1
|
Sirimaharaj N, Thiankhaw K, Chattipakorn N, Chattipakorn SC. Unveiling the Protective Roles of Melatonin on Glial Cells in the Battle Against Alzheimer's Disease-Insights from In Vivo and In Vitro Studies. Mol Neurobiol 2025:10.1007/s12035-025-04904-7. [PMID: 40208552 DOI: 10.1007/s12035-025-04904-7] [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: 11/09/2024] [Accepted: 03/31/2025] [Indexed: 04/11/2025]
Abstract
Alzheimer's disease (AD) is a chronic, progressive neurodegenerative disorder that predominantly affects the elderly. Characterized by amyloid-beta (Aβ) plaques and neurofibrillary tangles, AD leads to memory loss, cognitive decline, and severe behavioral changes. As the most common form of dementia, AD imposes a significant global health burden, highlighting the need for interventions that address underlying disease mechanisms rather than only symptomatic treatment. Glial cells, including microglia and astrocytes, play a crucial role in AD progression by mediating neuroinflammatory responses and modulating Aβ clearance and neuronal health. Dysfunction in these cells can exacerbate neuroinflammation and neuronal damage, making glial cells an important target for therapeutic intervention. This review synthesizes findings from in vivo and in vitro studies on melatonin's effects on glial cell dysfunction in AD, emphasizing the multi-mechanistic nature of its neuroprotective properties. Recent studies highlight melatonin's potential as a therapeutic agent that addresses AD-related mechanisms through its interactions with glial cells. Melatonin has demonstrated protective effects, including reducing oxidative stress, apoptosis, and inflammation, inhibiting Aβ fibrillogenesis, and modulating amyloid precursor proteins. Additionally, its influence on glial cell activity, through melatonin receptor pathways, suggests it can alleviate neuroinflammation, a key component of AD progression. The collective evidence points to melatonin's promise as a therapeutic tool with potential roles in both preventive and adjunctive treatments for AD. However, further research is necessary to establish its efficacy and safety in clinical settings.
Collapse
Affiliation(s)
- Nopdanai Sirimaharaj
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Kitti Thiankhaw
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
- The Academy of Science, The Royal Society of Thailand, Bangkok, Thailand
| | - Siriporn C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
2
|
Duggan MR, Morgan DG, Price BR, Rajbanshi B, Martin-Peña A, Tansey MG, Walker KA. Immune modulation to treat Alzheimer's disease. Mol Neurodegener 2025; 20:39. [PMID: 40165251 PMCID: PMC11956194 DOI: 10.1186/s13024-025-00828-x] [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: 10/17/2024] [Accepted: 03/11/2025] [Indexed: 04/02/2025] Open
Abstract
Immune mechanisms play a fundamental role in Alzheimer's disease (AD) pathogenesis, suggesting that approaches which target immune cells and immunologically relevant molecules can offer therapeutic opportunities beyond the recently approved amyloid beta monoclonal therapies. In this review, we provide an overview of immunomodulatory therapeutics in development, including their preclinical evidence and clinical trial results. Along with detailing immune processes involved in AD pathogenesis and highlighting how these mechanisms can be therapeutically targeted to modify disease progression, we summarize knowledge gained from previous trials of immune-based interventions, and provide a series of recommendations for the development of future immunomodulatory therapeutics to treat AD.
Collapse
Affiliation(s)
- Michael R Duggan
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Intramural Research Program, Baltimore, MD, 21224, USA
| | - David G Morgan
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, 49503, USA
| | | | - Binita Rajbanshi
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Alfonso Martin-Peña
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Malú Gámez Tansey
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Keenan A Walker
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Intramural Research Program, Baltimore, MD, 21224, USA.
| |
Collapse
|
3
|
Kour D, Khajuria P, Sharma K, Sharma A, Sharma A, Ali SM, Wazir P, Ramajayan P, Sawant SD, Nandi U, Ahmed Z, Kumar A. Isobavachalcone ameliorates Alzheimer disease pathology by autophagy-mediated clearance of amyloid beta and inhibition of NLRP3 inflammasome in primary astrocytes and 5x-FAD mice. Front Pharmacol 2025; 16:1525364. [PMID: 40183098 PMCID: PMC11965660 DOI: 10.3389/fphar.2025.1525364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 02/24/2025] [Indexed: 04/05/2025] Open
Abstract
Background and Aim Alzheimer's disease (AD) progresses with Aβ plaque deposition and neuroinflammation. Given the complexity of AD pathology, single-target therapies have frequently failed in clinical trials. We hypothesized that a multitarget approach could yield better therapeutic outcomes. To this end, we identified isobavachalcone (IBC), a natural compound with dual pharmacological activity in reducing Aβ plaques and neuroinflammation. Experimental Procedure Primary astrocytes were isolated from 3 to 4 days old C57BL/6J mice pups for in-vitro assays, while in-vivo studies were conducted on 5x-FAD mice. Protein alterations were evaluated using ELISA, western blotting, immunocytochemistry, and immunohistochemistry. Behavioral analyses included the radial arm maze, open field, and rotarod tests. Data from all in vitro and in vivo experiments were analyzed by using one-way ANOVA and post-hoc Bonferroni tests. Results In-vitro analyses in astrocytes demonstrated that IBC at 5 and 10 μM concentrations induce AMPK phosphorylation through CAMKK2, promoting autophagy and inhibiting the NLRP3 inflammasome in primary astrocytes. IBC-treated astrocytes exhibited significant clearance of extracellular amyloid beta. Mechanistic studies highlighted autophagy as a key factor in reducing both NLRP3 inflammasome activity and Aβ levels. Two months of treatment of 5x-FAD mice with IBC at 25 and 50 mg/kg significantly improved cognitive functions, as evidenced by enhanced memory and motor performance in behavioral tests. Subsequent brain tissue analysis revealed that IBC upregulated autophagic proteins to reduce the brain's amyloid beta levels, resulting in decreased expression of neuroinflammation markers. Conclusion IBC effectively ameliorates AD pathology through autophagy-mediated clearance of Aβ and suppressing neuroinflammation in 5x-FAD mice.
Collapse
Affiliation(s)
- Dilpreet Kour
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Parul Khajuria
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Kuhu Sharma
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Alpa Sharma
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Ankita Sharma
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Syed Mudassir Ali
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Priya Wazir
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - P. Ramajayan
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Sanghapal D. Sawant
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Organic Chemsitry Division, CSIR-National Chemical Laboratory, Pune, India
| | - Utpal Nandi
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Zabeer Ahmed
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Ajay Kumar
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
4
|
Mansour A, Eldin MH, El-Sherbiny IM. Metallic nanomaterials in Parkinson's disease: a transformative approach for early detection and targeted therapy. J Mater Chem B 2025; 13:3806-3830. [PMID: 40029109 DOI: 10.1039/d4tb02428a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by substantial loss of dopaminergic neurons in the substantia nigra, leading to both motor and non-motor symptoms that significantly impact quality of life. The prevalence of PD is expected to increase with the aging population, affecting millions globally. Current detection techniques, including clinical assays and neuroimaging, lack the sensitivity and specificity to sense PD in its earliest stages. Despite extensive research, there is no cure for PD, and available treatments primarily focus on symptomatic relief rather than halting disease progression. Conventional treatments, such as levodopa and dopamine agonists, provide limited and often temporary relief, with long-term use associated with significant side effects and diminished efficacy. Nanotechnology, particularly the use of metallic-based nanomaterials (MNMs), offers a promising approach to overcome these limitations. MNMs, due to their unique physicochemical properties, can be engineered to target specific cellular and molecular mechanisms involved in PD. These MNMs can improve drug delivery, enhance imaging and biosensing techniques, and provide neuroprotective effects. For example, gold and silver nanoparticles have shown potential in crossing the blood-brain barrier, providing real-time imaging for early diagnosis and delivering therapeutic agents directly to the affected neurons. This review aims to reveal the current advancements in the use of MNMs for the detection and treatment of PD. It will provide a comprehensive overview of the limitations of conventional detection techniques and therapies, followed by a detailed discussion on how nanotechnology can address these challenges. The review will also highlight recent preclinical research and examine the potential toxicity of MNMs. By emphasizing the potential of MNMs, this review article aims to underscore the transformative impact of nanotechnology in revolutionizing the detection and treatment of PD.
Collapse
Affiliation(s)
- Amira Mansour
- Nanomedicine Laboratories, Center for Materials Science, Zewail City of Science and Technology, 6th October City, 12578 Giza, Egypt.
| | - Mariam Hossam Eldin
- Nanomedicine Laboratories, Center for Materials Science, Zewail City of Science and Technology, 6th October City, 12578 Giza, Egypt.
| | - Ibrahim M El-Sherbiny
- Nanomedicine Laboratories, Center for Materials Science, Zewail City of Science and Technology, 6th October City, 12578 Giza, Egypt.
| |
Collapse
|
5
|
Sun Y, Liu Z, Zhang Z, Kang Y, Wang X, Zhang Y, Liu Y, Zhao P. Human induced pluripotent stem cell models for Alzheimer's disease research: a bibliometric analysis. Front Hum Neurosci 2025; 19:1548701. [PMID: 40177166 PMCID: PMC11962003 DOI: 10.3389/fnhum.2025.1548701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/06/2025] [Indexed: 04/05/2025] Open
Abstract
Introduction Alzheimer's disease (AD), the leading cause of dementia, remains without adequate treatment. Current models do not fully replicate human physiology and pathology. The advent of human induced pluripotent stem cell (hiPSC) technology offers a novel approach to studying AD. Methods Our study conducted a bibliometric analysis to assess the application and development of hiPSC technology in AD research. We retrieved 531 articles on hiPSC models of AD from the Web of Science Core Collection, published between January 2010 and June 2024. CiteSpace and VOSviewer were used to analyze authorship, geographic contributions, journal influence, and citation patterns. Results Our findings reveal a steady increase in publications over 14 years, with the United States leading in contributions, followed by China. Li-Huei Tsai from the Massachusetts Institute of Technology is a prominent researcher. PLoS One emerges as the most influential journal. Research trends have focused on inflammation, astrocytes, microglia, apolipoprotein E (ApoE), and tau. Discussion Bibliometric analysis is crucial in identifying research gaps and trends and guiding future studies to address unmet needs in understanding and modeling human physiology and pathology. Leveraging hiPSC models to investigate the molecular mechanisms of familial and sporadic AD is expected to provide a crucial foundation for developing future treatment strategies. Conclusion In summary, the bibliometric findings from this study provide a comprehensive overview of the current research landscape in hiPSC models for AD. It also highlights emerging trends and research gaps, crucial for guiding future research efforts, particularly in exploring novel therapeutic targets and improving understanding of disease mechanisms.
Collapse
Affiliation(s)
- Yuning Sun
- School of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, China
- Gansu Provincial People’s Hospital, Lanzhou, China
| | - Zhilong Liu
- Gansu Provincial People’s Hospital, Lanzhou, China
| | - Zongbo Zhang
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Yufeng Kang
- School of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, China
| | - Xinlian Wang
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Yiping Zhang
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Yan Liu
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Pei Zhao
- School of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, China
- Gansu Provincial People’s Hospital, Lanzhou, China
| |
Collapse
|
6
|
Sánchez SV, Otavalo GN, Gazeau F, Silva AKA, Morales JO. Intranasal delivery of extracellular vesicles: A promising new approach for treating neurological and respiratory disorders. J Control Release 2025; 379:489-523. [PMID: 39800240 DOI: 10.1016/j.jconrel.2025.01.018] [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: 09/13/2024] [Revised: 01/03/2025] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
BACKGROUND Extracellular vesicles (EVs) are membrane vesicles secreted by all types of cells, including bacteria, animals, and plants. These vesicles contain proteins, nucleic acids, and lipids from their parent cells and can transfer these components between cells. EVs have attracted attention for their potential use in diagnosis and therapy due to their natural properties, such as low immunogenicity, high biocompatibility, and ability to cross the blood-brain barrier. They can also be engineered to carry therapeutic molecules. EVs can be delivered via various routes. The intranasal route is particularly advantageous for delivering them to the central nervous system, making it a promising approach for treating neurological disorders. SCOPE OF REVIEW This review delves into the promising potential of intranasally administered EVs-based therapies for various medical conditions, with a particular focus on those affecting the brain and central nervous system. Additionally, the potential use of these therapies for pulmonary conditions, cancer, and allergies is examined, offering a hopeful outlook for the future of medical treatments. MAJOR CONCLUSIONS The intranasal administration of EVs offers significant advantages over other delivery methods. By directly delivering EVs to the brain, specifically targeting areas that have been injured, this administration proves to be highly efficient and effective, providing reassurance about the progress in medical treatments. Intranasal delivery is not limited to brain-related conditions. It can also benefit other organs like the lungs and stimulate a mucosal immune response against various pathogens due to the highly vascularized nature of the nasal cavity and airways. Moreover, it has the added benefit of minimizing toxicity to non-targeted organs and allows the EVs to remain longer in the body. As a result, there is a growing emphasis on conducting clinical trials for intranasal administration of EVs, particularly in treating respiratory tract pathologies such as coronavirus disease.
Collapse
Affiliation(s)
- Sofía V Sánchez
- Drug Delivery Laboratory, Departamento de Ciencias y Tecnología Farmacéuticas, Universidad de Chile, Santiago, Chile; Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile; Center of New Drugs for Hypertension and Heart Failure (CENDHY), Santiago, Chile
| | - Gabriela N Otavalo
- Drug Delivery Laboratory, Departamento de Ciencias y Tecnología Farmacéuticas, Universidad de Chile, Santiago, Chile; Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile; Center of New Drugs for Hypertension and Heart Failure (CENDHY), Santiago, Chile
| | - Florence Gazeau
- Université Paris Cité, CNRS UMR8175, INSERM U1334, Laboratory NABI (Nanomédecine, Biologie Extracellulaire, Intégratome et Innovations en santé), Paris, France
| | - Amanda K A Silva
- Université Paris Cité, CNRS UMR8175, INSERM U1334, Laboratory NABI (Nanomédecine, Biologie Extracellulaire, Intégratome et Innovations en santé), Paris, France
| | - Javier O Morales
- Drug Delivery Laboratory, Departamento de Ciencias y Tecnología Farmacéuticas, Universidad de Chile, Santiago, Chile; Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile; Center of New Drugs for Hypertension and Heart Failure (CENDHY), Santiago, Chile.
| |
Collapse
|
7
|
A E F Cardinali C, Martins YA, C M Moraes R, Costa AP, Torrão AS. Benfotiamine Ameliorates Streptozotocin-Induced Alzheimer's Disease in Rats by Modulating Neuroinflammation, Oxidative Stress, and Microglia. Mol Neurobiol 2025:10.1007/s12035-025-04811-x. [PMID: 40038195 DOI: 10.1007/s12035-025-04811-x] [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: 10/14/2024] [Accepted: 02/25/2025] [Indexed: 03/06/2025]
Abstract
Alzheimer's disease (AD) is the most prevalent cause of dementia, characterized by progressive memory loss and cognitive decline. Recent evidence indicates that inflammation plays a central role in AD pathogenesis, with elevated inflammatory markers and risk genes linked to innate immune functions. Glial cell dysfunction, particularly in astrocytes and microglia, is crucial to the neuroinflammatory process, contributing to oxidative stress, synaptic dysfunction, neuronal death, and impaired neurogenesis. This study aimed to investigate the therapeutic effects of benfotiamine (BFT), a vitamin B1 analogue, on microglial morphology, inflammation, and oxidative stress parameters in a sporadic Alzheimer-like disease model induced by intracerebroventricular injection of streptozotocin (STZ). Supplementation with 150 mg/kg of BFT for 7 days significantly reduced inflammation in the hippocampus and provided protection against oxidative damage in the entorhinal cortex by activating the Nrf-2 pathway and enhancing the expression of antioxidant enzymes such as SOD1 and CAT. These findings suggest that BFT exerts neuroprotective effects in AD, particularly impacting glial cell function and redox homeostasis.
