1
|
D'Alessandro MCB, Kanaan S, Geller M, Praticò D, Daher JPL. Mitochondrial dysfunction in Alzheimer's disease. Ageing Res Rev 2025; 107:102713. [PMID: 40023293 DOI: 10.1016/j.arr.2025.102713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 02/13/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disease characterized by progressive cognitive decline and distinct neuropathological features. The absence of a definitive cure presents a significant challenge in neurology and neuroscience. Early clinical manifestations, such as memory retrieval deficits and apathy, underscore the need for a deeper understanding of the disease's underlying mechanisms. While amyloid-β plaques and tau neurofibrillary tangles have dominated research efforts, accumulating evidence highlights mitochondrial dysfunction as a central factor in AD pathogenesis. Mitochondria, essential cellular organelles responsible for energy production necessary for neuronal function become impaired in AD, triggering several cellular consequences. Factors such as oxidative stress, disturbances in energy metabolism, failures in the mitochondrial quality control system, and dysregulation of calcium release are associated with mitochondrial dysfunction. These abnormalities are closely linked to the neurodegenerative processes driving AD development and progression. This review explores the intricate relationship between mitochondrial dysfunction and AD pathogenesis, emphasizing its role in disease onset and progression, while also considering its potential as a biomarker and a therapeutic target.
Collapse
Affiliation(s)
- Maria Clara Bila D'Alessandro
- Universidade Federal Fluminense, Faculty of Medicine, Desembargador Athayde Parreiras road 100, Niterói, Rio de Janeiro, Brazil.
| | - Salim Kanaan
- Universidade Federal Fluminense, Faculty of Medicine, Department of Pathology, Marquês do Paraná road, 303, 2nd floor, Niterói, Rio de Janeiro, Brazil.
| | - Mauro Geller
- Unifeso, Department of Immunology and Microbiology, Alberto Torres avenue 111, Teresópolis, Rio de Janeiro, Brazil
| | - Domenico Praticò
- Department of Neurosciences, Lewis Katz School of Medicine. Temple University, 3500 North Broad Street, Philadelphia, PA, United States.
| | - João Paulo Lima Daher
- Universidade Federal Fluminense, Faculty of Medicine, Department of Pathology, Marquês do Paraná road, 303, 2nd floor, Niterói, Rio de Janeiro, Brazil.
| |
Collapse
|
2
|
Zia‐Ur‐Rehman, Awang MK, Ali G, Faheem M. Recent Advancements in Neuroimaging-Based Alzheimer's Disease Prediction Using Deep Learning Approaches in e-Health: A Systematic Review. Health Sci Rep 2025; 8:e70802. [PMID: 40330773 PMCID: PMC12051440 DOI: 10.1002/hsr2.70802] [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: 12/01/2024] [Revised: 04/07/2025] [Accepted: 04/14/2025] [Indexed: 05/08/2025] Open
Abstract
Purpose Alzheimer's disease (AD) is a severe neurological disease that significantly impairs brain function. Timely identification of AD is essential for appropriate treatment and care. This comprehensive review intends to examine current developments in deep learning (DL) approaches with neuroimaging for AD diagnosis, where popular imaging types, reviews well-known online accessible data sets, and describes different algorithms used in DL for the correct initial evaluation of AD are presented. Significance Conventional diagnostic techniques, including medical evaluations and cognitive assessments, usually not identify the initial stages of Alzheimer's. Neuroimaging methods, when integrated with DL techniques, have demonstrated considerable potential in enhancing the diagnosis and categorization of AD. DL models have received significant interest due to their capability to identify AD in its early phases automatically, which reduces the mortality rate and treatment cost of AD. Method An extensive literature search was performed in leading scientific databases, concentrating on papers published from 2021 to 2025. Research leveraging DL models on different neuroimaging techniques such as magnetic resonance imaging (MRI), positron emission tomography, and functional magnetic resonance imaging (fMRI), and so forth. The review complies with Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Results Current developments show that CNN-based techniques, especially those utilizing hybrid and transfer learning frameworks, outperform conventional DL methods. Research employing the combination of multimodal neuroimaging data has demonstrated enhanced diagnostic precision. Still, challenges such as method interpretability, data heterogeneity, and limited data exist as significant issues. Conclusion DL has considerably improved the accuracy and reliability of AD diagnosis with neuroimaging. Regardless of issues with data accessibility and adaptability, current studies into the interpretability of models and multimodal fusion provide potential for clinical application. Further research should concentrate on standardized data sets, rigorous validation architectures, and understandable AI methodologies to enhance the effectiveness of DL methods in AD prediction.
Collapse
Affiliation(s)
- Zia‐Ur‐Rehman
- Faculty of Informatics and Computing (FIK)Universiti Sultan Zainal Abidin (UniSZA)BesutTerengganuMalaysia
| | - Mohd Khalid Awang
- Faculty of Informatics and Computing (FIK)Universiti Sultan Zainal Abidin (UniSZA)BesutTerengganuMalaysia
| | - Ghulam Ali
- Department of Computer ScienceUniversity of OkaraOkaraPakistan
| | - Muhammad Faheem
- School of Technology and InnovationsUniversity of VaasaVaasaFinland
- VTT Technical Research Centre of FinlandEspooFinland
| |
Collapse
|
3
|
Ponce-Lopez T. Peripheral Inflammation and Insulin Resistance: Their Impact on Blood-Brain Barrier Integrity and Glia Activation in Alzheimer's Disease. Int J Mol Sci 2025; 26:4209. [PMID: 40362446 PMCID: PMC12072112 DOI: 10.3390/ijms26094209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2025] [Revised: 04/22/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by cognitive decline, memory impairment, and synaptic dysfunction. The accumulation of amyloid beta (Aβ) plaques and hyperphosphorylated tau protein leads to neuronal dysfunction, neuroinflammation, and glial cell activation. Emerging evidence suggests that peripheral insulin resistance and chronic inflammation, often associated with type 2 diabetes (T2D) and obesity, promote increased proinflammatory cytokines, oxidative stress, and immune cell infiltration. These conditions further damage the blood-brain barrier (BBB) integrity and promote neurotoxicity and chronic glial cell activation. This induces neuroinflammation and impaired neuronal insulin signaling, reducing glucose metabolism and exacerbating Aβ accumulation and tau hyperphosphorylation. Indeed, epidemiological studies have linked T2D and obesity with an increased risk of developing AD, reinforcing the connection between metabolic disorders and neurodegeneration. This review explores the relationships between peripheral insulin resistance, inflammation, and BBB dysfunction, highlighting their role in glial activation and the exacerbation of AD pathology.
Collapse
Affiliation(s)
- Teresa Ponce-Lopez
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte, Huixquilucan 52786, Mexico
| |
Collapse
|
4
|
Vidil S, Singh-Manoux A, Landré B, Fayosse A, Sabia S, Machado-Fragua MD. Optimizing midlife metabolic syndrome thresholds for dementia: a prospective study of two UK population-based cohorts. Alzheimers Res Ther 2025; 17:89. [PMID: 40269987 PMCID: PMC12016442 DOI: 10.1186/s13195-025-01732-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 04/06/2025] [Indexed: 04/25/2025]
Abstract
BACKGROUND The concept of metabolic syndrome (MetS) was developed to identify individuals at higher risk of type 2 diabetes and cardiovascular disease, but its relevance for dementia remains unclear. We examined MetS in midlife for association with late-onset dementia, focusing on the thresholds of MetS components that carry risk for dementia. METHODS MetS components (waist circumference, blood pressure, triglycerides, HDL-C, and fasting glucose) were measured on 6,137 white participants < 60 years from the Whitehall II (WII) cohort study. A changepoint method in time-to-event analyses was used to identify optimal thresholds, and those exhibiting better performance for dementia were retained to develop a revised MetS definition. Results were validated on 171,886 participants in the UK Biobank (UKB) study. RESULTS Over a median follow-up of 22.6 years in WII and 13.8 years in UKB, 522 and 418 late-onset dementia cases were recorded, respectively. Optimized thresholds for triglycerides and fasting glucose performed better than original MetS thresholds in WII, and were used to develop a revised MetS definition. The MetS scale had a linear association with dementia, and 1-component increment (range 0 to 5) was associated with higher dementia risk using the revised MetS definition (HR, 95% CI: 1.11, 1.03-1.19) but not the original MetS definition (HR, 95% CI: 1.06, 0.98-1.14) in WII. In UKB, the revised MetS definition exhibited better performance for dementia risk than the original definition (p for HR comparison < 0.01). CONCLUSIONS MetS in midlife is potentially an important target for dementia prevention. However, the thresholds for triglycerides and glucose that carry risk need to be tailored specifically for dementia.
Collapse
Affiliation(s)
- Sam Vidil
- Epidemiology of Ageing and Neurodegenerative diseases (EpiAgeing), Université Paris Cité, Inserm U1153, Paris, France
| | - Archana Singh-Manoux
- Epidemiology of Ageing and Neurodegenerative diseases (EpiAgeing), Université Paris Cité, Inserm U1153, Paris, France
- Faculty of Brain Sciences, University College London, Alexandra House, 17 Queen Square, London, WC1N3AR, UK
| | - Benjamin Landré
- Epidemiology of Ageing and Neurodegenerative diseases (EpiAgeing), Université Paris Cité, Inserm U1153, Paris, France
| | - Aurore Fayosse
- Epidemiology of Ageing and Neurodegenerative diseases (EpiAgeing), Université Paris Cité, Inserm U1153, Paris, France
| | - Séverine Sabia
- Epidemiology of Ageing and Neurodegenerative diseases (EpiAgeing), Université Paris Cité, Inserm U1153, Paris, France.
