51
|
Liu W, Rao X, Sun W, Chen X, Yu L, Zhang J, Chen J, Zheng X. The neuroinflammatory role of microRNAs in Alzheimer's disease: pathological insights to therapeutic potential. Mol Cell Biochem 2024:10.1007/s11010-024-05164-0. [PMID: 39567427 DOI: 10.1007/s11010-024-05164-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 11/10/2024] [Indexed: 11/22/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease and the most common cause of dementia, contributing to around 60-80% of cases. The main pathophysiology of AD is characterized by an abnormal accumulation of protein aggregates extracellularly (beta-amyloid plaques) and intracellularly (neurofibrillary tangles of hyperphosphorylated tau). However, an increasing number of studies have also suggested neuroinflammation may have a crucial role in precipitating the cascade reactions that result in the development of AD neuropathology. In particular, several studies indicate microRNAs (miRNAs) can act as regulatory factors for neuroinflammation in AD, with potential to affect the occurrence and/or progression of AD inflammation by targeting the expression of multiple genes. Therefore, miRNAs may have potential as therapeutic targets for AD, which requires more research. This article will review the existing studies on miRNAs that have been identified to regulate neuroinflammation, aiming to gain further insights into the specific regulatory processes of miRNAs, highlight the diagnostic and therapeutic potential of miRNAs as biomarkers in AD, as well as current challenges, and suggest the further work to bridge the gap in knowledge to utilize miRNAs as therapeutic targets for AD.
Collapse
Affiliation(s)
- Wenjia Liu
- School of Electronics and Information, Hangzhou Dianzi University, Hangzhou, 310018, China
| | - Xin Rao
- School of Electronics and Information, Hangzhou Dianzi University, Hangzhou, 310018, China.
| | - Wen Sun
- School of Electronics and Information, Hangzhou Dianzi University, Hangzhou, 310018, China
| | - Xiaodong Chen
- School of Electronics and Information, Hangzhou Dianzi University, Hangzhou, 310018, China.
| | - Liyang Yu
- School of Electronics and Information, Hangzhou Dianzi University, Hangzhou, 310018, China
| | - Jiangtao Zhang
- Department of Geriatrics, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, China.
| | - Jiong Chen
- Department of Geriatrics, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, China
| | - Xiaorong Zheng
- Blood Purification Center, The Second Affiliated Hospital of Jiaxing University, Jiaxing, 314000, China
| |
Collapse
|
52
|
Xu J, Zhang C, Yan Z, Fan C, Yuan S, Wang J, Zhu Y, Luo L, Shi K, Deng J. Dental Pulp Stem Cell Lysate-Based Hydrogel Improves Diabetic Wound Healing via the Regulation of Anti-Inflammatory Macrophages and Keratinocytes. ACS APPLIED BIO MATERIALS 2024; 7:7684-7699. [PMID: 39503733 DOI: 10.1021/acsabm.4c01157] [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: 11/08/2024]
Abstract
The prolonged existence of chronic wounds heightens the risk of patients experiencing chronic pain, necrosis, and amputation. Dental pulp stem cells (DPSCs) have garnered attention due to their potential immunomodulatory and tissue repair regenerative effects in the management of chronic wounds. However, stem-cell-based therapy faces challenges such as malignant differentiation, immune rejection, and long-term effectiveness. To overcome these challenges, we proposed a chronic wound therapy using a hydrogel derived from human-originated dental pulp stem cell lysate (DPSCL). Our data indicate that, with the degradation of the dental pulp stem cell lysate-based hydrogel (DPSCLH), the slowly released cell lysates recruit anti-inflammatory M2 macrophages and promote the proliferation, migration, and keratinization of HacaT cells. In addition, in vivo studies revealed that DPSCLH avoids immune rejection reactions and induces a long-term accumulation of endogenous M2 macrophages. In a mouse model of diabetic wounds, DPSCLH effectively modulates the inflammatory microenvironment around diabetic wounds, promotes the formation of the stratum corneum, and facilitates the healing of wounds, thus holding tremendous potential for the treatment of diabetic wounds.
Collapse
Affiliation(s)
- Jianghua Xu
- Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
| | - Changhuan Zhang
- First People's Hospital of Linping District, Hangzhou, Hangzhou, Zhejiang 311100, China
| | - Zhuo Yan
- Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
| | - Chen Fan
- Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
| | - Shanshan Yuan
- Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
| | - Jilong Wang
- Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
| | - Yuting Zhu
- Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
| | - Lihua Luo
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Keqing Shi
- Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Junjie Deng
- Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
| |
Collapse
|
53
|
Sighencea MG, Popescu RȘ, Trifu SC. From Fundamentals to Innovation in Alzheimer's Disease: Molecular Findings and Revolutionary Therapies. Int J Mol Sci 2024; 25:12311. [PMID: 39596378 PMCID: PMC11594972 DOI: 10.3390/ijms252212311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/11/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Alzheimer's disease (AD) is a global health concern and the leading cause of dementia in the elderly. The prevalence of this neurodegenerative condition is projected to increase concomitantly with increased life expectancy, resulting in a significant economic burden. With very few FDA-approved disease-modifying drugs available for AD, there is an urgent need to develop new compounds capable of impeding the progression of the disease. Given the unclear etiopathogenesis of AD, this review emphasizes the underlying mechanisms of this condition. It explores not only well-studied aspects, such as the accumulation of Aβ plaques and neurofibrillary tangles, but also novel areas, including glymphatic and lymphatic pathways, microbiota and the gut-brain axis, serotoninergic and autophagy alterations, vascular dysfunction, the metal hypothesis, the olfactory pathway, and oral health. Furthermore, the potential molecular targets arising from all these mechanisms have been reviewed, along with novel promising approaches such as nanoparticle-based therapy, neural stem cell transplantation, vaccines, and CRISPR-Cas9-mediated genome editing techniques. Taking into account the overlap of these various mechanisms, individual and combination therapies emerge as the future direction in the AD strategy.
Collapse
Affiliation(s)
| | - Ramona Ștefania Popescu
- Department of Infectious Diseases, “Carol Davila” University of Medicine and Pharmacy Bucharest, 020021 Bucharest, Romania;
| | - Simona Corina Trifu
- Department of Psychiatry, “Carol Davila” University of Medicine and Pharmacy Bucharest, 020021 Bucharest, Romania
| |
Collapse
|
54
|
Paidlewar M, Kumari S, Dhapola R, Sharma P, HariKrishnaReddy D. Unveiling the role of astrogliosis in Alzheimer's disease Pathology: Insights into mechanisms and therapeutic approaches. Int Immunopharmacol 2024; 141:112940. [PMID: 39154532 DOI: 10.1016/j.intimp.2024.112940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/30/2024] [Accepted: 08/12/2024] [Indexed: 08/20/2024]
Abstract
Alzheimer's disease (AD) is one of the most debilitating age-related disorders that affect people globally. It impacts social and cognitive behavior of the individual and is characterized by phosphorylated tau and Aβ accumulation. Astrocytesmaintain a quiescent, anti-inflammatory state on anatomical level, expressing few cytokines and exhibit phagocytic activity to remove misfolded proteins. But in AD, in response to specific stimuli, astrocytes overstimulate their phagocytic character with overexpressing cytokine gene modules. Upon interaction with generated Aβ and neurofibrillary tangle, astrocytes that are continuously activated release a large number of inflammatory cytokines. This cytokine storm leads to neuroinflammation which is also one of the recognizable features of AD. Astrogliosis eventually promotes cholinergic dysfunction, calcium imbalance, oxidative stress and excitotoxicity. Furthermore, C5aR1, Lcn2/, BDNF/TrkB and PPARα/TFEB signaling dysregulation has a major impact on the disease progression. This review clarifies numerous ways that lead to astrogliosis, which is stimulated by a variety of processes that exacerbate AD pathology and make it a suitable target for AD treatment. Drugs under clinical and preclinical investigations that target several pathways managing astrogliosis and are efficacious in ameliorating the pathology of the disease are also included in this study. D-ALA2GIP, TRAM-34, Genistein, L-serine, MW150 and XPro1595 are examples of few drugs targeting astrogliosis. Therefore, this study may aid in the development of a potent therapeutic agent for ameliorating astrogliosis mediated AD progression.
Collapse
Affiliation(s)
- Mohit Paidlewar
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda-151401, Punjab, India
| | - Sneha Kumari
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda-151401, Punjab, India
| | - Rishika Dhapola
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda-151401, Punjab, India
| | - Prajjwal Sharma
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda-151401, Punjab, India
| | - Dibbanti HariKrishnaReddy
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda-151401, Punjab, India.
| |
Collapse
|
55
|
Wen X, Hu J. Targeting STAT3 signaling pathway in the treatment of Alzheimer's disease with compounds from natural products. Int Immunopharmacol 2024; 141:112936. [PMID: 39163684 DOI: 10.1016/j.intimp.2024.112936] [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: 06/12/2024] [Revised: 08/12/2024] [Accepted: 08/12/2024] [Indexed: 08/22/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that is difficult to cure and of global concern. Neuroinflammation is closely associated with the onset and progression of AD, making its treatment increasingly important. Compounds from natural products, with fewer side effects than synthetic drugs, are of high research interest. STAT3, a multifunctional transcription factor, is involved in various cellular processes including inflammation, cell growth, and apoptosis. Its activation and inhibition can have different effects under various pathological conditions. In AD, the STAT3 protein plays a crucial role in promoting neuroinflammation and contributing to disease progression. This occurs primarily through the JAK2-STAT3 signaling pathway, which impacts microglia, astrocytes, and hippocampal neurons. This paper reviews the STAT3 signaling pathway in AD and 25 compounds targeting STAT3 up to 2024. Notably, Rutin, Paeoniflorin, and Geniposide up-regulate STAT3 in hippocampal and cortex neurons, showing neuroprotective effects in various AD models. Other 23 compounds downregulate AD by suppressing neuroinflammation through inhibition of STAT3 activation in microglia and astrocytes. These findings highlight the potential of compounds from natural products in improving AD by targeting STAT3, offering insights into the prevention and management of AD.
Collapse
Affiliation(s)
- Xiyue Wen
- Department of Clinical Laboratory, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha 410004, China
| | - Jinyue Hu
- Medical Research Center, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha 410004, China.
| |
Collapse
|
56
|
Castolo-Sanchez S, Zamilpa A, Herrera-Ruiz M, Trejo-Espino JL, Domínguez-Mendoza BE, González-Cortazar M, Trejo-Tapia G. Phytochemicals from Passiflora coriacea Juss. Have Anti-Inflammatory and Neuroprotective Effects in Mouse Models. Pharmaceuticals (Basel) 2024; 17:1534. [PMID: 39598443 PMCID: PMC11597510 DOI: 10.3390/ph17111534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/05/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024] Open
Abstract
Background: Neuroinflammatory diseases trigger an inflammatory response and a state of oxidative stress. Passiflora coriacea Juss. has been used to treat conditions related to inflammatory processes in the central nervous system; however, to date, there has been no study on the anti-inflammatory and neuroprotective effects of this species. Methods: The anti-inflammatory effect of P. coriacea was evaluated in a TPA-induced auricular edema model, and the percentage of edema inhibition (Ei) was recorded. The Morris water maze was used to assess the neuroprotective effect, measuring the latency time (LT), and lipopolysaccharide was administered to induce neuroinflammation. The concentrations of cytokines (IL-6, IL-10, and TNF-α) and activities of antioxidant system components (CAT, SOD, GR, NO, and MDA) were measured in the mouse brains. The chemical composition was determined using chromatographic and nuclear magnetic resonance techniques. Results: T1.1, T2.1, and T3.1 showed anti-inflammatory (Ei = 92.5, 88.3, and 64.8%, respectively) and neuroprotective (LT = 27.2, 22.9, and 27.7 s, respectively) effects. T1.1 was identified as scopolin with immunomodulatory (IL-6 = 3307 pg/g) and antioxidant (CAT = 1198 mmol, SOD = 23%, GR = 5.34 units/mL, NO = 11.5 µM, MDA = 1526 nmol/mL) effects; T2.1 was a mixture of terpenes (fitone, 7-dehydrodiosgenin, tremulone) with immunomodulatory (TNF-α = 857 pg/g) and antioxidant (CAT = 1245 mmol, NO = 8.75 µM) effects; and T3.1 was a mixture of isoquercetin and astragalin with immunomodulatory (IL-6 = 3135 pg/g, IL-10 = 1300 pg/g, TNF-α = 751 pg/g) and antioxidant (SOD = 1204 nmol/mL, CAT = 1131 nmol/mL, NO = 6.37 µM, MDA = 1204 nmol/mL) effects. Conclusions: The administration of P. coriacea treatments generated anti-inflammatory, neuroprotective, immunomodulatory, and antioxidant effects. These effects are attributable to its chemical composition, comprising scopolin, terpenes, and a mixture of isoquercetin and astragalin, which have not previously been described in this species.
Collapse
Affiliation(s)
- Samir Castolo-Sanchez
- Centro de Desarrollo de Productos Bióticos (CEPROBI), Instituto Politécnico Nacional (IPN), Yautepec 62739, Morelos, Mexico; (S.C.-S.); (J.L.T.-E.)
- Centro de Investigación Biomédica del Sur (CIBIS), Instituto Mexicano del Seguro Social (IMSS), Xochitepec 62790, Morelos, Mexico (M.G.-C.)
| | - Alejandro Zamilpa
- Centro de Investigación Biomédica del Sur (CIBIS), Instituto Mexicano del Seguro Social (IMSS), Xochitepec 62790, Morelos, Mexico (M.G.-C.)
| | - Maribel Herrera-Ruiz
- Centro de Investigación Biomédica del Sur (CIBIS), Instituto Mexicano del Seguro Social (IMSS), Xochitepec 62790, Morelos, Mexico (M.G.-C.)
| | - José Luis Trejo-Espino
- Centro de Desarrollo de Productos Bióticos (CEPROBI), Instituto Politécnico Nacional (IPN), Yautepec 62739, Morelos, Mexico; (S.C.-S.); (J.L.T.-E.)
| | - Blanca Eda Domínguez-Mendoza
- Centro de Investigaciones Químicas (CIQ), Universidad Autónoma del Estado de Morelos (UAEM), Cuernavaca 62210, Morelos, Mexico;
| | - Manasés González-Cortazar
- Centro de Investigación Biomédica del Sur (CIBIS), Instituto Mexicano del Seguro Social (IMSS), Xochitepec 62790, Morelos, Mexico (M.G.-C.)
| | - Gabriela Trejo-Tapia
- Centro de Desarrollo de Productos Bióticos (CEPROBI), Instituto Politécnico Nacional (IPN), Yautepec 62739, Morelos, Mexico; (S.C.-S.); (J.L.T.-E.)
| |
Collapse
|
57
|
Liu M, Pan J, Li X, Zhang X, Tian F, Li M, Wu X, Zhang L, Qin C. Interleukin-6 deficiency reduces neuroinflammation by inhibiting the STAT3-cGAS-STING pathway in Alzheimer's disease mice. J Neuroinflammation 2024; 21:282. [PMID: 39487531 PMCID: PMC11529443 DOI: 10.1186/s12974-024-03277-3] [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: 09/01/2024] [Accepted: 10/26/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND The Interleukin-6 (IL-6)-signal transducer and activator of transcription 3 (STAT3) pathway, along with the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway, are critical contributors to neuroinflammation in Alzheimer's disease (AD). Although previous research outside the context of AD has indicated that the IL-6-STAT3 pathway may regulate the cGAS-STING pathway, the exact molecular mechanisms through which IL-6-STAT3 influences cGAS-STING in AD are still not well understood. METHODS The activation of the IL-6-STAT3 and cGAS-STING pathways in the hippocampus of 5×FAD and WT mice was analyzed using WB and qRT-PCR. To explore the effects of IL-6 deficiency, Il6+/- mice were crossed with 5×FAD mice, and the subsequent impact on hippocampal STAT3 pathway activity, cGAS-STING pathway activation, amyloid pathology, neuroinflammation, and cognitive function was evaluated through WB, qRT-PCR, immunohistochemistry, ThS staining, ELISA, and behavioral tests. The regulatory role of STAT3 in the transcription of the Cgas and Sting genes was further validated using ChIP-seq and ChIP-qPCR on hippocampal tissue from 5×FAD and Il6-/-: 5×FAD mice. Additionally, in the BV2 microglial cell line, the impact of STAT3 activation on the transcriptional regulation of Cgas and Sting genes, as well as the production of inflammatory mediators, was examined through WB and qRT-PCR. RESULTS We observed marked activation of the IL-6-STAT3 and cGAS-STING pathways in the hippocampus of AD mice, which was attenuated in the absence of IL-6. IL-6 deficiency reduced beta-amyloid deposition and neuroinflammation in the hippocampus of AD mice, contributing to cognitive improvements. Further analysis revealed that STAT3 directly regulates the transcription of both the Cgas and Sting genes. These findings suggest a potential mechanism involving the STAT3-cGAS-STING pathway, wherein IL-6 deficiency mitigates neuroinflammation in AD mice by modulating this pathway. CONCLUSION These findings indicate that the STAT3-cGAS-STING pathway is critical in mediating neuroinflammation associated with AD and may represent a potential therapeutic target for modulating this inflammatory process in AD.