Collapse
Affiliation(s)
- Camila A E F Cardinali
- Departamento de Fisiologia E Biofisica, Universidade de Sao Paulo, Av Professor Lineu Prestes 2415, Sao Paulo, 05508-000, Brazil.
| | - Yandara A Martins
- Departamento de Fisiologia E Biofisica, Universidade de Sao Paulo, Av Professor Lineu Prestes 2415, Sao Paulo, 05508-000, Brazil
| | - Ruan C M Moraes
- Departamento de Fisiologia E Biofisica, Universidade de Sao Paulo, Av Professor Lineu Prestes 2415, Sao Paulo, 05508-000, Brazil
- Department of Psychiatry & Behavioral Neurobiology, The University of Alabama at Birmingham, Alabama, USA
| | - Andressa P Costa
- Departamento de Fisiologia E Biofisica, Universidade de Sao Paulo, Av Professor Lineu Prestes 2415, Sao Paulo, 05508-000, Brazil
| | - Andréa S Torrão
- Departamento de Fisiologia E Biofisica, Universidade de Sao Paulo, Av Professor Lineu Prestes 2415, Sao Paulo, 05508-000, Brazil
| |
Collapse
|
8
|
Chen L, Zhao X, Sheng R, Lazarovici P, Zheng W. Artemisinin alleviates astrocyte overactivation and neuroinflammation by modulating the IRE1/NF-κB signaling pathway in in vitro and in vivo Alzheimer's disease models. Free Radic Biol Med 2025; 229:96-110. [PMID: 39826816 DOI: 10.1016/j.freeradbiomed.2025.01.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 12/25/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
Recent studies have shown that neuroinflammation and heightened glial activity, particularly astrocyte overactivation, are associated with Alzheimer's disease (AD). Abnormal accumulation of amyloid-beta (Aβ) induces endoplasmic reticulum (ER) stress and activates astrocytes. Artemisinin (ART), a frontline anti-malarial drug, has been found to have neuroprotective properties. However, its impact on astrocytes remains unclear. In this study, we used Aβ1-42 induced astrocyte cultures and 3 × Tg-AD mice as in vitro and in vivo models, respectively, to investigate the effects of ART on AD related astrocyte overactivation and its underlying mechanisms. ART attenuated Aβ1-42-induced astrocyte activation, ER stress, and inflammatory responses in astrocyte cultures by inhibiting IRE1 phosphorylation and the NF-κB pathway, as evidenced by the overexpression of IRE1 WT and IRE1-K599A (kinase activity invalidated), along with application of activators and inhibitors related to ER stress. Furthermore, ART alleviated the detrimental effects and restored neurotrophic function of astrocytes on co-cultured neurons, preventing neuronal apoptosis during Aβ1-42 treatment. In 3 × Tg-AD mice, ART treatment improved cognitive function and reduced astrocyte overactivation, neuroinflammation, ER stress, and neuronal apoptosis. Moreover, ART attenuated the upregulation of IRE1/NF-κB pathway activity in AD mice. Astrocyte-specific overexpression of IRE1 via adeno-associated virus in AD mice reversed the ameliorating effects of ART. Our findings suggest that ART inhibits astrocyte overactivation and neuroinflammation in both in vitro and in vivo AD models by modulating the IRE1/NF-κB signaling pathway, thereby enhancing neuronal functions. This study underscores the therapeutic potential of ART in AD and highlights the significance of modulating the ER stress-inflammatory cycle and normalizing astrocyte-neuron communication.
Collapse
Affiliation(s)
- Lei Chen
- Pharmaceutical Science, Faculty of Health Sciences, University of Macau, Macau, Taipa, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, China
| | - Xia Zhao
- Pharmaceutical Science, Faculty of Health Sciences, University of Macau, Macau, Taipa, China
| | - Rui Sheng
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, China.
| | - Philip Lazarovici
- School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 9112002, Israel
| | - Wenhua Zheng
- Pharmaceutical Science, Faculty of Health Sciences, University of Macau, Macau, Taipa, China.
| |
Collapse
|
9
|
Han X, Chen X, Zheng X, Yan F. Strawberry anthocyanin pelargonidin-3-glucoside attenuated OA-induced neurotoxicity by activating UPR mt. Food Funct 2025; 16:1330-1346. [PMID: 39873116 DOI: 10.1039/d4fo04639k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2025]
Abstract
In this study, network pharmacology analysis revealed that strawberry anthocyanins mainly interfered with lipid metabolism and nerve-related signaling pathways. Pelargonidin-3-glucoside (Pg3G), one of the main anthocyanins in strawberry, was screened as the most effective anthocyanin for attenuating excess lipid accumulation. Moreover, Pg3G decreased lipid levels, relieved oxidative stress, and restored abnormal behavioral activities in Caenorhabditis elegans under oleic acid (OA) exposure. Meanwhile, Pg3G increased the expression of HSP-6 and HSP-60 proteins and activated the mitochondrial unfolded protein response (UPRmt), while beneficial effects of Pg3G were impaired in the ubl-5 knockout strain, suggesting that ubl-5 may be a key target for improving OA-induced neurotoxicity. Expressions of neurotransmitter transmission-related genes showed great correlations with genes involved in lipid metabolism and UPRmt, further explaining the underlying mechanism of Pg3G in neuroprotection. Our findings emphasize the key role of UPRmt in alleviating OA-induced neurotoxicity of Pg3G, providing a theoretical basis for the research and development of strawberry anthocyanins as a dietary supplement for lipid reduction and treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Xiao Han
- Department of Food Science and Nutrition, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China.
| | - Xinyi Chen
- Department of Food Science and Nutrition, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China.
| | - Xiaodong Zheng
- Department of Food Science and Nutrition, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China.
| | - Fujie Yan
- Department of Food Science and Nutrition, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
10
|
Bjerkan J, Meglič B, Lancaster G, Kobal J, McClintock PVE, Crawford TJ, Stefanovska A. Neurovascular phase coherence is altered in Alzheimer's disease. Brain Commun 2025; 7:fcaf007. [PMID: 40008330 PMCID: PMC11852277 DOI: 10.1093/braincomms/fcaf007] [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: 08/15/2024] [Revised: 11/01/2024] [Accepted: 01/08/2025] [Indexed: 02/27/2025] Open
Abstract
Alzheimer's disease is the commonest form of dementia, but its cause still remains elusive. It is characterized by neurodegeneration, with amyloid-beta and tau aggregation. Recently, however, the roles of the vasculature and the neurovascular unit are being highlighted as important for disease progression. In particular, there is reduced microvascular density, and altered gene expression in vascular and glial cells. Structural changes naturally impact the functioning of the neurovascular unit, and the goal of the study was to quantify the corresponding changes in vivo, non-invasively. Our assessment is based on recordings of brain oxygenation, neuronal and cardiorespiratory activities, captured by functional near-infrared spectroscopy, electroencephalogram, electrocardiogram and respiration effort, respectively. Two groups were compared: an Alzheimer's disease group (N = 19) and a control group (N = 20) of similar age. The time-series were analysed using methods that can capture multi-scale and time-varying oscillations such as the wavelet transform power and wavelet phase coherence. The Alzheimer's disease group shows a significant decrease in the power of brain oxygenation oscillations compared to the control group. There is also a significant global reduction in the phase coherence between brain oxygenation time-series. The neurovascular phase coherence around 0.1 Hz is also significantly reduced in the Alzheimer's disease group. In addition, the average respiration rate is increased in the Alzheimer's disease group compared to the control group. We show that the phase coherence between vascular and neuronal activities is reduced in Alzheimer's disease compared to the control group, indicating altered functioning of the neurovascular unit. The brain oxygenation dynamics reveals reduced power and coordination of oscillations, especially in frequency ranges that are associated with vasomotion. This could lead to reduced oxygen delivery to the brain, which could affect ATP production, and potentially reduce amyloid-beta clearance. These changes in neurovascular dynamics have potential for early diagnosis, as a marker of disease progression, and for evaluating the effect of interventions.
Collapse
Affiliation(s)
- Juliane Bjerkan
- Department of Physics, Lancaster University, LA1 4YB Lancaster, UK
| | - Bernard Meglič
- Department of Neurology, University Medical Centre, 1525 Ljubljana, Slovenia
| | - Gemma Lancaster
- Department of Physics, Lancaster University, LA1 4YB Lancaster, UK
| | - Jan Kobal
- Department of Neurology, University Medical Centre, 1525 Ljubljana, Slovenia
| | | | - Trevor J Crawford
- Department of Psychology, Lancaster University, LA1 4YF Lancaster, UK
| | | |
Collapse
|
11
|
Radhakrishnan K, Zhang Y, Mustapha O, Weigel TK, Upchurch CM, Tian X, Herbert F, Huang W, Leitinger N, Eyo UB, Ai H, Ferris HA. 7-ketocholesterol contributes to microglia-driven increases in astrocyte reactive oxygen species in Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.19.633810. [PMID: 39868327 PMCID: PMC11761689 DOI: 10.1101/2025.01.19.633810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Oxidative stress is a prominent feature of Alzheimer's disease. Within this context, cholesterol undergoes oxidation, producing the pro-inflammatory product 7-ketocholesterol (7-KC). In this study, we observe elevated levels of 7-KC in the brains of the 3xTg mouse model of AD. To further understand the contribution of 7-KC on the oxidative environment, we developed a method to express a genetically encoded fluorescent hydrogen peroxide (H2O2) sensor in astrocytes, the primary source of cholesterol in the brain. With this sensor, we discovered that 7-KC increases H2O2 levels in astrocytes in vivo, but not when directly applied to astrocytes in vitro. Interestingly, when 7-KC was applied to a microglia cell line alone or mixed astrocyte and microglia cultures, it resulted in microglia activation and increased oxidative stress in astrocytes. Depletion of microglia from 3xTg mice resulted in reduced 7-KC in the brains of these mice. Taken together, these findings suggest that 7-KC, acting through microglia, contributes to increased astrocyte oxidative stress in AD. This study sheds light on the complex interplay between cholesterol oxidation, microglia activation, and astrocyte oxidative stress in the pathogenesis of AD.
Collapse
Affiliation(s)
- Kayalvizhi Radhakrishnan
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
- Division of Endocrinology and Metabolism, University of Virginia, Charlottesville, VA, USA
| | - Yiyu Zhang
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA
| | - Oluwaseun Mustapha
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA
| | - Thaddeus K. Weigel
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Clint M. Upchurch
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Xiaodong Tian
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA
| | - Franklin Herbert
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA
| | - Wenyuan Huang
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA
| | - Norbert Leitinger
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Ukpong B. Eyo
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Huiwang Ai
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA
| | - Heather A. Ferris
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
- Division of Endocrinology and Metabolism, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
12
|
la Torre A, Lo Vecchio F, Angelillis VS, Gravina C, D’Onofrio G, Greco A. Reinforcing Nrf2 Signaling: Help in the Alzheimer's Disease Context. Int J Mol Sci 2025; 26:1130. [PMID: 39940900 PMCID: PMC11818887 DOI: 10.3390/ijms26031130] [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: 12/24/2024] [Revised: 01/13/2025] [Accepted: 01/15/2025] [Indexed: 02/16/2025] Open
Abstract
Oxidative stress plays a role in various pathophysiological diseases, including neurogenerative diseases, such as Alzheimer's disease (AD), which is the most prevalent neuro-pathology in the aging population. Oxidative stress has been reported to be one of the earliest pathological alterations in AD. Additionally, it was demonstrated that in older adults, there is a loss of free radical scavenging ability. The Nrf2 transcription factor is a key regulator in antioxidant defense systems, but, with aging, both the amount and the transcriptional activity of Nrf2 decrease. With the available treatments for AD being poorly effective, reinforcing the antioxidant defense systems via the Nrf2 pathway may be a way to prevent and treat AD. To highlight the predominant role of Nrf2 signaling in defending against oxidative stress and, therefore, against neurotoxicity, we present an overview of the natural compounds that exert their own neuroprotective roles through the activation of the Nrf2 pathway. This review is an opportunity to promote a holistic approach in the treatment of AD and to highlight the need to further refine the development of new potential Nrf2-targeting drugs.
Collapse
Affiliation(s)
- Annamaria la Torre
- Laboratory of Gerontology and Geriatrics, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (F.L.V.); (C.G.)
| | - Filomena Lo Vecchio
- Laboratory of Gerontology and Geriatrics, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (F.L.V.); (C.G.)
| | - Valentina Soccorsa Angelillis
- Complex Unit of Geriatrics, Department of Medical Sciences, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (V.S.A.); (A.G.)
| | - Carolina Gravina
- Laboratory of Gerontology and Geriatrics, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (F.L.V.); (C.G.)
| | - Grazia D’Onofrio
- Clinical Psychology Service, Health Department, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy;
| | - Antonio Greco
- Complex Unit of Geriatrics, Department of Medical Sciences, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (V.S.A.); (A.G.)
| |
Collapse
|
13
|
Choi JJ, Cohen Kalafut N, Gruenloh T, Engelman CD, Lu T, Wang D. COSIME: Cooperative multi-view integration and Scalable and Interpretable Model Explainer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.11.632570. [PMID: 39868220 PMCID: PMC11761389 DOI: 10.1101/2025.01.11.632570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Single-omics approaches often provide a limited view of complex biological systems, whereas multiomics integration offers a more comprehensive understanding by combining diverse data views. However, integrating heterogeneous data types and interpreting the intricate relationships between biological features-both within and across different data views-remains a bottleneck. To address these challenges, we introduce COSIME (Cooperative Multi-view Integration and Scalable Interpretable Model Explainer). COSIME uses backpropagation of Learnable Optimal Transport (LOT) to deep neural networks, enabling the learning of latent features from multiple views to predict disease phenotypes. In addition, COSIME incorporates Monte Carlo sampling to efficiently estimate Shapley values and Shapley-Taylor indices, enabling the assessment of both feature importance and their pairwise interactions-synergistically or antagonistically-in predicting disease phenotypes. We applied COSIME to both simulated data and real-world datasets, including single-cell transcriptomics, single-cell spatial transcriptomics, epigenomics, and metabolomics, specifically for Alzheimer's disease-related phenotypes. Our results demonstrate that COSIME significantly improves prediction performance while offering enhanced interpretability of feature relationships. For example, we identified that synergistic interactions between microglia and astrocyte genes associated with AD are more likely to be active at the edges of the middle temporal gyrus as indicated by spatial locations. Finally, COSIME is open-source and available for general use.