- Faculty of Brain Sciences, University College London, Alexandra House, 17 Queen Square, London, WC1N3AR, UK.
| | - Marcos D Machado-Fragua
- Epidemiology of Ageing and Neurodegenerative diseases (EpiAgeing), Université Paris Cité, Inserm U1153, Paris, France
| |
Collapse
|
5
|
Vear A, Heneka MT, Clemmensen C. Incretin-based therapeutics for the treatment of neurodegenerative diseases. Nat Metab 2025; 7:679-696. [PMID: 40211045 DOI: 10.1038/s42255-025-01263-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 03/06/2025] [Indexed: 04/12/2025]
Abstract
Neurodegenerative diseases (NDDs) represent a heterogeneous group of disorders characterized by progressive neuronal loss, which results in significant deficits in memory, cognition, motor skills, and sensory functions. As the prevalence of NDDs rises, there is an urgent unmet need for effective therapies. Current drug development approaches primarily target single pathological features of the disease, which could explain the limited efficacy observed in late-stage clinical trials. Originally developed for the treatment of obesity and diabetes, incretin-based therapies, particularly long-acting GLP-1 receptor (GLP-1R) agonists and GLP-1R-gastric inhibitory polypeptide receptor (GIPR) dual agonists, are emerging as promising treatments for NDDs. Despite limited conclusive preclinical evidence, their pleiotropic ability to reduce neuroinflammation, enhance neuronal energy metabolism and promote synaptic plasticity positions them as potential disease-modifying NDD interventions. In anticipation of results from larger clinical trials, continued advances in next-generation incretin mimetics offer the potential for improved brain access and enhanced neuroprotection, paving the way for incretin-based therapies as a future cornerstone in the management of NDDs.
Collapse
Affiliation(s)
- Anika Vear
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Michael T Heneka
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
- Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany
- Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Christoffer Clemmensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
6
|
Yu B, Li M, Yu Z, Zhang H, Feng X, Gao A, Gao R, Gao R. Red blood cell distribution width to albumin ratio (RAR) is associated with low cognitive performance in American older adults: NHANES 2011-2014. BMC Geriatr 2025; 25:157. [PMID: 40055657 PMCID: PMC11887108 DOI: 10.1186/s12877-025-05800-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 02/17/2025] [Indexed: 05/13/2025] Open
Abstract
BACKGROUND The red blood cell distribution width to albumin ratio (RAR) is a novel comprehensive biomarker of inflammation and nutrition, which has emerged as a reliable prognostic indicator for adverse outcomes and mortality in patients with various diseases. However, the association between RAR and low cognitive performance in older adults remains unclear. This study aims to investigate the relationship between RAR and low cognitive performance among older adults in the United States. METHODS This study, a retrospective analysis, included 2,765 participants aged 60 years and older from the National Health and Nutrition Examination Survey (NHANES) conducted between 2011 and 2014. Low cognitive performance was assessed using word learning subset from the Consortium to Establish a Registry for Alzheimer's Disease (CERAD), the Digit Symbol Substitution Test (DSST), and the Animal Fluency Test (AFT). Low cognitive performance was defined as scores below the lowest quartile in each cognitive test. The association between RAR and low cognitive performance was evaluated using weighted multivariable logistic regression, restricted cubic splines (RCS), and subgroup analyses. RESULTS After adjusting for all potential confounders, RAR was independently and linearly positively associated with both low DSST performance and low AFT performance. Specifically, compared to participants in the first quartile of RAR, those in the fourth quartile had adjusted ORs (95% CIs) of 1.81 (1.03, 3.20) for low DSST performance and 1.68 (1.05, 2.67) for low AFT performance. Subgroup analysis did not reveal significant interactions between stratification variables. CONCLUSION RAR is significantly linearly positively associated with low cognitive performance. Maintaining a lower RAR may be a crucial strategy for mitigating the risk of cognitive decline in the elderly population.
Collapse
Affiliation(s)
- Binyang Yu
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, China
- Xiyuan Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Min Li
- School of Nursing, Xi 'an Jiaotong University Health Science Center, Xi 'an, 710061, China
| | - Zongliang Yu
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Haoling Zhang
- Xiyuan Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Xue Feng
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Anran Gao
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Rui Gao
- Xiyuan Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, 100091, China.
| | - Rui Gao
- School of Nursing, Xi 'an Jiaotong University Health Science Center, Xi 'an, 710061, China.
| |
Collapse
|
7
|
Li J, Wang M, Wang Y, Peng X, Lv G, Zheng T, Peng Y, Li J. Revealing Lingonberry's Neuroprotective Potential in Alzheimer's Disease Through Network Pharmacology and Molecular Docking. Int J Mol Sci 2025; 26:2363. [PMID: 40076984 PMCID: PMC11899733 DOI: 10.3390/ijms26052363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/28/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025] Open
Abstract
Alzheimer's disease is a progressive neurodegenerative disorder with limited treatment options. Lingonberry (Vaccinium vitis-idaea L.) has demonstrated neuroprotective and anti-inflammatory properties, yet the underlying mechanisms remain unclear. This study employed network pharmacology, molecular docking, and molecular dynamics simulations to explore the therapeutic potential in Alzheimer's disease. Pathway analysis identified monoamine oxidase B as a key target involved in serotonergic synapse dysfunction related to Alzheimer's disease. Molecular docking revealed that ferulic acid, a major bioactive compound in lingonberry, exhibits strong binding affinity to monoamine oxidase B. Further molecular dynamics simulations confirmed the stability of this interaction, highlighting the potential inhibitory effect of ferulic acid on monoamine oxidase B. These findings provide novel insights into the neuroprotective mechanisms of lingonberry and suggest its potential as a natural therapeutic intervention for Alzheimer's disease.
Collapse
Affiliation(s)
- Juncheng Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Harbin Medical University, Harbin 150086, China; (J.L.); (M.W.); (Y.W.); (X.P.); (G.L.); (T.Z.)
| | - Mian Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Harbin Medical University, Harbin 150086, China; (J.L.); (M.W.); (Y.W.); (X.P.); (G.L.); (T.Z.)
| | - Yi Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Harbin Medical University, Harbin 150086, China; (J.L.); (M.W.); (Y.W.); (X.P.); (G.L.); (T.Z.)
| | - Xichen Peng
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Harbin Medical University, Harbin 150086, China; (J.L.); (M.W.); (Y.W.); (X.P.); (G.L.); (T.Z.)
| | - Guixiang Lv
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Harbin Medical University, Harbin 150086, China; (J.L.); (M.W.); (Y.W.); (X.P.); (G.L.); (T.Z.)
- Translational Medicine Center of Northern China, Harbin 150000, China
| | - Tianhu Zheng
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Harbin Medical University, Harbin 150086, China; (J.L.); (M.W.); (Y.W.); (X.P.); (G.L.); (T.Z.)
- Translational Medicine Center of Northern China, Harbin 150000, China
| | - Yahui Peng
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Harbin Medical University, Harbin 150086, China; (J.L.); (M.W.); (Y.W.); (X.P.); (G.L.); (T.Z.)
- Translational Medicine Center of Northern China, Harbin 150000, China
| | - Jihong Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Harbin Medical University, Harbin 150086, China; (J.L.); (M.W.); (Y.W.); (X.P.); (G.L.); (T.Z.)
- Translational Medicine Center of Northern China, Harbin 150000, China
| |
Collapse
|
8
|
Liu H, Chen J, Ling J, Wu Y, Yang P, Liu X, Liu J, Zhang D, Yin X, Yu P, Zhang J. The association between diabetes mellitus and postoperative cognitive dysfunction: a systematic review and meta-analysis. Int J Surg 2025; 111:2633-2650. [PMID: 39728730 DOI: 10.1097/js9.0000000000002156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 11/06/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND Postoperative cognitive dysfunction (POCD) is a typical consequence following surgery, particularly in cardiac surgeries. Despite its high incidence, the underlying etiology remains unclear. While diabetes mellitus (DM) has been associated with cognitive impairment, its specific function in POCD development remains unidentified. This study aims to evaluate the connection between DM and the risk of POCD. METHODS We conducted a comprehensive search of PubMed, Embase, Web of Science, and the Cochrane Library databases for studies of DM and risk with POCD, collecting data up to 14 September 2023. We assessed publication bias, heterogeneity, and study quality, adhering to PRISMA and AMSTAR guidelines. RESULTS Our study comprised 38 trials involving 8748 individuals, with 7734 patients undergoing follow-up. The pooled results showed that individuals with DM had an increased incidence of POCD compared to nondiabetic individuals (RR: 1.44, 95% CI: 1.26-1.65). The incidence of POCD was significantly higher in the group of patients with an average age older than 65 years (RR: 1.69, 95% CI: 1.30-2.20) compared with diabetic patients with an average age younger than 65 years (RR: 1.29, 95% CI: 1.09-1.64). Compared with diabetic patients undergoing cardiac surgery (RR: 1.33, 95% CI: 1.15-1.53), patients receiving non-cardiac surgery showed a greater incidence of POCD (RR: 2.01, 95% CI: 1.43-2.84). CONCLUSION Current evidence underscores that diabetic patients face a significantly higher risk of POCD compared to their nondiabetic counterparts. Further research is warranted to clarify the precise mechanisms of this relationship and explore potential preventive strategies for diabetic patients.