Collapse
Affiliation(s)
- Min Liu
- Institute of Laboratory Animal Science, CAMS & Comparative Medicine Center, PUMC, Beijing, China
- National Human Diseases Animal Model Resource Center, Beijing, China
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing, China
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Beijing, China
- National Center of Technology Innovation for Animal Model, Beijing, China
| | - Jirong Pan
- Institute of Laboratory Animal Science, CAMS & Comparative Medicine Center, PUMC, Beijing, China
- National Human Diseases Animal Model Resource Center, Beijing, China
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing, China
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Beijing, China
- National Center of Technology Innovation for Animal Model, Beijing, China
| | - Xiaomeng Li
- Institute of Laboratory Animal Science, CAMS & Comparative Medicine Center, PUMC, Beijing, China
- National Human Diseases Animal Model Resource Center, Beijing, China
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing, China
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Beijing, China
- National Center of Technology Innovation for Animal Model, Beijing, China
| | - Xueling Zhang
- Institute of Laboratory Animal Science, CAMS & Comparative Medicine Center, PUMC, Beijing, China
- National Human Diseases Animal Model Resource Center, Beijing, China
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing, China
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Beijing, China
- National Center of Technology Innovation for Animal Model, Beijing, China
| | - Fan Tian
- Institute of Laboratory Animal Science, CAMS & Comparative Medicine Center, PUMC, Beijing, China
- National Human Diseases Animal Model Resource Center, Beijing, China
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing, China
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Beijing, China
- National Center of Technology Innovation for Animal Model, Beijing, China
| | - Mingfeng Li
- Institute of Laboratory Animal Science, CAMS & Comparative Medicine Center, PUMC, Beijing, China
- National Human Diseases Animal Model Resource Center, Beijing, China
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing, China
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Beijing, China
- National Center of Technology Innovation for Animal Model, Beijing, China
| | - Xinghan Wu
- Institute of Laboratory Animal Science, CAMS & Comparative Medicine Center, PUMC, Beijing, China
- National Human Diseases Animal Model Resource Center, Beijing, China
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing, China
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Beijing, China
- National Center of Technology Innovation for Animal Model, Beijing, China
| | - Ling Zhang
- Institute of Laboratory Animal Science, CAMS & Comparative Medicine Center, PUMC, Beijing, China.
- National Human Diseases Animal Model Resource Center, Beijing, China.
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing, China.
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Beijing, China.
- National Center of Technology Innovation for Animal Model, Beijing, China.
| | - Chuan Qin
- Institute of Laboratory Animal Science, CAMS & Comparative Medicine Center, PUMC, Beijing, China.
- Changping National Laboratory, Beijing, China.
- National Human Diseases Animal Model Resource Center, Beijing, China.
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing, China.
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Beijing, China.
- National Center of Technology Innovation for Animal Model, Beijing, China.
| |
Collapse
|
58
|
Sharma M, Pal P, Gupta SK. Deciphering the role of miRNAs in Alzheimer's disease: Predictive targeting and pathway modulation - A systematic review. Ageing Res Rev 2024; 101:102483. [PMID: 39236856 DOI: 10.1016/j.arr.2024.102483] [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: 01/23/2024] [Revised: 08/12/2024] [Accepted: 08/29/2024] [Indexed: 09/07/2024]
Abstract
Alzheimer's Disease (AD), a multifaceted neurodegenerative disorder, is increasingly understood through the regulatory lens of microRNAs (miRNAs). This review comprehensively examines the pivotal roles of miRNAs in AD pathogenesis, shedding light on their influence across various pathways. We delve into the biogenesis and mechanisms of miRNAs, emphasizing their significant roles in brain function and regulation. The review then navigates the complex landscape of AD pathogenesis, identifying key genetic, environmental, and molecular factors, with a focus on hallmark pathological features like amyloid-beta accumulation and tau protein hyperphosphorylation. Central to our discussion is the intricate involvement of miRNAs in these processes, highlighting their altered expression patterns in AD and subsequent functional implications, from amyloid-beta metabolism to tau pathology, neuroinflammation, oxidative stress, and synaptic dysfunction. The predictive analysis of miRNA targets using computational methods, complemented by experimental validations, forms a crucial part of our discourse, unraveling the contributions of specific miRNAs to AD. Moreover, we explore the therapeutic potential of miRNAs as biomarkers and in miRNA-based interventions, while addressing the challenges in translating these findings into clinical practice. This review aims to enhance understanding of miRNAs in AD, offering a foundation for future research directions and novel therapeutic strategies.
Collapse
Affiliation(s)
- Monika Sharma
- Department of Pharmacy, Banasthali Vidyapith, Rajasthan, India
| | - Pankaj Pal
- IIMT College of Pharmacy, IIMT Group of Colleges, Greater Noida, Uttar Pradesh, India.
| | - Sukesh Kumar Gupta
- KIET School of Pharmacy, KIET Group of Institutions, Ghaziabad, Uttar Pradesh, India; Department of Ophthalmology, Visual and Anatomical Sciences (OVAS), School of Medicine, Wayne State University, USA.
| |
Collapse
|
59
|
Peña-Bautista C, Álvarez-Sánchez L, Balaguer Á, Raga L, García-Vallés L, Baquero M, Cháfer-Pericás C. Defining Alzheimer's Disease through Proteomic CSF Profiling. J Proteome Res 2024; 23:5096-5106. [PMID: 39373095 DOI: 10.1021/acs.jproteome.4c00590] [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: 10/08/2024]
Abstract
Alzheimer disease (AD) is the main cause of dementia, and its complexity is not yet completely understood. Proteomic profiles can provide useful information to explore the pathways involved and the heterogeneity among AD patients. A proteomic analysis was performed in cerebrospinal fluid (CSF) samples from mild cognitive impairment due to AD (MCI-AD) and control individuals; both groups were classified by amyloid β42/amyloid β40 levels in CSF (data available in BioStudies database (S-BSST1456)). The analysis based on PLS regression and volcano plot identified 7 proteins (FOLR2, PPP3CA, SMOC2, STMN1, TAGLN3, TMEM132B, and UCHL1) mainly related to protein phosphorylation, structure maintenance, inflammation, and protein degradation. Enrichment analysis revealed the involvement of different biological processes related to neuronal mechanisms and synapses, lipid and carbohydrate metabolism, immune system and inflammation, vascular, hormones, and response to stimuli, and cell signaling and adhesion. In addition, the proteomic profile showed some association with the levels of AD biomarkers in CSF. Regarding the subtypes, two MCI-AD subgroups were identified: one could be related to synapsis and neuronal functions and the other to innate immunity. The study of the proteomic profile in the CSF of AD patients reflects the heterogeneity of biochemical pathways involved in AD.
Collapse
Affiliation(s)
- Carmen Peña-Bautista
- Alzheimer's Disease Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | - Lourdes Álvarez-Sánchez
- Alzheimer's Disease Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | - Ángel Balaguer
- Faculty of Mathematical Sciences, University of Valencia, 46100 Burjassot, Valencia, Spain
| | - Luis Raga
- Alzheimer's Disease Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | - Lorena García-Vallés
- Alzheimer's Disease Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | - Miguel Baquero
- Division of Neurology, Hospital Universitari I Politècnic La Fe, 46026 Valencia, Spain
| | - Consuelo Cháfer-Pericás
- Alzheimer's Disease Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| |
Collapse
|
60
|
Li X, Ding Z. Cognitive dysfunction induced by cranial radiotherapy: mechanisms and therapeutic methods. Brain Res Bull 2024; 218:111106. [PMID: 39447765 DOI: 10.1016/j.brainresbull.2024.111106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 10/19/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024]
Abstract
Cranial radiotherapy can damage normal brain tissues, inducing cognitive dysfunction in patients. Radiotherapy-induced cognitive dysfunction is associated with hippocampal injury, white matter damage and microvascular injury. In this study, the mechanisms of cognitive dysfunction induced by cranial radiotherapy and combined chemoradiotherapy are reviewed, and the advances in therapeutic methods for radiotherapy-induced brain injury are summarized. The mechanisms of radiotherapy-induced brain injury include a decline of neurogenesis, impairment of neurons and glial cells, vascular injury, oxidative stress and DNA damage, cell death, and inflammatory response. Disruption of the bloodbrain barrier (BBB) increases the exposure of the brain to chemotherapeutic agents, thus exacerbating radiotherapy-induced brain damage. The current methods used to prevent radiotherapy-induced brain injury mainly include precision radiotherapy, stem cell transplantation, and treatment with neuroprotective drugs. The combined application of precision radiotherapy and neuroprotective drugs, including antioxidants, anti-inflammatory agents and other drugs, might exert better neuroprotective effects. To resolve the issues of neuroprotective drugs, such as difficulty in crossing the BBB, nanoenzymes and drug delivery nano-systems could be applied in the future.
Collapse
Affiliation(s)
- Xuejiao Li
- Department of Radiology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, China
| | - Zhongxiang Ding
- Department of Radiology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, China.
| |
Collapse
|
61
|
Pan Y, Sun X, Tian Y, Yu M, Luo Y, Sun X. L-NRB alleviates amyotrophic lateral sclerosis by regulating P11-Htr4 signaling pathway. Biomed Pharmacother 2024; 180:117588. [PMID: 39427550 DOI: 10.1016/j.biopha.2024.117588] [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: 08/08/2024] [Revised: 10/03/2024] [Accepted: 10/14/2024] [Indexed: 10/22/2024] Open
Abstract
INTRODUCTION L-NRB is a compound formed as a ring cleavage product of butylphthalide and borneol in a molar ratio 1:2. This study aimed to explore the therapeutic effect of L-NRB on amyotrophic lateral sclerosis (ALS) and its possible mechanism. METHODS SOD1-G93A mice were used as an ALS model. Behavioral tests, histopathological staining, Nissl staining, immunohistochemistry, enzyme-linked immunosorbent assays, and Western blotting were used to analyze the therapeutic effect. The underlying mechanism of L-NRB in treating ALS was investigated using transcriptomic analyses. RESULTS It was found that L-NRB alleviated motor dysfunction, pathological changes in the gastrocnemius muscle, and motor neuron injuries. The results indicated that L-NRB had a neuroprotective function associated with the inhibition of neuroinflammation. The anti-apoptotic effect of L-NRB was found to be related to the regulation of the P11-Htr4 signaling pathway. CONCLUSION In summary, the results demonstrated the therapeutic effect of L-NRB on ALS and suggest a promising new therapeutic candidate for ALS.
Collapse
Affiliation(s)
- Yunfeng Pan
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; Key Laboratory of new drug discovery based on Classic Chinese medicine prescription, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiao Sun
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; Key Laboratory of new drug discovery based on Classic Chinese medicine prescription, Chinese Academy of Medical Sciences, Beijing, China
| | - Yu Tian
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; Key Laboratory of new drug discovery based on Classic Chinese medicine prescription, Chinese Academy of Medical Sciences, Beijing, China
| | - Miao Yu
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; Key Laboratory of new drug discovery based on Classic Chinese medicine prescription, Chinese Academy of Medical Sciences, Beijing, China
| | - Yun Luo
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; Key Laboratory of new drug discovery based on Classic Chinese medicine prescription, Chinese Academy of Medical Sciences, Beijing, China.
| | - Xiaobo Sun
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; Key Laboratory of new drug discovery based on Classic Chinese medicine prescription, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
62
|
Toledano A, Rodríguez-Casado A, Älvarez MI, Toledano-Díaz A. Alzheimer's Disease, Obesity, and Type 2 Diabetes: Focus on Common Neuroglial Dysfunctions (Critical Review and New Data on Human Brain and Models). Brain Sci 2024; 14:1101. [PMID: 39595866 PMCID: PMC11591712 DOI: 10.3390/brainsci14111101] [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: 09/19/2024] [Revised: 10/17/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Obesity, type 2 diabetes (T2D), and Alzheimer's disease (AD) are pathologies that affect millions of people worldwide. They have no effective therapy and are difficult to prevent and control when they develop. It has been known for many years that these diseases have many pathogenic aspects in common. We highlight in this review that neuroglial cells (astroglia, oligodendroglia, and microglia) play a vital role in the origin, clinical-pathological development, and course of brain neurodegeneration. Moreover, we include the new results of a T2D-AD mouse model (APP+PS1 mice on a high-calorie diet) that we are investigating. METHODS Critical bibliographic revision and biochemical neuropathological study of neuroglia in a T2D-AD model. RESULTS T2D and AD are not only "connected" by producing complex pathologies in the same individual (obesity, T2D, and AD), but they also have many common pathogenic mechanisms. These include insulin resistance, hyperinsulinemia, hyperglycemia, oxidative stress, mitochondrial dysfunction, and inflammation (both peripheral and central-or neuroinflammation). Cognitive impairment and AD are the maximum exponents of brain neurodegeneration in these pathological processes. both due to the dysfunctions induced by metabolic changes in peripheral tissues and inadequate neurotoxic responses to changes in the brain. In this review, we first analyze the common pathogenic mechanisms of obesity, T2D, and AD (and/or cerebral vascular dementia) that induce transcendental changes and responses in neuroglia. The relationships between T2D and AD discussed mainly focus on neuroglial responses. Next, we present neuroglial changes within their neuropathological context in diverse scenarios: (a) aging involution and neurodegenerative disorders, (b) human obesity and diabetes and obesity/diabetes models, (c) human AD and in AD models, and (d) human AD-T2D and AD-T2D models. An important part of the data presented comes from our own studies on humans and experimental models over the past few years. In the T2D-AD section, we included the results of a T2D-AD mouse model (APP+PS1 mice on a high-calorie diet) that we investigated, which showed that neuroglial dysfunctions (astrocytosis and microgliosis) manifest before the appearance of amyloid neuropathology, and that the amyloid pathology is greater than that presented by mice fed a normal, non-high-caloric diet A broad review is finally included on pharmacological, cellular, genic, and non-pharmacological (especially diet and lifestyle) neuroglial-related treatments, as well as clinical trials in a comparative way between T2D and AD. These neuroglial treatments need to be included in the multimodal/integral treatments of T2D and AD to achieve greater therapeutic efficacy in many millions of patients. CONCLUSIONS Neuroglial alterations (especially in astroglia and microglia, cornerstones of neuroinflammation) are markedly defining brain neurodegeneration in T2D and A, although there are some not significant differences between each of the studied pathologies. Neuroglial therapies are a very important and p. promising tool that are being developed to prevent and/or treat brain dysfunction in T2D-AD. The need for further research in two very different directions is evident: (a) characterization of the phenotypic changes of astrocytes and microglial cells in each region of the brain and in each phase of development of each isolated and associated pathology (single-cell studies are mandatory) to better understand the pathologies and define new therapeutic targets; (b) studying new therapeutic avenues to normalize the function of neuroglial cells (preventing neurotoxic responses and/or reversing them) in these pathologies, as well as the phenotypic characteristics in each moment of the course and place of the neurodegenerative process.
Collapse
Affiliation(s)
- Adolfo Toledano
- Instituto Cajal, CSIC, 28002 Madrid, Spain; (A.R.-C.); (M.I.Ä.)
| | | | | | | |
Collapse
|
63
|
Ortí-Casañ N, Bethea JR, Eisel ULM. Mapping Alzheimer's disease: exploring cellular vulnerability and resilience. Signal Transduct Target Ther 2024; 9:299. [PMID: 39468007 PMCID: PMC11519876 DOI: 10.1038/s41392-024-02014-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/04/2024] [Accepted: 10/08/2024] [Indexed: 10/30/2024] Open
Affiliation(s)
- Natalia Ortí-Casañ
- Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - John R Bethea
- Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Ross Hall, Washington, DC, USA
| | - Ulrich L M Eisel
- Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
64
|
Hao Z, Zhang X, Wang Y. Evidence of the Long-Term Protective Effect of Moderate-Intensity Physical Activity on Cognitive Function in Middle-Aged and Elderly Individuals: A Predictive Analysis of Longitudinal Studies. Life (Basel) 2024; 14:1343. [PMID: 39459642 PMCID: PMC11509916 DOI: 10.3390/life14101343] [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: 10/01/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
OBJECTIVE To investigate the effects of different intensities of physical activity (PA) on cognitive function in middle-aged and elderly individuals, and to predict future trends in cognitive ability using longitudinal data to assess the long-term role of PA in cognitive preservation. METHODS Data from the China Health and Retirement Longitudinal Study (CHARLS) were utilized. Mixed-effects models were employed to analyze the impacts of low-intensity PA (LPA), moderate-intensity PA (MPA), and vigorous-intensity PA (VPA) on overall cognition, episodic memory, and mental intactness. Random forest and XGBoost machine learning methods were employed to further validate the effects of PA. ARIMA models predicted future cognitive trends under the influence of PA. RESULTS MPA demonstrated significant advantages in preserving cognitive function, particularly in overall cognition and episodic memory. While LPA had some protective effects, they were less significant than those of MPA, and VPA did not show advantages. Machine learning methods confirmed these findings. ARIMA model predictions indicated that the protective effects of MPA on cognitive function are likely to persist in the future. CONCLUSIONS Moderate-intensity physical activity is associated with the preservation of cognitive ability in middle-aged and elderly individuals and may continue to provide this benefit in the future; however, further in-depth research is needed for confirmation.