Collapse
|
14
|
Inamdar A, Gurupadayya B, Halagali P, Nandakumar S, Pathak R, Singh H, Sharma H. Cutting-edge Strategies for Overcoming Therapeutic Barriers in Alzheimer's Disease. Curr Pharm Des 2025; 31:598-618. [PMID: 39492772 DOI: 10.2174/0113816128344571241018154506] [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: 08/04/2024] [Revised: 09/13/2024] [Accepted: 09/16/2024] [Indexed: 11/05/2024]
Abstract
Alzheimer's disease (AD) remains one of the hardest neurodegenerative diseases to treat due to its enduring cognitive deterioration and memory loss. Despite extensive research, few viable treatment approaches have been found; these are mostly due to several barriers, such as the disease's complex biology, limited pharmaceutical efficacy, and the BBB. This presentation discusses current strategies for addressing these therapeutic barriers to enhance AD treatment. Innovative drug delivery methods including liposomes, exosomes, and nanoparticles may be able to pass the blood-brain barrier and allow medicine to enter specific brain regions. These innovative strategies of medicine distribution reduce systemic side effects by improving absorption. Moreover, the development of disease-modifying treatments that target tau protein tangles, amyloid-beta plaques, and neuroinflammation offers the chance to influence the course of the illness rather than only treat its symptoms. Furthermore, gene therapy and CRISPR-Cas9 technologies have surfaced as potentially groundbreaking methods for addressing the underlying genetic defects associated with AD. Furthermore, novel approaches to patient care may involve the utilization of existing medications having neuroprotective properties, such as those for diabetes and cardiovascular conditions. Furthermore, biomarker research and personalized medicine have made individualized therapy approaches possible, ensuring that patients receive the best care possible based on their unique genetic and molecular profiles.
Collapse
Affiliation(s)
- Aparna Inamdar
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, Karnataka, India
| | - Bannimath Gurupadayya
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, Karnataka, India
| | - Prashant Halagali
- Department of Pharmaceutical Quality Assurance, KLE College of Pharmacy, KLE Academy of Higher Education and Research, Belagavi 590010, Karnataka, India
| | - S Nandakumar
- Associate Scientist, Corteva Agriscience, Hyderabad 500081, Telangana, India
| | - Rashmi Pathak
- Department of Pharmacy, Invertis University, Bareilly (UP) 243123, India
| | - Himalaya Singh
- Department of Medicine, Government Institute of Medical Sciences, Greater Noida (UP) 201312, India
| | - Himanshu Sharma
- Department of Pharmacy, Teerthanker Mahaveer College of Pharmacy, Teerthanker Mahaveer University, Moradabad (UP) 244001, India
| |
Collapse
|
15
|
Zhang Y, Wang Z, Xu F, Liu Z, Zhao Y, Yang LZ, Fang W. Progress of Astrocyte-Neuron Crosstalk in Central Nervous System Diseases. Neurochem Res 2024; 49:3187-3207. [PMID: 39292330 DOI: 10.1007/s11064-024-04241-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/08/2024] [Accepted: 09/04/2024] [Indexed: 09/19/2024]
Abstract
Neurons are the primary cells responsible for information processing in the central nervous system (CNS). However, they are vulnerable to damage and insult in a variety of neurological disorders. As the most abundant glial cells in the brain, astrocytes provide crucial support to neurons and participate in synapse formation, synaptic transmission, neurotransmitter recycling, regulation of metabolic processes, and the maintenance of the blood-brain barrier integrity. Though astrocytes play a significant role in the manifestation of injury and disease, they do not work in isolation. Cellular interactions between astrocytes and neurons are essential for maintaining the homeostasis of the CNS under both physiological and pathological conditions. In this review, we explore the diverse interactions between astrocytes and neurons under physiological conditions, including the exchange of neurotrophic factors, gliotransmitters, and energy substrates, and different CNS diseases such as Alzheimer's disease, Parkinson's disease, stroke, traumatic brain injury, and multiple sclerosis. This review sheds light on the contribution of astrocyte-neuron crosstalk to the progression of neurological diseases to provide potential therapeutic targets for the treatment of neurological diseases.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Mailbox 207, Tongjiaxiang 24, Nanjing, 210009, P. R. China
| | - Ziyu Wang
- Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Mailbox 207, Tongjiaxiang 24, Nanjing, 210009, P. R. China
| | - Fenglian Xu
- Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Mailbox 207, Tongjiaxiang 24, Nanjing, 210009, P. R. China
| | - Zijun Liu
- Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Mailbox 207, Tongjiaxiang 24, Nanjing, 210009, P. R. China
| | - Yu Zhao
- Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Mailbox 207, Tongjiaxiang 24, Nanjing, 210009, P. R. China
| | - Lele Zixin Yang
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, PA, 19107, USA
| | - Weirong Fang
- Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Mailbox 207, Tongjiaxiang 24, Nanjing, 210009, P. R. China.
| |
Collapse
|
16
|
Niu Q, Li D, Zhang J, Piao Z, Xu B, Xi Y, Mohamed Kamal NNSN, Lim V, Li P, Yin Y. The new perspective of Alzheimer's Disease Research: Mechanism and therapeutic strategy of neuronal senescence. Ageing Res Rev 2024; 102:102593. [PMID: 39566741 DOI: 10.1016/j.arr.2024.102593] [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: 10/16/2024] [Accepted: 11/16/2024] [Indexed: 11/22/2024]
Abstract
Alzheimer's disease (AD), commonly known as senile dementia, is a neurodegenerative disease with insidious onset and gradually worsening course. The brain is particularly sensitive to senescence, and neuronal senescence is an important risk factor for the occurrence of AD. However, the exact pathogenesis between neuronal senescence and AD has not been fully elucidated so far. Neuronal senescence is characterized by the permanent stagnation of the cell cycle, and the changes in its structure, function, and microenvironment are closely related to the pathogenesis and progression of AD. In recent years, studies such as the Aβ cascade hypothesis and Tau protein phosphorylation have provided new strategies for the therapy of AD, but due to the complexity of the etiology of AD, there are still no effective treatment measures. This article aims to deeply analyze the pathogenesis between AD and neuronal senescence, and sort out various existing therapeutic methods, to provide new ideas and references for the clinical treatment of AD.
Collapse
Affiliation(s)
- Qianqian Niu
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China; Department of Toxicology, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang 13200, Malaysia
| | - Danjie Li
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, School of Pharmacy, Xinxiang 453003, China
| | - Jiayin Zhang
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, School of Pharmacy, Xinxiang 453003, China
| | - Zhengji Piao
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, School of Pharmacy, Xinxiang 453003, China
| | - Bo Xu
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, School of Pharmacy, Xinxiang 453003, China
| | - Yuting Xi
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, School of Pharmacy, Xinxiang 453003, China
| | - Nik Nur Syazni Nik Mohamed Kamal
- Department of Toxicology, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang 13200, Malaysia; Dementia Multidisciplinary Research Program of IPPT (DMR-IPPT), Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang 13200, Malaysia.
| | - Vuanghao Lim
- Department of Toxicology, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang 13200, Malaysia.
| | - Peng Li
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, School of Pharmacy, Xinxiang 453003, China.
| | - Yaling Yin
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China.
| |
Collapse
|
17
|
Zeng M, Peng M, Liang J, Sun H. The Role of Gut Microbiota in Blood-Brain Barrier Disruption after Stroke. Mol Neurobiol 2024; 61:9735-9755. [PMID: 37498481 DOI: 10.1007/s12035-023-03512-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 07/13/2023] [Indexed: 07/28/2023]
Abstract
Growing evidence has proved that alterations in the gut microbiota have been linked to neurological disorders including stroke. Structural and functional disruption of the blood-brain barrier (BBB) is observed after stroke. In this context, there is pioneering evidence supporting that gut microbiota may be involved in the pathogenesis of stroke by regulating the BBB function. However, only a few experimental studies have been performed on stroke models to observe the BBB by altering the structure of gut microbiota, which warrant further exploration. Therefore, in order to provide a novel mechanism for stroke and highlight new insights into BBB modification as a stroke intervention, this review summarizes existing evidence of the relationship between gut microbiota and BBB integrity and discusses the mechanisms of gut microbiota on BBB dysfunction and its role in stroke.
Collapse
Affiliation(s)
- Meiqin Zeng
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China On Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory On Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Meichang Peng
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China On Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory On Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Jianhao Liang
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China On Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory On Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Haitao Sun
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, China.
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China On Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory On Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China.
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Centre for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, China.
| |
Collapse
|
18
|
Zhu J, Wu C, Yang L. Cellular senescence in Alzheimer's disease: from physiology to pathology. Transl Neurodegener 2024; 13:55. [PMID: 39568081 PMCID: PMC11577763 DOI: 10.1186/s40035-024-00447-4] [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: 05/30/2024] [Accepted: 10/12/2024] [Indexed: 11/22/2024] Open
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative disorders, characterized by the accumulation of Aβ and abnormal tau hyperphosphorylation. Despite substantial efforts in development of drugs targeting Aβ and tau pathologies, effective therapeutic strategies for AD remain elusive. Recent attention has been paid to the significant role of cellular senescence in AD progression. Mounting evidence suggests that interventions targeting cellular senescence hold promise in improving cognitive function and ameliorating hallmark pathologies in AD. This narrative review provides a comprehensive summary and discussion of the physiological roles, characteristics, biomarkers, and commonly employed in vivo and in vitro models of cellular senescence, with a particular focus on various cell types in the brain, including astrocytes, microglia, oligodendrocyte precursor cells, neurons, and endothelial cells. The review further delves into factors influencing cellular senescence in AD and emphasizes the significance of targeting cellular senescence as a promising approach for AD treatment, which includes the utilization of senolytics and senomorphics.
Collapse
Affiliation(s)
- Jing Zhu
- Department of Pulmonary and Critical Care Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430014, Hubei, China
| | - Chongyun Wu
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, Guangdong, China
| | - Luodan Yang
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, Guangdong, China.
| |
Collapse
|
19
|
Mitroshina EV, Kalinina EP, Kalyakulina AI, Teplyakova AV, Vedunova MV. The Effect of the Optogenetic Stimulation of Astrocytes on Neural Network Activity in an In Vitro Model of Alzheimer's Disease. Int J Mol Sci 2024; 25:12237. [PMID: 39596305 PMCID: PMC11594756 DOI: 10.3390/ijms252212237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/07/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
Optogenetics is a combination of optical and genetic technologies used to activate or, conversely, inhibit specific cells in living tissues. The possibilities of using optogenetics approaches for the treatment of epilepsy, Parkinson's and Alzheimer's disease (AD) are being actively researched. In recent years, it has become clear that one of the most important players in the development of AD is astrocytes. Astrocytes affect amyloid clearance, participate in the development of neuroinflammation, and regulate the functioning of neural networks. We used an adeno-associated virus carrying the glial fibrillary acidic protein (GFAP) promoter driving the optogenetic channelrhodopsin-2 (ChR2) gene to transduce astrocytes in primary mouse hippocampal cultures. We recorded the bioelectrical activity of neural networks from day 14 to day 21 of cultivation using multielectrode arrays. A single optogenetic stimulation of astrocytes at 14 day of cultivation (DIV14) did not cause significant changes in neural network bioelectrical activity. Chronic optogenetic stimulation from DIV14 to DIV21 exerts a stimulatory effect on the bioelectrical activity of primary hippocampal cultures (the proportion of spikes included in network bursts significantly increased since DIV19). Moreover, chronic optogenetic stimulation over seven days partially preserved the activity and functional architecture of neuronal network in amyloidosis modeling. These results suggest that the selective optogenetic activation of astrocytes may represent a promising novel therapeutic strategy for combating AD.
Collapse
Affiliation(s)
- Elena V. Mitroshina
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Avenue, 603022 Nizhny Novgorod, Russia
| | - Elizaveta P. Kalinina
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Avenue, 603022 Nizhny Novgorod, Russia
| | - Alena I. Kalyakulina
- Institute of Information Technologies, Mathematics and Mechanics, Lobachevsky State University, 603022 Nizhny Novgorod, Russia
- Institute of Biogerontology, Lobachevsky State University, 603022 Nizhny Novgorod, Russia
| | - Alexandra V. Teplyakova
- Federal Center of Brain Research and Neurotechnologies, Federal Medical-Biological Agency, 119330 Moscow, Russia
| | - Maria V. Vedunova
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Avenue, 603022 Nizhny Novgorod, Russia
| |
Collapse
|
20
|
Zhang S, Gao Z, Feng L, Li M. Prevention and Treatment Strategies for Alzheimer's Disease: Focusing on Microglia and Astrocytes in Neuroinflammation. J Inflamm Res 2024; 17:7235-7259. [PMID: 39421566 PMCID: PMC11484773 DOI: 10.2147/jir.s483412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024] Open
Abstract
Alzheimer's disease (AD) is a fatal neurodegenerative disease characterized by its insidious onset and progressive development, making it the most common form of dementia. Despite its prevalence, the exact causes and mechanisms responsible for AD remain unclear. Recent studies have highlighted that inflammation in the central nervous system (CNS) plays a crucial role in both the initiation and progression of AD. Neuroinflammation, an immune response within the CNS triggered by glial cells in response to various stimuli, such as nerve injury, infection, toxins, or autoimmune reactions, has emerged as a significant factor alongside amyloid deposition and neurofibrillary tangles (NFTs) commonly associated with AD. This article aims to provide an overview of the most recent research regarding the involvement of neuroinflammation in AD, with a particular focus on elucidating the specific mechanisms involving microglia and astrocytes. By exploring these intricate processes, a new theoretical framework can be established to further probe the impact of neuroinflammation on the development and progression of AD. Through a deeper understanding of these underlying mechanisms, potential targets for therapeutic interventions and novel treatment strategies can be identified in the ongoing battle against AD.
Collapse
Affiliation(s)
- Shenghao Zhang
- Department of Neurology, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin Province, 130021, People’s Republic of China
| | - Zhejianyi Gao
- Department of Orthopaedics, Fushun Hospital of Chinese Medicine, Fushun, Liaoning Province, 113008, People’s Republic of China
| | - Lina Feng
- Department of Neurology, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin Province, 130021, People’s Republic of China
- Shandong Key Laboratory of TCM Multi-Targets Intervention and Disease Control, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, 271000, People’s Republic of China
| | - Mingquan Li
- Department of Neurology, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin Province, 130021, People’s Republic of China
| |
Collapse
|
21
|
Furman S, Green K, Lane TE. COVID-19 and the impact on Alzheimer's disease pathology. J Neurochem 2024; 168:3415-3429. [PMID: 37850241 PMCID: PMC11024062 DOI: 10.1111/jnc.15985] [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: 08/18/2023] [Revised: 09/17/2023] [Accepted: 09/20/2023] [Indexed: 10/19/2023]
Abstract
Coronavirus disease 2019 (COVID-19) has rapidly escalated into a global pandemic that primarily affects older and immunocompromised individuals due to underlying clinical conditions and suppressed immune responses. Furthermore, COVID-19 patients exhibit a spectrum of neurological symptoms, indicating that COVID-19 can affect the brain in a variety of manners. Many studies, past and recent, suggest a connection between viral infections and an increased risk of neurodegeneration, raising concerns about the neurological effects of COVID-19 and the possibility that it may contribute to Alzheimer's disease (AD) onset or worsen already existing AD pathology through inflammatory processes given that both COVID-19 and AD share pathological features and risk factors. This leads us to question whether COVID-19 is a risk factor for AD and how these two conditions might influence each other. Considering the extensive reach of the COVID-19 pandemic and the devastating impact of the ongoing AD pandemic, their combined effects could have significant public health consequences worldwide.