Collapse
Affiliation(s)
- Hongbo Liu
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, China
- The First Clinical Medical College, Jiangxi Medical College, Nanchang University, Jiangxi, Nanchang, China
| | - Jiali Chen
- The First Clinical Medical College, Jiangxi Medical College, Nanchang University, Jiangxi, Nanchang, China
| | - Jitao Ling
- School of Stomatology, Jiangxi Medical College, Nanchang University, Jiangxi, Nanchang, China
| | - Yuting Wu
- School of Stomatology, Jiangxi Medical College, Nanchang University, Jiangxi, Nanchang, China
| | - Pingping Yang
- School of Stomatology, Jiangxi Medical College, Nanchang University, Jiangxi, Nanchang, China
| | - Xiao Liu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, Nanchang, China
| | - Jianping Liu
- School of Stomatology, Jiangxi Medical College, Nanchang University, Jiangxi, Nanchang, China
| | - Deju Zhang
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Food and Nutritional Sciences, School of Biological Sciences, The University of Hong Kong, Hong Kong, China
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi, China
| | - Xiaoping Yin
- Food and Nutritional Sciences, School of Biological Sciences, The University of Hong Kong, Hong Kong, China
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi, China
| | - Peng Yu
- School of Stomatology, Jiangxi Medical College, Nanchang University, Jiangxi, Nanchang, China
| | - Jing Zhang
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| |
Collapse
|
9
|
Shehbaz A, Afzaal M, Akram N, Saeed F, Khan W, Ahmed F, Ahmed A, Asghar A, Faisal Z. Intermittent Fasting and Probiotics: Synergistic Modulation of Gut Health for Therapeutic Advantages. Probiotics Antimicrob Proteins 2025; 17:479-486. [PMID: 39261391 DOI: 10.1007/s12602-024-10358-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/29/2024] [Indexed: 09/13/2024]
Abstract
Intermittent fasting (IF) is an increasingly popular dietary approach involving alternating fasting and eating periods. This review aims to summarize the growing body of literature demonstrating that IF is a potential nutritional practice that involves alternating periods of fasting and eating and the numerous benefits of IF, especially in the modulation of healthy gut microbiota. The positive impact of intermittent fasting on gut microbiota not only promotes gastrointestinal health but also has far-reaching effects on critical systems throughout the body. Additionally, the evidence presented in this review highlights the significant preventive and therapeutic effects of intermittent fasting on a wide range of disorders. This includes reducing the risk of diabetes, and neurological disorders, alleviating obesity, and enhancing the functioning of the liver, ultimately contributing to the maintenance of metabolic equilibrium. Perhaps most notably, these effects play a substantial role in preventing diabetes, a global health concern of increasing significance. This comprehensive investigation delves into the scientific foundations of intermittent fasting's impact on gut microbiota and its implications for averting chronic diseases, providing valuable insights for future research and therapeutic applications.
Collapse
Affiliation(s)
- Amna Shehbaz
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan
| | - Muhammad Afzaal
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan.
| | - Noor Akram
- Food Safety & Biotechnology Lab, Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan
| | - Farhan Saeed
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan.
| | - Warda Khan
- Department of Nutritional Sciences, Government College University Faisalabad, Faisalabad, Pakistan
| | - Faiyaz Ahmed
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, P.O. Box 6666, 51452, Buraydah, Saudi Arabia
| | - Aftab Ahmed
- Department of Nutritional Sciences, Government College University Faisalabad, Faisalabad, Pakistan
| | - Aasma Asghar
- Department of Nutritional Sciences, Government College University Faisalabad, Faisalabad, Pakistan
| | - Zargham Faisal
- Department of Human Nutrition and Dietetics, Iqra University Karachi, Karachi, Pakistan
| |
Collapse
|
10
|
Knezovic A, Salkovic-Petrisic M. Cholinergic neurotransmission in the brain of streptozotocin-induced rat model of sporadic Alzheimer's disease: long-term follow up. J Neural Transm (Vienna) 2025:10.1007/s00702-025-02887-2. [PMID: 39891708 DOI: 10.1007/s00702-025-02887-2] [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: 12/17/2024] [Accepted: 01/22/2025] [Indexed: 02/03/2025]
Abstract
Rats treated intracerebroventricularly with streptozotocin (STZ-icv) develop pathologic features, which resemble those in Alzheimer's disease and have been proposed as a non-transgenic model for sporadic type of the disease (sAD). We aimed to characterize cholinergic transmission in the rat brain as a function of STZ-icv dose and time after the treatment. Acetylcholinesterase (AChE) activity and expression of muscarinic (M1, M4) and nicotinic (α7) receptors, cholin acetyltransferase (ChAT) and glial fibrillary acidic protein (GFAP) were measured in hippocampus (HPC) and parietotemporal cortex (CTX) of STZ-icv and age-matched control rats one week, and one, three, six and nine months after the icv administration of STZ (0.3, 1 and 3 mg/kg), respectively. Cholinergic and astroglial changes were found most pronounced with a highest STZ dose in time-dependent manner. The cortex and hippocampus exhibited specific alterations in cholinergic transmission following STZ-icv administration, with either similar or distinct patterns depending on the parameter observed: increased AChE activity in HPC and invariable in CTX; increased M4 and ChAT levels in both regions; substantial cortical M1 level increment and moderate hippocampal M1 decrement; and decreased α7 levels in both regions, with subsequent increase observed only in HPC. Alterations in cerebral cholinergic neurotransmission in STZ-icv rat model were mostly following a threephasic time pattern: acute response (Phase I), complete/partial compensation (Phase II), and reappearance/progression of changes (Phase III). Staging structure of cholinergic changes in STZ-icv rat model might be speculated to partly correlate with cholinergic pathology in clinical AD stages.
Collapse
Affiliation(s)
- Ana Knezovic
- Department of Pharmacology, University of Zagreb School of Medicine, Salata 11, Zagreb, 10 000, Croatia.
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Salata 12, Zagreb, 10 000, Croatia.
| | - Melita Salkovic-Petrisic
- Department of Pharmacology, University of Zagreb School of Medicine, Salata 11, Zagreb, 10 000, Croatia
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Salata 12, Zagreb, 10 000, Croatia
| |
Collapse
|
11
|
Szablewski L. Associations Between Diabetes Mellitus and Neurodegenerative Diseases. Int J Mol Sci 2025; 26:542. [PMID: 39859258 PMCID: PMC11765393 DOI: 10.3390/ijms26020542] [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: 10/31/2024] [Revised: 01/03/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
Diabetes mellitus (DM) and neurodegenerative diseases/disturbances are worldwide health problems. The most common chronic conditions diagnosed in persons 60 years and older are type 2 diabetes mellitus (T2DM) and cognitive impairment. It was found that diabetes mellitus is a major risk for cognitive decline, dementia, Parkinson's disease (PD), Alzheimer's disease (AD), Huntington's disease (HD), amyotrophic lateral sclerosis (ALS) and other neurodegenerative disorders. Different mechanisms of associations between these diseases and diabetes mellitus have been suggested. For example, it is postulated that an impaired intracellular insulin signaling pathway, together with hyperglycemia and hyperinsulinemia, may cause pathological changes, such as dysfunction of the mitochondria, oxidative stress inflammatory responses, etc. The association between diabetes mellitus and neurodegenerative diseases, as well as the mechanisms of these associations, needs further investigation. The aim of this review is to describe the associations between diabetes mellitus, especially type 1 (T1DM) and type 2 diabetes mellitus, and selected neurodegenerative diseases, i.e., Alzheimer's disease, Parkinson's disease, Huntington's disease and amyotrophic lateral sclerosis. Suggested mechanisms of these associations are also described.
Collapse
Affiliation(s)
- Leszek Szablewski
- Chair and Department of General Biology and Parasitology, Medical University of Warsaw, Chałubińskiego 5, 02-004 Warsaw, Poland
| |
Collapse
|
12
|
Yang Y, Shi G, Ge Y, Huang S, Cui N, Tan L, Liu R, Yang X. Accumulated BCAAs and BCKAs contribute to the HFD-induced deterioration of Alzheimer's disease via a dysfunctional TREM2-related reduction in microglial β-amyloid clearance. J Neuroinflammation 2024; 21:327. [PMID: 39716292 DOI: 10.1186/s12974-024-03314-1] [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: 08/08/2024] [Accepted: 11/27/2024] [Indexed: 12/25/2024] Open
Abstract
A high-fat diet (HFD) induces obesity and insulin resistance, which may exacerbate amyloid-β peptide (Aβ) pathology during Alzheimer's disease (AD) progression. Branched-chain amino acids (BCAAs) accumulate in obese or insulin-resistant patients and animal models. However, roles of accumulated BCAAs and their metabolites, branched-chain keto acids (BCKAs), in the HFD-induced deterioration of AD and the underlying mechanisms remains largely unclear. In this study, APPswe/PSEN1dE9 (APP/PS1) transgenic mice were fed a HFD for 6 months, and the BCAAs content of the HFD was adjusted to 200% or 50% to determine the effects of BCAAs. The HFD-fed APP/PS1 mice accumulated BCAAs and BCKAs in the serum and cortex, which was accompanied by more severe cognitive deficits and AD-related pathology. The additional or restricted intake of BCAAs aggravated or reversed these phenomena. Importantly, BCAAs and BCKAs repressed microglial phagocytosis of Aβ in vivo and in BV2 cells, which might be relevant for triggering receptor expressed on myeloid cells 2 (TREM2) dysfunction and autophagy deficiency. We found that BCAAs and BCKAs could bind to TREM2 in silico, in pure protein solutions and in the cellular environment. These molecules competed with Aβ for binding to TREM2 so that the response of TREM2 to Aβ was impaired. Moreover, BCAAs and BCKAs decreased TREM2 recycling in an mTOR-independent manner, which might also lead to TREM2 dysfunction. Our findings suggest that accumulated BCAAs and BCKAs contribute to the HFD-induced acceleration of AD progression through hypofunctional TREM2-mediated disturbances in Aβ clearance in microglia. Lowering BCAAs and BCKAs levels may become a potential dietary intervention for AD.
Collapse
Affiliation(s)
- Yang Yang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People's Republic of China
| | - Guanjin Shi
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People's Republic of China
| | - Yanyan Ge
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People's Republic of China
| | - Shanshan Huang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People's Republic of China
| | - Ningning Cui
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People's Republic of China
| | - Le Tan
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People's Republic of China
| | - Rui Liu
- Department of Public Health and Preventive Medicine, School of Medicine, Jianghan University, Wuhan, Hubei, 430010, People's Republic of China.
| | - Xuefeng Yang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People's Republic of China.
| |
Collapse
|
13
|
Khandayataray P, Murthy MK. Dietary interventions in mitigating the impact of environmental pollutants on Alzheimer's disease - A review. Neuroscience 2024; 563:148-166. [PMID: 39542342 DOI: 10.1016/j.neuroscience.2024.11.020] [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: 08/25/2024] [Revised: 10/23/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
Numerous studies linking environmental pollutants to oxidative stress, inflammation, and neurotoxicity have assigned pollutants to several neurodegenerative disorders, including Alzheimer's disease (AD). Heavy metals, pesticides, air pollutants, and endocrine disruptor chemicals have been shown to play important roles in AD development, with some traditional functions in amyloid-β formation, tau kinase action, and neuronal degeneration. However, pharmacological management and supplementation have resulted in limited improvement. This raises the interesting possibility that activities usually considered preventive, including diet, exercise, or mental activity, might be more similar to treatment or therapy for AD. This review focuses on the effects of diet on the effects of environmental pollutants on AD. One of the primary issues addressed in this review is a group of specific diets, including the Mediterranean diet (MeDi), Dietary Approaches to Stop Hypertension (DASH), and Mediterranean-DASH intervention for Neurodegenerative Delay (MIND), which prevent exposure to these toxins. Such diets have been proven to decrease oxidative stress and inflammation, which are unfavorable for neuronal growth. Furthermore, they contribute to positive changes in the composition of the human gut microbiota and thus encourage interactions in the Gut-Brain Axis, reducing inflammation caused by pollutants. This review emphasizes a multi-professional approach with reference to nutritional activities that would lower the neurotoxic load in populations with a high level of exposure to pollutants. Future studies focusing on diet and environment association plans may help identify preventive measures aimed at enhancing current disease deceleration.