Collapse
Affiliation(s)
- Zikang Hao
- School of Physical Education, Shandong University, Jinan 250061, China
- Exercise Science Laboratory, Department of Physical Education, Ocean University of China, Qingdao 266005, China
| | - Xianliang Zhang
- School of Physical Education, Shandong University, Jinan 250061, China
| | - Yu Wang
- Department of Physical Education, Moscow State University of Sport and Tourism, Kirovogradskaya Street, 21, Moscow 117519, Russia
| |
Collapse
|
65
|
Cafferata EA, Ramanauskaite A, Cuypers A, Obreja K, Dohle E, Ghanaati S, Schwarz F. Experimental peri-implantitis induces neuroinflammation: An exploratory study in rats. BMC Oral Health 2024; 24:1238. [PMID: 39425138 PMCID: PMC11490110 DOI: 10.1186/s12903-024-04995-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: 06/05/2024] [Accepted: 10/01/2024] [Indexed: 10/21/2024] Open
Abstract
PURPOSE Cumulating evidence supports the close association between periodontal diseases, neuroinflammation and neurodegenerative pathologies, except for peri-implantitis (PI). Thus, this study explored the association between experimental PI and neuropathological changes in the rat brain. MATERIALS AND METHODS After bilateral first molars extraction, experimental PI was induced at titanium implants placed in the maxillae by lipopolysaccharide injections and ligature placement. Following 28-weeks of disease progression, the maxillae and brains were retrieved from 6 rats. Healthy brains from 3 rats were used as control. Brains were analyzed by immunohistochemistry to detect signs of neuroinflammation (interleukin (IL)-6 and tumor necrosis factor (TNF)-α)), microglial activation (IBA-1) and astrogliosis (GFAP). To explore signs of neurodegeneration, hematoxylin/eosin and Nissl stainings were used. Also, four different antibodies against amyloid beta (Aβ 1-42) were tested. RESULTS Chronic PI lesions showed peri-implant bone resorption accompanied by large inflammatory infiltrates. IL-6+ and TNF-α+ cells were found within the CA1 and Dentate Gyrus regions of the hippocampus of the PI-affected group, while almost no immune-positivity was detected in the control (p < 0.05). Detection of activated GFAP+ microglia and IBA-1+ astrocytes surface were significantly higher at the CA areas, and cerebral cortex of the PI-affected group, in comparison with control (p < 0.05). Shrunk neurons with pyknotic nuclei were inconsistently found among the PI-affected group, and these were almost not detected in control. No positive Aβ reactivity was detected in any of the samples. CONCLUSION Chronic experimental PI lesions led to an increased detection of IL-6 and TNF-α, GFAP+ microgliosis and IBA-1+ astrocytosis, and in some cases, neurodegeneration, in the rat brain.
Collapse
Affiliation(s)
- Emilio A Cafferata
- Department of Oral Surgery and Implantology, Goethe University, Carolinum, Frankfurt am Main, Germany.
- Department of Periodontology, School of Dentistry, Universidad Científica del Sur, Lima, Perú.
| | - Ausra Ramanauskaite
- Department of Oral Surgery and Implantology, Goethe University, Carolinum, Frankfurt am Main, Germany
| | - Astrid Cuypers
- Department of Oral Surgery and Implantology, Goethe University, Carolinum, Frankfurt am Main, Germany
| | - Karina Obreja
- Department of Oral Surgery and Implantology, Goethe University, Carolinum, Frankfurt am Main, Germany
| | - Eva Dohle
- Frankfurt Oral Regenerative Medicine (FORM-Lab), Clinic for Maxillofacial and Plastic Surgery, Goethe University, Frankfurt am Main, Germany
| | - Shahram Ghanaati
- Frankfurt Oral Regenerative Medicine (FORM-Lab), Clinic for Maxillofacial and Plastic Surgery, Goethe University, Frankfurt am Main, Germany
| | - Frank Schwarz
- Department of Oral Surgery and Implantology, Goethe University, Carolinum, Frankfurt am Main, Germany
| |
Collapse
|
66
|
Diniz DG, Bento-Torres J, da Costa VO, Carvalho JPR, Tomás AM, Galdino de Oliveira TC, Soares FC, de Macedo LDED, Jardim NYV, Bento-Torres NVO, Anthony DC, Brites D, Picanço Diniz CW. The Hidden Dangers of Sedentary Living: Insights into Molecular, Cellular, and Systemic Mechanisms. Int J Mol Sci 2024; 25:10757. [PMID: 39409085 PMCID: PMC11476792 DOI: 10.3390/ijms251910757] [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: 08/16/2024] [Revised: 09/23/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
With the aging of the global population, neurodegenerative diseases are emerging as a major public health issue. The adoption of a less sedentary lifestyle has been shown to have a beneficial effect on cognitive decline, but the molecular mechanisms responsible are less clear. Here we provide a detailed analysis of the complex molecular, cellular, and systemic mechanisms underlying age-related cognitive decline and how lifestyle choices influence these processes. A review of the evidence from animal models, human studies, and postmortem analyses emphasizes the importance of integrating physical exercise with cognitive, multisensory, and motor stimulation as part of a multifaceted approach to mitigating cognitive decline. We highlight the potential of these non-pharmacological interventions to address key aging hallmarks, such as genomic instability, telomere attrition, and neuroinflammation, and underscore the need for comprehensive and personalized strategies to promote cognitive resilience and healthy aging.
Collapse
Affiliation(s)
- Daniel Guerreiro Diniz
- Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Seção de Hepatologia, Belém 66.093-020, Pará, Brazil;
- Núcleo de Pesquisas em Oncologia, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66.073-005, Pará, Brazil;
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66.073-005, Pará, Brazil; (J.B.-T.); (V.O.d.C.); (J.P.R.C.); (A.M.T.); (T.C.G.d.O.); (F.C.S.); (L.D.e.D.d.M.); (N.Y.V.J.)
| | - João Bento-Torres
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66.073-005, Pará, Brazil; (J.B.-T.); (V.O.d.C.); (J.P.R.C.); (A.M.T.); (T.C.G.d.O.); (F.C.S.); (L.D.e.D.d.M.); (N.Y.V.J.)
- Programa de Pós-Graduação em Ciências do Movimento Humano, Universidade Federal do Pará, Belém 66.050-160, Pará, Brazil
| | - Victor Oliveira da Costa
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66.073-005, Pará, Brazil; (J.B.-T.); (V.O.d.C.); (J.P.R.C.); (A.M.T.); (T.C.G.d.O.); (F.C.S.); (L.D.e.D.d.M.); (N.Y.V.J.)
| | - Josilayne Patricia Ramos Carvalho
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66.073-005, Pará, Brazil; (J.B.-T.); (V.O.d.C.); (J.P.R.C.); (A.M.T.); (T.C.G.d.O.); (F.C.S.); (L.D.e.D.d.M.); (N.Y.V.J.)
- Programa de Pós-Graduação em Ciências do Movimento Humano, Universidade Federal do Pará, Belém 66.050-160, Pará, Brazil
| | - Alessandra Mendonça Tomás
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66.073-005, Pará, Brazil; (J.B.-T.); (V.O.d.C.); (J.P.R.C.); (A.M.T.); (T.C.G.d.O.); (F.C.S.); (L.D.e.D.d.M.); (N.Y.V.J.)
- Campus Samabaia, Universidade Federal de Goiás (EBTT), CEPAE, Goiânia 74.001-970, Goiás, Brazil
| | - Thaís Cristina Galdino de Oliveira
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66.073-005, Pará, Brazil; (J.B.-T.); (V.O.d.C.); (J.P.R.C.); (A.M.T.); (T.C.G.d.O.); (F.C.S.); (L.D.e.D.d.M.); (N.Y.V.J.)
- Faculdade de Ceilândia, Ceilândia, Universidade de Brasília, Brasília 72.220-900, Brazil
| | - Fernanda Cabral Soares
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66.073-005, Pará, Brazil; (J.B.-T.); (V.O.d.C.); (J.P.R.C.); (A.M.T.); (T.C.G.d.O.); (F.C.S.); (L.D.e.D.d.M.); (N.Y.V.J.)
| | - Liliane Dias e Dias de Macedo
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66.073-005, Pará, Brazil; (J.B.-T.); (V.O.d.C.); (J.P.R.C.); (A.M.T.); (T.C.G.d.O.); (F.C.S.); (L.D.e.D.d.M.); (N.Y.V.J.)
- Campus Tucurui, Universidade do Estado do Pará, Tucurui 68.455-210, Pará, Brazil
| | - Naina Yuki Vieira Jardim
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66.073-005, Pará, Brazil; (J.B.-T.); (V.O.d.C.); (J.P.R.C.); (A.M.T.); (T.C.G.d.O.); (F.C.S.); (L.D.e.D.d.M.); (N.Y.V.J.)
- Campus Tucurui, Universidade do Estado do Pará, Tucurui 68.455-210, Pará, Brazil
- Programa de Pós-Graduação em Neurociências e Biologia Celular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66.075-110, Pará, Brazil
| | - Natáli Valim Oliver Bento-Torres
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66.073-005, Pará, Brazil; (J.B.-T.); (V.O.d.C.); (J.P.R.C.); (A.M.T.); (T.C.G.d.O.); (F.C.S.); (L.D.e.D.d.M.); (N.Y.V.J.)
- Programa de Pós-Graduação em Ciências do Movimento Humano, Universidade Federal do Pará, Belém 66.050-160, Pará, Brazil
| | - Daniel Clive Anthony
- Laboratory of Experimental Neuropathology, Department of Pharmacology, University of Oxford, Oxford OX1 2JD, UK;
| | - Dora Brites
- Faculty of Pharmacy, Department of Pharmaceutical Sciences and Medicines, Universidade de Lisboa, 1649-003 Lisbon, Portugal;
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Cristovam Wanderley Picanço Diniz
- Núcleo de Pesquisas em Oncologia, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66.073-005, Pará, Brazil;
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66.073-005, Pará, Brazil; (J.B.-T.); (V.O.d.C.); (J.P.R.C.); (A.M.T.); (T.C.G.d.O.); (F.C.S.); (L.D.e.D.d.M.); (N.Y.V.J.)
- Programa de Pós-Graduação em Neurociências e Biologia Celular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66.075-110, Pará, Brazil
| |
Collapse
|
67
|
Bhardwaj S, Grewal AK, Singh S, Dhankar V, Jindal A. An insight into the concept of neuroinflammation and neurodegeneration in Alzheimer's disease: targeting molecular approach Nrf2, NF-κB, and CREB. Inflammopharmacology 2024; 32:2943-2960. [PMID: 38951436 DOI: 10.1007/s10787-024-01502-2] [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: 01/03/2024] [Accepted: 06/04/2024] [Indexed: 07/03/2024]
Abstract
Alzheimer's disease (AD) is a most prevalent neurologic disorder characterized by cognitive dysfunction, amyloid-β (Aβ) protein accumulation, and excessive neuroinflammation. It affects various life tasks and reduces thinking, memory, capability, reasoning and orientation ability, decision, and language. The major parts responsible for these abnormalities are the cerebral cortex, amygdala, and hippocampus. Excessive inflammatory markers release, and microglial activation affect post-synaptic neurotransmission. Various mechanisms of AD pathogenesis have been explored, but still, there is a need to debate the role of NF-κB, Nrf2, inflammatory markers, CREB signaling, etc. In this review, we have briefly discussed the signaling mechanisms and function of the NF-ĸB signaling pathway, inflammatory mediators, microglia activation, and alteration of autophagy. NF-κB inhibition is a current strategy to counter neuroinflammation and neurodegeneration in the brain of individuals with AD. In clinical trials, numbers of NF-κB modulators are being examined. Recent reports revealed that molecular and cellular pathways initiate complex pathological competencies that cause AD. Moreover, this review will provide extensive knowledge of the cAMP response element binding protein (CREB) and how these nuclear proteins affect neuronal plasticity.
Collapse
Affiliation(s)
- Shaveta Bhardwaj
- G.H.G. Khalsa College of Pharmacy, Gurusar Sudhar, Ludhiana, India
| | - Amarjot Kaur Grewal
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India.
| | - Shamsher Singh
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India.
| | - Vaibhav Dhankar
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Anu Jindal
- G.H.G. Khalsa College of Pharmacy, Gurusar Sudhar, Ludhiana, India
| |
Collapse
|
68
|
Yan Y, Gao Y, Kumar G, Fang Q, Yan H, Zhang N, Zhang Y, Song L, Li J, Zheng Y, Zhang N, Zhang P, Ma C. Exosomal MicroRNAs modulate the cognitive function in fasudil treated APPswe/PSEN1dE9 transgenic (APP/PS1) mice model of Alzheimer's disease. Metab Brain Dis 2024; 39:1335-1351. [PMID: 39088109 PMCID: PMC11513711 DOI: 10.1007/s11011-024-01395-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 07/15/2024] [Indexed: 08/02/2024]
Abstract
Alzheimer's disease (AD) is characterized by cognitive decline stemming from the accumulation of beta-amyloid (Aβ) plaques and the propagation of tau pathology through synapses. Exosomes, crucial mediators in neuronal development, maintenance, and intercellular communication, have gained attention in AD research. Yet, the molecular mechanisms involving exosomal miRNAs in AD remain elusive. In this study, we treated APPswe/PSEN1dE9 transgenic (APP/PS1) mice, a model for AD, with either vehicle (ADNS) or fasudil (ADF), while C57BL/6 (control) mice received vehicle (WT). Cognitive function was evaluated using the Y-maze test, and AD pathology was confirmed through immunostaining and western blot analysis of Aβ plaques and phosphorylated tau. Exosomal RNAs were extracted, sequenced, and analyzed from each mouse group. Our findings revealed that fasudil treatment improved cognitive function in AD mice, as evidenced by increased spontaneous alternation in the Y-maze test and reduced Aβ plaque load and phosphorylated tau protein expression in the hippocampus. Analysis of exosomal miRNAs identified three miRNAs (mmu-let-7i-5p, mmu-miR-19a-3p, mmu-miR-451a) common to both ADNS vs ADF and WT vs ADNS groups. Utilizing miRTarBase software, we predicted and analyzed target genes associated with these miRNAs. Gene ontology (GO) annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis of miRNA target genes indicated that mmu-miR-19a-3p and mmu-miR-451a are implicated in signal transduction, immune response, cellular communication, and nervous system pathways. Specifically, mmu-miR-19a-3p targeted genes involved in the sphingolipid signaling pathway, such as Pten and Tnf, while mmu-miR-451a targeted Nsmaf, Gnai3, and Akt3. Moreover, mmu-miR-451a targeted Myc in signaling pathways regulating the pluripotency of stem cells. In conclusion, fasudil treatment enhanced cognitive function by modulating exosomal MicroRNAs, particularly mmu-miR-451a and mmu-miR-19a-3p. These miRNAs hold promise as potential biomarkers and therapeutic targets for novel AD treatments.
Collapse
Affiliation(s)
- Yuqing Yan
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China.
| | - Ye Gao
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China
| | - Gajendra Kumar
- Department of Neuroscience, City University of Hong Kong, Kowloon, Hong Kong.
| | - Qingli Fang
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China
| | - Hailong Yan
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China
| | - Nianping Zhang
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China
| | - Yuna Zhang
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China
| | - Lijuan Song
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple, Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Jiehui Li
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China
| | - Yucheng Zheng
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China
| | - Nan Zhang
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China
| | - Peijun Zhang
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China
| | - Cungen Ma
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China.
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple, Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Taiyuan, China.
| |
Collapse
|
69
|
Yang JY, Baek SE, Yoon JW, Kim HS, Kwon Y, Yeom E. Nesfatin-1 ameliorates pathological abnormalities in Drosophila hTau model of Alzheimer's disease. Biochem Biophys Res Commun 2024; 727:150311. [PMID: 38950494 DOI: 10.1016/j.bbrc.2024.150311] [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: 06/03/2024] [Revised: 06/07/2024] [Accepted: 06/23/2024] [Indexed: 07/03/2024]
Abstract
In human Alzheimer's disease (AD), the aggregation of tau protein is considered a significant hallmark, along with amyloid-beta. The formation of neurofibrillary tangles due to aberrant phosphorylation of tau disrupts microtubule stability, leading to neuronal toxicity, dysfunction, and subsequent cell death. Nesfatin-1 is a neuropeptide primarily known for regulating appetite and energy homeostasis. However, the function of Nesfatin-1 in a neuroprotective role has not been investigated. In this study, we aimed to elucidate the effect of Nesfatin-1 on tau pathology using the Drosophila model system. Our findings demonstrate that Nesfatin-1 effectively mitigates the pathological phenotypes observed in Drosophila human Tau overexpression models. Nesfatin-1 overexpression rescued the neurodegenerative phenotypes in the adult fly's eye and bristle. Additionally, Nesfatin-1 improved locomotive behavior, neuromuscular junction formation, and lifespan in the hTau AD model. Moreover, Nesfatin-1 controls tauopathy by reducing the protein level of hTau. Overall, this research highlights the potential therapeutic applications of Nesfatin-1 in ameliorating the pathological features associated with Alzheimer's disease.
