the prevention and treatment of AD. However, current research focused on the use of these functional factors for the prevention and treatment of AD is in its initial stages, and a complete theoretical and application system remains to be determined. An increasing number of recent studies have found that functional factors such as polyphenols, polysaccharides, unsaturated fatty acids, melatonin, and caffeine have positive effects in delaying the progression of AD and improving cognitive function. For example, polyphenols exhibit antioxidant, anti-inflammatory, and neuroprotective effects, and polysaccharides promote neuronal growth and inhibit inflammation and oxidative stress. Additionally, unsaturated fatty acids inhibit Aβ production and Tau protein phosphorylation and reduce neuroinflammation, and melatonin has been shown to protect nerve cells and improve cognitive function by regulating mitochondrial homeostasis and autophagy. Caffeine has also been shown to inhibit inflammation and reduce neuronal damage. Future research should further explore the mechanisms of action of these functional factors and develop relevant functional foods or nutritional supplements to provide new strategies and support for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Rong-Zu Nie
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Henan Key Laboratory of Cold Chain Food Quality and Safety Control, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou University of Light Industry, Zhengzhou 450001, China
| | - Huo-Min Luo
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Henan Key Laboratory of Cold Chain Food Quality and Safety Control, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou University of Light Industry, Zhengzhou 450001, China
| | - Ya-Ping Liu
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Henan Key Laboratory of Cold Chain Food Quality and Safety Control, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou University of Light Industry, Zhengzhou 450001, China
| | - Shuang-Shuang Wang
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Henan Key Laboratory of Cold Chain Food Quality and Safety Control, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou University of Light Industry, Zhengzhou 450001, China
| | - Yan-Jie Hou
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Henan Key Laboratory of Cold Chain Food Quality and Safety Control, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou University of Light Industry, Zhengzhou 450001, China
| | - Chen Chen
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Henan Key Laboratory of Cold Chain Food Quality and Safety Control, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou University of Light Industry, Zhengzhou 450001, China
| | - Hang Wang
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Henan Key Laboratory of Cold Chain Food Quality and Safety Control, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou University of Light Industry, Zhengzhou 450001, China
| | - Hui-Lin Lv
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Henan Key Laboratory of Cold Chain Food Quality and Safety Control, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou University of Light Industry, Zhengzhou 450001, China
| | - Xing-Yue Tao
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Henan Key Laboratory of Cold Chain Food Quality and Safety Control, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou University of Light Industry, Zhengzhou 450001, China
| | - Zhao-Hui Jing
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Henan Key Laboratory of Cold Chain Food Quality and Safety Control, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou University of Light Industry, Zhengzhou 450001, China
| | - Hao-Kun Zhang
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Henan Key Laboratory of Cold Chain Food Quality and Safety Control, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou University of Light Industry, Zhengzhou 450001, China
| | - Pei-Feng Li
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Henan Key Laboratory of Cold Chain Food Quality and Safety Control, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou University of Light Industry, Zhengzhou 450001, China
| |
Collapse
|
48
|
Cómitre-Mariano B, Vellila-Alonso G, Segura-Collar B, Mondéjar-Ruescas L, Sepulveda JM, Gargini R. Sentinels of neuroinflammation: the crucial role of myeloid cells in the pathogenesis of gliomas and neurodegenerative diseases. J Neuroinflammation 2024; 21:304. [PMID: 39578808 PMCID: PMC11583668 DOI: 10.1186/s12974-024-03298-y] [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: 03/04/2024] [Accepted: 11/13/2024] [Indexed: 11/24/2024] Open
Abstract
The inflammatory processes that drive pathologies of the central nervous system (CNS) are complex and involve significant contributions from the immune system, particularly myeloid cells. Understanding the shared and distinct pathways of myeloid cell regulation in different CNS diseases may offer critical insights into therapeutic development. This review aims to elucidate the mechanisms underlying myeloid cell dysfunction and neuroinflammation in two groups of neurological pathologies with significant social impact and a limited efficacy of their treatments: the most common primary brain tumors -gliomas-, and the most prevalent neurodegenerative disorders -Alzheimer's and Parkinson's disease. Despite their distinct clinical manifestations, these diseases share key pathological features, including chronic inflammation and immune dysregulation. The role of myeloid cells in neuroinflammation has garnered special interest in recent years in both groups, as evidenced by the growing focus on therapeutic research centred on myeloid cells. By examining the cellular and molecular dynamics that govern these conditions, we hope to identify common and unique therapeutic targets that can inform the development of more effective treatments. Recent advances in single-cell technologies have revolutionized our understanding of myeloid cell heterogeneity, revealing diverse phenotypes and molecular profiles across different disease stages and microenvironments. Here, we present a comprehensive analysis of myeloid cell involvement in gliomas, Alzheimer's and Parkinson's disease, with a focus on phenotypic acquisition, molecular alterations, and therapeutic strategies targeting myeloid cells. This integrated approach not only addresses the limitations of current treatments but also suggests new avenues for therapeutic intervention, aimed at modulating the immune landscape to improve patient outcomes.