Collapse
Affiliation(s)
- Pratima Khandayataray
- Department of Biotechnology, Academy of Management and Information Technology, Utkal University, Bhubaneswar, Odisha 752057, India
| | - Meesala Krishna Murthy
- Department of Allied Health Sciences, Chitkara School of Health Sciences, Chitkara University, Rajpura, Punjab 140401, India.
| |
Collapse
|
14
|
Medina-Vera D, López-Gambero AJ, Verheul-Campos J, Navarro JA, Morelli L, Galeano P, Suárez J, Sanjuan C, Pacheco-Sánchez B, Rivera P, Pavon-Morón FJ, Rosell-Valle C, Fonseca FRD. Therapeutic Efficacy of the Inositol D-Pinitol as a Multi-Faceted Disease Modifier in the 5×FAD Humanized Mouse Model of Alzheimer's Amyloidosis. Nutrients 2024; 16:4186. [PMID: 39683582 DOI: 10.3390/nu16234186] [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: 11/06/2024] [Revised: 11/27/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND/OBJECTIVES Alzheimer's disease (AD), a leading cause of dementia, lacks effective long-term treatments. Current therapies offer temporary relief or fail to halt its progression and are often inaccessible due to cost. AD involves multiple pathological processes, including amyloid beta (Aβ) deposition, insulin resistance, tau protein hyperphosphorylation, and systemic inflammation accelerated by gut microbiota dysbiosis originating from a leaky gut. Given this context, exploring alternative therapeutic interventions capable of addressing the multifaceted components of AD etiology is essential. METHODS This study suggests D-Pinitol (DPIN) as a potential treatment modifier for AD. DPIN, derived from carob pods, demonstrates insulin-sensitizing, tau hyperphosphorylation inhibition, and antioxidant properties. To test this hypothesis, we studied whether chronic oral administration of DPIN (200 mg/kg/day) could reverse the AD-like disease progression in the 5×FAD mice. RESULTS Results showed that treatment of 5×FAD mice with DPIN improved cognition, reduced hippocampal Aβ and hyperphosphorylated tau levels, increased insulin-degrading enzyme (IDE) expression, enhanced pro-cognitive hormone circulation (such as ghrelin and leptin), and normalized the PI3K/Akt insulin pathway. This enhancement may be mediated through the modulation of cyclin-dependent kinase 5 (CDK5). DPIN also protected the gut barrier and microbiota, reducing the pro-inflammatory impact of the leaky gut observed in 5×FAD mice. DPIN reduced bacterial lipopolysaccharide (LPS) and LPS-associated inflammation, as well as restored intestinal proteins such as Claudin-3. This effect was associated with a modulation of gut microbiota towards a more balanced bacterial composition. CONCLUSIONS These findings underscore DPIN's promise in mitigating cognitive decline in the early AD stages, positioning it as a potential disease modifier.
Collapse
Affiliation(s)
- Dina Medina-Vera
- Grupo de Neuropsicofarmacología, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Unidades Clínicas de Neurología y Salud Mental, 29010 Málaga, Spain
- Facultad de Ciencias, Universidad de Málaga, 29010 Málaga, Spain
- Unidad de Gestión Clínica del Corazón-CIBERCV (Enfermedades Cardiovasculares), Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain
| | - Antonio J López-Gambero
- Grupo de Neuropsicofarmacología, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Unidades Clínicas de Neurología y Salud Mental, 29010 Málaga, Spain
- INSERM, Neurocentre Magendie, University of Bordeaux, 33000 Bordeaux, France
| | - Julia Verheul-Campos
- Grupo de Neuropsicofarmacología, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Unidades Clínicas de Neurología y Salud Mental, 29010 Málaga, Spain
| | - Juan A Navarro
- Grupo de Neuropsicofarmacología, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Unidades Clínicas de Neurología y Salud Mental, 29010 Málaga, Spain
- Facultad de Medicina, Universidad de Málaga, 29010 Málaga, Spain
| | - Laura Morelli
- Laboratory of Brain Aging and Neurodegeneration, Fundación Instituto Leloir (IIBBA-CONICET), Av. Patricias Argentinas 435, Ciudad Autónoma de Buenos Aires C1405BWE, Argentina
| | - Pablo Galeano
- Laboratory of Brain Aging and Neurodegeneration, Fundación Instituto Leloir (IIBBA-CONICET), Av. Patricias Argentinas 435, Ciudad Autónoma de Buenos Aires C1405BWE, Argentina
| | - Juan Suárez
- Grupo de Neuropsicofarmacología, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Unidades Clínicas de Neurología y Salud Mental, 29010 Málaga, Spain
- Departamento de Anatomía Humana, Medicina Legal e Historia de la Ciencia, Facultad de Medicina, Universidad de Málaga, 29071 Málaga, Spain
- Andalusian Network for Clinical and Translational Research in Neurology [NEURO-RECA], 29001 Málaga, Spain
| | - Carlos Sanjuan
- Euronutra S.L. Calle Johannes Kepler, 3, 29590 Málaga, Spain
| | - Beatriz Pacheco-Sánchez
- Grupo de Neuropsicofarmacología, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Unidades Clínicas de Neurología y Salud Mental, 29010 Málaga, Spain
| | - Patricia Rivera
- Grupo de Neuropsicofarmacología, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Unidades Clínicas de Neurología y Salud Mental, 29010 Málaga, Spain
| | - Francisco J Pavon-Morón
- Grupo de Neuropsicofarmacología, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Unidades Clínicas de Neurología y Salud Mental, 29010 Málaga, Spain
- Unidad de Gestión Clínica del Corazón-CIBERCV (Enfermedades Cardiovasculares), Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain
| | - Cristina Rosell-Valle
- Grupo de Neuropsicofarmacología, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Unidades Clínicas de Neurología y Salud Mental, 29010 Málaga, Spain
| | - Fernando Rodríguez de Fonseca
- Grupo de Neuropsicofarmacología, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Unidades Clínicas de Neurología y Salud Mental, 29010 Málaga, Spain
- Andalusian Network for Clinical and Translational Research in Neurology [NEURO-RECA], 29001 Málaga, Spain
| |
Collapse
|
15
|
Titisari N, Fauzi A, Abdul Razak IS, Mohd Noor MH, Samsulrizal N, Ahmad H. Dietary menhaden fish oil supplementation suppresses lipopolysaccharide-induced neuroinflammation and cognitive impairment in diabetic rats. PHARMACEUTICAL BIOLOGY 2024; 62:447-455. [PMID: 38753370 PMCID: PMC11100436 DOI: 10.1080/13880209.2024.2351933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 04/28/2024] [Indexed: 05/18/2024]
Abstract
CONTEXT Menhaden fish oil (FO) is widely recognized for inhibiting neuroinflammatory responses and preserving brain function. Nevertheless, the mechanisms of FO influencing brain cognitive function in diabetic states remain unclear. OBJECTIVE This study examines the potential role of FO in suppressing LPS-induced neuroinflammation and cognitive impairment in diabetic animals (DA). MATERIALS AND METHODS Thirty male Wistar rats were divided into 5 groups: i) DA received LPS induction (DA-LPS); ii) DA received LPS induction and 1 g/kg FO (DA-LPS-1FO); iii) DA received LPS induction and 3 g/kg FO (DA-LPS-3FO); iv) animals received normal saline and 3 g/kg FO (NS-3FO) and v) control animals received normal saline (CTRL). Y-maze test was used to measure cognitive performance, while brain samples were collected for inflammatory markers and morphological analysis. RESULTS DA received LPS induction, and 1 or 3 g/kg FO significantly inhibited hyperglycaemia and brain inflammation, as evidenced by lowered levels of pro-inflammatory mediators. Additionally, both DA-LPS-1FO and DA-LPS-3FO groups exhibited a notable reduction in neuronal damage and glial cell migration compared to the other groups. These results were correlated with the increasing number of entries and time spent in the novel arm of the Y-maze test. DISCUSSION AND CONCLUSION This study indicates that supplementation of menhaden FO inhibits the LPS signaling pathway and protects against neuroinflammation, consequently maintaining cognitive performance in diabetic animals. Thus, the current study suggested that fish oil may be effective as a supporting therapy option for diabetes to avoid diabetes-cognitive impairment.