Collapse
Affiliation(s)
- Jae-Yoon Yang
- School of Life Science and Biotechnology, College of Natural Sciences, Kyungpook National University, Daegu, 41566, South Korea; School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, South Korea
| | - Si-Eun Baek
- School of Life Science and Biotechnology, College of Natural Sciences, Kyungpook National University, Daegu, 41566, South Korea; School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, South Korea
| | - Jong-Won Yoon
- School of Life Science and Biotechnology, College of Natural Sciences, Kyungpook National University, Daegu, 41566, South Korea; School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, South Korea
| | - Hyo-Sung Kim
- School of Life Science and Biotechnology, College of Natural Sciences, Kyungpook National University, Daegu, 41566, South Korea; School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, South Korea
| | - Younghwi Kwon
- KNU-G LAMP Project Group, KNU-Institute of Basic Sciences, School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu, 41566, South Korea
| | - Eunbyul Yeom
- School of Life Science and Biotechnology, College of Natural Sciences, Kyungpook National University, Daegu, 41566, South Korea; School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, South Korea; KNU-G LAMP Project Group, KNU-Institute of Basic Sciences, School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu, 41566, South Korea.
| |
Collapse
|
70
|
Li L, Chen R, Zhang H, Li J, Huang H, Weng J, Tan H, Guo T, Wang M, Xie J. The epigenetic modification of DNA methylation in neurological diseases. Front Immunol 2024; 15:1401962. [PMID: 39376563 PMCID: PMC11456496 DOI: 10.3389/fimmu.2024.1401962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 09/03/2024] [Indexed: 10/09/2024] Open
Abstract
Methylation, a key epigenetic modification, is essential for regulating gene expression and protein function without altering the DNA sequence, contributing to various biological processes, including gene transcription, embryonic development, and cellular functions. Methylation encompasses DNA methylation, RNA methylation and histone modification. Recent research indicates that DNA methylation is vital for establishing and maintaining normal brain functions by modulating the high-order structure of DNA. Alterations in the patterns of DNA methylation can exert significant impacts on both gene expression and cellular function, playing a role in the development of numerous diseases, such as neurological disorders, cardiovascular diseases as well as cancer. Our current understanding of the etiology of neurological diseases emphasizes a multifaceted process that includes neurodegenerative, neuroinflammatory, and neurovascular events. Epigenetic modifications, especially DNA methylation, are fundamental in the control of gene expression and are critical in the onset and progression of neurological disorders. Furthermore, we comprehensively overview the role and mechanism of DNA methylation in in various biological processes and gene regulation in neurological diseases. Understanding the mechanisms and dynamics of DNA methylation in neural development can provide valuable insights into human biology and potentially lead to novel therapies for various neurological diseases.
Collapse
Affiliation(s)
- Linke Li
- The Center of Obesity and Metabolic Diseases, Department of General Surgery, The Third People’s Hospital of Chengdu and The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
- College of Medicine, Southwest Jiaotong University, Chengdu, China
| | - Rui Chen
- The Center of Obesity and Metabolic Diseases, Department of General Surgery, The Third People’s Hospital of Chengdu and The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
- College of Medicine, Southwest Jiaotong University, Chengdu, China
- Department of Stomatology, The Third People’s Hospital of Chengdu and The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Hui Zhang
- Department of Stomatology, The Third People’s Hospital of Chengdu and The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
- College of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Jinsheng Li
- College of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Hao Huang
- College of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Jie Weng
- College of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Huan Tan
- College of Medicine, Southwest Jiaotong University, Chengdu, China
| | - Tailin Guo
- College of Medicine, Southwest Jiaotong University, Chengdu, China
| | - Mengyuan Wang
- The Center of Obesity and Metabolic Diseases, Department of General Surgery, The Third People’s Hospital of Chengdu and The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
- College of Medicine, Southwest Jiaotong University, Chengdu, China
- Department of Stomatology, The Third People’s Hospital of Chengdu and The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Jiang Xie
- Key Laboratory of Drug Targeting and Drug Delivery of Ministry of Education (MOE), Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, West China School of Pharmacy, Sichuan University, Chengdu, China
- Department of Pediatrics, Chengdu Third People’s Hospital, Chengdu, China
| |
Collapse
|
71
|
Kuang Y, Zhu M, Gu H, Tao Y, Huang H, Chen L. Alkaloids in Uncaria rhynchophylla improves AD pathology by restraining CD4 + T cell-mediated neuroinflammation via inhibition of glycolysis in APP/PS1 mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 331:118273. [PMID: 38703874 DOI: 10.1016/j.jep.2024.118273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 04/18/2024] [Accepted: 04/29/2024] [Indexed: 05/06/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Uncaria rhynchophylla (Miq.) Miq.ex Havil. was a classical medicinal plant exhibiting the properties of extinguishing wind, arresting convulsions, clearing heat and pacifying the liver. Clinically, it could be utilized for the treatment of central nervous system-related diseases, such as Alzheimer's disease. U. rhynchophylla (UR) and its major ingredient alkaloid compounds (URA) have been proved to exert significant neuroprotective effects. However, the potential mechanism aren't fully understood. AIM OF THE STUDY This study systematically examined the therapeutic effects of URA on AD pathology in APP-PS1 mice, and revealed the potential mechanism of action. MATERIALS AND METHODS The cognitive ability was evaluated by morris water maze test in APP-PS1 mice. The H&E staining was used to observe the tissue pathological changes. The ELISA kits were used to detect the level of inflammatory factors. The flow cytometry was used to analyze the percentage of CD4+ effector T cells (Teffs) in spleen. The immunofluorescent staining was performed to count the Teffs and microglia in brain. The protein expression was analyzed by western blot. In vitro, the lymphocyte proliferation induced by ConA was performed by CCK-8 kits. The IFN-γ, IL-17, and TNF-α production were detected by ELISA kits. The effects of URA on glycolysis and the involvement of PI3K/Akt/mTOR signaling pathway was analyzed by Lactic Acid assay kit and western blot in ConA-induced naive T cell. RESULTS URA treatment improved AD pathology effectively as demonstrated by enhanced cognitive ability, decreased Aβ deposit and Tau phosphorylation, as well as reduced neuron apoptosis. Also, the neuroinflammation was significantly alleviated as evidenced by decreased IFN-γ, IL-17 and increased IL-10, TGF-β. Notably, URA treatment down-regulated the percentage of Teffs (Th1 and Th17) in spleen, and reduced the infiltration of Teffs and microglia in brain. Meanwhile, the Treg cell was up-regulated both in spleen and brain. In vitro, URA was capable of attenuating the spleen lymphocyte proliferation and release of inflammatory factors provoked by ConA. Interestingly, glycolysis was inhibited by URA treatment as evidenced by the decrease in Lactic Acid production and expression of HK2 and GLUT1 via regulating PI3K/Akt/mTOR signaling pathway in ConA-induced naive T cell. CONCLUSION This study proved that URA could improve AD pathology which was possibly attributable to the restraints of CD4+ T cell mediated neuroinflammation via inhibiting glycolysis.
Collapse
Affiliation(s)
- Ying Kuang
- National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou, 341000, China
| | - Mengyu Zhu
- National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou, 341000, China
| | - Hongting Gu
- National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou, 341000, China
| | - Yue Tao
- National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou, 341000, China
| | - Hao Huang
- National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou, 341000, China
| | - Lei Chen
- National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou, 341000, China.
| |
Collapse
|
72
|
Detka J, Płachtij N, Strzelec M, Manik A, Sałat K. p38α Mitogen-Activated Protein Kinase-An Emerging Drug Target for the Treatment of Alzheimer's Disease. Molecules 2024; 29:4354. [PMID: 39339348 PMCID: PMC11433989 DOI: 10.3390/molecules29184354] [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: 08/10/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder, characterized by the formation of amyloid β and tau protein aggregates in the brain, neuroinflammation, impaired cholinergic neurotransmission, and oxidative stress, resulting in the gradual loss of neurons and neuronal function, which leads to cognitive and memory deficits in AD patients. Chronic neuroinflammation plays a particularly important role in the progression of AD since the excessive release of proinflammatory cytokines from glial cells (microglia and astrocytes) induces neuronal damage, which subsequently causes microglial activation, thus facilitating further neurodegenerative changes. Mitogen-activated protein kinase (MAPK) p38α is one of the key enzymes involved in the control of innate immune response. The increased activation of the p38α MAPK pathway, observed in AD, has been for a long time associated not only with the maintenance of excessive inflammatory process but is also linked with pathophysiological hallmarks of this disease, and therefore is currently considered an attractive drug target for novel AD therapeutics. This review aims to summarize the current state of knowledge about the involvement of p38α MAPK in different aspects of AD pathophysiology and also provides insight into the possible therapeutic effects of novel p38α MAPK inhibitors, which are currently studied as potential drug candidates for AD treatment.
Collapse
Affiliation(s)
- Jan Detka
- Department of Pharmacodynamics, Chair of Pharmacodynamics, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland; (J.D.); (N.P.); (A.M.)
| | - Natalia Płachtij
- Department of Pharmacodynamics, Chair of Pharmacodynamics, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland; (J.D.); (N.P.); (A.M.)
| | - Martyna Strzelec
- Department of Transplantation, Institute of Pediatrics, Faculty of Medicine, Jagiellonian University Medical College, 265 Wielicka St., 30-663 Krakow, Poland;
| | - Aleksandra Manik
- Department of Pharmacodynamics, Chair of Pharmacodynamics, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland; (J.D.); (N.P.); (A.M.)
| | - Kinga Sałat
- Department of Pharmacodynamics, Chair of Pharmacodynamics, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland; (J.D.); (N.P.); (A.M.)
| |
Collapse
|
73
|
Wang J, Du L, Zhang T, Chu Y, Wang Y, Wang Y, Ji X, Kang Y, Cui R, Zhang G, Liu J, Shi G. Edaravone Dexborneol ameliorates the cognitive deficits of APP/PS1 mice by inhibiting TLR4/MAPK signaling pathway via upregulating TREM2. Neuropharmacology 2024; 255:110006. [PMID: 38763325 DOI: 10.1016/j.neuropharm.2024.110006] [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: 01/30/2024] [Revised: 05/06/2024] [Accepted: 05/16/2024] [Indexed: 05/21/2024]
Abstract
Currently, there are no effective therapeutic agents available to treat Alzheimer's disease (AD). However, edaravone dexborneol (EDB), a novel composite agent used to treat acute ischemic stroke, has recently been shown to exert efficacious neuroprotective effects. However, whether EDB can ameliorate cognitive deficits in AD currently remains unclear. To this end, we explored the effects of EDB on AD and its potential mechanisms using an AD animal model (male APP/PS1 mice) treated with EDB for 10 weeks starting at 6 months of age. Subsequent analyses revealed that EDB-treated APP/PS1 mice exhibited improved cognitive abilities compared to untreated APP/PS1 mice. Administration of EDB in APP/PS1 mice further alleviated neuropathological alterations of the hippocampus, including Aβ deposition, pyramidal cell karyopyknosis, and oxidative damage, and significantly decreased the levels of inflammatory cytokines (IL-1β, IL-6 and TNF-α) and COX-2 in the hippocampus of APP/PS1 mice. Transcriptome sequencing analysis demonstrated the critical role of the inflammatory reaction in EDB treatment in APP/PS1 mice, indicating that the alleviation of the inflammatory reaction by EDB in the hippocampus of APP/PS1 mice was linked to the action of the TREM2/TLR4/MAPK signaling pathway. Further in vitro investigations showed that EDB suppressed neuroinflammation in LPS-stimulated BV2 cells by inhibiting the TLR4/MAPK signaling pathway and upregulating TREM2 expression. Thus, the findings of the present study demonstrate that EDB is a promising therapeutic agent for AD-related cognitive dysfunction.
Collapse
Affiliation(s)
- Jinyang Wang
- Department of Neurology, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China; Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Longyuan Du
- Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Tianyun Zhang
- Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China; Neuroscience Research Center, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yun Chu
- Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yue Wang
- Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yu Wang
- Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China; Neuroscience Research Center, Hebei Medical University, Shijiazhuang, 050017, China
| | - Xiaoming Ji
- Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China; Neuroscience Research Center, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yunxiao Kang
- Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China; Neuroscience Research Center, Hebei Medical University, Shijiazhuang, 050017, China
| | - Rui Cui
- Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China; Neuroscience Research Center, Hebei Medical University, Shijiazhuang, 050017, China
| | - Guoliang Zhang
- Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China; Neuroscience Research Center, Hebei Medical University, Shijiazhuang, 050017, China
| | - Junyan Liu
- Department of Neurology, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
| | - Geming Shi
- Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China; Neuroscience Research Center, Hebei Medical University, Shijiazhuang, 050017, China.
| |
Collapse
|
74
|
Sharma P, Kumari P, Sharma M, Sharma R, Paliwal A, Srivastava S, Ashique S, Bhowmick M, Adnan M, Mir RH. Therapeutic potential of Aloe vera-coated curcumin encapsulated nanoparticles in an Alzheimer-induced mice model: behavioural, biochemical and histopathological evidence. J Microencapsul 2024; 41:403-418. [PMID: 39007845 DOI: 10.1080/02652048.2024.2373715] [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: 11/14/2023] [Accepted: 06/20/2024] [Indexed: 07/16/2024]
Abstract
OBJECTIVE The main purpose of the present study was to evaluate the therapeutic efficacy of Aloe vera-coated curcumin encapsulated nanoparticles in mitigating Alzheimer's disease progression in mice, by examining behavioural changes, biochemical markers, and histopathological alterations, thus elucidating its potential as a treatment strategy. METHODS The green synthesis method was used to synthesise this nanoformulation, which was then characterised using a variety of techniques, including percentage encapsulation efficacy, UV-visible spectroscopy, DLS, FT-IR, FESEM, and EDX. Several in-vivo assessments, including behavioural evaluations, dose optimisation studies, oxidative stress marker estimation, and histological studies, were conducted to determine the potential therapeutic impact of nanoformulation on the Alzheimer-induced mice model. RESULTS The synthesised nanoparticles show a mean diameter of 76.12 nm ±1.23, a PDI of 0.313 ± 0.02, a zeta potential of 6.27 ± 0.65 mV, and the percentage encapsulation efficiency between 90% and 95% indicating good stability of synthesised nanoformulation. With the help of Morris water maze, Y-maze, and novel object recognition assay, the learning capacity and memory were assessed, and the results show that the synthesised nanoformulation significantly decreased the transfer latency to reach baited arm or to the hidden platform within 7 days. CONCLUSION The formulation demonstrated significant biochemical benefits and remarkable cognitive advantages, establishing it as a prospective therapeutic intervention option that is both safe and effective.
Collapse
Affiliation(s)
- Preeti Sharma
- Department of Bioscience and Biotechnology, Banasthali Vidyapith, Jaipur, Rajasthan, India
| | - Pooja Kumari
- Department of Bioscience and Biotechnology, Banasthali Vidyapith, Jaipur, Rajasthan, India
| | - Mansi Sharma
- Department of Chemistry, Banasthali Vidyapith, Jaipur, Rajasthan, India
| | - Rekha Sharma
- Department of Chemistry, Banasthali Vidyapith, Jaipur, Rajasthan, India
| | - Ajita Paliwal
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Shriyansh Srivastava
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi, India
| | - Sumel Ashique
- Department of Pharmaceutical Sciences, Bengal College of Pharmaceutical Sciences and Research, Durgapur, West Bengal, India
| | - Mithun Bhowmick
- Department of Pharmaceutical Sciences, Bengal College of Pharmaceutical Sciences and Research, Durgapur, West Bengal, India
| | - Mohd Adnan
- Department of Biology, College of Science, University of Ha'il, Ha'il, Saudi Arabia
| | - Reyaz Hassan Mir
- Division of Pharmaceutical Chemistry, Department of Pharmaceutical Sciences, University of Kashmir, Srinagar, Jammu and Kashmir, India
| |
Collapse
|
75
|
He R, Cheng J, Qiu Y, Hu Y, Liu J, Wang TH, Cao X. IGF1R and FLT1 in female endothelial cells and CHD2 in male microglia play important roles in Alzheimer's disease based on gender difference analysis. Exp Gerontol 2024; 194:112512. [PMID: 38971545 DOI: 10.1016/j.exger.2024.112512] [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: 11/24/2023] [Revised: 06/29/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
OBJECTIVE This study investigated sex-specific pathogenesis mechanisms in Alzheimer's disease (AD) using single-nucleus RNA sequencing (snRNA-seq) data. METHODS Data from the Gene Expression Omnibus (GEO) were searched using terms "Alzheimer's Disease", "single cell", and "Homo sapiens". Studies excluding APOE E4 and including comprehensive gender information with 10× sequencing methods were selected, resulting in GSE157827 and GSE174367 datasets from human prefrontal cortex samples. Sex-stratified analyses were conducted on these datasets, and the outcomes of the analysis for GSE157827 were compared with those of GSE174367. The findings were validated using expression profiling from the mouse dataset GSE85162. Furthermore, real-time PCR experiments in mice further confirmed these findings. The Seurat R package was used to identify cell types, and batch effects were mitigated using the Harmony R package. Cell proportions by sex were compared using the Mann-Whitney-Wilcoxon test, and gene expression variability was displayed with an empirical cumulative distribution plot. Differentially expressed genes were identified using the FindMarkers function with the MAST test. Transcription factors were analyzed using the RcisTarget R package. RESULTS Seven cell types were identified: astrocytes, endothelial cells, excitatory neurons, inhibitory neurons, microglia, oligodendrocytes, and oligodendrocyte progenitor cells. Additionally, five distinct subpopulations of both endothelial and microglial cells were also identified, respectively. Key findings included: (1) In endothelial cells, genes involved in synapse organization, such as Insulin Like Growth Factor 1 Receptor (IGF1R) and Fms Related Receptor Tyrosine Kinase 1(FLT1), showed higher expression in females with AD. (2) In microglial cells, genes in the ribosome pathway exhibited higher expression in males without AD compared to females (with or without AD) and males with AD. (3) Chromodomain Helicase DNA Binding Protein 2 (CHD2) negatively regulated gene expression in the ribosome pathway in male microglia, suppressing AD, this finding was further validated in mice. (4) Differences between Asians and Caucasians were observed based on sex and disease status stratification. CONCLUSIONS IGF1R and FLT1 in endothelial cells contribute to AD in females, while CHD2 negatively regulates ribosome pathway gene expression in male microglia, suppressing AD in humans and mice.