Collapse
Affiliation(s)
- Susana Furman
- Department of Neurobiology & Behavior, School of Biological Sciences, University of California, Irvine 92697
| | - Kim Green
- Department of Neurobiology & Behavior, School of Biological Sciences, University of California, Irvine 92697
| | - Thomas E. Lane
- Department of Neurobiology & Behavior, School of Biological Sciences, University of California, Irvine 92697
- Department of Molecular Biology & Biochemistry, School of Biological Sciences, University of California, Irvine 92697, USA
- Center for Virus Research, University of California, Irvine 92697, USA
| |
Collapse
|
22
|
Wan M, Liu Y, Li D, Snyder R, Elkin L, Day C, Rodriguez J, Grunseich C, Mahley R, Watts J, Cheung V. The enhancer RNA, AANCR, regulates APOE expression in astrocytes and microglia. Nucleic Acids Res 2024; 52:10235-10254. [PMID: 39162226 PMCID: PMC11417409 DOI: 10.1093/nar/gkae696] [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: 05/01/2024] [Revised: 07/26/2024] [Accepted: 08/01/2024] [Indexed: 08/21/2024] Open
Abstract
Enhancers, critical regulatory elements within the human genome, are often transcribed into enhancer RNAs. The dysregulation of enhancers leads to diseases collectively termed enhanceropathies. While it is known that enhancers play a role in diseases by regulating gene expression, the specific mechanisms by which individual enhancers cause diseases are not well understood. Studies of individual enhancers are needed to fill this gap. This study delves into the role of APOE-activating noncoding RNA, AANCR, in the central nervous system, elucidating its function as a genetic modifier in Alzheimer's Disease. We employed RNA interference, RNaseH-mediated degradation, and single-molecule RNA fluorescence in situ hybridization to demonstrate that mere transcription of AANCR is insufficient; rather, its transcripts are crucial for promoting APOE expression. Our findings revealed that AANCR is induced by ATM-mediated ERK phosphorylation and subsequent AP-1 transcription factor activation. Once activated, AANCR enhances APOE expression, which in turn imparts an inflammatory phenotype to astrocytes. These findings demonstrate that AANCR is a key enhancer RNA in some cell types within the nervous system, pivotal for regulating APOE expression and influencing inflammatory responses, underscoring its potential as a therapeutic target in neurodegenerative diseases.
Collapse
Affiliation(s)
- Ma Wan
- Epigenetics and Stem Cell Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Yaojuan Liu
- Department of Pediatrics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Dongjun Li
- Department of Pediatrics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ryan J Snyder
- Epigenetics and Stem Cell Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Lillian B Elkin
- Epigenetics and Stem Cell Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Christopher R Day
- Epigenetics and Stem Cell Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Joseph Rodriguez
- Epigenetics and Stem Cell Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Christopher Grunseich
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Robert W Mahley
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
- Department of Pathology and Medicine, University of California, San Francisco, CA, USA
| | - Jason A Watts
- Epigenetics and Stem Cell Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Vivian G Cheung
- Department of Pediatrics, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
23
|
Di Domenico F, Lanzillotta C, Perluigi M. Redox imbalance and metabolic defects in the context of Alzheimer disease. FEBS Lett 2024; 598:2047-2066. [PMID: 38472147 DOI: 10.1002/1873-3468.14840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 02/12/2024] [Accepted: 02/12/2024] [Indexed: 03/14/2024]
Abstract
Redox reactions play a critical role for intracellular processes, including pathways involved in metabolism and signaling. Reactive oxygen species (ROS) act either as second messengers or generators of protein modifications, fundamental mechanisms for signal transduction. Disturbance of redox homeostasis is associated with many disorders. Among these, Alzheimer's disease is a neurodegenerative pathology that presents hallmarks of oxidative damage such as increased ROS production, decreased activity of antioxidant enzymes, oxidative modifications of macromolecules, and changes in mitochondrial homeostasis. Interestingly, alteration of redox homeostasis is closely associated with defects of energy metabolism, involving both carbohydrates and lipids, the major energy fuels for the cell. As the brain relies exclusively on glucose metabolism, defects of glucose utilization represent a harmful event for the brain. During aging, a progressive perturbation of energy metabolism occurs resulting in brain hypometabolism. This condition contributes to increase neuronal cell vulnerability ultimately resulting in cognitive impairment. The current review discusses the crosstalk between alteration of redox homeostasis and brain energy defects that seems to act in concert in promoting Alzheimer's neurodegeneration.
Collapse
Affiliation(s)
- Fabio Di Domenico
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Italy
| | - Chiara Lanzillotta
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Italy
| | - Marzia Perluigi
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Italy
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| |
Collapse
|
24
|
Jin W, Pei J, Roy JR, Jayaraman S, Ahalliya RM, Kanniappan GV, Mironescu M, Palanisamy CP. Comprehensive review on single-cell RNA sequencing: A new frontier in Alzheimer's disease research. Ageing Res Rev 2024; 100:102454. [PMID: 39142391 DOI: 10.1016/j.arr.2024.102454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/07/2024] [Accepted: 08/09/2024] [Indexed: 08/16/2024]
Abstract
Alzheimer's disease (AD) is a multifaceted neurodegenerative condition marked by gradual cognitive deterioration and the loss of neurons. While conventional bulk RNA sequencing techniques have shed light on AD pathology, they frequently obscure the cellular diversity within brain tissues. The advent of single-cell RNA sequencing (scRNA-seq) has transformed our capability to analyze the cellular composition of AD, allowing for the detection of unique cell populations, rare cell types, and gene expression alterations at an individual cell level. This review examines the use of scRNA-seq in AD research, focusing on its contributions to understanding cellular diversity, disease progression, and potential therapeutic targets. We discuss key technological innovations, data analysis techniques, and challenges associated with scRNA-seq in studying AD. Furthermore, we highlight recent studies that have utilized scRNA-seq to identify novel biomarkers, uncover disease-associated pathways, and elucidate the role of non-neuronal cells, such as microglia and astrocytes, in AD pathogenesis. By providing a comprehensive overview of advancements in scRNA-seq for unraveling cellular heterogeneity in AD, this review highlights the transformative impact of scRNA-seq on our comprehension of disease mechanisms and the creation of targeted treatments.
Collapse
Affiliation(s)
- Wengang Jin
- Qinba State Key Laboratory of Biological Resources and Ecological Environment, 2011 QinLing-Bashan Mountains Bioresources Comprehensive Development C. I. C, Shaanxi Province Key Laboratory of Bio-Resources, College of Bioscience and Bioengineering, Shaanxi University of Technology, Hanzhong 723001, China
| | - JinJin Pei
- Qinba State Key Laboratory of Biological Resources and Ecological Environment, 2011 QinLing-Bashan Mountains Bioresources Comprehensive Development C. I. C, Shaanxi Province Key Laboratory of Bio-Resources, College of Bioscience and Bioengineering, Shaanxi University of Technology, Hanzhong 723001, China
| | - Jeane Rebecca Roy
- Department of Anatomy, Bhaarath Medical College and hospital, Bharath Institute of Higher Education and Research (BIHER), Chennai, Tamil Nadu 600073, India
| | - Selvaraj Jayaraman
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospital, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai 600077, India
| | - Rathi Muthaiyan Ahalliya
- Department of Biochemistry and Cancer Research Centre, FASCM, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu 641021, India
| | - Gopalakrishnan Velliyur Kanniappan
- Center for Global Health Research, Saveetha Medical College & Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Thandalam, Chennai, Tamil Nadu 602105, India.
| | - Monica Mironescu
- Faculty of Agricultural Sciences Food Industry and Environmental Protection, Lucian Blaga University of Sibiu, Bv. Victoriei 10, Sibiu 550024, Romania.
| | - Chella Perumal Palanisamy
- Department of Chemical Technology, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand.
| |
Collapse
|
25
|
Kim Y, Lim J, Oh J. Taming neuroinflammation in Alzheimer's disease: The protective role of phytochemicals through the gut-brain axis. Biomed Pharmacother 2024; 178:117277. [PMID: 39126772 DOI: 10.1016/j.biopha.2024.117277] [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: 05/24/2024] [Revised: 08/05/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive degenerative neurological condition characterized by cognitive decline, primarily affecting memory and logical thinking, attributed to amyloid-β plaques and tau protein tangles in the brain, leading to neuronal loss and brain atrophy. Neuroinflammation, a hallmark of AD, involves the activation of microglia and astrocytes in response to pathological changes, potentially exacerbating neuronal damage. The gut-brain axis is a bidirectional communication pathway between the gastrointestinal and central nervous systems, crucial for maintaining brain health. Phytochemicals, natural compounds found in plants with antioxidant and anti-inflammatory properties, such as flavonoids, curcumin, resveratrol, and quercetin, have emerged as potential modulators of this axis, suggesting implications for AD prevention. Intake of phytochemicals influences the gut microbial composition and its metabolites, thereby impacting neuroinflammation and oxidative stress in the brain. Consumption of phytochemical-rich foods may promote a healthy gut microbiota, fostering the production of anti-inflammatory and neuroprotective substances. Early dietary incorporation of phytochemicals offers a non-invasive strategy for modulating the gut-brain axis and potentially reducing AD risk or delaying its onset. The exploration of interventions targeting the gut-brain axis through phytochemical intake represents a promising avenue for the development of preventive or therapeutic strategies against AD initiation and progression.
Collapse
Affiliation(s)
- Yoonsu Kim
- Department of Integrative Biology, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Jinkyu Lim
- School of Food Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea.
| | - Jisun Oh
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea.
| |
Collapse
|
26
|
Nunes YC, Mendes NM, Pereira de Lima E, Chehadi AC, Lamas CB, Haber JFS, dos Santos Bueno M, Araújo AC, Catharin VCS, Detregiachi CRP, Laurindo LF, Tanaka M, Barbalho SM, Marin MJS. Curcumin: A Golden Approach to Healthy Aging: A Systematic Review of the Evidence. Nutrients 2024; 16:2721. [PMID: 39203857 PMCID: PMC11357524 DOI: 10.3390/nu16162721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 09/03/2024] Open
Abstract
Aging-related disorders pose significant challenges due to their complex interplay of physiological and metabolic factors, including inflammation, oxidative stress, and mitochondrial dysfunction. Curcumin, a natural compound with potent antioxidant and anti-inflammatory properties, has emerged as a promising candidate for mitigating these age-related processes. However, gaps in understanding the precise mechanisms of curcumin's effects and the optimal dosages for different conditions necessitate further investigation. This systematic review synthesizes current evidence on curcumin's potential in addressing age-related disorders, emphasizing its impact on cognitive function, neurodegeneration, and muscle health in older adults. By evaluating the safety, efficacy, and mechanisms of action of curcumin supplementation, this review aims to provide insights into its therapeutic potential for promoting healthy aging. A systematic search across three databases using specific keywords yielded 2256 documents, leading to the selection of 15 clinical trials for synthesis. Here, we highlight the promising potential of curcumin as a multifaceted therapeutic agent in combating age-related disorders. The findings of this review suggest that curcumin could offer a natural and effective approach to enhancing the quality of life of aging individuals. Further research and well-designed clinical trials are essential to validate these findings and optimize the use of curcumin in personalized medicine approaches for age-related conditions.
Collapse
Affiliation(s)
- Yandra Cervelim Nunes
- Faculdade de Medicina de Marília (FAMEMA), Marília 17519-030, SP, Brazil; (Y.C.N.); (L.F.L.)
| | - Nathalia M. Mendes
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil; (N.M.M.); (E.P.d.L.); (A.C.C.); (J.F.S.H.); (M.d.S.B.); (A.C.A.)
| | - Enzo Pereira de Lima
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil; (N.M.M.); (E.P.d.L.); (A.C.C.); (J.F.S.H.); (M.d.S.B.); (A.C.A.)
| | - Amanda Chabrour Chehadi
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil; (N.M.M.); (E.P.d.L.); (A.C.C.); (J.F.S.H.); (M.d.S.B.); (A.C.A.)
| | - Caroline Barbalho Lamas
- Department of Gerontology, School of Gerontology, Universidade Federal de São Carlos (UFSCar), São Carlos 13565-905, SP, Brazil;
| | - Jesselina F. S. Haber
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil; (N.M.M.); (E.P.d.L.); (A.C.C.); (J.F.S.H.); (M.d.S.B.); (A.C.A.)
| | - Manoela dos Santos Bueno
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil; (N.M.M.); (E.P.d.L.); (A.C.C.); (J.F.S.H.); (M.d.S.B.); (A.C.A.)
| | - Adriano Cressoni Araújo
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil; (N.M.M.); (E.P.d.L.); (A.C.C.); (J.F.S.H.); (M.d.S.B.); (A.C.A.)
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil; (V.C.S.C.); (C.R.P.D.)
| | - Vitor C. Strozze Catharin
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil; (V.C.S.C.); (C.R.P.D.)
| | - Claudia Rucco P. Detregiachi
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil; (V.C.S.C.); (C.R.P.D.)
| | - Lucas Fornari Laurindo
- Faculdade de Medicina de Marília (FAMEMA), Marília 17519-030, SP, Brazil; (Y.C.N.); (L.F.L.)
| | - Masaru Tanaka
- Danube Neuroscience Research Laboratory, HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Tisza Lajos krt. 113, H-6725 Szeged, Hungary
| | - Sandra Maria Barbalho
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil; (N.M.M.); (E.P.d.L.); (A.C.C.); (J.F.S.H.); (M.d.S.B.); (A.C.A.)
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil; (V.C.S.C.); (C.R.P.D.)
- Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), Marília 17500-000, SP, Brazil
- Research Coordination, Hospital Beneficente (HBU), University of Marília (UNIMAR), Marília 17525-160, SP, Brazil
| | | |
Collapse
|
27
|
Liu CF, Young ZY, Shih TW, Pan TM, Lee CL. Lactocaseibacillus-deglycosylated isoflavones prevent Aβ 40-induced Alzheimer's disease in a rat model. AMB Express 2024; 14:90. [PMID: 39105988 PMCID: PMC11303605 DOI: 10.1186/s13568-024-01735-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 06/25/2024] [Indexed: 08/07/2024] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease, with symptoms appearing in the cerebral cortex and hippocampus. amyloid β peptide (Aβ) has been shown to deposit in the brain, causing oxidative stress and inflammation, leading to impaired memory and learning. Lactocaseibacillus fermentation can produce deglycosylated isoflavones with high physiological activity, which can scavenge free radicals, enhance total antioxidant capacity and inhibit oxidative inflammatory responses. Therefore, in this study, Lactocaseibacillus paracasei subsp. paracasei NTU101 (NTU101) fermented soybean milk and its extracts were used as test substances, and AD model rats were established by infusion of Aβ40 in the brain for 28 days, and the preventive and ameliorating effects of NTU 101 fermented soymilk were discussed. Effects of soymilk and unfermented soymilk on AD, and explore its effects on AD. Main functional ingredients. The results showed that deglycosylated isoflavones in NTU101 fermented soybean milk improved AD symptoms. Mechanisms of actions include the inhibition of oxidative inflammation; reduction in the expression of risk factors for tau protein and apo E protein production, the deposition of Aβ40 around the hippocampus, and the expression of TLR-2 and RAGE proteins in astrocytes and microglia; and improvement in the memory and learning ability.
Collapse
Affiliation(s)
- Chin-Feng Liu
- Continuing Education Program of Food Biotechnology Applications, National Taitung University, Taitung, Taiwan, ROC
| | - Zong-Yang Young
- Department of Life Science, National Taitung University, 369, Sec. 2, University Rd., Taitung, 95092, Taiwan, ROC
| | | | - Tzu-Ming Pan
- SunWay Biotech Co. LTD., Taipei, Taiwan, ROC.