Collapse
Affiliation(s)
- Nurina Titisari
- Department of Veterinary Preclinical Sciences, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Selangor, Malaysia
- Department of Veterinary Physiology, Faculty of Veterinary Medicine, Universitas Brawijaya, East Java, Indonesia
| | - Ahmad Fauzi
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Universitas Brawijaya, East Java, Indonesia
| | - Intan Shameha Abdul Razak
- Department of Veterinary Preclinical Sciences, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Selangor, Malaysia
| | - Mohd Hezmee Mohd Noor
- Department of Veterinary Preclinical Sciences, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Selangor, Malaysia
| | | | - Hafandi Ahmad
- Department of Veterinary Preclinical Sciences, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Selangor, Malaysia
- Institute of Tropical Agriculture and Food Security, Universiti Putra Malaysia, Selangor, Malaysia
| |
Collapse
|
16
|
Xiao Y, Fan Y, Feng Z. A meta-analysis of the efficacy of physical exercise interventions on activities of daily living in patients with Alzheimer's disease. Front Public Health 2024; 12:1485807. [PMID: 39664530 PMCID: PMC11631704 DOI: 10.3389/fpubh.2024.1485807] [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: 08/24/2024] [Accepted: 11/05/2024] [Indexed: 12/13/2024] Open
Abstract
Objective This study aimed to systematically review published randomized controlled trials on the effects of physical exercise on activities of daily living (ADL) in Alzheimer's patients through meta-analysis, thereby synthesizing existing evidence to provide scientific intervention recommendations for exercise prescriptions in Alzheimer's patients. Methods Based on strict literature inclusion and exclusion criteria, a systematic search was conducted in databases including PubMed and Web of Science from their inception to July 1, 2024. The Cochrane risk assessment tool was used to evaluate the design of randomized controlled trials. Studies reporting on physical exercise interventions for ADL in Alzheimer's patients were systematically identified. Subgroup analyses and meta-regression were performed to explore sources of heterogeneity. Results Nineteen articles, for analysis, providing 27 randomized controlled trials (RCTs). A random-effects model was used to calculate the effect size and 95% confidence interval for each independent study, and meta-analysis was performed using Stata 16.0 and RevMan 5.4 software. The results showed that physical exercise might significantly improve ADL in Alzheimer's patients (SMD = 0.33, 95% CI: 0.12-0.54, I 2 = 81.7%). Sensitivity analysis confirmed the robustness of the results (p > 0.05). Egger's test did not reveal significant publication bias (p = 0.145). Samples were divided into different subgroups based on intervention content, duration, frequency, and session length. Subgroup analysis based on intervention characteristics showed that resistance training or aerobic exercise (SMD = 0.83, 95% CI: 0.60-1.05), long-term interventions (>6 months, SMD = 0.31, 95% CI: 0.13-0.49), medium-frequency interventions (4-5 times per week, SMD=0.39, 95% CI: 0.23-0.55), and short-duration training ( ≤ 30 min, SMD = 0.96, 95% CI: 0.71-1.21) might be most effective in enhancing ADL in Alzheimer's patients. These improvements were not only statistically significant but also had substantial impact in clinical practice. Conclusion Resistance training or aerobic exercise lasting more than 6 months, 4-5 times per week, and lasting no more than 30 min per session may be most effective in improving ADLs in patients with Alzheimer's disease.
Collapse
Affiliation(s)
- Yang Xiao
- College of Physical Education and Sports, Beijing Normal University, Beijing, China
| | - Yu Fan
- Department of Physical Education, Nanjing University of Science and Technology, Nanjing, China
| | - Zhengteng Feng
- China Athletics College, Beijing Sport University, Beijing, China
| |
Collapse
|
17
|
Mani V, Arfeen M. In Vivo and Computational Studies on Sitagliptin's Neuroprotective Role in Type 2 Diabetes Mellitus: Implications for Alzheimer's Disease. Brain Sci 2024; 14:1191. [PMID: 39766390 PMCID: PMC11674309 DOI: 10.3390/brainsci14121191] [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: 11/02/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND/OBJECTIVES Diabetes mellitus (DM), a widespread endocrine disorder characterized by chronic hyperglycemia, can cause nerve damage and increase the risk of neurodegenerative diseases such as Alzheimer's disease (AD). Effective blood glucose management is essential, and sitagliptin (SITG), a dipeptidyl peptidase-4 (DPP-4) inhibitor, may offer neuroprotective benefits in type 2 diabetes mellitus (T2DM). METHODS T2DM was induced in rats using nicotinamide (NICO) and streptozotocin (STZ), and biomarkers of AD and DM-linked enzymes, inflammation, oxidative stress, and apoptosis were evaluated in the brain. Computational studies supported the in vivo findings. RESULTS SITG significantly reduced the brain enzyme levels of acetylcholinesterase (AChE), beta-secretase-1 (BACE-1), DPP-4, and glycogen synthase kinase-3β (GSK-3β) in T2DM-induced rats. It also reduced inflammation by lowering cyclooxygenase-2 (COX-2), prostaglandin E2 (PGE2), tumor necrosis factor-α (TNF-α), and nuclear factor-κB (NF-κB). Additionally, SITG improved oxidative stress markers by reducing malondialdehyde (MDA) and enhancing glutathione (GSH). It increased anti-apoptotic B-cell lymphoma protein-2 (Bcl-2) while reducing pro-apoptotic markers such as Bcl-2-associated X (BAX) and Caspace-3. SITG also lowered blood glucose levels and improved plasma insulin levels. To explore potential molecular level mechanisms, docking was performed on AChE, COX-2, GSK-3β, BACE-1, and Caspace-3. The potential binding affinity of SITG for the above-mentioned target enzymes were 10.8, 8.0, 9.7, 7.7, and 7.9 kcal/mol, respectively, comparable to co-crystallized ligands. Further binding mode analysis of the lowest energy conformation revealed interactions with the critical residues. CONCLUSIONS These findings highlight SITG's neuroprotective molecular targets in T2DM-associated neurodegeneration and its potential as a therapeutic approach for AD, warranting further clinical investigations.
Collapse
Affiliation(s)
- Vasudevan Mani
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah 51452, Saudi Arabia
| | - Minhajul Arfeen
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraydah 51452, Saudi Arabia;
| |
Collapse
|
18
|
Sanz-Martos AB, Roca M, Plaza A, Merino B, Ruiz-Gayo M, Olmo ND. Long-term saturated fat-enriched diets impair hippocampal learning and memory processes in a sex-dependent manner. Neuropharmacology 2024; 259:110108. [PMID: 39128582 DOI: 10.1016/j.neuropharm.2024.110108] [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: 05/28/2024] [Revised: 07/29/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
Consumption of saturated fat-enriched diets during adolescence has been closely associated with the reduction of hippocampal synaptic plasticity and the impairment of cognitive function. Nevertheless, the effect of long-term intake of these foods has not yet been studied. In the present study, we have investigated the effect of a treatment, lasting for 40 weeks, with a diet enriched in saturated fat (SOLF) on i) spatial learning and memory, ii) hippocampal synaptic transmission and plasticity, and iii) hippocampal gene expression levels in aged male and female mice. Our findings reveal that SOLF has a detrimental impact on spatial memory and synaptic plasticity mechanisms, such as long-term potentiation (LTP), and downregulates Gria1 expression specifically in males. In females, SOLF downregulates the gene expression of Gria1/2/3 and Grin1/2A/2B glutamate receptor subunits as well as some proinflammatory interleukins. These findings highlight the importance of considering sex-specific factors when assessing the long-term effects of high-fat diets on cognition and brain plasticity.
Collapse
Affiliation(s)
- Ana Belén Sanz-Martos
- Department of Psychobiology, School of Psychology, UNED, C/ Juan del Rosal 10, 28040, Madrid, Spain.
| | - María Roca
- Department of Psychobiology, School of Psychology, UNED, C/ Juan del Rosal 10, 28040, Madrid, Spain
| | - Adrián Plaza
- Department of Health and Pharmaceutical Sciences, School of Pharmacy, Universidad CEU-San Pablo, CEU Universities, 28668, Madrid, Spain
| | - Beatriz Merino
- Department of Health and Pharmaceutical Sciences, School of Pharmacy, Universidad CEU-San Pablo, CEU Universities, 28668, Madrid, Spain
| | - Mariano Ruiz-Gayo
- Department of Health and Pharmaceutical Sciences, School of Pharmacy, Universidad CEU-San Pablo, CEU Universities, 28668, Madrid, Spain
| | - Nuria Del Olmo
- Department of Psychobiology, School of Psychology, UNED, C/ Juan del Rosal 10, 28040, Madrid, Spain
| |
Collapse
|
19
|
Ali M, Gupta A, Verma RD, Akhtar S, Ghosh JK. A peptide derived from the amino terminus of leptin improves glucose metabolism and energy homeostasis in myotubes and db/db mice. J Biol Chem 2024; 300:107919. [PMID: 39490585 PMCID: PMC11625344 DOI: 10.1016/j.jbc.2024.107919] [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: 05/11/2024] [Revised: 10/10/2024] [Accepted: 10/14/2024] [Indexed: 11/05/2024] Open
Abstract
Leptin is an adipokine, which plays key roles in regulation of glucose metabolism and energy homeostasis. Therefore, identification of a short peptide from leptin which improves glucose-metabolism and energy-homeostasis could be of significant therapeutic importance. Mutational studies demonstrated that N-terminal of human leptin hormone is crucial for activation of leptin-receptor while its C-terminal seems to have lesser effects in it. Thus, for finding a metabolically active peptide and complimenting the mutational studies on leptin, we have identified a 17-mer (leptin-1) and a 16-mer (leptin-2) segment from its N-terminal and C-terminal, respectively. Consistent with the mutational studies, leptin-1 improved glucose-metabolism by increasing glucose-uptake, GLUT4 expression and its translocation to the plasma membrane in L6-myotubes, while leptin-2 was mostly inactive. Leptin-1-induced glucose-uptake is mediated through activation of AMPK, PI3K, and AKT proteins since inhibitors of these proteins inhibited the event. Leptin-1 activated leptin-receptor immediate downstream target protein, JAK2 reflecting its possible interaction with leptin-receptor while leptin-2 was less active. Furthermore, leptin-1 increased mitochondrial-biogenesis and ATP-production, and increased expression of PGC1α, NRF1, and Tfam proteins, that are important regulators of mitochondrial biogenesis. The results suggested that leptin-1 improved energy-homeostasis in L6-myotubes, whereas, leptin-2 showed much lesser effects. In diabetic, db/db mice, leptin-1 significantly decreased blood glucose level and improved glucose-tolerance. Leptin-1 also increased serum adiponectin and decreased serum TNF-α and IL-6 level signifying the improvement in insulin-sensitivity and decrease in insulin-resistance, respectively in db/db mice. Overall, the results show the identification of a short peptide from the N-terminal of human leptin hormone which significantly improves glucose-metabolism and energy-homeostasis.