Collapse
Affiliation(s)
- Rong He
- Laboratory Animal Department, Kunming Medical University, Kunming 650500, Yunnan, China
| | - Jishuai Cheng
- Laboratory Animal Department, Kunming Medical University, Kunming 650500, Yunnan, China
| | - Yue Qiu
- Dermatology Department of Xiangya Hospital, Central South University, Changsha, China
| | - Yiwen Hu
- Laboratory Animal Department, Kunming Medical University, Kunming 650500, Yunnan, China
| | - Jia Liu
- Laboratory Animal Department, Kunming Medical University, Kunming 650500, Yunnan, China.
| | - Ting-Hua Wang
- Laboratory Animal Department, Kunming Medical University, Kunming 650500, Yunnan, China.
| | - Xue Cao
- Laboratory Animal Department, Kunming Medical University, Kunming 650500, Yunnan, China.
| |
Collapse
|
76
|
Sharma P, Kumari P, Sharma M, Sharma R, Paliwal A, Srivastava S, Ashique S, Bhowmick M, Adnan M, Mir RH. Therapeutic potential of Aloe vera-coated curcumin encapsulated nanoparticles in an Alzheimer-induced mice model: behavioural, biochemical and histopathological evidence. J Microencapsul 2024; 41:403-418. [DOI: https:/doi.org/10.1080/02652048.2024.2373715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 06/20/2024] [Indexed: 04/05/2025]
Affiliation(s)
- Preeti Sharma
- Department of Bioscience and Biotechnology, Banasthali Vidyapith, Jaipur, Rajasthan, India
| | - Pooja Kumari
- Department of Bioscience and Biotechnology, Banasthali Vidyapith, Jaipur, Rajasthan, India
| | - Mansi Sharma
- Department of Chemistry, Banasthali Vidyapith, Jaipur, Rajasthan, India
| | - Rekha Sharma
- Department of Chemistry, Banasthali Vidyapith, Jaipur, Rajasthan, India
| | - Ajita Paliwal
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Shriyansh Srivastava
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi, India
| | - Sumel Ashique
- Department of Pharmaceutical Sciences, Bengal College of Pharmaceutical Sciences and Research, Durgapur, West Bengal, India
| | - Mithun Bhowmick
- Department of Pharmaceutical Sciences, Bengal College of Pharmaceutical Sciences and Research, Durgapur, West Bengal, India
| | - Mohd Adnan
- Department of Biology, College of Science, University of Ha’il, Ha’il, Saudi Arabia
| | - Reyaz Hassan Mir
- Division of Pharmaceutical Chemistry, Department of Pharmaceutical Sciences, University of Kashmir, Srinagar, Jammu and Kashmir, India
| |
Collapse
|
77
|
Zhang J, Zhang Y, Wang J, Xia Y, Zhang J, Chen L. Recent advances in Alzheimer's disease: Mechanisms, clinical trials and new drug development strategies. Signal Transduct Target Ther 2024; 9:211. [PMID: 39174535 PMCID: PMC11344989 DOI: 10.1038/s41392-024-01911-3] [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: 11/09/2023] [Revised: 03/18/2024] [Accepted: 07/02/2024] [Indexed: 08/24/2024] Open
Abstract
Alzheimer's disease (AD) stands as the predominant form of dementia, presenting significant and escalating global challenges. Its etiology is intricate and diverse, stemming from a combination of factors such as aging, genetics, and environment. Our current understanding of AD pathologies involves various hypotheses, such as the cholinergic, amyloid, tau protein, inflammatory, oxidative stress, metal ion, glutamate excitotoxicity, microbiota-gut-brain axis, and abnormal autophagy. Nonetheless, unraveling the interplay among these pathological aspects and pinpointing the primary initiators of AD require further elucidation and validation. In the past decades, most clinical drugs have been discontinued due to limited effectiveness or adverse effects. Presently, available drugs primarily offer symptomatic relief and often accompanied by undesirable side effects. However, recent approvals of aducanumab (1) and lecanemab (2) by the Food and Drug Administration (FDA) present the potential in disrease-modifying effects. Nevertheless, the long-term efficacy and safety of these drugs need further validation. Consequently, the quest for safer and more effective AD drugs persists as a formidable and pressing task. This review discusses the current understanding of AD pathogenesis, advances in diagnostic biomarkers, the latest updates of clinical trials, and emerging technologies for AD drug development. We highlight recent progress in the discovery of selective inhibitors, dual-target inhibitors, allosteric modulators, covalent inhibitors, proteolysis-targeting chimeras (PROTACs), and protein-protein interaction (PPI) modulators. Our goal is to provide insights into the prospective development and clinical application of novel AD drugs.
Collapse
Affiliation(s)
- Jifa Zhang
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yinglu Zhang
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jiaxing Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, 38163, TN, USA
| | - Yilin Xia
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jiaxian Zhang
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Lei Chen
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
78
|
Zhang Y, Lin X, Xia L, Xiong S, Xia B, Xie J, Lin Y, Lin L, Wu P. Progress on the Anti-Inflammatory Activity and Structure-Efficacy Relationship of Polysaccharides from Medical and Edible Homologous Traditional Chinese Medicines. Molecules 2024; 29:3852. [PMID: 39202931 PMCID: PMC11356930 DOI: 10.3390/molecules29163852] [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: 07/02/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 09/03/2024] Open
Abstract
Medicinal food varieties developed according to the theory of medical and edible homologues are effective at preventing and treating chronic diseases and in health care. As of 2022, 110 types of traditional Chinese medicines from the same source of medicine and food have been published by the National Health Commission. Inflammation is the immune system's first response to injury, infection, and stress. Chronic inflammation is closely related to many diseases such as atherosclerosis and cancer. Therefore, timely intervention for inflammation is the mainstay treatment for other complex diseases. However, some traditional anti-inflammatory drugs on the market are commonly associated with a number of adverse effects, which seriously affect the health and safety of patients. Therefore, the in-depth development of new safe, harmless, and effective anti-inflammatory drugs has become a hot topic of research and an urgent clinical need. Polysaccharides, one of the main active ingredients of medical and edible homologous traditional Chinese medicines (MEHTCMs), have been confirmed by a large number of studies to exert anti-inflammatory effects through multiple targets and are considered potential natural anti-inflammatory drugs. In addition, the structure of medical and edible homologous traditional Chinese medicines' polysaccharides (MEHTCMPs) may be the key factor determining their anti-inflammatory activity, which makes the underlying the anti-inflammatory effects of polysaccharides and their structure-efficacy relationship hot topics of domestic and international research. However, due to the limitations of the current analytical techniques and tools, the structures have not been fully elucidated and the structure-efficacy relationship is relatively ambiguous, which are some of the difficulties in the process of developing and utilizing MEHTCMPs as novel anti-inflammatory drugs in the future. For this reason, this paper summarizes the potential anti-inflammatory mechanisms of MEHTCMPs, such as the regulation of the Toll-like receptor-related signaling pathway, MAPK signaling pathway, JAK-STAT signaling pathway, NLRP3 signaling pathway, PI3K-AKT signaling pathway, PPAR-γ signaling pathway, Nrf2-HO-1 signaling pathway, and the regulation of intestinal flora, and it systematically analyzes and evaluates the relationships between the anti-inflammatory activity of MEHTCMPs and their structures.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.Z.); (X.L.); (L.X.); (S.X.); (B.X.); (J.X.); (Y.L.)
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Xiulian Lin
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.Z.); (X.L.); (L.X.); (S.X.); (B.X.); (J.X.); (Y.L.)
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Li Xia
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.Z.); (X.L.); (L.X.); (S.X.); (B.X.); (J.X.); (Y.L.)
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Suhui Xiong
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.Z.); (X.L.); (L.X.); (S.X.); (B.X.); (J.X.); (Y.L.)
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Bohou Xia
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.Z.); (X.L.); (L.X.); (S.X.); (B.X.); (J.X.); (Y.L.)
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Jingchen Xie
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.Z.); (X.L.); (L.X.); (S.X.); (B.X.); (J.X.); (Y.L.)
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Yan Lin
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.Z.); (X.L.); (L.X.); (S.X.); (B.X.); (J.X.); (Y.L.)
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Limei Lin
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.Z.); (X.L.); (L.X.); (S.X.); (B.X.); (J.X.); (Y.L.)
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Ping Wu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.Z.); (X.L.); (L.X.); (S.X.); (B.X.); (J.X.); (Y.L.)
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha 410208, China
| |
Collapse
|
79
|
Mei J, Li Y, Niu L, Liang R, Tang M, Cai Q, Xu J, Zhang D, Yin X, Liu X, Shen Y, Liu J, Xu M, Xia P, Ling J, Wu Y, Liang J, Zhang J, Yu P. SGLT2 inhibitors: a novel therapy for cognitive impairment via multifaceted effects on the nervous system. Transl Neurodegener 2024; 13:41. [PMID: 39123214 PMCID: PMC11312905 DOI: 10.1186/s40035-024-00431-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: 03/14/2024] [Accepted: 07/11/2024] [Indexed: 08/12/2024] Open
Abstract
The rising prevalence of diabetes mellitus has casted a spotlight on one of its significant sequelae: cognitive impairment. Sodium-glucose cotransporter-2 (SGLT2) inhibitors, originally developed for diabetes management, are increasingly studied for their cognitive benefits. These benefits may include reduction of oxidative stress and neuroinflammation, decrease of amyloid burdens, enhancement of neuronal plasticity, and improved cerebral glucose utilization. The multifaceted effects and the relatively favorable side-effect profile of SGLT2 inhibitors render them a promising therapeutic candidate for cognitive disorders. Nonetheless, the application of SGLT2 inhibitors for cognitive impairment is not without its limitations, necessitating more comprehensive research to fully determine their therapeutic potential for cognitive treatment. In this review, we discuss the role of SGLT2 in neural function, elucidate the diabetes-cognition nexus, and synthesize current knowledge on the cognitive effects of SGLT2 inhibitors based on animal studies and clinical evidence. Research gaps are proposed to spur further investigation.
Collapse
Affiliation(s)
- Jiaqi Mei
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Huan Kui College of Nanchang University, Nanchang, China
| | - Yi Li
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Huan Kui College of Nanchang University, Nanchang, China
| | - Liyan Niu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Huan Kui College of Nanchang University, Nanchang, China
| | - Ruikai Liang
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Mingyue Tang
- Queen Mary College of Nanchang University, Nanchang, China
| | - Qi Cai
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jingdong Xu
- Queen Mary College of Nanchang University, Nanchang, China
| | - Deju Zhang
- Food and Nutritional Sciences, School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Xiaoping Yin
- Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, China
| | - Xiao Liu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yunfeng Shen
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jianping Liu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Minxuan Xu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Panpan Xia
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jitao Ling
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yuting Wu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jianqi Liang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.
| | - Peng Yu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China.
| |
Collapse
|
80
|
Chand Dakal T, Choudhary K, Tiwari I, Yadav V, Kumar Maurya P, Kumar Sharma N. Unraveling the Triad: Hypoxia, Oxidative Stress and Inflammation in Neurodegenerative Disorders. Neuroscience 2024; 552:126-141. [PMID: 38936458 DOI: 10.1016/j.neuroscience.2024.06.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 06/07/2024] [Accepted: 06/22/2024] [Indexed: 06/29/2024]
Abstract
The mammalian brain's complete dependence on oxygen for ATP production makes it highly susceptible to hypoxia, at high altitudes or in clinical scenarios including anemia or pulmonary disease. Hypoxia plays a crucial role in the development of various brain disorders, such as Alzheimer's, Parkinson's, and other age-related neurodegenerative diseases. On the other hand, a decrease in environmental oxygen levels, such as prolonged stays at high elevations, may have beneficial impacts on the process of ageing and the likelihood of death. Additionally, the utilization of controlled hypoxia exposure could potentially serve as a therapeutic approach for age-related brain diseases. Recent findings indicate that the involvement of HIF-1α and the NLRP3 inflammasome is of significant importance in the development of Alzheimer's disease. HIF-1α serves as a pivotal controller of various cellular reactions to oxygen deprivation, exerting influence on a multitude of physiological mechanisms such as energy metabolism and inflammatory responses. The NLRP3 plays a crucial role in the innate immune system by coordinating the initiation of inflammatory reactions through the assembly of the inflammasome complex. This review examines the information pertaining to the contrasting effects of hypoxia on the brain, highlighting both its positive and deleterious effects and molecular pathways that are involved in mediating these different effects. This study explores potential strategies for therapeutic intervention that focus on restoring cellular balance and reducing neuroinflammation, which are critical aspects in addressing this severe neurodegenerative condition and addresses crucial inquiries that warrant further future investigations.
Collapse
Affiliation(s)
- Tikam Chand Dakal
- Genome and Computational Biology Lab, Mohanlal Sukhadia University, Udaipur 313001, Rajasthan, India
| | - Kanika Choudhary
- School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Isha Tiwari
- Department of Bioscience and Biotechnology, Banasthali Vidyapith, Tonk 304022, Rajasthan, India
| | - Vikas Yadav
- School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Pawan Kumar Maurya
- Department of Biochemistry, Central University of Haryana, Mahendergarh 123031, India
| | - Narendra Kumar Sharma
- Department of Bioscience and Biotechnology, Banasthali Vidyapith, Tonk 304022, Rajasthan, India.
| |
Collapse
|
81
|
Prajapati SK, Pathak A, Samaiya PK. Alzheimer's disease: from early pathogenesis to novel therapeutic approaches. Metab Brain Dis 2024; 39:1231-1254. [PMID: 39046584 DOI: 10.1007/s11011-024-01389-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 07/15/2024] [Indexed: 07/25/2024]
Abstract
The mainstay behind Alzheimer's disease (AD) remains unknown due to the elusive pathophysiology of the disease. Beta-amyloid and phosphorylated Tau is still widely incorporated in various research studies while studying AD. However, they are not sufficient. Therefore, many scientists and researchers have dug into AD studies to deliver many innovations in this field. Many novel biomarkers, such as phosphoglycerate-dehydrogenase, clusterin, microRNA, and a new peptide ratio (Aβ37/Aβ42) in cerebral-spinal fluid, plasma glial-fibrillary-acidic-protein, and lipid peroxidation biomarkers, are mushrooming. They are helping scientists find breakthroughs and substantiating their research on the early detection of AD. Neurovascular unit dysfunction in AD is a significant discovery that can help us understand the relationship between neuronal activity and cerebral blood flow. These new biomarkers are promising and can take these AD studies to another level. There have also been big steps forward in diagnosing and finding AD. One example is self-administered-gerocognitive-examination, which is less expensive and better at finding AD early on than mini-mental-state-examination. Quantum brain sensors and electrochemical biosensors are innovations in the detection field that must be explored and incorporated into the studies. Finally, novel innovations in AD studies like nanotheranostics are the future of AD treatment, which can not only diagnose and detect AD but also offer treatment. Non-pharmacological strategies to treat AD have also yielded interesting results. Our literature review spans from 1957 to 2022, capturing research and trends in the field over six decades. This review article is an update not only on the recent advances in the search for credible biomarkers but also on the newer detection techniques and therapeutic approaches targeting AD.