- Department of Biochemical Science and Technology, National Taiwan University, Taipei, Taiwan, ROC.
| | - Chun-Lin Lee
- Department of Life Science, National Taitung University, 369, Sec. 2, University Rd., Taitung, 95092, Taiwan, ROC.
| |
Collapse
|
28
|
Wirth S, Schlößer A, Beiersdorfer A, Schweizer M, Woo MS, Friese MA, Lohr C, Grochowska KM. Astrocytic uptake of posttranslationally modified amyloid-β leads to endolysosomal system disruption and induction of pro-inflammatory signaling. Glia 2024; 72:1451-1468. [PMID: 38629411 DOI: 10.1002/glia.24539] [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: 09/07/2023] [Revised: 04/05/2024] [Accepted: 04/09/2024] [Indexed: 06/29/2024]
Abstract
The disruption of astrocytic catabolic processes contributes to the impairment of amyloid-β (Aβ) clearance, neuroinflammatory signaling, and the loss of synaptic contacts in late-onset Alzheimer's disease (AD). While it is known that the posttranslational modifications of Aβ have significant implications on biophysical properties of the peptides, their consequences for clearance impairment are not well understood. It was previously shown that N-terminally pyroglutamylated Aβ3(pE)-42, a significant constituent of amyloid plaques, is efficiently taken up by astrocytes, leading to the release of pro-inflammatory cytokine tumor necrosis factor α and synapse loss. Here we report that Aβ3(pE)-42, but not Aβ1-42, gradually accumulates within the astrocytic endolysosomal system, disrupting this catabolic pathway and inducing the formation of heteromorphous vacuoles. This accumulation alters lysosomal kinetics, lysosome-dependent calcium signaling, and upregulates the lysosomal stress response. These changes correlate with the upregulation of glial fibrillary acidic protein (GFAP) and increased activity of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). Treatment with a lysosomal protease inhibitor, E-64, rescues GFAP upregulation, NF-κB activation, and synapse loss, indicating that abnormal lysosomal protease activity is upstream of pro-inflammatory signaling and related synapse loss. Collectively, our data suggest that Aβ3(pE)-42-induced disruption of the astrocytic endolysosomal system leads to cytoplasmic leakage of lysosomal proteases, promoting pro-inflammatory signaling and synapse loss, hallmarks of AD-pathology.
Collapse
Affiliation(s)
- Sarah Wirth
- Leibniz Group 'Dendritic Organelles and Synaptic Function', Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Annika Schlößer
- Leibniz Group 'Dendritic Organelles and Synaptic Function', Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Antonia Beiersdorfer
- Institute of Cell and Systems Biology of Animals, Department of Biology, University of Hamburg, Hamburg, Germany
| | - Michaela Schweizer
- Core Facility of Electron Microscopy, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marcel S Woo
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Manuel A Friese
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Lohr
- Institute of Cell and Systems Biology of Animals, Department of Biology, University of Hamburg, Hamburg, Germany
| | - Katarzyna M Grochowska
- Leibniz Group 'Dendritic Organelles and Synaptic Function', Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany
| |
Collapse
|
29
|
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
|
30
|
Jambi EJ, Alamri A, Afzal M, Al-Abbasi FA, Al-Qahtani SD, Almalki NAR, Bawadood AS, Alzarea SI, Sayyed N, Kazmi I. 6-shogaol against 3-Nitropropionic acid-induced Huntington's disease in rodents: Based on molecular docking/targeting pro-inflammatory cytokines/NF-κB-BDNF-Nrf2 pathway. PLoS One 2024; 19:e0305358. [PMID: 39008492 PMCID: PMC11249262 DOI: 10.1371/journal.pone.0305358] [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: 02/17/2024] [Accepted: 05/27/2024] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND Huntington's disease (HD) is an extremely harmful autosomal inherited neurodegenerative disease. Motor dysfunction, mental disorder, and cognitive deficits are the characteristic features of this disease. The current study examined whether 6-shogaol has a protective effect against 3-Nitropropionic Acid (3-NPA)-induced HD in rats. METHODS A total of thirty male Wistar rats received 6-shogaol (10 and 20 mg/kg, per oral) an hour before injection of 3-NPA (10 mg/kg i.p.) for 15 days. Behavioral tests were performed, including narrow beam walk, rotarod test, and grip strength test. Biochemical tests promoting oxidative stress were evaluated [superoxide dismutase (SOD), reduced glutathione (GSH), catalase (CAT) and malondialdehyde (MDA)], including changes to neurotransmitters serotonin (5-HT), dopamine (DA), norepinephrine (NE), homovanillic acid (HVA), (3,4-dihydroxyphenylacetic acid (DOPAC), γ-aminobutyric acid (GABA), and 5-hydroxy indole acetic acid (5-HIAA), nuclear factor kappa-B (NF-κB), tumor necrosis factor-α (TNF-α), interleukins-1β (IL-1β), IL-6, brain-derived neurotrophic factor (BDNF), and nuclear factor erythroid 2-related factor 2 (Nrf2). The 6-shogaol was docked to the active site of TNF-α (2AZ5), NF-κB (1SVC), BDNF) [1B8M], and Nrf2 [5FZN] proteins using AutoDock tools. RESULTS The 6-shogaol group significantly improved behavioral activity over the 3-NPA-injected control rats. Moreover, 3-NPA-induced significantly altered neurotransmitters, biochemical and neuroinflammatory indices, which could efficiently be reversed by 6-shogaol. The 6-shogaol showed favorable negative binding energies at -9.271 (BDNF) kcal/mol. CONCLUSIONS The present investigation demonstrated the neuroprotective effects of 6-shogaol in an experimental animal paradigm against 3-NPA-induced HD in rats. The suggested mechanism is supported by immunohistochemical analysis and western blots, although more research is necessary for definite confirmation.
Collapse
Affiliation(s)
- Ebtihaj J. Jambi
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Experimental Biochemistry Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdulaziz Alamri
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, Jeddah, Saudi Arabia
| | - Fahad A. Al-Abbasi
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Salwa D. Al-Qahtani
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah, Saudi Arabia
| | - Naif A. R. Almalki
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Experimental Biochemistry Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Azizah Salim Bawadood
- Basic Medical Sciences Department, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Sami I. Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Aljouf, Sakaka, Saudi Arabia
| | - Nadeem Sayyed
- School of Pharmacy, Glocal University, Saharanpur, India
| | - Imran Kazmi
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
31
|
Kumar M, Swanson N, Ray S, Buch S, Saraswathi V, Sil S. Astrocytes in Amyloid Generation and Alcohol Metabolism: Implications of Alcohol Use in Neurological Disorder(s). Cells 2024; 13:1173. [PMID: 39056755 PMCID: PMC11274690 DOI: 10.3390/cells13141173] [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: 05/20/2024] [Revised: 06/29/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
As per the National Survey on Drug Use and Health, 10.5% of Americans aged 12 years and older are suffering from alcohol use disorder, with a wide range of neurological disorders. Alcohol-mediated neurological disorders can be linked to Alzheimer's-like pathology, which has not been well studied. We hypothesize that alcohol exposure can induce astrocytic amyloidosis, which can be corroborated by the neurological disorders observed in alcohol use disorder. In this study, we demonstrated that the exposure of astrocytes to ethanol resulted in an increase in Alzheimer's disease markers-the amyloid precursor protein, Aβ1-42, and the β-site-cleaving enzyme; an oxidative stress marker-4HNE; proinflammatory cytokines-TNF-α, IL1β, and IL6; lncRNA BACE1-AS; and alcohol-metabolizing enzymes-alcohol dehydrogenase, aldehyde dehydrogenase-2, and cytochrome P450 2E1. A gene-silencing approach confirmed the regulatory role of lncRNA BACE1-AS in amyloid generation, alcohol metabolism, and neuroinflammation. This report is the first to suggest the involvement of lncRNA BACE1-AS in alcohol-induced astrocytic amyloid generation and alcohol metabolism. These findings will aid in developing therapies targeting astrocyte-mediated neurological disorders and cognitive deficits in alcohol users.
Collapse
Affiliation(s)
- Mohit Kumar
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Natalie Swanson
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Sudipta Ray
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Shilpa Buch
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Viswanathan Saraswathi
- VA Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
- Department of Internal Medicine, Division of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Susmita Sil
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
32
|
Jothi D, Kulka LAM. Strategies for modeling aging and age-related diseases. NPJ AGING 2024; 10:32. [PMID: 38987252 PMCID: PMC11237002 DOI: 10.1038/s41514-024-00161-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 06/18/2024] [Indexed: 07/12/2024]
Abstract
The ability to reprogram patient-derived-somatic cells to IPSCs (Induced Pluripotent Stem Cells) has led to a better understanding of aging and age-related diseases like Parkinson's, and Alzheimer's. The established patient-derived disease models mimic disease pathology and can be used to design drugs for aging and age-related diseases. However, the age and genetic mutations of the donor cells, the employed reprogramming, and the differentiation protocol might often pose challenges in establishing an appropriate disease model. In this review, we will focus on the various strategies for the successful reprogramming and differentiation of patient-derived cells to disease models for aging and age-related diseases, emphasizing the accuracy in the recapitulation of disease pathology and ways to overcome the limitations of its potential application in cell replacement therapy and drug development.
Collapse
Affiliation(s)
- D Jothi
- Department of Biochemistry II, Friedrich Schiller University, Jena, Germany.
| | | |
Collapse
|
33
|
Farris T, González-Ochoa S, Mohammed M, Rajakaruna H, Tonello J, Kanagasabai T, Korolkova O, Shimamoto A, Ivanova A, Shanker A. Loss of Mitochondrial Tusc2/Fus1 Triggers a Brain Pro-Inflammatory Microenvironment and Early Spatial Memory Impairment. Int J Mol Sci 2024; 25:7406. [PMID: 39000512 PMCID: PMC11242373 DOI: 10.3390/ijms25137406] [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: 05/02/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/16/2024] Open
Abstract
Brain pathological changes impair cognition early in disease etiology. There is an urgent need to understand aging-linked mechanisms of early memory loss to develop therapeutic strategies and prevent the development of cognitive impairment. Tusc2 is a mitochondrial-resident protein regulating Ca2+ fluxes to and from mitochondria impacting overall health. We previously reported that Tusc2-/- female mice develop chronic inflammation and age prematurely, causing age- and sex-dependent spatial memory deficits at 5 months old. Therefore, we investigated Tusc2-dependent mechanisms of memory impairment in 4-month-old mice, comparing changes in resident and brain-infiltrating immune cells. Interestingly, Tusc2-/- female mice demonstrated a pro-inflammatory increase in astrocytes, expression of IFN-γ in CD4+ T cells and Granzyme-B in CD8+T cells. We also found fewer FOXP3+ T-regulatory cells and Ly49G+ NK and Ly49G+ NKT cells in female Tusc2-/- brains, suggesting a dampened anti-inflammatory response. Moreover, Tusc2-/- hippocampi exhibited Tusc2- and sex-specific protein changes associated with brain plasticity, including mTOR activation, and Calbindin and CamKII dysregulation affecting intracellular Ca2+ dynamics. Overall, the data suggest that dysregulation of Ca2+-dependent processes and a heightened pro-inflammatory brain microenvironment in Tusc2-/- mice could underlie cognitive impairment. Thus, strategies to modulate the mitochondrial Tusc2- and Ca2+- signaling pathways in the brain should be explored to improve cognitive health.
Collapse
Affiliation(s)
- Tonie Farris
- Department of Biomedical Sciences, School of Graduate Studies, Meharry Medical College, Nashville, TN 37208, USA; (T.F.); (M.M.); (T.K.)
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA; (S.G.-O.); (J.T.); (O.K.); (A.S.)
| | - Salvador González-Ochoa
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA; (S.G.-O.); (J.T.); (O.K.); (A.S.)
| | - Muna Mohammed
- Department of Biomedical Sciences, School of Graduate Studies, Meharry Medical College, Nashville, TN 37208, USA; (T.F.); (M.M.); (T.K.)
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA; (S.G.-O.); (J.T.); (O.K.); (A.S.)
| | - Harshana Rajakaruna
- The Office for Research and Innovation, Meharry Medical College, Nashville, TN 37208, USA;
| | - Jane Tonello
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA; (S.G.-O.); (J.T.); (O.K.); (A.S.)
| | - Thanigaivelan Kanagasabai
- Department of Biomedical Sciences, School of Graduate Studies, Meharry Medical College, Nashville, TN 37208, USA; (T.F.); (M.M.); (T.K.)
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA; (S.G.-O.); (J.T.); (O.K.); (A.S.)
| | - Olga Korolkova
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA; (S.G.-O.); (J.T.); (O.K.); (A.S.)
| | - Akiko Shimamoto
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA; (S.G.-O.); (J.T.); (O.K.); (A.S.)
| | - Alla Ivanova
- Department of Biomedical Sciences, School of Graduate Studies, Meharry Medical College, Nashville, TN 37208, USA; (T.F.); (M.M.); (T.K.)
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA; (S.G.-O.); (J.T.); (O.K.); (A.S.)
| | - Anil Shanker
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA; (S.G.-O.); (J.T.); (O.K.); (A.S.)
- The Office for Research and Innovation, Meharry Medical College, Nashville, TN 37208, USA;
| |
Collapse
|
34
|
Sanjay, Sood R, Jaiswal V, Kang SU, Park M, Lee HJ. Nobiletin regulates intracellular Ca 2+ levels via IP 3R and ameliorates neuroinflammation in Aβ42-induced astrocytes. Redox Biol 2024; 73:103197. [PMID: 38781730 PMCID: PMC11145555 DOI: 10.1016/j.redox.2024.103197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 05/12/2024] [Accepted: 05/15/2024] [Indexed: 05/25/2024] Open
Abstract
Astrocytes are the major glial cells in the human brain and provide crucial metabolic and trophic support to neurons. The amyloid-β peptide (Aβ) alter the morphological and functional properties of astrocytes and induce inflammation and calcium dysregulation, contributing to Alzheimer's disease (AD) pathology. Recent studies highlight the role of Toll-like receptor (TLR) 4/nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling in inflammation. Reactive oxygen species (ROS) generated due to Aβ, induce apoptosis in the brain cells worsening AD progression. Astrocytic cell surface receptors, such as purinergic receptors (P2Y1 and P2Y2), metabotropic glutamate receptor (mGLUR)5, α7 nicotinic acetylcholine receptor (α7nAChR), and N-methyl-d-aspartate receptors (NMDARs), have been suggested to interact with inositol trisphosphate receptor (IP3R) on the endoplasmic reticulum (ER) to induce Ca2+ movement from ER to cytoplasm, causing Ca2+ dysregulation. We found that the citrus flavonoid nobiletin (NOB) protected primary astrocytes from Aβ42-induced cytotoxicity and inhibited TLR4/NF-κB signaling in Aβ42-induced primary rat astrocytes. NOB was found to regulate Aβ42-induced ROS levels through Keap1-Nrf2 pathway. The receptors P2Y1, P2Y2, mGLUR5, α7nAChR, and NMDARs induced intracellular Ca2+ levels by activating IP3R and NOB regulated them, thereby regulating intracellular Ca2+ levels. Molecular docking analysis revealed a possible interaction between NOB and IP3R in IP3R regulation. Furthermore, RNA sequencing revealed various NOB-mediated biological signaling pathways, such as the AD-presenilin, AD-amyloid secretase, and Wnt signaling pathway, suggesting possible neuroprotective roles of NOB. To conclude, NOB is a promising therapeutic agent for AD and works by modulating AD pathology at various levels in Aβ42-induced primary rat astrocytes.