Collapse
Affiliation(s)
- Mehmood Ali
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Arvind Gupta
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Rahul Dev Verma
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Sariyah Akhtar
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Jimut Kanti Ghosh
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.
| |
Collapse
|
20
|
Oumeddour DZ, Al-Dalali S, Zhao L, Zhao L, Wang C. Recent advances on cyanidin-3-O-glucoside in preventing obesity-related metabolic disorders: A comprehensive review. Biochem Biophys Res Commun 2024; 729:150344. [PMID: 38976946 DOI: 10.1016/j.bbrc.2024.150344] [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: 05/16/2024] [Revised: 06/25/2024] [Accepted: 07/02/2024] [Indexed: 07/10/2024]
Abstract
Anthocyanins, found in various pigmented plants as secondary metabolites, represent a class of dietary polyphenols known for their bioactive properties, demonstrating health-promoting effects against several chronic diseases. Among these, cyanidin-3-O-glucoside (C3G) is one of the most prevalent types of anthocyanins. Upon consumption, C3G undergoes phases I and II metabolism by oral epithelial cells, absorption in the gastric epithelium, and gut transformation (phase II & microbial metabolism), with limited amounts reaching the bloodstream. Obesity, characterized by excessive body fat accumulation, is a global health concern associated with heightened risks of disability, illness, and mortality. This comprehensive review delves into the biodegradation and absorption dynamics of C3G within the gastrointestinal tract. It meticulously examines the latest research findings, drawn from in vitro and in vivo models, presenting evidence underlining C3G's bioactivity. Notably, C3G has demonstrated significant efficacy in combating obesity, by regulating lipid metabolism, specifically decreasing lipid synthesis, increasing fatty acid oxidation, and reducing lipid accumulation. Additionally, C3G enhances energy homeostasis by boosting energy expenditure, promoting the activity of brown adipose tissue, and stimulating mitochondrial biogenesis. Furthermore, C3G shows potential in managing various prevalent obesity-related conditions. These include cardiovascular diseases (CVD) and hypertension through the suppression of reactive oxygen species (ROS) production, enhancement of endogenous antioxidant enzyme levels, and inhibition of the nuclear factor-kappa B (NF-κB) signaling pathway and by exercising its cardioprotective and vascular effects by decreasing pulmonary artery thickness and systolic pressure which enhances vascular relaxation and angiogenesis. Type 2 diabetes mellitus (T2DM) and insulin resistance (IR) are also managed by reducing gluconeogenesis via AMPK pathway activation, promoting autophagy, protecting pancreatic β-cells from oxidative stress and enhancing glucose-stimulated insulin secretion. Additionally, C3G improves insulin sensitivity by upregulating GLUT-1 and GLUT-4 expression and regulating the PI3K/Akt pathway. C3G exhibits anti-inflammatory properties by inhibiting the NF-κB pathway, reducing pro-inflammatory cytokines, and shifting macrophage polarization from the pro-inflammatory M1 phenotype to the anti-inflammatory M2 phenotype. C3G demonstrates antioxidative effects by enhancing the expression of antioxidant enzymes, reducing ROS production, and activating the Nrf2/AMPK signaling pathway. Moreover, these mechanisms also contribute to attenuating inflammatory bowel disease and regulating gut microbiota by decreasing Firmicutes and increasing Bacteroidetes abundance, restoring colon length, and reducing levels of inflammatory cytokines. The therapeutic potential of C3G extends beyond metabolic disorders; it has also been found effective in managing specific cancer types and neurodegenerative disorders. The findings of this research can provide an important reference for future investigations that seek to improve human health through the use of naturally occurring bioactive compounds.
Collapse
Affiliation(s)
- Dounya Zad Oumeddour
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Ministry of Education, Beijing, 100048, China; Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing, 100048, China.
| | - Sam Al-Dalali
- School of Food and Health, Guilin Tourism University, Guilin, 541006, China; Department of Food Science and Technology, Faculty of Agriculture and Food Science, Ibb University, Ibb, 70270, Yemen.
| | - Liang Zhao
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Ministry of Education, Beijing, 100048, China; Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing, 100048, China.
| | - Lei Zhao
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Ministry of Education, Beijing, 100048, China; Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing, 100048, China.
| | - Chengtao Wang
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Ministry of Education, Beijing, 100048, China; Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing, 100048, China.
| |
Collapse
|
21
|
Tran J, Parekh S, Rockcole J, Wilson D, Parmar MS. Repurposing antidiabetic drugs for Alzheimer's disease: A review of preclinical and clinical evidence and overcoming challenges. Life Sci 2024; 355:123001. [PMID: 39173996 DOI: 10.1016/j.lfs.2024.123001] [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: 07/05/2024] [Revised: 08/13/2024] [Accepted: 08/19/2024] [Indexed: 08/24/2024]
Abstract
Repurposing antidiabetic drugs for the treatment of Alzheimer's disease (AD) has emerged as a promising therapeutic strategy. This review examines the potential of repurposing antidiabetic drugs for AD treatment, focusing on preclinical evidence, clinical trials, and observational studies. In addition, the review aims to explore challenges and opportunities in repurposing antidiabetic drugs for AD, emphasizing the importance of well-designed clinical trials that consider patient selection criteria, refined outcome measures, adverse effects, and combination therapies to enhance therapeutic efficacy. Preclinical evidence suggests that glucagon-like peptide-1 (GLP-1) analogs, dipeptidyl peptidase-4 (DPP4) inhibitors, metformin, thiazolidinediones, and sodium-glucose co-transporter-2 (SGLT2) inhibitors exhibit neuroprotective effects in AD preclinical models. In preclinical studies, antidiabetic drugs have demonstrated neuroprotective effects by reducing amyloid beta (Aβ) plaques, tau hyperphosphorylation, neuroinflammation, and cognitive impairment. Antidiabetic drug classes, notably GLP-1 analogs and SGLT2 inhibitors, and a reduced risk of dementia in patients with diabetes mellitus. While the evidence for DPP4 inhibitors is mixed, some studies suggest a potential protective effect. On the other hand, alpha-glucosidase inhibitors (AGIs) and sulfonylureas may potentially increase the risk, especially in those experiencing recurrent hypoglycemic events. Repurposing antidiabetic drugs for AD is a promising therapeutic strategy, but challenges such as disease heterogeneity, limited biomarkers, and benefits versus risk evaluation need to be addressed. Ongoing clinical trials in mild cognitive impairment (MCI) and early AD patients without diabetes will be crucial in determining the clinical efficacy and safety of the antidiabetic drugs, paving the way for potential treatments for AD.
Collapse
Affiliation(s)
- Jacky Tran
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Clearwater, FL, USA
| | - Sneh Parekh
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Clearwater, FL, USA
| | - Julia Rockcole
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Clearwater, FL, USA
| | - Danielle Wilson
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Clearwater, FL, USA
| | - Mayur S Parmar
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Clearwater, FL, USA.
| |
Collapse
|
22
|
Rhea EM, Babin A, Thomas P, Omer M, Weaver R, Hansen K, Banks WA, Talbot K. Brain uptake pharmacokinetics of albiglutide, dulaglutide, tirzepatide, and DA5-CH in the search for new treatments of Alzheimer's and Parkinson's diseases. Tissue Barriers 2024; 12:2292461. [PMID: 38095516 PMCID: PMC11583597 DOI: 10.1080/21688370.2023.2292461] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/29/2023] [Accepted: 12/02/2023] [Indexed: 06/29/2024] Open
Abstract
BACKGROUND A number of peptide incretin receptor agonists (IRAs) show promise as therapeutics for Alzheimer's disease (AD) and Parkinson's disease (PD). Transport across the blood-brain barrier (BBB) is one way for IRAs to act directly within the brain. To determine which IRAs are high priority candidates for treating these disorders, we have studied their brain uptake pharmacokinetics. METHODS We quantitatively measure the ability of four IRAs to cross the BBB. We injected adult male CD-1 mice intravenously with 125I- or 14C-labeled albiglutide, dulaglutide, DA5-CH, or tirzepatide and used multiple-time regression analyses to measure brain kinetics up to 1 hour. For those IRAs failing to enter the brain 1 h after intravenous injection, we also investigated their ability to enter over a longer time frame (i.e., 6 h). RESULTS Albiglutide and dulaglutide had the fastest brain uptake rates within 1 hour. DA5-CH appears to enter the brain rapidly, reaching equilibrium quickly. Tirzepatide does not appear to cross the BBB within 1 h after iv injection but like albumin, did so slowly over 6 h, presumably via the extracellular pathways. CONCLUSIONS We find that IRAs can cross the BBB by two separate processes; one that is fast and one that is slow. Three of the four IRAs investigated here have fast rates of transport and should be taken into consideration for testing as AD and PD therapeutics as they would have the ability to act quickly and directly on the brain as a whole.
Collapse
Affiliation(s)
- Elizabeth M Rhea
- Veterans Affairs Puget Sound Health Care System, Geriatrics Research Education and Clinical Center, Seattle, WA, USA
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Alice Babin
- Veterans Affairs Puget Sound Health Care System, Geriatrics Research Education and Clinical Center, Seattle, WA, USA
| | - Peter Thomas
- Veterans Affairs Puget Sound Health Care System, Geriatrics Research Education and Clinical Center, Seattle, WA, USA
| | - Mohamed Omer
- Veterans Affairs Puget Sound Health Care System, Geriatrics Research Education and Clinical Center, Seattle, WA, USA
| | - Riley Weaver
- Veterans Affairs Puget Sound Health Care System, Geriatrics Research Education and Clinical Center, Seattle, WA, USA
| | - Kim Hansen
- Veterans Affairs Puget Sound Health Care System, Geriatrics Research Education and Clinical Center, Seattle, WA, USA
| | - William A Banks
- Veterans Affairs Puget Sound Health Care System, Geriatrics Research Education and Clinical Center, Seattle, WA, USA
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Konrad Talbot
- Departments of Neurosurgery, Pathology and Human Anatomy, and Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA
| |
Collapse
|
23
|
Sulangi AJ, Lyons SE, Abdou AA, Patel H, Nagliya D, Joseph E, Joseph C, Kumar D, Patel S, Jinwala I, Parmar MS. Exploring the Therapeutic Potential of DPP4 Inhibitors in Alzheimer's Disease: Molecular Insight and Clinical Outcome. Cureus 2024; 16:e72648. [PMID: 39610591 PMCID: PMC11604213 DOI: 10.7759/cureus.72648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 10/22/2024] [Indexed: 11/30/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by amyloid-beta (Aβ) plaques, neurofibrillary tangles, and cognitive decline. Given the shared neuropathophysiological traits between AD and type 2 diabetes mellitus (T2DM), repurposing antidiabetic medications, such as dipeptidyl peptidase 4 inhibitors (DPP4i), has emerged as a promising therapeutic strategy. This review comprehensively evaluates the preclinical and clinical evidence supporting the potential of DPP4i in preventing or treating AD by modulating Aβ and tau pathology, improving cognitive function, reducing neuroinflammation and oxidative stress, and promoting neuronal survival. The beneficial effects of DPP4i are likely mediated through the modulation of insulin signaling, anti-inflammatory and antioxidant properties, glucagon-like peptide-1 (GLP-1) upregulation, and modulation of the amyloidogenic pathway. While further research is needed to establish their clinical efficacy in AD patients, DPP4i offers a promising avenue for therapeutic intervention for this devastating neurodegenerative disease.