Collapse
Affiliation(s)
- Santosh Kumar Prajapati
- Bhavdiya Institute of Pharmaceutical Sciences and Research, Ayodhya, UP, India
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, 33613, USA
| | - Arjit Pathak
- Department of Pharmacy Shri G.S. Institute of Technology and Science, Indore, 452003, Madhya Pradesh, India
| | - Puneet K Samaiya
- Department of Pharmacy Shri G.S. Institute of Technology and Science, Indore, 452003, Madhya Pradesh, India.
| |
Collapse
|
82
|
Hernández-Contreras KA, Martínez-Díaz JA, Hernández-Aguilar ME, Herrera-Covarrubias D, Rojas-Durán F, Chi-Castañeda LD, García-Hernández LI, Aranda-Abreu GE. Alterations of mRNAs and Non-coding RNAs Associated with Neuroinflammation in Alzheimer's Disease. Mol Neurobiol 2024; 61:5826-5840. [PMID: 38236345 DOI: 10.1007/s12035-023-03908-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 12/27/2023] [Indexed: 01/19/2024]
Abstract
Alzheimer's disease is a neurodegenerative pathology whose pathognomonic hallmarks are increased generation of β-amyloid (Aβ) peptide, production of hyperphosphorylated (pTau), and neuroinflammation. The last is an alteration closely related to the progression of AD and although it is present in multiple neurodegenerative diseases, the pathophysiological events that characterize neuroinflammatory processes vary depending on the disease. In this article, we focus on mRNA and non-coding RNA alterations as part of the pathophysiological events characteristic of neuroinflammation in AD and the influence of these alterations on the course of the disease through interaction with multiple RNAs related to the generation of Aβ, pTau, and neuroinflammation itself.
Collapse
Affiliation(s)
- Karla Aketzalli Hernández-Contreras
- Doctorado en Investigaciones Cerebrales/Universidad Veracruzana, Av. Luis Castelazo Ayala S/N, Carr. Xalapa-Veracruz, Km 3.5, C.P. 91190, Xalapa, Veracruz, México
| | - Jorge Antonio Martínez-Díaz
- Instituto de Investigaciones Cerebrales/Universidad Veracruzana, Av. Luis Castelazo Ayala S/N, Carr. Xalapa-Veracruz, Km 3.5, C.P. 91190, Xalapa, Veracruz, México
| | - María Elena Hernández-Aguilar
- Instituto de Investigaciones Cerebrales/Universidad Veracruzana, Av. Luis Castelazo Ayala S/N, Carr. Xalapa-Veracruz, Km 3.5, C.P. 91190, Xalapa, Veracruz, México
| | - Deissy Herrera-Covarrubias
- Instituto de Investigaciones Cerebrales/Universidad Veracruzana, Av. Luis Castelazo Ayala S/N, Carr. Xalapa-Veracruz, Km 3.5, C.P. 91190, Xalapa, Veracruz, México
| | - Fausto Rojas-Durán
- Instituto de Investigaciones Cerebrales/Universidad Veracruzana, Av. Luis Castelazo Ayala S/N, Carr. Xalapa-Veracruz, Km 3.5, C.P. 91190, Xalapa, Veracruz, México
| | - Lizbeth Donají Chi-Castañeda
- Instituto de Investigaciones Cerebrales/Universidad Veracruzana, Av. Luis Castelazo Ayala S/N, Carr. Xalapa-Veracruz, Km 3.5, C.P. 91190, Xalapa, Veracruz, México
| | - Luis Isauro García-Hernández
- Instituto de Investigaciones Cerebrales/Universidad Veracruzana, Av. Luis Castelazo Ayala S/N, Carr. Xalapa-Veracruz, Km 3.5, C.P. 91190, Xalapa, Veracruz, México
| | - Gonzalo Emiliano Aranda-Abreu
- Instituto de Investigaciones Cerebrales/Universidad Veracruzana, Av. Luis Castelazo Ayala S/N, Carr. Xalapa-Veracruz, Km 3.5, C.P. 91190, Xalapa, Veracruz, México.
| |
Collapse
|
83
|
Hou J, Wang X, Zhang J, Shen Z, Li X, Yang Y. Chuanxiong Renshen Decoction Inhibits Alzheimer's Disease Neuroinflammation by Regulating PPARγ/NF-κB Pathway. Drug Des Devel Ther 2024; 18:3209-3232. [PMID: 39071817 PMCID: PMC11283787 DOI: 10.2147/dddt.s462266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 07/12/2024] [Indexed: 07/30/2024] Open
Abstract
Background and Aim Previous studies of our research group have shown that Chuanxiong Renshen Decoction (CRD) has the effect of treating AD, but the exact mechanism of its effect is still not clarified. The aim of this study was to investigate the effect and mechanism of CRD on AD neuroinflammation. Materials and Methods Morris Water Maze (MWM) tests were employed to assess the memory and learning capacity of AD mice. HE and Nissl staining were used to observe the neural cells of mice. The expression of Iba-1 and CD86 were detected by immunohistochemical staining. Utilize UHPLC-MS/MS metabolomics techniques and the KEGG to analyze the metabolic pathways of CRD against AD. Lipopolysaccharide (LPS) induced BV2 microglia cells to construct a neuroinflammatory model. The expression of Iba-1 and CD86 were detected by immunofluorescence and flow cytometry. The contents of TNF-α and IL-1β were detected by ELISA. Western blot assay was used to detect the expression of PPARγ, p-NF-κB p65, NF-κB p65 proteins and inflammatory cytokines iNOS and COX-2 in PPARγ/NF-κB pathway with and without PPARγ inhibitor GW9662. Results CRD ameliorated the learning and memory ability of 3×Tg-AD mice, repaired the damaged nerve cells in the hippocampus, reduced the area of Iba-1 and CD86 positive areas in both the hippocampus and cortex regions, as well as attenuated serum levels of IL-1β and TNF-α in mice. CRD-containing serum significantly decreased the expression level of Iba-1, significantly reduced the levels of TNF-α and IL-1β, significantly increased the protein expression of PPARγ, and significantly decreased the proteins expression of iNOS, COX-2 and p-NF-κB p65 in BV2 microglia cells. After addition of PPARγ inhibitor GW9662, the inhibitory effect of CRD-containing serum on NF-κB activation was significantly weakened. Conclusion CRD can activate PPARγ, regulating PPARγ/NF-κB signaling pathway, inhibiting microglia over-activation and reducing AD neuroinflammation.
Collapse
Affiliation(s)
- Jinling Hou
- School of Pharmacy, Hangzhou Medical College, Hangzhou, People’s Republic of China
| | - Xiaoyan Wang
- School of Pharmacy, Hangzhou Medical College, Hangzhou, People’s Republic of China
| | - Jian Zhang
- School of Pharmacy, Hangzhou Medical College, Hangzhou, People’s Republic of China
| | - Zhuojun Shen
- School of Pharmacy, Hangzhou Medical College, Hangzhou, People’s Republic of China
| | - Xiang Li
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, People’s Republic of China
| | - Yuanxiao Yang
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, People’s Republic of China
| |
Collapse
|
84
|
Sun W, Gong J, Li S, Wang P, Han X, Xu C, Luan H, Li R, Wen B, Wei C. Bibliometric analysis of neuroinflammation and Alzheimer's disease. Front Aging Neurosci 2024; 16:1423139. [PMID: 39076205 PMCID: PMC11284157 DOI: 10.3389/fnagi.2024.1423139] [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: 05/01/2024] [Accepted: 07/02/2024] [Indexed: 07/31/2024] Open
Abstract
Background Alzheimer's disease (AD) is the predominant cause of dementia on a global scale, significantly impacting the health of the elderly population. The pathogenesis of AD is closely linked to neuroinflammation. The present study employs a bibliometric analysis to examine research pertaining to neuroinflammation and AD within the last decade, with the objective of providing a comprehensive overview of the current research profile, hotspots and trends. Methods This research conducted a comprehensive review of publications within the Science Citation Index Expanded of the Web of Science Core Collection Database spanning the years 2014 to 2024. Bibliometric analyses were performed using VOSviewer (version 1.6.19) and CiteSpace (version 6.3.R1) software to visualize data on countries, institutions, authors, journals, keywords, and references. Results A total of 3,833 publications on neuroinflammation and AD were included from January 2014 to January 2024. Publications were mainly from the United States and China. Zetterberg, Henrik emerged as the author with the highest publication output, while Edison, Paul was identified as the most cited author. The most productive journal was Journal of Alzheimers Disease, and the most co-cited was Journal of Neuroinflammation. Research hotspot focused on microglia, mouse models, oxidative stress, and amyloid-beta through keyword analysis. Additionally, keywords such as blood-brain barrier and tau protein exhibited prolonged citation bursts from 2022 to 2024. Conclusion This study provides a comprehensive review of the last 10 years of research on neuroinflammation and AD, including the number and impact of research findings, research hotspots, and future trends. The quantity of publications in this field is increasing, mainly in the United States and China, and there is a need to further strengthen close cooperation with different countries and institutions worldwide. Presently, research hotspots are primarily concentrated on microglia, with a focus on inhibiting their pro-inflammatory responses and promoting their anti-inflammatory functions as a potential direction for future investigations.
Collapse
Affiliation(s)
- Wenxian Sun
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Jin Gong
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Shaoqi Li
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Pin Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Xiaodong Han
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Chang Xu
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Heya Luan
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Ruina Li
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Boye Wen
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Cuibai Wei
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| |
Collapse
|
85
|
Xia N, Xu L, Huang M, Xu D, Li Y, Wu H, Mei Z, Yu Z. Neuroprotection of macamide in a mouse model of Alzheimer's disease involves Nrf2 signaling pathway and gut microbiota. Eur J Pharmacol 2024; 975:176638. [PMID: 38734297 DOI: 10.1016/j.ejphar.2024.176638] [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: 03/13/2024] [Revised: 05/03/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024]
Abstract
The underlying mechanisms of macamide's neuroprotective effects in Alzheimer's disease (AD) were investigated in the paper. Macamides are considered as unique ingredients in maca. Improvement effects and mechanisms of macamide on cognitive impairment have not been revealed. In this study, Vina 1.1.2 was used for docking to evaluate the binding abilities of 12 main macamides to acetylcholinesterase (AChE). N-benzyl-(9Z,12Z)-octadecadienamide (M 18:2) was selected to study the following experiments because it can stably bind to AChE with a strong binding energy. The animal experiments showed that M 18:2 prevented the scopolamine (SCP)-induced cognitive impairment and neurotransmitter disorders, increased the positive rates of Nrf2 and HO-1 in hippocampal CA1, improved the synaptic plasticity by maintaining synaptic morphology and increasing the synapse density. Moreover, the contents of IL-1β, IL-6, and TNF-α in the hippocampus, serum, and colon were reduced by M 18:2. Furthermore, M 18:2 promoted colonic epithelial integrity and partially restored the composition of the gut microbiota to normal, including decreased genera Clostridiales_unclassified and Lachnospiraceae_unclassified, as well as increased genera Muribaculaceae_unclassified, Muribaculum, Alistipes, and Bacteroides, which may be the possible biomarkers of cognitive aging. In summary, M 18:2 exerted neuroprotective effects on SCP-induced AD mice possibly via activating the Nrf2/HO-1 signaling pathway and modulating the gut microbiota.
Collapse
Affiliation(s)
- Nengyin Xia
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Lingyun Xu
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Mengyuan Huang
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Dengrui Xu
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Yang Li
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Haoming Wu
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Zhinan Mei
- College of Plant Science & Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zejun Yu
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan, 430023, China.
| |
Collapse
|
86
|
Yang S, Tian M, Dai Y, Wang R, Yamada S, Feng S, Wang Y, Chhangani D, Ou T, Li W, Guo X, McAdow J, Rincon-Limas DE, Yin X, Tai W, Cheng G, Johnson A. Infection and chronic disease activate a systemic brain-muscle signaling axis. Sci Immunol 2024; 9:eadm7908. [PMID: 38996009 DOI: 10.1126/sciimmunol.adm7908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 04/18/2024] [Accepted: 06/18/2024] [Indexed: 07/14/2024]
Abstract
Infections and neurodegenerative diseases induce neuroinflammation, but affected individuals often show nonneural symptoms including muscle pain and muscle fatigue. The molecular pathways by which neuroinflammation causes pathologies outside the central nervous system (CNS) are poorly understood. We developed multiple models to investigate the impact of CNS stressors on motor function and found that Escherichia coli infections and SARS-CoV-2 protein expression caused reactive oxygen species (ROS) to accumulate in the brain. ROS induced expression of the cytokine Unpaired 3 (Upd3) in Drosophila and its ortholog, IL-6, in mice. CNS-derived Upd3/IL-6 activated the JAK-STAT pathway in skeletal muscle, which caused muscle mitochondrial dysfunction and impaired motor function. We observed similar phenotypes after expressing toxic amyloid-β (Aβ42) in the CNS. Infection and chronic disease therefore activate a systemic brain-muscle signaling axis in which CNS-derived cytokines bypass the connectome and directly regulate muscle physiology, highlighting IL-6 as a therapeutic target to treat disease-associated muscle dysfunction.
Collapse
Affiliation(s)
- Shuo Yang
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
- Department of Genetics and Genetics Engineering, School of Life Science, Fudan University, Shanghai 200438, China
| | - Meijie Tian
- Genetics Branch, Oncogenomics Section, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Yulong Dai
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen 518000, China
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Rong Wang
- Department of Genetics and Genetics Engineering, School of Life Science, Fudan University, Shanghai 200438, China
| | - Shigehiro Yamada
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Shengyong Feng
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Yunyun Wang
- Department of Forensic Medicine, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Deepak Chhangani
- Department of Neurology and McKnight Brain Institute, Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease, Genetics Institute, and Norman Fixel Institute for Neurological Diseases, University of Florida College of Medicine, Gainesville, FL 32611, USA
| | - Tiffany Ou
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Wenle Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Xuan Guo
- Life Science Institute, Jinzhou Medical University, Jinzhou 121001, China
| | - Jennifer McAdow
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Diego E Rincon-Limas
- Department of Neurology and McKnight Brain Institute, Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease, Genetics Institute, and Norman Fixel Institute for Neurological Diseases, University of Florida College of Medicine, Gainesville, FL 32611, USA
| | - Xin Yin
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Wanbo Tai
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen 518000, China
| | - Gong Cheng
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen 518000, China
- Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
- Southwest United Graduate School, Kunming 650092, China
| | - Aaron Johnson
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| |
Collapse
|
87
|
Zheng S, Ma R, Yang Y, Li G. Psilocybin for the treatment of Alzheimer's disease. Front Neurosci 2024; 18:1420601. [PMID: 39050672 PMCID: PMC11266071 DOI: 10.3389/fnins.2024.1420601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 06/21/2024] [Indexed: 07/27/2024] Open
Abstract
Alzheimer's disease (AD) stands as a formidable neurodegenerative ailment and a prominent contributor to dementia. The scarcity of available therapies for AD accentuates the exigency for innovative treatment modalities. Psilocybin, a psychoactive alkaloid intrinsic to hallucinogenic mushrooms, has garnered attention within the neuropsychiatric realm due to its established safety and efficacy in treating depression. Nonetheless, its potential as a therapeutic avenue for AD remains largely uncharted. This comprehensive review endeavors to encapsulate the pharmacological effects of psilocybin while elucidating the existing evidence concerning its potential mechanisms contributing to a positive impact on AD. Specifically, the active metabolite of psilocybin, psilocin, elicits its effects through the modulation of the 5-hydroxytryptamine 2A receptor (5-HT2A receptor). This modulation causes heightened neural plasticity, diminished inflammation, and improvements in cognitive functions such as creativity, cognitive flexibility, and emotional facial recognition. Noteworthy is psilocybin's promising role in mitigating anxiety and depression symptoms in AD patients. Acknowledging the attendant adverse reactions, we proffer strategies aimed at tempering or mitigating its hallucinogenic effects. Moreover, we broach the ethical and legal dimensions inherent in psilocybin's exploration for AD treatment. By traversing these avenues, We propose therapeutic potential of psilocybin in the nuanced management of Alzheimer's disease.
Collapse
Affiliation(s)
- Siyi Zheng
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rong Ma
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Yang
- Department of General Medicine, Binzhou Medical University Hospital, Binzhou, China
| | - Gang Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
88
|
Liao Z, Zhang Q, Ren N, Zhao H, Zheng X. Progress in mitochondrial and omics studies in Alzheimer's disease research: from molecular mechanisms to therapeutic interventions. Front Immunol 2024; 15:1418939. [PMID: 39040111 PMCID: PMC11260616 DOI: 10.3389/fimmu.2024.1418939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 06/21/2024] [Indexed: 07/24/2024] Open
Abstract
Alzheimer's disease (Alzheimer's disease, AD) is a progressive neurological disorder characterized by memory loss and cognitive impairment. It is characterized by the formation of tau protein neurofibrillary tangles and β-amyloid plaques. Recent studies have found that mitochondria in neuronal cells of AD patients exhibit various dysfunctions, including reduced numbers, ultrastructural changes, reduced enzyme activity, and abnormal kinetics. These abnormal mitochondria not only lead to the loss of normal neuronal cell function, but are also a major driver of AD progression. In this review, we will focus on the advances of mitochondria and their multi-omics in AD research, with particular emphasis on how mitochondrial dysfunction in AD drives disease progression. At the same time, we will focus on summarizing how mitochondrial genomics technologies have revealed specific details of these dysfunctions and how therapeutic strategies targeting mitochondria may provide new directions for future AD treatments. By delving into the key mechanisms of mitochondria in AD related to energy metabolism, altered kinetics, regulation of cell death, and dysregulation of calcium-ion homeostasis, and how mitochondrial multi-omics technologies can be utilized to provide us with a better understanding of these processes. In the future, mitochondria-centered therapeutic strategies will be a key idea in the treatment of AD.