Collapse
Affiliation(s)
- Sanjay
- Institute for Aging and Clinical Nutrition Research, Gachon University, Seongnam, Gyeonggi-do, 13120, Republic of Korea.
| | - Rachit Sood
- Institute for Aging and Clinical Nutrition Research, Gachon University, Seongnam, Gyeonggi-do, 13120, Republic of Korea; Department of Food and Nutrition, College of BioNano Technology, Gachon University, Seongnam, Gyeonggi-do, 13120, Republic of Korea.
| | - Varun Jaiswal
- Institute for Aging and Clinical Nutrition Research, Gachon University, Seongnam, Gyeonggi-do, 13120, Republic of Korea; Department of Food and Nutrition, College of BioNano Technology, Gachon University, Seongnam, Gyeonggi-do, 13120, Republic of Korea.
| | - Sung-Ung Kang
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| | - Miey Park
- Institute for Aging and Clinical Nutrition Research, Gachon University, Seongnam, Gyeonggi-do, 13120, Republic of Korea; Department of Food and Nutrition, College of BioNano Technology, Gachon University, Seongnam, Gyeonggi-do, 13120, Republic of Korea.
| | - Hae-Jeung Lee
- Institute for Aging and Clinical Nutrition Research, Gachon University, Seongnam, Gyeonggi-do, 13120, Republic of Korea; Department of Food and Nutrition, College of BioNano Technology, Gachon University, Seongnam, Gyeonggi-do, 13120, Republic of Korea; Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, Republic of Korea.
| |
Collapse
|
35
|
Matthews DC, Kinney JW, Ritter A, Andrews RD, Toledano Strom EN, Lukic AS, Koenig LN, Revta C, Fillit HM, Zhong K, Tousi B, Leverenz JB, Feldman HH, Cummings J. Relationships between plasma biomarkers, tau PET, FDG PET, and volumetric MRI in mild to moderate Alzheimer's disease patients. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2024; 10:e12490. [PMID: 38988416 PMCID: PMC11233274 DOI: 10.1002/trc2.12490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 05/24/2024] [Accepted: 05/25/2024] [Indexed: 07/12/2024]
Abstract
INTRODUCTION The "A/T/N" (amyloid/tau/neurodegeneration) framework provides a biological basis for Alzheimer's disease (AD) diagnosis and can encompass additional changes such as inflammation ("I"). A spectrum of T/N/I imaging and plasma biomarkers was acquired in a phase 2 clinical trial of rasagiline in mild to moderate AD patients. We evaluated these to understand biomarker distributions and relationships within this population. METHODS Plasma biomarkers of pTau-181, neurofilament light chain (NfL), glial fibrillary acidic protein (GFAP), other inflammation-related proteins, imaging measures including fluorodeoxyglucose (FDG) positron emission tomography (PET), flortaucipir PET, and volumetric magnetic resonance imaging (MRI), and cognitive endpoints were analyzed to assess characteristics and relationships for the overall population (N = 47 at baseline and N = 21 for longitudinal cognitive comparisons) and within age-decade subgroups (57-69, 70-79, 80-90 years). RESULTS Data demonstrate wide clinical and biomarker heterogeneity in this population influenced by age and sex. Plasma pTau-181 and GFAP correlate with tau PET, most strongly in left inferior temporal cortex (p = 0.0002, p = 0.0006, respectively). In regions beyond temporal cortex, tau PET uptake decreased with age for the same pTau-181 or GFAP concentrations. FDG PET and brain volumes correlate with tau PET in numerous regions (such as inferior temporal: p = 0.0007, p = 0.00001, respectively). NfL, GFAP, and all imaging modalities correlate with baseline MMSE; subsequent MMSE decline is predicted by baseline parahippocampal and lateral temporal tau PET (p = 0.0007) and volume (p = 0.0006). Lateral temporal FDG PET (p = 0.006) and volume (p = 0.0001) are most strongly associated with subsequent ADAS-cog decline. NfL correlates with FDG PET and baseline MMSE but not tau PET. Inflammation biomarkers are intercorrelated but correlated with other biomarkers in only the youngest group. DISCUSSION Associations between plasma biomarkers, imaging biomarkers, and cognitive status observed in this study provide insight into relationships among biological processes in mild to moderate AD. Findings show the potential to characterize AD patients regarding likely tau pathology, neurodegeneration, prospective clinical decline, and the importance of covariates such as age. Highlights Plasma pTau-181 and GFAP correlated with regional and global tau PET in mild to moderate AD.NfL correlated with FDG PET and cognitive endpoints but not plasma pTau-181 or tau PET.Volume and FDG PET showed strong relationships to tau PET, one another, and cognitive status.Temporal volumes most strongly predicted decline in both MMSE and ADAS-cog.Volume and plasma biomarkers can enrich for elevated tau PET with age a significant covariate.
Collapse
Affiliation(s)
| | | | - Aaron Ritter
- Hoag Pickup Family Neurosciences InstituteNewport BeachCaliforniaUSA
| | | | - Erin N. Toledano Strom
- Chambers‐Grundy Center for Transformative NeuroscienceDepartment of Brain HealthSchool of Integrated Health Sciences, University of Nevada Las VegasLas VegasNevadaUSA
| | | | | | - Carolyn Revta
- Alzheimer's Disease Cooperative StudyUniversity of California, San Diego, School of MedicineLa JollaCaliforniaUSA
| | | | | | - Babak Tousi
- Cleveland Clinical Lous Ruvo Center for Brain HealthClevelandOhioUSA
| | | | - Howard H. Feldman
- Alzheimer's Disease Cooperative StudyUniversity of California, San Diego, School of MedicineLa JollaCaliforniaUSA
- Department of NeurosciencesUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Jeffrey Cummings
- Chambers‐Grundy Center for Transformative NeuroscienceDepartment of Brain HealthSchool of Integrated Health Sciences, University of Nevada Las VegasLas VegasNevadaUSA
| |
Collapse
|
36
|
Zhang J, Xie D, Jiao D, Zhou S, Liu S, Ju Z, Hu L, Qi L, Yao C, Zhao C. From inflammatory signaling to neuronal damage: Exploring NLR inflammasomes in ageing neurological disorders. Heliyon 2024; 10:e32688. [PMID: 38975145 PMCID: PMC11226848 DOI: 10.1016/j.heliyon.2024.e32688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 06/06/2024] [Indexed: 07/09/2024] Open
Abstract
The persistence of neuronal degeneration and damage is a major obstacle in ageing medicine. Nucleotide-binding oligomerization domain (NOD)-like receptors detect environmental stressors and trigger the maturation and secretion of pro-inflammatory cytokines that can cause neuronal damage and accelerate cell death. NLR (NOD-like receptors) inflammasomes are protein complexes that contain NOD-like receptors. Studying the role of NLR inflammasomes in ageing-related neurological disorders can provide valuable insights into the mechanisms of neurodegeneration. This includes investigating their activation of inflammasomes, transcription, and capacity to promote or inhibit inflammatory signaling, as well as exploring strategies to regulate NLR inflammasomes levels. This review summarizes the use of NLR inflammasomes in guiding neuronal degeneration and injury during the ageing process, covering several neurological disorders such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, stroke, and peripheral neuropathies. To improve the quality of life and slow the progression of neurological damage, NLR-based treatment strategies, including inhibitor-related therapies and physical therapy, are presented. Additionally, important connections between age-related neurological disorders and NLR inflammasomes are highlighted to guide future research and facilitate the development of new treatment options.
Collapse
Affiliation(s)
- Jingwen Zhang
- School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Dong Xie
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Danli Jiao
- School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Shuang Zhou
- School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Shimin Liu
- School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Shanghai Research Institute of Acupuncture and Meridian, Shanghai, 200030, China
| | - Ziyong Ju
- School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Li Hu
- School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Li Qi
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Chongjie Yao
- School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Chen Zhao
- School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| |
Collapse
|
37
|
Zhong S, Zhou Q, Yang J, Zhang Z, Zhang X, Liu J, Chang X, Wang H. Relationship between the cGAS-STING and NF-κB pathways-role in neurotoxicity. Biomed Pharmacother 2024; 175:116698. [PMID: 38713946 DOI: 10.1016/j.biopha.2024.116698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/16/2024] [Accepted: 05/01/2024] [Indexed: 05/09/2024] Open
Abstract
Neurotoxicity can cause a range of symptoms and disorders in humans, including neurodegenerative diseases, neurodevelopmental disorders, nerve conduction abnormalities, neuroinflammation, autoimmune disorders, and cognitive deficits. The cyclic guanosine-adenosine synthase (cGAS)-stimulator of interferon genes (STING) pathway and NF-κB pathway are two important signaling pathways involved in the innate immune response. The cGAS-STING pathway is activated by the recognition of intracellular DNA, which triggers the production of type I interferons and pro-inflammatory cytokines, such as tumor necrosis factor, IL-1β, and IL-6. These cytokines play a role in oxidative stress and mitochondrial dysfunction in neurons. The NF-κB pathway is activated by various stimuli, such as bacterial lipopolysaccharide, viral particle components, and neurotoxins. NF-κB activation may lead to the production of pro-inflammatory cytokines, which promote neuroinflammation and cause neuronal damage. A potential interaction exists between the cGAS-STING and NF-κB pathways, and NF-κB activation blocks STING degradation by inhibiting microtubule-mediated STING transport. This review examines the progress of research on the roles of these pathways in neurotoxicity and their interrelationships. Understanding the mechanisms of these pathways will provide valuable therapeutic insights for preventing and controlling neurotoxicity.
Collapse
Affiliation(s)
- Shiyin Zhong
- Department of Toxicology, School of Public Health, Lanzhou University, Gansu 730000, China
| | - Qiongli Zhou
- Department of Toxicology, School of Public Health, Lanzhou University, Gansu 730000, China
| | - Jirui Yang
- Department of Toxicology, School of Public Health, Lanzhou University, Gansu 730000, China
| | - Zhimin Zhang
- Department of Toxicology, School of Public Health, Lanzhou University, Gansu 730000, China
| | - Xin Zhang
- Department of Toxicology, School of Public Health, Lanzhou University, Gansu 730000, China
| | - Jingjing Liu
- Department of Toxicology, School of Public Health, Lanzhou University, Gansu 730000, China
| | - Xuhong Chang
- Department of Toxicology, School of Public Health, Lanzhou University, Gansu 730000, China
| | - Hui Wang
- Department of Toxicology, School of Public Health, Lanzhou University, Gansu 730000, China.
| |
Collapse
|
38
|
Barsoum S, Latimer CS, Nolan AL, Barrett A, Chang K, Troncoso J, Keene CD, Benjamini D. Resiliency to Alzheimer's disease neuropathology can be distinguished from dementia using cortical astrogliosis imaging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.06.592719. [PMID: 38766087 PMCID: PMC11100587 DOI: 10.1101/2024.05.06.592719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Despite the presence of significant Alzheimer's disease (AD) pathology, characterized by amyloid β (Aβ) plaques and phosphorylated tau (pTau) tangles, some cognitively normal elderly individuals do not inevitably develop dementia. These findings give rise to the notion of cognitive 'resilience', suggesting maintained cognitive function despite the presence of AD neuropathology, highlighting the influence of factors beyond classical pathology. Cortical astroglial inflammation, a ubiquitous feature of symptomatic AD, shows a strong correlation with cognitive impairment severity, potentially contributing to the diversity of clinical presentations. However, noninvasively imaging neuroinflammation, particularly astrogliosis, using MRI remains a significant challenge. Here we sought to address this challenge and to leverage multidimensional (MD) MRI, a powerful approach that combines relaxation with diffusion MR contrasts, to map cortical astrogliosis in the human brain by accessing sub-voxel information. Our goal was to test whether MD-MRI can map astroglial pathology in the cerebral cortex, and if so, whether it can distinguish cognitive resiliency from dementia in the presence of hallmark AD neuropathological changes. We adopted a multimodal approach by integrating histological and MRI analyses using human postmortem brain samples. Ex vivo cerebral cortical tissue specimens derived from three groups comprised of non-demented individuals with significant AD pathology postmortem, individuals with both AD pathology and dementia, and non-demented individuals with minimal AD pathology postmortem as controls, underwent MRI at 7 T. We acquired and processed MD-MRI, diffusion tensor, and quantitative T 1 and T 2 MRI data, followed by histopathological processing on slices from the same tissue. By carefully co-registering MRI and microscopy data, we performed quantitative multimodal analyses, leveraging targeted immunostaining to assess MD-MRI sensitivity and specificity towards Aβ, pTau, and glial fibrillary acidic protein (GFAP), a marker for astrogliosis. Our findings reveal a distinct MD-MRI signature of cortical astrogliosis, enabling the creation of predictive maps for cognitive resilience amid AD neuropathological changes. Multiple linear regression linked histological values to MRI changes, revealing that the MD-MRI cortical astrogliosis biomarker was significantly associated with GFAP burden (standardized β=0.658, pFDR<0.0001), but not with Aβ (standardized β=0.009, p FDR =0.913) or pTau (standardized β=-0.196, p FDR =0.051). Conversely, none of the conventional MRI parameters showed significant associations with GFAP burden in the cortex. While the extent to which pathological glial activation contributes to neuronal damage and cognitive impairment in AD is uncertain, developing a noninvasive imaging method to see its affects holds promise from a mechanistic perspective and as a potential predictor of cognitive outcomes.
Collapse
|
39
|
Qin P, Sun Y, Li L. Mitochondrial dysfunction in chronic neuroinflammatory diseases (Review). Int J Mol Med 2024; 53:47. [PMID: 38577947 PMCID: PMC10999227 DOI: 10.3892/ijmm.2024.5371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 03/14/2024] [Indexed: 04/06/2024] Open
Abstract
Chronic neuroinflammation serves a key role in the onset and progression of neurodegenerative disorders. Mitochondria serve as central regulators of neuroinflammation. In addition to providing energy to cells, mitochondria also participate in the immunoinflammatory response of neurodegenerative disorders including Alzheimer's disease, Parkinson's disease, multiple sclerosis and epilepsy, by regulating processes such as cell death and inflammasome activation. Under inflammatory conditions, mitochondrial oxidative stress, epigenetics, mitochondrial dynamics and calcium homeostasis imbalance may serve as underlying regulatory mechanisms for these diseases. Therefore, investigating mechanisms related to mitochondrial dysfunction may result in therapeutic strategies against chronic neuroinflammation and neurodegeneration. The present review summarizes the mechanisms of mitochondria in chronic neuroinflammatory diseases and the current treatment approaches that target mitochondrial dysfunction in these diseases.
Collapse
Affiliation(s)
- Pei Qin
- Department of Anesthesiology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, P.R. China
| | - Ye Sun
- Department of Anesthesiology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, P.R. China
| | - Liya Li
- Department of Anesthesiology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, P.R. China
| |
Collapse
|
40
|
Assis-de-Lemos G, Moura-do-Nascimento R, Amaral-do-Nascimento M, Miceli AC, Vieira TCRG. Interactions between Cytokines and the Pathogenesis of Prion Diseases: Insights and Implications. Brain Sci 2024; 14:413. [PMID: 38790392 PMCID: PMC11117815 DOI: 10.3390/brainsci14050413] [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: 03/20/2024] [Revised: 04/16/2024] [Accepted: 04/21/2024] [Indexed: 05/26/2024] Open
Abstract
Transmissible Spongiform Encephalopathies (TSEs), including prion diseases such as Bovine Spongiform Encephalopathy (Mad Cow Disease) and variant Creutzfeldt-Jakob Disease, pose unique challenges to the scientific and medical communities due to their infectious nature, neurodegenerative effects, and the absence of a cure. Central to the progression of TSEs is the conversion of the normal cellular prion protein (PrPC) into its infectious scrapie form (PrPSc), leading to neurodegeneration through a complex interplay involving the immune system. This review elucidates the current understanding of the immune response in prion diseases, emphasizing the dual role of the immune system in both propagating and mitigating the disease through mechanisms such as glial activation, cytokine release, and blood-brain barrier dynamics. We highlight the differential cytokine profiles associated with various prion strains and stages of disease, pointing towards the potential for cytokines as biomarkers and therapeutic targets. Immunomodulatory strategies are discussed as promising avenues for mitigating neuroinflammation and delaying disease progression. This comprehensive examination of the immune response in TSEs not only advances our understanding of these enigmatic diseases but also sheds light on broader neuroinflammatory processes, offering hope for future therapeutic interventions.