Collapse
Affiliation(s)
- Albert Joseph Sulangi
- Osteopathic Medicine, Nova Southeastern University Dr. Kiran C. Patel College of Osteopathic Medicine, Clearwater, USA
| | - Sarah E Lyons
- Osteopathic Medicine, Nova Southeastern University Dr. Kiran C. Patel College of Osteopathic Medicine, Fort Lauderdale, USA
| | - Amy A Abdou
- Osteopathic Medicine, Nova Southeastern University Dr. Kiran C. Patel College of Osteopathic Medicine, Fort Lauderdale, USA
| | - Hemangi Patel
- Osteopathic Medicine, Nova Southeastern University Dr. Kiran C. Patel College of Osteopathic Medicine, Fort Lauderdale, USA
| | - Deepika Nagliya
- Osteopathic Medicine, Nova Southeastern University Dr. Kiran C. Patel College of Osteopathic Medicine, Fort Lauderdale, USA
| | - Eileen Joseph
- Osteopathic Medicine, Nova Southeastern University Dr. Kiran C. Patel College of Osteopathic Medicine, Fort Lauderdale, USA
| | - Carmel Joseph
- Osteopathic Medicine, Nova Southeastern University Dr. Kiran C. Patel College of Osteopathic Medicine, Fort Lauderdale, USA
| | - Divya Kumar
- Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, USA
| | - Shivani Patel
- Osteopathic Medicine, Nova Southeastern University Dr. Kiran C. Patel College of Osteopathic Medicine, Fort Lauderdale, USA
| | - Isha Jinwala
- Osteopathic Medicine, Nova Southeastern University Dr. Kiran C. Patel College of Osteopathic Medicine, Fort Lauderdale, USA
| | - Mayur S Parmar
- Foundational Sciences, Nova Southeastern University Dr. Kiran C. Patel College of Osteopathic Medicine, Clearwater, USA
| |
Collapse
|
24
|
Vashishth S, Ambasta RK, Kumar P. Deciphering the microbial map and its implications in the therapeutics of neurodegenerative disorder. Ageing Res Rev 2024; 100:102466. [PMID: 39197710 DOI: 10.1016/j.arr.2024.102466] [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: 07/31/2024] [Revised: 08/19/2024] [Accepted: 08/19/2024] [Indexed: 09/01/2024]
Abstract
Every facet of biological anthropology, including development, ageing, diseases, and even health maintenance, is influenced by gut microbiota's significant genetic and metabolic capabilities. With current advancements in sequencing technology and with new culture-independent approaches, researchers can surpass older correlative studies and develop mechanism-based studies on microbiome-host interactions. The microbiota-gut-brain axis (MGBA) regulates glial functioning, making it a possible target for the improvement of development and advancement of treatments for neurodegenerative diseases (NDDs). The gut-brain axis (GBA) is accountable for the reciprocal communication between the gastrointestinal and central nervous system, which plays an essential role in the regulation of physiological processes like controlling hunger, metabolism, and various gastrointestinal functions. Lately, studies have discovered the function of the gut microbiome for brain health-different microbiota through different pathways such as immunological, neurological and metabolic pathways. Additionally, we review the involvement of the neurotransmitters and the gut hormones related to gut microbiota. We also explore the MGBA in neurodegenerative disorders by focusing on metabolites. Further, targeting the blood-brain barrier (BBB), intestinal barrier, meninges, and peripheral immune system is investigated. Lastly, we discuss the therapeutics approach and evaluate the pre-clinical and clinical trial data regarding using prebiotics, probiotics, paraprobiotics, fecal microbiota transplantation, personalised medicine, and natural food bioactive in NDDs. A comprehensive study of the GBA will felicitate the creation of efficient therapeutic approaches for treating different NDDs.
Collapse
Affiliation(s)
- Shrutikirti Vashishth
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi 110042, India
| | - Rashmi K Ambasta
- Department of Medicine, School of Medicine, VUMC, Vanderbilt University, TN, USA
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi 110042, India.
| |
Collapse
|
25
|
Pandit M, Frishman WH. The Association Between Cardiovascular Disease and Dementia: A Review of Trends in Epidemiology, Risk Factors, Pathophysiologic Mechanisms, and Clinical Implications. Cardiol Rev 2024; 32:463-467. [PMID: 36946920 DOI: 10.1097/crd.0000000000000523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
With increases in life expectancy and the size of the aging population, cognitive decline and neurodegenerative pathologies are expected to increase in the next few decades. Age-related increases in risk for dementia and cardiovascular disease have been researched widely. Epidemiology trends reveal a predicted increase of neurodegenerative disease to more than 65 million by 2030 in the United States. There are several risk factors for the development of cardiovascular disease that have been widely studied for their impact on dementia; such as: diabetes, hypertension, and hyperlipidemia. Several pathophysiologic mechanisms exist by which cardiovascular disease could impact dementia including cerebral hypoperfusion, reactive oxidative species, and increased cleavage of amyloid precursor protein into amyloid beta plaques and accumulation of neurofibrillary tangles. Emerging evidence also suggests that treatment of cardiovascular disease risk factors could reduce the risk of dementia development. In this review, we seek to examine the relationship between cardiovascular disease and dementia by examining epidemiologic trends, common risk factors, pathophysiologic mechanisms and implications for clinical management.
Collapse
Affiliation(s)
- Maya Pandit
- From the New York Medical College, Valhalla, NY
| | - William H Frishman
- Department of Medicine, New York Medical College/Westchester Medical Center, Valhalla, NY
| |
Collapse
|
26
|
Hu Y, Zhang Y, Guo J, Chen S, Jin J, Li P, Pan Y, Lei S, Li J, Wu S, Bu B, Fu L. Synthesis and anti-proliferative effect of novel 4-Aryl-1, 3-Thiazole-TPP conjugates via mitochondrial uncoupling process. Bioorg Chem 2024; 150:107588. [PMID: 38936051 DOI: 10.1016/j.bioorg.2024.107588] [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: 05/11/2024] [Revised: 06/18/2024] [Accepted: 06/22/2024] [Indexed: 06/29/2024]
Abstract
With the advent of mitochondrial targeting moiety such as triphenlyphosphonium cation (TPP+), targeting mitochondria in cancer cells has become a promising strategy for combating tumors. Herein, a series of novel 4-aryl-1,3-thiazole derivatives linked to TPP+ moiety were designed and synthesized. The cytotoxicity against a panel of four cancer cell lines was evaluated by CCK-8 assay. Most of these compounds exhibited moderate to good inhibitory activity over HeLa, PC-3 and HCT-15 cells while MCF-7 cells were less sensitive to most compounds. Among them, compound 12a exhibited a significant anti-proliferative activity against HeLa cells, and prompted for further investigation. Specifically, 12a decreased mitochondrial membrane potential and enhanced levels of reactive oxygen species (ROS). The flow cytometry analysis revealed that compound 12a could induce apoptosis and cell cycle arrest at G0/G1 phase in HeLa cells. In addition, mitochondrial bioenergetics assay revealed that 12a displayed mild mitochondrial uncoupling effect. Taken together, these findings suggest the therapeutic potential of compound 12a as an antitumor agent targeting mitochondria.
Collapse
Affiliation(s)
- Yixin Hu
- Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Yang Zhang
- Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Jie Guo
- Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Shihao Chen
- Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Jie Jin
- Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Pengyu Li
- Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Yuchen Pan
- Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Shuwen Lei
- Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Jiaqi Li
- Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Suheng Wu
- Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Buzhou Bu
- Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Lei Fu
- Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China; Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, China.
| |
Collapse
|
27
|
Ríos JA, Bórquez JC, Godoy JA, Zolezzi JM, Furrianca MC, Inestrosa NC. Emerging role of Metformin in Alzheimer's disease: A translational view. Ageing Res Rev 2024; 100:102439. [PMID: 39074563 DOI: 10.1016/j.arr.2024.102439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/23/2024] [Accepted: 07/23/2024] [Indexed: 07/31/2024]
Abstract
Alzheimer's disease (AD) constitutes a major public-health issue of our time. Regrettably, despite our considerable understanding of the pathophysiological aspects of this disease, current interventions lead to poor outcomes. Furthermore, experimentally promising compounds have continuously failed when translated to clinical trials. Along with increased population ageing, Type 2 Diabetes Mellitus (T2DM) has become an extremely common condition, mainly due to unbalanced dietary habits. Substantial epidemiological evidence correlates T2DM with cognitive impairment as well. Considering that brain insulin resistance, mitochondrial dysfunction, oxidative stress, and amyloidogenesis are common phenomena, further approaching the common features among these pathological conditions. Metformin constitutes the first-choice drug to preclude insulin resistance in T2DM clinical management. Experimental evidence suggests that its functions might include neuroprotective effects, in addition to its hypoglycemic activity. This review aims to summarize and discuss current knowledge of experimental data on metformin on this path towards translational medicine. Finally, we discuss the controversial data of responses to metformin in vitro, and in vivo, animal models and human studies.