Collapse
Affiliation(s)
- Zuning Liao
- Department of Neurology, Fourth People’s Hospital of Jinan, Jinan, China
| | - Qiying Zhang
- Department of Internal Medicine, Jinan Municipal Government Hospital, Jinan, China
| | - Na Ren
- Pharmacy Department, Jinan Municipal People’s Government Organs Outpatient Department, Jinan, China
| | - Haiyan Zhao
- Department of Pharmacy, Qihe County People’s Hospital, Dezhou, China
| | - Xueyan Zheng
- Department of Pharmacy, Jinan Second People’s Hospital, Jinan, China
| |
Collapse
|
89
|
Rodrigues FDS, Newton WR, Tassinari ID, da Cunha Xavier FH, Marx A, de Fraga LS, Wright K, Guedes RP, Bambini-Jr V. Cannabidiol prevents LPS-induced inflammation by inhibiting the NLRP3 inflammasome and iNOS activity in BV2 microglia cells via CB2 receptors and PPARγ. Neurochem Int 2024; 177:105769. [PMID: 38761855 DOI: 10.1016/j.neuint.2024.105769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/10/2024] [Accepted: 05/15/2024] [Indexed: 05/20/2024]
Abstract
Neuroinflammation stands as a critical player in the pathogenesis of diverse neurological disorders, with microglial cells playing a central role in orchestrating the inflammatory landscape within the central nervous system. Cannabidiol (CBD) has gained attention for its potential to elicit anti-inflammatory responses in microglia, offering promising perspectives for conditions associated with neuroinflammation. Here we investigated whether the NLRP3 inflammasome and inducible nitric oxide synthase (iNOS) are involved in the protective effects of CBD, and if their modulation is dependent on cannabinoid receptor 2 (CB2) and PPARγ signalling pathways. We found that treatment with CBD attenuated pro-inflammatory markers in lipopolysaccharide (LPS)-challenged BV2 microglia in a CB2- and PPARγ-dependent manner. At a molecular level, CBD inhibited the LPS-induced pro-inflammatory responses by suppressing iNOS and NLRP3/Caspase-1-dependent signalling cascades, resulting in reduced nitric oxide (NO), interleukin-1β (IL-1β), and tumour necrosis factor-alpha (TNF-α) concentrations. Notably, the protective effects of CBD on NLRP3 expression, Caspase-1 activity, and IL-1β concentration were partially hindered by the antagonism of both CB2 receptors and PPARγ, while iNOS expression and NO secretion were dependent exclusively on PPARγ activation, with no CB2 involvement. Interestingly, CBD exhibited a protective effect against TNF-α increase, regardless of CB2 or PPARγ activation. Altogether, these findings indicate that CB2 receptors and PPARγ mediate the anti-inflammatory effects of CBD on the NLRP3 inflammasome complex, iNOS activity and, ultimately, on microglial phenotype. Our results highlight the specific components responsible for the potential therapeutic applications of CBD on neuroinflammatory conditions.
Collapse
Affiliation(s)
- Fernanda da Silva Rodrigues
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil; Division of Biomedical and Life Sciences, Lancaster University, Lancaster, Lancashire, United Kingdom.
| | - William Robert Newton
- Division of Biomedical and Life Sciences, Lancaster University, Lancaster, Lancashire, United Kingdom; MRC Centre for Medical Mycology, Exeter University, Exeter, United Kingdom.
| | - Isadora D'Ávila Tassinari
- Division of Biomedical and Life Sciences, Lancaster University, Lancaster, Lancashire, United Kingdom; Graduate Program in Physiology, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil.
| | | | - Adél Marx
- Division of Biomedical and Life Sciences, Lancaster University, Lancaster, Lancashire, United Kingdom.
| | - Luciano Stürmer de Fraga
- Graduate Program in Physiology, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil.
| | - Karen Wright
- Division of Biomedical and Life Sciences, Lancaster University, Lancaster, Lancashire, United Kingdom.
| | - Renata Padilha Guedes
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil; Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil.
| | - Victorio Bambini-Jr
- Division of Biomedical and Life Sciences, Lancaster University, Lancaster, Lancashire, United Kingdom.
| |
Collapse
|
90
|
Maxwell JR, Noor S, Pavlik N, Rodriguez DE, Enriquez Marquez L, DiDomenico J, Blossom SJ, Bakhireva LN. Moderate Prenatal Alcohol Exposure Increases Toll-like Receptor Activity in Umbilical Cord Blood at Birth: A Pilot Study. Int J Mol Sci 2024; 25:7019. [PMID: 39000127 PMCID: PMC11241342 DOI: 10.3390/ijms25137019] [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: 05/10/2024] [Revised: 06/20/2024] [Accepted: 06/22/2024] [Indexed: 07/16/2024] Open
Abstract
The prevalence of prenatal alcohol exposure (PAE) is increasing, with evidence suggesting that PAE is linked to an increased risk of infections. PAE is hypothesized to affect the innate immune system, which identifies pathogens through pattern recognition receptors, of which toll-like receptors (TLRs) are key components. We hypothesized that light-to-moderate PAE would impair immune responses, as measured by a heightened response in cytokine levels following TLR stimulation. Umbilical cord samples (10 controls and 8 PAE) from a subset of the Ethanol, Neurodevelopment, Infant and Child Health Study-2 cohort were included. Peripheral blood mononuclear cells (PMBCs) were stimulated with one agonist (TLR2, TLR3, TLR4, or TLR9). TLR2 agonist stimulation significantly increased pro-inflammatory interleukin-1-beta in the PAE group after 24 h. Pro- and anti-inflammatory cytokines were increased following stimulation with the TLR2 agonists. Stimulation with TLR3 or TLR9 agonists displayed minimal impact overall, but there were significant increases in the percent change of the control compared to PAE after 24 h. The results of this pilot investigation support further work into the impact on TLR2 and TLR4 response following PAE to delineate if alterations in levels of pro- and anti-inflammatory cytokines have clinical significance that could be used in patient management and/or attention to follow-up.
Collapse
Affiliation(s)
- Jessie R. Maxwell
- Department of Pediatrics, University of New Mexico, Albuquerque, NM 87131, USA
- Department of Neurosciences, University of New Mexico, Albuquerque, NM 87131, USA
| | - Shahani Noor
- Department of Neurosciences, University of New Mexico, Albuquerque, NM 87131, USA
| | - Nathaniel Pavlik
- Department of Pediatrics, University of New Mexico, Albuquerque, NM 87131, USA
| | | | | | - Jared DiDomenico
- College of Pharmacy, University of New Mexico, Albuquerque, NM 87131, USA
| | - Sarah J. Blossom
- College of Pharmacy, University of New Mexico, Albuquerque, NM 87131, USA
| | - Ludmila N. Bakhireva
- Substance Use Research and Education (SURE) Center, College of Pharmacy, University of New Mexico, Albuquerque, NM 87131, USA
| |
Collapse
|
91
|
Chen N, Xu X, Guo Y, Zhao M, Li Y, Zhou T, Zhang X, Gao J, Zhu F, Guo C, Shi Y, Wang Q, Wu W, Zhang L, Li Y. Brain Short-Chain Fatty Acids Induce ACSS2 to Ameliorate Depressive-Like Behavior via PPARγ-TPH2 Axis. RESEARCH (WASHINGTON, D.C.) 2024; 7:0400. [PMID: 38939042 PMCID: PMC11210491 DOI: 10.34133/research.0400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/10/2024] [Indexed: 06/29/2024]
Abstract
Short-chain fatty acids (SCFAs) have been increasingly evidenced to be important bioactive metabolites of the gut microbiota and transducers in controlling diverse psychiatric or neurological disorders via the microbiota-gut-brain axis. However, the precise mechanism by which brain SCFAs extert multiple beneficial effects is not completely understood. Our previous research has demonstrated that the acetyl-coenzyme A synthetase short-chain family member 2 (ACSS2) is a novel target of the rapid and long-lasting antidepressant responses. Here, we show that micromolar SCFAs significantly augment both total cellular and nuclear ACSS2 to trigger tryptophan hydroxylase 2 (TPH2) promoter histone acetylation and its transcription in SH-SY5Y cells. In chronic-restraint-stress-induced depression mice, neuronal ACSS2 knockdown by stereotaxic injection of adeno-associated virus in the hippocampus abolished SCFA-mediated improvements in depressive-like behaviors of mice, supporting that ACSS2 is required for SCFA-mediated antidepressant responses. Mechanistically, the peroxisome-proliferator-activated receptor gamma (PPARγ) is identified as a novel partner of ACSS2 to activate TPH2 transcription. Importantly, PPARγ is also responsible for SCFA-mediated antidepressant-like effects via ACSS2-TPH2 axis. To further support brain SCFAs as a therapeutic target for antidepressant effects, d-mannose, which is a naturally present hexose, can significantly reverse the dysbiosis of gut microbiota in the chronic-restraint-stress-exposure mice and augment brain SCFAs to protect against the depressive-like behaviors via ACSS2-PPARγ-TPH2 axis. In summary, brain SCFAs can activate ACSS2-PPARγ-TPH2 axis to play the antidepressive-like effects, and d-mannose is suggested to be an inducer of brain SCFAs in resisting depression.
Collapse
Affiliation(s)
- Nuo Chen
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| | - Xinyi Xu
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| | - Yaxin Guo
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| | - Ming Zhao
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| | - Yubin Li
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| | - Tian Zhou
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| | - Xinyue Zhang
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| | - Jie Gao
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| | - Faliang Zhu
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| | - Chun Guo
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| | - Yongyu Shi
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| | - Qun Wang
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| | - Wenxian Wu
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Department of Neurology,
Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Lining Zhang
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| | - Yan Li
- Department of Pathogen Biology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| |
Collapse
|
92
|
Catumbela CSG, Morales R. Elderly mice with history of acetaminophen intoxication display worsened cognitive impairment and persistent elevation of astrocyte and microglia burden. Sci Rep 2024; 14:14205. [PMID: 38902507 PMCID: PMC11190293 DOI: 10.1038/s41598-024-65185-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: 03/16/2024] [Accepted: 06/18/2024] [Indexed: 06/22/2024] Open
Abstract
Acetaminophen (APAP) is a leading cause of acute liver failure. The effect of APAP metabolite's effects in the periphery are well characterized; however, associated consequences in the brain remain poorly understood. Animal studies on this subject are few and reveal that frequent APAP intake can trigger cerebral abnormalities that vary depending on the subject's age. Alarmingly, experimental efforts have yet to examine associated consequences in elderly hosts, who correspond to the highest risk of medication overload, impaired drug clearance, and cognitive deficits. Here, we interrogated the cerebral and peripheral pathology of elderly mice submitted to monthly episodes of APAP intoxication since a young adult age. We found that weeks after the final episode of recurrent APAP exposure, mice exhibited worsened non-spatial memory deficit whereas spatial memory performance was unaltered. Interestingly, one month after the period of APAP intoxication, these mice showed increased glial burden without associated drivers, namely, blood-brain barrier disruption, cholesterol accumulation, and elevation of inflammatory molecules in the brain and/or periphery. Our experimental study reveals how recurrent APAP exposure affects the cognitive performance and cellular events in elderly brains. These data suggest that APAP-containing pharmacological interventions may foreshadow the elevated risk of neuropsychiatric disorders that afflict elderly populations.
Collapse
Affiliation(s)
- Celso S G Catumbela
- Department of Neurology, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Rodrigo Morales
- Department of Neurology, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
- Centro Integrativo de Biologia y Quimica Aplicada (CIBQA), Universidad Bernardo O'Higgins, Santiago, Chile.
| |
Collapse
|
93
|
Dhapola R, Sharma P, Kumari S, Bhatti JS, HariKrishnaReddy D. Environmental Toxins and Alzheimer's Disease: a Comprehensive Analysis of Pathogenic Mechanisms and Therapeutic Modulation. Mol Neurobiol 2024; 61:3657-3677. [PMID: 38006469 DOI: 10.1007/s12035-023-03805-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 11/13/2023] [Indexed: 11/27/2023]
Abstract
Alzheimer's disease is a leading cause of mortality worldwide. Inorganic and organic hazards, susceptibility to harmful metals, pesticides, agrochemicals, and air pollution are major environmental concerns. As merely 5% of AD cases are directly inherited indicating that these environmental factors play a major role in disease development. Long-term exposure to environmental toxins is believed to progress neuropathology, which leads to the development of AD. Numerous in-vitro and in-vivo studies have suggested the harmful impact of environmental toxins at cellular and molecular level. Common mechanisms involved in the toxicity of these environmental pollutants include oxidative stress, neuroinflammation, mitochondrial dysfunction, abnormal tau, and APP processing. Increased expression of GSK-3β, BACE-1, TNF-α, and pro-apoptotic molecules like caspases is observed upon exposure to these environmental toxins. In addition, the expression of neurotrophins like BDNF and GAP-43 have been found to be reduced as a result of toxicity. Further, modulation of signaling pathways involving PARP-1, PGC-1α, and MAPK/ERK induced by toxins have been reported to contribute in AD pathogenesis. These pathways are a promising target for developing novel AD therapeutics. Drugs like epigallocatechin-gallate, neflamapimod, salsalate, dexmedetomidine, and atabecestat are in different phases of clinical trials targeting the pathways for possible treatment of AD. This review aims to culminate the correlation between environmental toxicants and AD development. We emphasized upon the signaling pathways involved in the progression of the disease and the therapeutics under clinical trial targeting the altered pathways for possible treatment of AD.
Collapse
Affiliation(s)
- Rishika Dhapola
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, Central University of Punjab, Bathinda, 151 401, India
| | - Prajjwal Sharma
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, Central University of Punjab, Bathinda, 151 401, India
| | - Sneha Kumari
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, Central University of Punjab, Bathinda, 151 401, India
| | - Jasvinder Singh Bhatti
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, 151 401, India
| | - Dibbanti HariKrishnaReddy
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, Central University of Punjab, Bathinda, 151 401, India.
| |
Collapse
|
94
|
Umar M, Rehman Y, Ambreen S, Mumtaz SM, Shaququzzaman M, Alam MM, Ali R. Innovative approaches to Alzheimer's therapy: Harnessing the power of heterocycles, oxidative stress management, and nanomaterial drug delivery system. Ageing Res Rev 2024; 97:102298. [PMID: 38604453 DOI: 10.1016/j.arr.2024.102298] [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: 02/01/2024] [Revised: 03/10/2024] [Accepted: 04/05/2024] [Indexed: 04/13/2024]
Abstract
Alzheimer's disease (AD) presents a complex pathology involving amyloidogenic proteolysis, neuroinflammation, mitochondrial dysfunction, and cholinergic deficits. Oxidative stress exacerbates AD progression through pathways like macromolecular peroxidation, mitochondrial dysfunction, and metal ion redox potential alteration linked to amyloid-beta (Aβ). Despite limited approved medications, heterocyclic compounds have emerged as promising candidates in AD drug discovery. This review highlights recent advancements in synthetic heterocyclic compounds targeting oxidative stress, mitochondrial dysfunction, and neuroinflammation in AD. Additionally, it explores the potential of nanomaterial-based drug delivery systems to overcome challenges in AD treatment. Nanoparticles with heterocyclic scaffolds, like polysorbate 80-coated PLGA and Resveratrol-loaded nano-selenium, show improved brain transport and efficacy. Micellar CAPE and Melatonin-loaded nano-capsules exhibit enhanced antioxidant properties, while a tetra hydroacridine derivative (CHDA) combined with nano-radiogold particles demonstrates promising acetylcholinesterase inhibition without toxicity. This comprehensive review underscores the potential of nanotechnology-driven drug delivery for optimizing the therapeutic outcomes of novel synthetic heterocyclic compounds in AD management. Furthermore, the inclusion of various promising heterocyclic compounds with detailed ADMET (Absorption, Distribution, Metabolism, Excretion, and Toxicity) data provides valuable insights for planning the development of novel drug delivery treatments for AD.