Collapse
Affiliation(s)
| | | | | | | | - Tuane C. R. G. Vieira
- Institute of Medical Biochemistry Leopoldo de Meis and National Institute of Science and Technology for Structural Biology and Bioimaging, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (G.A.-d.-L.); (R.M.-d.-N.); (M.A.-d.-N.); (A.C.M.)
| |
Collapse
|
41
|
Del Pozo A, Knox KM, Lehmann LM, Davidson S, Rho SL, Jayadev S, Barker-Haliski M. Chronic evoked seizures in young pre-symptomatic APP/PS1 mice induce serotonin changes and accelerate onset of Alzheimer's disease-related neuropathology. Prog Neurobiol 2024; 235:102591. [PMID: 38484965 PMCID: PMC11015961 DOI: 10.1016/j.pneurobio.2024.102591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 02/29/2024] [Accepted: 03/05/2024] [Indexed: 03/18/2024]
Abstract
OBJECTIVE Hyperexcitability is intimately linked to Alzheimer's disease (AD) pathology, but the precise timing and contributions of neuronal hyperexcitability to disease progression is unclear. Seizure induction in rodent AD models can uncover new therapeutic targets. Further, investigator-evoked seizures can directly establish how hyperexcitability and AD-associated risk factors influence neuropathological hallmarks and disease course at presymptomatic stages. METHODS Corneal kindling is a well-characterized preclinical epilepsy model that allows for precise control of seizure history to pair to subsequent behavioral assessments. 2-3-month-old APP/PS1, PSEN2-N141I, and transgenic control male and female mice were thus sham or corneal kindled for 2 weeks. Seizure-induced changes in glia, serotonin pathway proteins, and amyloid β levels in hippocampus and prefrontal cortex were quantified. RESULTS APP/PS1 females were more susceptible to corneal kindling. However, regardless of sex, APP/PS1 mice experienced extensive seizure-induced mortality versus kindled Tg- controls. PSEN2-N141I mice were not negatively affected by corneal kindling. Mortality correlated with a marked downregulation of hippocampal tryptophan hydroxylase 2 and monoamine oxidase A protein expression versus controls; these changes were not detected in PSEN2-N141I mice. Kindled APP/PS1 mice also exhibited soluble amyloid β upregulation and glial reactivity without plaque deposition. SIGNIFICANCE Evoked convulsive seizures and neuronal hyperexcitability in pre-symptomatic APP/PS1 mice promoted premature mortality without pathological Aβ plaque deposition, whereas PSEN2-N141I mice were unaffected. Disruptions in serotonin pathway metabolism in APP/PS1 mice was associated with increased glial reactivity without Aβ plaque deposition, demonstrating that neuronal hyperexcitability in early AD causes pathological Aβ overexpression and worsens long-term outcomes through a serotonin-related mechanism.
Collapse
Affiliation(s)
- Aaron Del Pozo
- Center for Epilepsy Drug Discovery (CEDD), Department of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Kevin M Knox
- Center for Epilepsy Drug Discovery (CEDD), Department of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Leanne M Lehmann
- Center for Epilepsy Drug Discovery (CEDD), Department of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Stephanie Davidson
- Center for Epilepsy Drug Discovery (CEDD), Department of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Seongheon Leo Rho
- Center for Epilepsy Drug Discovery (CEDD), Department of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Suman Jayadev
- Department of Neurology, University of Washington, Seattle, WA 98195, USA
| | - Melissa Barker-Haliski
- Center for Epilepsy Drug Discovery (CEDD), Department of Pharmacy, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
42
|
Edison P. Astroglial activation: Current concepts and future directions. Alzheimers Dement 2024; 20:3034-3053. [PMID: 38305570 PMCID: PMC11032537 DOI: 10.1002/alz.13678] [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: 08/24/2023] [Revised: 11/27/2023] [Accepted: 12/11/2023] [Indexed: 02/03/2024]
Abstract
Astrocytes are abundantly and ubiquitously expressed cell types with diverse functions throughout the central nervous system. Astrocytes show remarkable plasticity and exhibit morphological, molecular, and functional remodeling in response to injury, disease, or infection of the central nervous system, as evident in neurodegenerative diseases. Astroglial mediated inflammation plays a prominent role in the pathogenesis of neurodegenerative diseases. This review focus on the role of astrocytes as essential players in neuroinflammation and discuss their morphological and functional heterogeneity in the normal central nervous system and explore the spatial and temporal variations in astroglial phenotypes observed under different disease conditions. This review discusses the intimate relationship of astrocytes to pathological hallmarks of neurodegenerative diseases. Finally, this review considers the putative therapeutic strategies that can be deployed to modulate the astroglial functions in neurodegenerative diseases. HIGHLIGHTS: Astroglia mediated neuroinflammation plays a key role in the pathogenesis of neurodegenerative diseases. Activated astrocytes exhibit diverse phenotypes in a region-specific manner in brain and interact with β-amyloid, tau, and α-synuclein species as well as with microglia and neuronal circuits. Activated astrocytes are likely to influence the trajectory of disease progression of neurodegenerative diseases, as determined by the stage of disease, individual susceptibility, and state of astroglial priming. Modulation of astroglial activation may be a therapeutic strategy at various stages in the trajectory of neurodegenerative diseases to modify the disease course.
Collapse
Affiliation(s)
- Paul Edison
- Division of NeurologyDepartment of Brain SciencesFaculty of Medicine, Imperial College LondonLondonUK
- Division of Psychological medicine and clinical neurosciencesSchool of Medicine, Cardiff UniversityWalesUK
| |
Collapse
|
43
|
Jain SK, Stevens CM, Margret JJ, Levine SN. Alzheimer's Disease: A Review of Pathology, Current Treatments, and the Potential Therapeutic Effect of Decreasing Oxidative Stress by Combined Vitamin D and l-Cysteine Supplementation. Antioxid Redox Signal 2024; 40:663-678. [PMID: 37756366 PMCID: PMC11001507 DOI: 10.1089/ars.2023.0245] [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: 02/22/2023] [Revised: 08/26/2023] [Accepted: 08/27/2023] [Indexed: 09/29/2023]
Abstract
Significance: Excess oxidative stress and neuroinflammation are risk factors in the onset and progression of Alzheimer's disease (AD) and its association with amyloid-β plaque accumulation. Oxidative stress impairs acetylcholine (ACH) and N-methyl-d-aspartate receptor signaling in brain areas that function in memory and learning. Glutathione (GSH) antioxidant depletion positively correlates with the cognitive decline in AD subjects. Treatments that upregulate GSH and ACH levels, which simultaneously decrease oxidative stress and inflammation, may be beneficial for AD. Recent Advances: Some clinical trials have shown a benefit of monotherapy with vitamin D (VD), whose deficiency is linked to AD or with l-cysteine (LC), a precursor of GSH biosynthesis, in reducing mild cognitive impairment. Animal studies have shown a simultaneous decrease in ACH esterase (AChE) and increase in GSH; combined supplementation with VD and LC results in a greater decrease in oxidative stress and inflammation, and increase in GSH levels compared with monotherapy with VD or LC. Therefore, cosupplementation with VD and LC has the potential of increasing GSH, downregulation of oxidative stress, and decreased inflammation and AChE levels. Future Directions: Clinical trials are needed to determine whether safe low-cost dietary supplements, using combined VD+LC, have the potential to alleviate elevated AChE, oxidative stress, and inflammation levels, thereby halting the onset of AD. Goal of Review: The goal of this review is to highlight the pathological hallmarks and current Food and Drug Administration-approved treatments for AD, and discuss the potential therapeutic effect that cosupplementation with VD+LC could manifest by increasing GSH levels in patients. Antioxid. Redox Signal. 40, 663-678.
Collapse
Affiliation(s)
- Sushil K. Jain
- Department of Pediatrics and Medicine, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana, USA
| | - Christopher M. Stevens
- Department of Pediatrics and Medicine, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana, USA
| | - Jeffrey Justin Margret
- Department of Pediatrics and Medicine, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana, USA
| | - Steven N. Levine
- Department of Pediatrics and Medicine, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana, USA
| |
Collapse
|
44
|
Soni N, Hohsfield LA, Tran KM, Kawauchi S, Walker A, Javonillo D, Phan J, Matheos D, Da Cunha C, Uyar A, Milinkeviciute G, Gomez‐Arboledas A, Tran K, Kaczorowski CC, Wood MA, Tenner AJ, LaFerla FM, Carter GW, Mortazavi A, Swarup V, MacGregor GR, Green KN. Genetic diversity promotes resilience in a mouse model of Alzheimer's disease. Alzheimers Dement 2024; 20:2794-2816. [PMID: 38426371 PMCID: PMC11032575 DOI: 10.1002/alz.13753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 03/02/2024]
Abstract
INTRODUCTION Alzheimer's disease (AD) is a neurodegenerative disorder with multifactorial etiology, including genetic factors that play a significant role in disease risk and resilience. However, the role of genetic diversity in preclinical AD studies has received limited attention. METHODS We crossed five Collaborative Cross strains with 5xFAD C57BL/6J female mice to generate F1 mice with and without the 5xFAD transgene. Amyloid plaque pathology, microglial and astrocytic responses, neurofilament light chain levels, and gene expression were assessed at various ages. RESULTS Genetic diversity significantly impacts AD-related pathology. Hybrid strains showed resistance to amyloid plaque formation and neuronal damage. Transcriptome diversity was maintained across ages and sexes, with observable strain-specific variations in AD-related phenotypes. Comparative gene expression analysis indicated correlations between mouse strains and human AD. DISCUSSION Increasing genetic diversity promotes resilience to AD-related pathogenesis, relative to an inbred C57BL/6J background, reinforcing the importance of genetic diversity in uncovering resilience in the development of AD. HIGHLIGHTS Genetic diversity's impact on AD in mice was explored. Diverse F1 mouse strains were used for AD study, via the Collaborative Cross. Strain-specific variations in AD pathology, glia, and transcription were found. Strains resilient to plaque formation and plasma neurofilament light chain (NfL) increases were identified. Correlations with human AD transcriptomics were observed.
Collapse
Affiliation(s)
- Neelakshi Soni
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
| | - Lindsay A. Hohsfield
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | - Kristine M. Tran
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
| | - Shimako Kawauchi
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
- Transgenic Mouse Facility, ULAROffice of ResearchUniversity of CaliforniaIrvineCaliforniaUSA
| | - Amber Walker
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
- Transgenic Mouse Facility, ULAROffice of ResearchUniversity of CaliforniaIrvineCaliforniaUSA
| | - Dominic Javonillo
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
| | - Jimmy Phan
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | - Dina Matheos
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
| | - Celia Da Cunha
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | - Asli Uyar
- The Jackson LaboratoryBar HarborMaineUSA
| | - Giedre Milinkeviciute
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | - Angela Gomez‐Arboledas
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | - Katelynn Tran
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | | | - Marcelo A. Wood
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | - Andrea J. Tenner
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
- Department of Molecular Biology and BiochemistryUniversity of CaliforniaIrvineCaliforniaUSA
- Department of Pathology and Laboratory MedicineUniversity of CaliforniaIrvineCaliforniaUSA
| | - Frank M. LaFerla
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | | | - Ali Mortazavi
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
- Department of Developmental and Cellular BiologyUniversity of CaliforniaIrvineCaliforniaUSA
- Center for Complex Biological SystemsUniversity of CaliforniaIrvineCaliforniaUSA
| | - Vivek Swarup
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | - Grant R. MacGregor
- Transgenic Mouse Facility, ULAROffice of ResearchUniversity of CaliforniaIrvineCaliforniaUSA
- Department of Developmental and Cellular BiologyUniversity of CaliforniaIrvineCaliforniaUSA
| | - Kim N. Green
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| |
Collapse
|
45
|
Pak V, Adewale Q, Bzdok D, Dadar M, Zeighami Y, Iturria-Medina Y. Distinctive whole-brain cell types predict tissue damage patterns in thirteen neurodegenerative conditions. eLife 2024; 12:RP89368. [PMID: 38512130 PMCID: PMC10957173 DOI: 10.7554/elife.89368] [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] [Indexed: 03/22/2024] Open
Abstract
For over a century, brain research narrative has mainly centered on neuron cells. Accordingly, most neurodegenerative studies focus on neuronal dysfunction and their selective vulnerability, while we lack comprehensive analyses of other major cell types' contribution. By unifying spatial gene expression, structural MRI, and cell deconvolution, here we describe how the human brain distribution of canonical cell types extensively predicts tissue damage in 13 neurodegenerative conditions, including early- and late-onset Alzheimer's disease, Parkinson's disease, dementia with Lewy bodies, amyotrophic lateral sclerosis, mutations in presenilin-1, and 3 clinical variants of frontotemporal lobar degeneration (behavioral variant, semantic and non-fluent primary progressive aphasia) along with associated three-repeat and four-repeat tauopathies and TDP43 proteinopathies types A and C. We reconstructed comprehensive whole-brain reference maps of cellular abundance for six major cell types and identified characteristic axes of spatial overlapping with atrophy. Our results support the strong mediating role of non-neuronal cells, primarily microglia and astrocytes, in spatial vulnerability to tissue loss in neurodegeneration, with distinct and shared across-disorder pathomechanisms. These observations provide critical insights into the multicellular pathophysiology underlying spatiotemporal advance in neurodegeneration. Notably, they also emphasize the need to exceed the current neuro-centric view of brain diseases, supporting the imperative for cell-specific therapeutic targets in neurodegeneration.
Collapse
Affiliation(s)
- Veronika Pak
- Department of Neurology and Neurosurgery, McGill UniversityMontrealCanada
- McConnell Brain Imaging Centre, Montreal Neurological InstituteMontrealCanada
- Ludmer Centre for Neuroinformatics & Mental HealthMontrealCanada
| | - Quadri Adewale
- Department of Neurology and Neurosurgery, McGill UniversityMontrealCanada
- McConnell Brain Imaging Centre, Montreal Neurological InstituteMontrealCanada
- Ludmer Centre for Neuroinformatics & Mental HealthMontrealCanada
| | - Danilo Bzdok
- McConnell Brain Imaging Centre, Montreal Neurological InstituteMontrealCanada
- Department of Biomedical Engineering, McGill UniversityMontrealCanada
- School of Computer Science, McGill UniversityMontrealCanada
- Mila – Quebec Artificial Intelligence InstituteMontrealCanada
| | | | | | - Yasser Iturria-Medina
- Department of Neurology and Neurosurgery, McGill UniversityMontrealCanada
- McConnell Brain Imaging Centre, Montreal Neurological InstituteMontrealCanada
- Ludmer Centre for Neuroinformatics & Mental HealthMontrealCanada
- Department of Biomedical Engineering, McGill UniversityMontrealCanada
- McGill Centre for Studies in AgingMontrealCanada
| |
Collapse
|
46
|
García-Juan M, Ordóñez-Gutiérrez L, Wandosell F. Clearance of β-amyloid mediated by autophagy is enhanced by MTORC1 inhibition but not AMPK activation in APP/PSEN1 astrocytes. Glia 2024; 72:588-606. [PMID: 38009275 DOI: 10.1002/glia.24492] [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: 02/08/2023] [Revised: 11/07/2023] [Accepted: 11/13/2023] [Indexed: 11/28/2023]
Abstract
Proteostasis mechanisms mediated by macroautophagy/autophagy are altered in neurodegenerative diseases such as Alzheimer disease (AD) and their recovery/enhancement has been proposed as a therapeutic approach. From the two central nodes in the anabolism-catabolism balance, it is generally accepted that mechanistic target of rapamycin kinase complex 1 (MTORC1)_ activation leads to the inhibition of autophagy, whereas adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) has the opposite role. In AD, amyloid beta (Aβ) production disturbs the optimal neuronal/glial proteostasis. As astrocytes are essential for brain homeostasis, the purpose of this work was to analyze if the upregulation of autophagy in this cell type, either by MTORC1 inhibition or AMPK activation, could modulate the generation/degradation of β-amyloid. By using primary astrocytes from amyloid beta precursor protein (APP)/Presenilin 1 (PSEN1) mouse model of AD, we confirmed that MTORC1 inhibition reduced Aβ secretion through moderate autophagy induction. Surprisingly, pharmacologically increased activity of AMPK did not enhance autophagy but had different effects on Aβ secretion. Conversely, AMPK inhibition did not affect autophagy but reduced Aβ secretion. These puzzling data were confirmed through the overexpression of different mutant AMPK isoforms: while only the constitutively active AMPK increased autophagy, all versions augmented Aβ secretion. We conclude that AMPK has a significantly different role in primary astrocytes than in other reported cells, similar to our previous findings in neurons. Our data support that perhaps only a basal AMPK activity is needed to maintain autophagy whereas the increased activity, either physiologically or pharmacologically, has no direct effect on autophagy-dependent amyloidosis. These results shed light on the controversy about the therapeutic effect of AMPK activation on autophagy induction.