Collapse
Affiliation(s)
- Blanca Cómitre-Mariano
- Instituto de Investigación Biomédicas I+12, Hospital Universitario 12 de Octubre, Madrid, 28041, Spain
- Pathology and Neurooncology Unit, Hospital Universitario 12 de Octubre, Av. de Córdoba, S/N, Madrid, 28041, Spain
| | - Gabriel Vellila-Alonso
- Pathology and Neurooncology Unit, Hospital Universitario 12 de Octubre, Av. de Córdoba, S/N, Madrid, 28041, Spain
- Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, 28041, Spain
| | - Berta Segura-Collar
- Instituto de Investigación Biomédicas I+12, Hospital Universitario 12 de Octubre, Madrid, 28041, Spain
- Pathology and Neurooncology Unit, Hospital Universitario 12 de Octubre, Av. de Córdoba, S/N, Madrid, 28041, Spain
| | - Lucía Mondéjar-Ruescas
- Instituto de Investigación Biomédicas I+12, Hospital Universitario 12 de Octubre, Madrid, 28041, Spain
- Pathology and Neurooncology Unit, Hospital Universitario 12 de Octubre, Av. de Córdoba, S/N, Madrid, 28041, Spain
| | - Juan M Sepulveda
- Instituto de Investigación Biomédicas I+12, Hospital Universitario 12 de Octubre, Madrid, 28041, Spain.
- Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, 28041, Spain.
| | - Ricardo Gargini
- Instituto de Investigación Biomédicas I+12, Hospital Universitario 12 de Octubre, Madrid, 28041, Spain.
- Pathology and Neurooncology Unit, Hospital Universitario 12 de Octubre, Av. de Córdoba, S/N, Madrid, 28041, Spain.
| |
Collapse
|
49
|
Shah S, Jain H. Microglia-Associated Neuroinflammation in Alzheimer’s Disease and Its Therapeutic Potential. NEUROGLIA 2024; 5:452-466. [DOI: 10.3390/neuroglia5040029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Background: Neuroinflammation has long been implicated in the progression of amyloid beta (Aβ) accumulation and the decline of cognitive function in Alzheimer’s disease (AD). The phenotype balance between A1 (toxic) and A2 (safe) microglial phenotypes to toxic illness in AD has become a hot research topic at present. Currently, many transcription factors, downstream signaling pathways, and molecular mechanisms that regulate the polarization of microglia are being explored. Furthermore, microglia may also exert a complex role in AD through the transformation of Aβ plaques or debris clearance, reflected in Aβ phagocytosis. One of the mediators of neuroinflammation in AD is the activated microglia. Therefore, the regulation of microglial function may be the key to successfully treating AD. Methods: This paper is a review article. PubMed, Embase, Scopus, and research meeting abstracts were searched up to 2024 for studies of microglia and neuroinflammation in Alzheimer’s Disease. Systematic information retrieval was performed, and appropriate studies were isolated based on important information available in the studies. The information from each of the articles was understood and extracted to form a database. Results: The similar neuropathological results between several animals and AD cases show the possibility of implementing microglia-related changes as an earlier diagnostic marker for AD in humans. The gene sets identified in various transcriptomic studies further foster this avenue of research by offering potential targets for therapeutic development. Substantial evidence, both in vitro and in vivo, has suggested that the loss of the normal A2 phenotype and the activation of toxic A1 microglia contribute to neurodegeneration in AD. Conclusions: Promoting or restoring the polarization of microglia towards the A2 phenotype may thus represent an effective therapeutic strategy for ameliorating neuroinflammation and progressive neurocognitive impairments. Multiple studies suggest that microglia-associated neuroinflammation at a special stage could also be protective, and, therefore, intervention should be delicate so that a beneficial response is retained.
Collapse
Affiliation(s)
- Siddharth Shah
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA
| | - Hritvik Jain
- Department of Internal Medicine, All India Institute of Medical Sciencies, Jodhpur 342005, India
| |
Collapse
|
50
|
Dai M, Sun S, Dai Y, Dou X, Yang J, Chen X, Yang D, Lin Y. Maresin-1 Ameliorates Sepsis-Induced Microglial Activation Through Modulation of the P38 MAPK Pathway. Neurochem Res 2024; 50:26. [PMID: 39565476 DOI: 10.1007/s11064-024-04280-z] [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: 03/31/2024] [Revised: 07/10/2024] [Accepted: 11/05/2024] [Indexed: 11/21/2024]
Abstract
Sepsis is a life-threatening disease characterized by a dysregulated immune response to infection, often leading to neuroinflammation. As a known immunomodulator, Maresin-1 (MaR1) may have potential applications in the treatment of sepsis-induced neuroinflammation, but its effects in this context are unknown. We used a mouse cecum ligation and puncture (CLP)-induced sepsis model and an in vitro lipopolysaccharide (LPS)-induced neuroinflammatory model of BV2 microglia. Expression of microglial cell markers (IBA1, CD11B, CD68, CD86 and CD206) and pro-inflammatory markers (iNOS and COX2) was assessed. The role of MaR1 in regulating the P38 MAPK pathway was explored using the P38 MAPK inhibitor SB203580. In the CLP model, an increased proportion of M1-type microglia was observed, and MaR1 was able to reverse it. However, the combination of SB203580 and MaR1 did not enhance the therapeutic effect compared to SB20580 alone. In vitro experiments, MaR1 inhibited LPS-induced P38 MAPK nuclear translocation and decreased the expression of pro-inflammatory markers such as iNOS and COX2. As with the animal results, no stacking effect could be obtained with the co-administration of SB203580 and MaR1. Our findings suggest that MaR1 attenuates sepsis-induced neuroinflammation mainly by inhibiting phosphorylation of P38 MAPK in microglial cells. This suggests that MaR1 may have a potential therapeutic role in the treatment of sepsis neuroinflammation.
Collapse
Affiliation(s)
- Maosha Dai
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Shujun Sun
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
- Department of Pain, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
| | - Yan Dai
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Xiaoke Dou
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Juexi Yang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Xiangdong Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China.
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China.
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology), Ministry of Education, Wuhan, China.
| | - Dong Yang
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China.
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology), Ministry of Education, Wuhan, China.
- Department of Pain, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China.
| | - Yun Lin
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China.
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China.
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology), Ministry of Education, Wuhan, China.
| |
Collapse
|