Collapse
Affiliation(s)
- Mohammad Umar
- Department of Pharmaceutical Chemistry, Delhi Pharmaceutical Sciences and Research University, Pushp Vihar, New Delhi 110017, India
| | - Yasir Rehman
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, Hamdard Nagar, New Delhi 110062, India
| | - Subiya Ambreen
- Department of Pharmaceutical Chemistry, Delhi Pharmaceutical Sciences and Research University, Pushp Vihar, New Delhi 110017, India
| | - Sayed Md Mumtaz
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, Hamdard Nagar, New Delhi 110062, India
| | - Mohd Shaququzzaman
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, Hamdard Nagar, New Delhi 110062, India
| | - Mohammad Mumtaz Alam
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, Hamdard Nagar, New Delhi 110062, India
| | - Ruhi Ali
- Department of Pharmaceutical Chemistry, Delhi Pharmaceutical Sciences and Research University, Pushp Vihar, New Delhi 110017, India.
| |
Collapse
|
95
|
Zhang B, Deng L, Liu X, Hu Y, Wang W, Li M, Xu T, Pang L, Lv M. Transcranial direct current stimulation combined with swimming exercise improves the learning and memory abilities of vascular dementia rats by regulating microglia through miR-223-3p/PRMT8. Neurol Res 2024; 46:525-537. [PMID: 38563325 DOI: 10.1080/01616412.2024.2337517] [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: 09/23/2023] [Accepted: 03/26/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND Vascular dementia (VD) is the second most common type of dementia worldwide. Previous studies have proven that transcranial direct current stimulation (tDCS) has potential applications in relieving cognitive impairment in VD animal models. The purpose of this study was to probe the mechanism by which tDCS combined with swimming exercise improves the learning and memory abilities of VD model rats. METHOD The VD rat model was induced using the permanent bilateral common carotid artery occlusion (2-VO) method; tDCS was applied to the rats and then they took part in swimming exercises. Rat memory, platform crossing time, and platform crossing frequency were analyzed via a water maze experiment. Nerve damage in the cortex and hippocampal CA1 area of the rats was observed using Nissl staining. Western blotting, immunohistochemistry, immunofluorescence staining and reverse transcription quantitative polymerase chain reaction (RT - qPCR) were used to determine the expression of related proteins and genes. The levels of oxidative stress were detected by kits. RESULTS We demonstrated that VD model rats treated with tDCS combined with swimming exercise exhibited significant improvement in memory, and VD model rats exhibited significantly reduced neuronal loss in the hippocampus, and reduced microglial activation and M1 polarization. tDCS combined with swimming exercise protects VD model rats from oxidative stress through the miR-223-3p/protein arginine methyltransferase 8 (PRMT8) axis and inhibits the activation of the TLR4/NF-κB signaling pathway. CONCLUSION Our results suggest that tDCS combined with swimming exercise improved the learning and memory ability of VD model rats by regulating the expression of PRMT8 through miR-223-3p to affect microglial activation and M1 polarization.
Collapse
Affiliation(s)
- Bingxue Zhang
- Rehabilitation Medicine, Qujing No.1 Hospital, Qujing, Yunnan, China
| | - Li Deng
- Rehabilitation Medicine, Qujing No.1 Hospital, Qujing, Yunnan, China
| | - Xiaodan Liu
- Rehabilitation Medicine, Qujing No.1 Hospital, Qujing, Yunnan, China
| | - Yao Hu
- Rehabilitation Medicine, Qujing No.1 Hospital, Qujing, Yunnan, China
| | - Wenyi Wang
- Rehabilitation Medicine, Qujing No.1 Hospital, Qujing, Yunnan, China
| | - Minghua Li
- Department of Neurology, Luoping County People's Hospital, Luoping, Yunnan, China
| | - Ting Xu
- Rehabilitation Medicine, Qujing No.1 Hospital, Qujing, Yunnan, China
| | - Li Pang
- Rehabilitation Medicine, Qujing No.1 Hospital, Qujing, Yunnan, China
| | - Meifen Lv
- Rehabilitation Medicine, Qujing No.1 Hospital, Qujing, Yunnan, China
| |
Collapse
|
96
|
He C, Jiang J, Liu J, Zhou L, Ge Y, Yang Z. Pseudostellaria heterophylla polysaccharide mitigates Alzheimer's-like pathology via regulating the microbiota-gut-brain axis in 5 × FAD mice. Int J Biol Macromol 2024; 270:132372. [PMID: 38750854 DOI: 10.1016/j.ijbiomac.2024.132372] [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: 03/22/2024] [Revised: 05/09/2024] [Accepted: 05/12/2024] [Indexed: 05/26/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterised by neuroinflammation, for which gut dysbiosis may be implicated. Our previous study showed that treatment with Pseudostellaria heterophylla aqueous extract and one of its cyclopeptides, heterophyllin B, attenuate memory deficits via immunomodulation and neurite regeneration. However, whether Pseudostellaria heterophylla polysaccharide (PH-PS) exerts neuroprotective effects against AD and its underlying mechanisms remain unclear. The infrared spectrum, molecular weight, and carbohydrate composition of the PH-PS were determined. The results showed that PH-PS (Mw 8.771 kDa) was composed of glucose (57.78 %), galactose (41.52 %), and arabinose (0.70 %). PH-PS treatment ameliorated learning and spatial memory deficits, reduced amyloid β build-up, and suppressed reactive glia and astrocytes in 5 × FAD mice. 16S rRNA sequencing further showed that PH-PS remodelled the intestinal flora composition by promoting probiotic microbiota, such as Lactobacillus, Muribaculum, Monoglobus, and [Eubacterium]_siraeum_group, and suppressing inflammation-related UCG-009 and Blautia. Additionally, PH-PS restored intestinal barrier function; ameliorated peripheral inflammation by reducing the secretion of inflammatory cytokines, thereby converting M1 microglia and A1 astrocyte toward beneficial M2 and A2 phenotypes; and contributed to Aβ plaques clearance by upregulation of insulin degradation enzyme and neprilysin. Collectively, our findings demonstrate that PH-PS may prevent the progression of AD via modulation of the gut microbiota and regulation of glial polarisation, which could provide evidence to design a potential diet therapy for preventing or curing AD.
Collapse
Affiliation(s)
- Chuantong He
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Zhanjiang Municipal Key laboratory of Marine Drugs and Nutrition for Brain Health, Zhanjiang 524088, China
| | - Jiahui Jiang
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Zhanjiang Municipal Key laboratory of Marine Drugs and Nutrition for Brain Health, Zhanjiang 524088, China
| | - Junxin Liu
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Zhanjiang Municipal Key laboratory of Marine Drugs and Nutrition for Brain Health, Zhanjiang 524088, China
| | - Longjian Zhou
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Zhanjiang Municipal Key laboratory of Marine Drugs and Nutrition for Brain Health, Zhanjiang 524088, China
| | - Yuewei Ge
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Zhiyou Yang
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Zhanjiang Municipal Key laboratory of Marine Drugs and Nutrition for Brain Health, Zhanjiang 524088, China.
| |
Collapse
|
97
|
Zhang Z, Hou L, Liu D, Luan S, Huang M, Zhao L. Directly targeting BAX for drug discovery: Therapeutic opportunities and challenges. Acta Pharm Sin B 2024; 14:2378-2401. [PMID: 38828138 PMCID: PMC11143528 DOI: 10.1016/j.apsb.2024.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/25/2024] [Accepted: 02/04/2024] [Indexed: 06/05/2024] Open
Abstract
For over two decades, the development of B-cell lymphoma-2 (Bcl-2) family therapeutics has primarily focused on anti-apoptotic proteins, resulting in the first-in-class drugs called BH3 mimetics, especially for Bcl-2 inhibitor Venetoclax. The pro-apoptotic protein Bcl-2-associated X protein (BAX) plays a crucial role as the executioner protein of the mitochondrial regulated cell death, contributing to organismal development, tissue homeostasis, and immunity. The dysregulation of BAX is closely associated with the onset and progression of diseases characterized by pathologic cell survival or death, such as cancer, neurodegeneration, and heart failure. In addition to conducting thorough investigations into the physiological modulation of BAX, research on the regulatory mechanisms of small molecules identified through biochemical screening approaches has prompted the identification of functional and potentially druggable binding sites on BAX, as well as diverse all-molecule BAX modulators. This review presents recent advancements in elucidating the physiological and pharmacological modulation of BAX and in identifying potentially druggable binding sites on BAX. Furthermore, it highlights the structural and mechanistic insights into small-molecule modulators targeting diverse binding surfaces or conformations of BAX, offering a promising avenue for developing next-generation apoptosis modulators to treat a wide range of diseases associated with dysregulated cell death by directly targeting BAX.
Collapse
Affiliation(s)
- Zhenwei Zhang
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Linghui Hou
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Dan Liu
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Shenglin Luan
- China Resources Sanjiu Medical & Pharmaceutical Co., Ltd., Shenzhen 518000, China
| | - Min Huang
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Linxiang Zhao
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
98
|
Zhang W, Zhao X, Bhuiyan P, Liu H, Wei H. Neuroprotective effects of dantrolene in neurodegenerative disease: Role of inhibition of pathological inflammation. JOURNAL OF ANESTHESIA AND TRANSLATIONAL MEDICINE 2024; 3:27-35. [PMID: 38826587 PMCID: PMC11138240 DOI: 10.1016/j.jatmed.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Neurodegenerative diseases (NDs) refer to a group of diseases in which slow, continuous cell death is the main pathogenic event in the nervous system. Most NDs are characterized by cognitive dysfunction or progressive motor dysfunction. Treatments of NDs mainly target alleviating symptoms, and most NDs do not have disease-modifying drugs. The pathogenesis of NDs involves inflammation and apoptosis mediated by mitochondrial dysfunction. Dantrolene, approved by the US Food and Drug Administration, acts as a RyRs antagonist for the treatment of malignant hyperthermia, spasticity, neuroleptic syndrome, ecstasy intoxication and exertional heat stroke with tolerable side effects. Recently, dantrolene has also shown therapeutic effects in some NDs. Its neuroprotective mechanisms include the reduction of excitotoxicity, apoptosis and neuroinflammation. In summary, dantrolene can be considered as a potential therapeutic candidate for NDs.
Collapse
Affiliation(s)
- Wenjia Zhang
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Anesthesiology, Shandong Provincial Hospital, Shandong First Medical University, Jinan, Shandong 250021, China
| | - Xu Zhao
- Department of Anesthesiology, Shandong Provincial Hospital, Shandong First Medical University, Jinan, Shandong 250021, China
| | - Piplu Bhuiyan
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Henry Liu
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Huafeng Wei
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
99
|
Tondo G, De Marchi F, Bonardi F, Menegon F, Verrini G, Aprile D, Anselmi M, Mazzini L, Comi C. Novel Therapeutic Strategies in Alzheimer's Disease: Pitfalls and Challenges of Anti-Amyloid Therapies and Beyond. J Clin Med 2024; 13:3098. [PMID: 38892809 PMCID: PMC11172489 DOI: 10.3390/jcm13113098] [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: 04/20/2024] [Revised: 05/21/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Alzheimer's disease (AD) causes a significant challenge to global healthcare systems, with limited effective treatments available. This review examines the landscape of novel therapeutic strategies for AD, focusing on the shortcomings of traditional therapies against amyloid-beta (Aβ) and exploring emerging alternatives. Despite decades of research emphasizing the role of Aβ accumulation in AD pathogenesis, clinical trials targeting Aβ have obtained disappointing results, highlighting the complexity of AD pathophysiology and the need for investigating other therapeutic approaches. In this manuscript, we first discuss the challenges associated with anti-Aβ therapies, including limited efficacy and potential adverse effects, underscoring the necessity of exploring alternative mechanisms and targets. Thereafter, we review promising non-Aβ-based strategies, such as tau-targeted therapies, neuroinflammation modulation, and gene and stem cell therapy. These approaches offer new avenues for AD treatment by addressing additional pathological hallmarks and downstream effects beyond Aβ deposition.
Collapse
Affiliation(s)
- Giacomo Tondo
- Neurology Unit, Department of Translational Medicine, Maggiore della Carità Hospital, University of Piemonte Orientale, 28100 Novara, Italy; (G.T.); (F.B.); (F.M.); (G.V.); (D.A.); (M.A.); (L.M.)
| | - Fabiola De Marchi
- Neurology Unit, Department of Translational Medicine, Maggiore della Carità Hospital, University of Piemonte Orientale, 28100 Novara, Italy; (G.T.); (F.B.); (F.M.); (G.V.); (D.A.); (M.A.); (L.M.)
| | - Francesca Bonardi
- Neurology Unit, Department of Translational Medicine, Maggiore della Carità Hospital, University of Piemonte Orientale, 28100 Novara, Italy; (G.T.); (F.B.); (F.M.); (G.V.); (D.A.); (M.A.); (L.M.)
| | - Federico Menegon
- Neurology Unit, Department of Translational Medicine, Maggiore della Carità Hospital, University of Piemonte Orientale, 28100 Novara, Italy; (G.T.); (F.B.); (F.M.); (G.V.); (D.A.); (M.A.); (L.M.)
| | - Gaia Verrini
- Neurology Unit, Department of Translational Medicine, Maggiore della Carità Hospital, University of Piemonte Orientale, 28100 Novara, Italy; (G.T.); (F.B.); (F.M.); (G.V.); (D.A.); (M.A.); (L.M.)
| | - Davide Aprile
- Neurology Unit, Department of Translational Medicine, Maggiore della Carità Hospital, University of Piemonte Orientale, 28100 Novara, Italy; (G.T.); (F.B.); (F.M.); (G.V.); (D.A.); (M.A.); (L.M.)
| | - Matteo Anselmi
- Neurology Unit, Department of Translational Medicine, Maggiore della Carità Hospital, University of Piemonte Orientale, 28100 Novara, Italy; (G.T.); (F.B.); (F.M.); (G.V.); (D.A.); (M.A.); (L.M.)
| | - Letizia Mazzini
- Neurology Unit, Department of Translational Medicine, Maggiore della Carità Hospital, University of Piemonte Orientale, 28100 Novara, Italy; (G.T.); (F.B.); (F.M.); (G.V.); (D.A.); (M.A.); (L.M.)
| | - Cristoforo Comi
- Neurology Unit, Department of Translational Medicine, Sant’Andrea Hospital, University of Piemonte Orientale, Corso Abbiate 21, 13100 Vercelli, Italy;
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, 28100 Novara, Italy
| |
Collapse
|
100
|
Guo J, Kong Z, Yang S, Da J, Chu L, Han G, Liu J, Tan Y, Zhang J. Therapeutic effects of orexin-A in sepsis-associated encephalopathy in mice. J Neuroinflammation 2024; 21:131. [PMID: 38760784 PMCID: PMC11102217 DOI: 10.1186/s12974-024-03111-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: 02/12/2024] [Accepted: 04/25/2024] [Indexed: 05/19/2024] Open
Abstract
BACKGROUND Sepsis-associated encephalopathy (SAE) causes acute and long-term cognitive deficits. However, information on the prevention and treatment of cognitive dysfunction after sepsis is limited. The neuropeptide orexin-A (OXA) has been shown to play a protective role against neurological diseases by modulating the inflammatory response through the activation of OXR1 and OXR2 receptors. However, the role of OXA in mediating the neuroprotective effects of SAE has not yet been reported. METHODS A mouse model of SAE was induced using cecal ligation perforation (CLP) and treated via intranasal administration of exogenous OXA after surgery. Mouse survival, in addition to cognitive and anxiety behaviors, were assessed. Changes in neurons, cerebral edema, blood-brain barrier (BBB) permeability, and brain ultrastructure were monitored. Levels of pro-inflammatory factors (IL-1β, TNF-α) and microglial activation were also measured. The underlying molecular mechanisms were investigated by proteomics analysis and western blotting. RESULTS Intranasal OXA treatment reduced mortality, ameliorated cognitive and emotional deficits, and attenuated cerebral edema, BBB disruption, and ultrastructural brain damage in mice. In addition, OXA significantly reduced the expression of the pro-inflammatory factors IL-1β and TNF-α, and inhibited microglial activation. In addition, OXA downregulated the expression of the Rras and RAS proteins, and reduced the phosphorylation of P-38 and JNK, thus inhibiting activation of the MAPK pathway. JNJ-10,397,049 (an OXR2 blocker) reversed the effect of OXA, whereas SB-334,867 (an OXR1 blocker) did not. CONCLUSION This study demonstrated that the intranasal administration of moderate amounts of OXA protects the BBB and inhibits the activation of the OXR2/RAS/MAPK pathway to attenuate the outcome of SAE, suggesting that OXA may be a promising therapeutic approach for the management of SAE.
Collapse
Affiliation(s)
- Jing Guo
- GuiZhou University Medical College, Guiyang, 550025, Guizhou Province, China
| | - Zhuo Kong
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Sha Yang
- GuiZhou University Medical College, Guiyang, 550025, Guizhou Province, China
| | - Jingjing Da
- Department of Nephrology, Guizhou Provincial People's Hospital, Guiyang, China
| | - Liangzhao Chu
- Department of Neurosurgery, the Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Guoqiang Han
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Jian Liu
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang, China.
| | - Ying Tan
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang, China.
| | - Jiqin Zhang
- Department of Anesthesiology, Guizhou Provincial People's Hospital, Guiyang, China.
| |
Collapse
|