Collapse
Affiliation(s)
- Marta García-Juan
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Nicolas Cabrera 1, Universidad Autónoma de Madrid, Madrid, Spain
| | - Lara Ordóñez-Gutiérrez
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Nicolas Cabrera 1, Universidad Autónoma de Madrid, Madrid, Spain
- Departamento de Bioquímica ry Biología Molecular, Universidad Complutense de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Francisco Wandosell
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Nicolas Cabrera 1, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
47
|
Wang X, Gan W, Kang M, Lv C, Zhao Z, Wu Y, Zhang X, Wang R. Asthma aggravates alzheimer's disease by up-regulating NF- κB signaling pathway through LTD4. Brain Res 2024; 1825:148711. [PMID: 38092296 DOI: 10.1016/j.brainres.2023.148711] [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: 08/22/2023] [Revised: 11/17/2023] [Accepted: 12/09/2023] [Indexed: 12/25/2023]
Abstract
Clinical studies have shown that asthma is a risk factor for dementia or Alzheimer's disease (AD). To investigate whether asthma aggravates AD in APP/PS1 mice and explore the potential mechanisms, an asthma model was established using six-month-old APP/PS1 mice, and montelukast was used as a therapeutic agent in APP/PS1 mice with asthma. The Morris water maze test showed that asthma aggravates spatial learning and memory abilities. Asthma also upregulates the NF-κB inflammatory pathway in APP/PS1 mice and promotes the expression of beta-site amyloid precursor protein cleaving enzyme 1 (BACE1), amyloid-β (Aβ) deposition, neuronal damage, synaptic plasticity deficiency, activation of microglia and astrocytes. The level of LTD4 and its receptor CysLT1R in the hippocampus of APP/PS1 mice after the asthma modeling was established was higher than that in APP/PS1 mice, suggesting that asthma may affect the pathology of AD through LTD4 and its receptor Cys-LT1R. Montelukast ameliorates these pathological changes and cognitive impairment. These results suggest that asthma aggravates AD pathology and cognitive impairment of APP/PS1 mice via upregulation of the NF-κB inflammatory pathway, and montelukast ameliorates these pathological changes.
Collapse
Affiliation(s)
- Xiaozhen Wang
- Central Laboratory, Xuanwu Hospital, Capital Medical University, Beijing Geriatric Medical Research Center, Key Laboratory for Neurodegenerative Disease of Ministry of Education, Beijing, PR China
| | - Wenjing Gan
- Central Laboratory, Xuanwu Hospital, Capital Medical University, Beijing Geriatric Medical Research Center, Key Laboratory for Neurodegenerative Disease of Ministry of Education, Beijing, PR China
| | - Meimei Kang
- Central Laboratory, Xuanwu Hospital, Capital Medical University, Beijing Geriatric Medical Research Center, Key Laboratory for Neurodegenerative Disease of Ministry of Education, Beijing, PR China
| | - Caizhen Lv
- Central Laboratory, Xuanwu Hospital, Capital Medical University, Beijing Geriatric Medical Research Center, Key Laboratory for Neurodegenerative Disease of Ministry of Education, Beijing, PR China
| | - Zhiwei Zhao
- Central Laboratory, Xuanwu Hospital, Capital Medical University, Beijing Geriatric Medical Research Center, Key Laboratory for Neurodegenerative Disease of Ministry of Education, Beijing, PR China
| | - Yanchuan Wu
- Central Laboratory, Xuanwu Hospital, Capital Medical University, Beijing Geriatric Medical Research Center, Key Laboratory for Neurodegenerative Disease of Ministry of Education, Beijing, PR China
| | - Xu Zhang
- Central Laboratory, Xuanwu Hospital, Capital Medical University, Beijing Geriatric Medical Research Center, Key Laboratory for Neurodegenerative Disease of Ministry of Education, Beijing, PR China
| | - Rong Wang
- Central Laboratory, Xuanwu Hospital, Capital Medical University, Beijing Geriatric Medical Research Center, Key Laboratory for Neurodegenerative Disease of Ministry of Education, Beijing, PR China; Beijing Institute for Brain Disorders, Beijing, PR China; National Clinical Research Center for Geriatric Disorders, Beijing, PR China.
| |
Collapse
|
48
|
Gordon-Blake J, Ratia K, Weidig V, Velma GR, Ackerman-Berrier M, Penton C, Musku SR, Alves ET, Driver T, Tai L, Thatcher GRJ. Nicotinamide Phosphoribosyltransferase Positive Allosteric Modulators Attenuate Neuronal Oxidative Stress. ACS Med Chem Lett 2024; 15:205-214. [PMID: 38352833 PMCID: PMC10860701 DOI: 10.1021/acsmedchemlett.3c00391] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 12/02/2023] [Accepted: 01/13/2024] [Indexed: 02/16/2024] Open
Abstract
Evidence supports boosting nicotinamide adenine dinucleotide (NAD+) to counteract oxidative stress in aging and neurodegenerative disease. One approach is to enhance the activity of nicotinamide phosphoribosyltransferase (NAMPT). Novel NAMPT positive allosteric modulators (N-PAMs) were identified. A cocrystal structure confirmed N-PAM binding to the NAMPT rear channel. Early hit-to-lead efforts led to a 1.88-fold maximum increase in the level of NAD+ in human THP-1 cells. Select N-PAMs were assessed for mitigation of reactive oxygen species (ROS) in HT-22 neuronal cells subject to inflammatory stress using tumor necrosis factor alpha (TNFα). N-PAMs that increased NAD+ more effectively in THP-1 cells attenuated TNFα-induced ROS more effectively in HT-22 cells. The most efficacious N-PAM completely attenuated ROS elevation in glutamate-stressed HT-22 cells, a model of neuronal excitotoxicity. This work demonstrates for the first time that N-PAMs are capable of mitigating elevated ROS in neurons stressed with TNFα and glutamate and provides support for further N-PAM optimization for treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Jesse Gordon-Blake
- Department
of Pharmaceutical Sciences, Research Resources Center, Department of Chemistry, and Department of
Anatomy and Cell Biology, University of
Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Kiira Ratia
- Department
of Pharmaceutical Sciences, Research Resources Center, Department of Chemistry, and Department of
Anatomy and Cell Biology, University of
Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Victoria Weidig
- Department
of Pharmaceutical Sciences, Research Resources Center, Department of Chemistry, and Department of
Anatomy and Cell Biology, University of
Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Ganga Reddy Velma
- Department
of Pharmacology & Toxicology, R Ken Coit College of Pharmacy, University of Arizona, Tucson, Arizona 85721, United States
| | - Martha Ackerman-Berrier
- Department
of Pharmacology & Toxicology, R Ken Coit College of Pharmacy, University of Arizona, Tucson, Arizona 85721, United States
| | - Christopher Penton
- Department
of Pharmacology & Toxicology, R Ken Coit College of Pharmacy, University of Arizona, Tucson, Arizona 85721, United States
| | - Soumya Reddy Musku
- Department
of Pharmacology & Toxicology, R Ken Coit College of Pharmacy, University of Arizona, Tucson, Arizona 85721, United States
| | - Erick T.M. Alves
- Department
of Pharmacology & Toxicology, R Ken Coit College of Pharmacy, University of Arizona, Tucson, Arizona 85721, United States
| | - Tom Driver
- Department
of Pharmaceutical Sciences, Research Resources Center, Department of Chemistry, and Department of
Anatomy and Cell Biology, University of
Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Leon Tai
- Department
of Pharmaceutical Sciences, Research Resources Center, Department of Chemistry, and Department of
Anatomy and Cell Biology, University of
Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Gregory R. J. Thatcher
- Department
of Pharmacology & Toxicology, R Ken Coit College of Pharmacy, University of Arizona, Tucson, Arizona 85721, United States
| |
Collapse
|
49
|
Li K, Ling H, Wang X, Xie Q, Gu C, Luo W, Qiu P. The role of NF-κB signaling pathway in reactive astrocytes among neurodegeneration after methamphetamine exposure by integrated bioinformatics. Prog Neuropsychopharmacol Biol Psychiatry 2024; 129:110909. [PMID: 38061485 DOI: 10.1016/j.pnpbp.2023.110909] [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: 09/06/2023] [Revised: 11/30/2023] [Accepted: 11/30/2023] [Indexed: 12/19/2023]
Abstract
BACKGROUND Methamphetamine (METH) is a highly addictive stimulant that has become one of the top five risk substances cause deaths from substance abuse. METH exposure increases the risk of neurodegenerative disease (ND), such as Parkinson's disease (PD), leading to disability and death. Activation of reactive astrocytes is an essential factor in neurodegeneration, and their complex role in METH exposure remains unclear. This study explored the role of reactive astrocyte overactivation in neurodegeneration after METH exposure. METHODS METH bulk RNA sequencing data (GSE107015 and GSE98793) and single-cell RNA sequencing data (GSE119861) were obtained from the GEO database. We performed immune infiltration analysis on the bulk RNA data. After cell clustering using the single-cell RNA data, astrocytes were extracted for downstream analysis. Differentially expressed genes (DEGs) were identified from the bulk and single-cell RNA sequencing datasets, and GO, KEGG, and GSEA pathway analyses were performed. The PPI network and random forest methods were performed on the overlapping genes of the DEGs to screen hub genes. To explore the common ground between METH exposure and neurodegenerative diseases, we applied a random forest algorithm to PD chip data (GSE99039 and GSE72267) to establish a diagnostic model using the hub genes in METH. New object recognition and the Morris water maze were used to examine cognitive function in mice exposed to METH for 14 days in vivo. Astrocytes were cocultured with neurons for the detection of intercellular crosstalk. RESULTS DEGs in the METH group significantly enriched pathways related to NDs, inflammation, and the NF-κB signaling pathway. Immune infiltration analysis revealed significantly increased infiltration of monocytes, T cells, and NK cells and decreased infiltration of neutrophils in the METH group. An intersection of 44 hub genes was screened based on the PPI network and random forest algorithm. These genes suggest that there might be similar pathogenesis between METH exposure and PD. METH exposure resulted in learning memory impairment, hippocampal astrocyte activation, and upregulation of NF-κB expression in mice. Activation of reactive astrocytes cocultured with neurons causes neural damage. CONCLUSIONS This study explored the crosstalk between astrocytes and neurons in METH exposure, providing a potential pathogenesis to explore the altered immune microenvironment involving reactive astrocytes after METH exposure.
Collapse
Affiliation(s)
- Kuan Li
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Haosen Ling
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Xiaohan Wang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Qiqian Xie
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Cihang Gu
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Wenyu Luo
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Pingming Qiu
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China.; Department of Thyroid Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
| |
Collapse
|
50
|
Ali M, Huarte OU, Heurtaux T, Garcia P, Rodriguez BP, Grzyb K, Halder R, Skupin A, Buttini M, Glaab E. Single-Cell Transcriptional Profiling and Gene Regulatory Network Modeling in Tg2576 Mice Reveal Gender-Dependent Molecular Features Preceding Alzheimer-Like Pathologies. Mol Neurobiol 2024; 61:541-566. [PMID: 35980567 PMCID: PMC10861719 DOI: 10.1007/s12035-022-02985-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 07/29/2022] [Indexed: 11/26/2022]
Abstract
Alzheimer's disease (AD) onset and progression is influenced by a complex interplay of several environmental and genetic factors, one of them gender. Pronounced gender differences have been observed both in the relative risk of developing AD and in clinical disease manifestations. A molecular level understanding of these gender disparities is still missing, but could provide important clues on cellular mechanisms modulating the disease and reveal new targets for gender-oriented disease-modifying precision therapies. We therefore present here a comprehensive single-cell analysis of disease-associated molecular gender differences in transcriptomics data from the neocortex, one of the brain regions most susceptible to AD, in one of the most widely used AD mouse models, the Tg2576 model. Cortical areas are also most commonly used in studies of post-mortem AD brains. To identify disease-linked molecular processes that occur before the onset of detectable neuropathology, we focused our analyses on an age with no detectable plaques and microgliosis. Cell-type specific alterations were investigated at the level of individual genes, pathways, and gene regulatory networks. The number of differentially expressed genes (DEGs) was not large enough to build context-specific gene regulatory networks for each individual cell type, and thus, we focused on the study of cell types with dominant changes and included analyses of changes across the combination of cell types. We observed significant disease-associated gender differences in cellular processes related to synapse organization and reactive oxygen species metabolism, and identified a limited set of transcription factors, including Egr1 and Klf6, as key regulators of many of the disease-associated and gender-dependent gene expression changes in the model. Overall, our analyses revealed significant cell-type specific gene expression changes in individual genes, pathways and sub-networks, including gender-specific and gender-dimorphic changes in both upstream transcription factors and their downstream targets, in the Tg2576 AD model before the onset of overt disease. This opens a window into molecular events that could determine gender-susceptibility to AD, and uncovers tractable target candidates for potential gender-specific precision medicine for AD.
Collapse
Affiliation(s)
- Muhammad Ali
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg
- School for Mental Health and Neuroscience (MHeNs), Department of Psychiatry and Neuropsychology, Maastricht University, 6200, Maastricht, the Netherlands
| | - Oihane Uriarte Huarte
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg
- Luxembourg Center of Neuropathology (LCNP), L-3555, Dudelange, Luxembourg
| | - Tony Heurtaux
- Luxembourg Center of Neuropathology (LCNP), L-3555, Dudelange, Luxembourg
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, L‑4362, Esch-Sur-Alzette, Luxembourg
| | - Pierre Garcia
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg
- Luxembourg Center of Neuropathology (LCNP), L-3555, Dudelange, Luxembourg
| | - Beatriz Pardo Rodriguez
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg
- Luxembourg Center of Neuropathology (LCNP), L-3555, Dudelange, Luxembourg
- University of the Basque Country, Cell Biology and Histology Department, 48940, Leioa, Vizcaya, Basque Country, Spain
| | - Kamil Grzyb
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg
| | - Rashi Halder
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg
| | - Alexander Skupin
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg
- Department of Physics and Materials Science, University of Luxembourg, 162a av. de la Faïencerie, 1511, Luxembourg, Luxembourg
- Department of Neuroscience, University of California San Diego, 9500 Gilman Dr, La Jolla, CA, 92093, USA
| | - Manuel Buttini
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg
- Luxembourg Center of Neuropathology (LCNP), L-3555, Dudelange, Luxembourg
| | - Enrico Glaab
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg.
| |
Collapse
|