Collapse
Affiliation(s)
- Juvenal A Ríos
- Facultad de Medicina y Ciencia, Escuela de Medicina, Universidad San Sebastián, Santiago, Chile
| | - Juan Carlos Bórquez
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Escuela de Medicina, Universidad de Magallanes, Punta Arenas, Chile; Facultad de Ciencias de la Salud, Universidad de Magallanes, Punta Arenas, Chile
| | - Juan A Godoy
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan M Zolezzi
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Escuela de Medicina, Universidad de Magallanes, Punta Arenas, Chile
| | | | - Nibaldo C Inestrosa
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile; Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Escuela de Medicina, Universidad de Magallanes, Punta Arenas, Chile.
| |
Collapse
|
28
|
Gholami M, Coleman-Fuller N, Salehirad M, Darbeheshti S, Motaghinejad M. Neuroprotective Effects of Sodium-Glucose Cotransporter-2 (SGLT2) Inhibitors (Gliflozins) on Diabetes-Induced Neurodegeneration and Neurotoxicity: A Graphical Review. Int J Prev Med 2024; 15:28. [PMID: 39239308 PMCID: PMC11376549 DOI: 10.4103/ijpvm.ijpvm_5_23] [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: 01/03/2023] [Accepted: 02/20/2024] [Indexed: 09/07/2024] Open
Abstract
Diabetes is a chronic endocrine disorder that negatively affects various body systems, including the nervous system. Diabetes can cause or exacerbate various neurological disorders, and diabetes-induced neurodegeneration can involve several mechanisms such as mitochondrial dysfunction, activation of oxidative stress, neuronal inflammation, and cell death. In recent years, the management of diabetes-induced neurodegeneration has relied on several types of drugs, including sodium-glucose cotransporter-2 (SGLT2) inhibitors, also called gliflozins. In addition to exerting powerful effects in reducing blood glucose, gliflozins have strong anti-neuro-inflammatory characteristics that function by inhibiting oxidative stress and cell death in the nervous system in diabetic subjects. This review presents the molecular pathways involved in diabetes-induced neurodegeneration and evaluates the clinical and laboratory studies investigating the neuroprotective effects of gliflozins against diabetes-induced neurodegeneration, with discussion about the contributing roles of diverse molecular pathways, such as mitochondrial dysfunction, oxidative stress, neuro-inflammation, and cell death. Several databases-including Web of Science, Scopus, PubMed, Google Scholar, and various publishers, such as Springer, Wiley, and Elsevier-were searched for keywords regarding the neuroprotective effects of gliflozins against diabetes-triggered neurodegenerative events. Additionally, anti-neuro-inflammatory, anti-oxidative stress, and anti-cell death keywords were applied to evaluate potential neuronal protection mechanisms of gliflozins in diabetes subjects. The search period considered valid peer-reviewed studies published from January 2000 to July 2023. The current body of literature suggests that gliflozins can exert neuroprotective effects against diabetes-induced neurodegenerative events and neuronal dysfunction, and these effects are mediated via activation of mitochondrial function and prevention of cell death processes, oxidative stress, and inflammation in neurons affected by diabetes. Gliflozins can confer neuroprotective properties in diabetes-triggered neurodegeneration, and these effects are mediated by inhibiting oxidative stress, inflammation, and cell death.
Collapse
Affiliation(s)
- Mina Gholami
- Chronic Respiratory Disease Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Natalie Coleman-Fuller
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, USA
| | - Mahsa Salehirad
- Chronic Respiratory Disease Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sepideh Darbeheshti
- Chronic Respiratory Disease Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Majid Motaghinejad
- Chronic Respiratory Disease Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
29
|
Lardaro A, Quarta L, Pagnotta S, Sodero G, Mariani S, Del Ben M, Desideri G, Ettorre E, Baratta F. Impact of Sodium Glucose Cotransporter 2 Inhibitors (SGLT2i) Therapy on Dementia and Cognitive Decline. Biomedicines 2024; 12:1750. [PMID: 39200215 PMCID: PMC11351143 DOI: 10.3390/biomedicines12081750] [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: 06/21/2024] [Revised: 07/28/2024] [Accepted: 07/29/2024] [Indexed: 09/02/2024] Open
Abstract
Dementia is an age-related syndrome characterized by the progressive deterioration of cognition and capacity for independent living. Diabetes is often associated with cognitive decline and shares similar pathophysiological mechanisms with dementia, such as systemic inflammation, oxidative stress, insulin resistance, and advanced glycation end-products formation. Therefore, adequate diabetes management may reduce the risk of cognitive decline, especially in patients with other comorbidities and risk factors. The sodium glucose cotransporter inhibitors (SGLT2i) regulate renal glucose reabsorption by blocking the SGLT2 cotransporters located in the proximal tubules, causing glycosuria and intraglomerular pressure reduction. Their use helps to lower blood pressure by modifying sodium and water homeostasis; these drugs are also commonly used in the treatment of heart failure and chronic kidney disease, while recently, a potential neuroprotective role in the central nervous system has been suggested. The aim of our scoping review is to analyze current evidence about the potential neuroprotective effects of SGLT2i in adult patients. We performed a scoping literature review to evaluate the effect of SGLT2i on dementia, mild cognitive impairment (MCI) and Alzheimer's disease incidence and progression. The screening process was performed through different searches on PubMed and EMBASE, evaluating original works published up to January 2024. In conclusion, the use of SGLT2i could be associated with a neuroprotective effect in patients with diabetes, reducing the incidence or the progression of MCI and dementia. Further prospective studies are needed to validate this hypothesis and to evaluate the effectiveness of this class of drugs in normal glycemic profile patients.
Collapse
Affiliation(s)
- Antonio Lardaro
- Department of Clinical Internal, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy; (A.L.); (L.Q.); (S.P.); (M.D.B.); (G.D.); (E.E.)
| | - Ludovica Quarta
- Department of Clinical Internal, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy; (A.L.); (L.Q.); (S.P.); (M.D.B.); (G.D.); (E.E.)
| | - Stefania Pagnotta
- Department of Clinical Internal, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy; (A.L.); (L.Q.); (S.P.); (M.D.B.); (G.D.); (E.E.)
| | - Giorgio Sodero
- Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00136 Rome, Italy
| | - Sandro Mariani
- Department of Internal Medicine and Medical Specialties, Policlinico Umberto I University Hospital, 00161 Rome, Italy;
| | - Maria Del Ben
- Department of Clinical Internal, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy; (A.L.); (L.Q.); (S.P.); (M.D.B.); (G.D.); (E.E.)
| | - Giovambattista Desideri
- Department of Clinical Internal, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy; (A.L.); (L.Q.); (S.P.); (M.D.B.); (G.D.); (E.E.)
| | - Evaristo Ettorre
- Department of Clinical Internal, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy; (A.L.); (L.Q.); (S.P.); (M.D.B.); (G.D.); (E.E.)
| | - Francesco Baratta
- Department of Clinical Internal, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy; (A.L.); (L.Q.); (S.P.); (M.D.B.); (G.D.); (E.E.)
| |
Collapse
|
30
|
Barros-Aragão FG, Pinto TP, Carregari VC, Rezende NB, Pinheiro TL, Reis-de-Oliveira G, Cabral-Castro MJ, Queiroz DC, Fonseca PL, Gonçalves AL, de Freitas GR, Sudo FK, Mattos P, Bozza FA, Rodrigues EC, Aguiar RS, Rodrigues RS, Brandão CO, Souza AS, Martins-de-Souza D, De Felice FG, Tovar-Moll F. Changes in neuroinflammatory biomarkers correlate with disease severity and neuroimaging alterations in patients with COVID-19 neurological complications. Brain Behav Immun Health 2024; 39:100805. [PMID: 39022627 PMCID: PMC11253226 DOI: 10.1016/j.bbih.2024.100805] [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: 12/07/2023] [Revised: 05/15/2024] [Accepted: 06/10/2024] [Indexed: 07/20/2024] Open
Abstract
COVID-19 induces acute and persistent neurological symptoms in mild and severe cases. Proposed concomitant mechanisms include direct viral infection and strain, coagulopathy, hypoxia, and neuroinflammation. However, underlying molecular alterations associated with multiple neurological outcomes in both mild and severe cases are majorly unexplored. To illuminate possible mechanisms leading to COVID-19 neurological disease, we retrospectively investigated in detail a cohort of 35 COVID-19 mild and severe hospitalized patients presenting neurological alterations subject to clinically indicated cerebrospinal fluid (CSF) sampling. Clinical and neurological investigation, brain imaging, viral sequencing, and cerebrospinal CSF analyses were carried out. We found that COVID-19 patients presented heterogeneous neurological symptoms dissociated from lung burden. Nasal swab viral sequencing revealed a dominant strain at the time of the study, and we could not detect traces of SARS-CoV-2's spike protein in patients' CSF by multiple reaction monitoring analysis. Patients presented ubiquitous systemic hyper-inflammation and broad alterations in CSF proteomics related to inflammation, innate immunity, and hemostasis, irrespective of COVID-19 severity or neuroimaging alterations. Elevated CSF interleukin-6 (IL6) correlated with disease severity (sex-, age-, and comorbidity-adjusted mean Severe 24.5 pg/ml, 95% confidence interval (CI) 9.62-62.23 vs. Mild 3.91 pg/mL CI 1.5-10.3 patients, p = 0.019). CSF tumor necrosis factor-alpha (TNFα) and IL6 levels were higher in patients presenting pronounced neuroimaging alterations compared to those who did not (sex-, age-, and comorbidity-adjusted mean TNFα Pronounced 3.4, CI 2.4-4.4 vs. Non-Pronounced 2.0, CI 1.4-2.5, p = 0.022; IL6 Pronounced 33.11, CI 8.89-123.31 vs Non-Pronounced 6.22, CI 2.9-13.34, p = 0.046). Collectively, our findings put neuroinflammation as a possible driver of COVID-19 acute neurological disease in mild and severe cases.
Collapse
Affiliation(s)
- Fernanda G.Q. Barros-Aragão
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil, 22281-100
- Institute of Medical Biochemistry Leopoldo De Meis (IBqM), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil, 21941-902
- Centre for Neuroscience Studies, Department of Biomedical and Molecular Sciences & Department of Psychiatry, Queen's University, Kingston, Ontario, Canada, K7L 3N6
| | - Talita P. Pinto
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil, 22281-100
| | - Victor C. Carregari
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil, 13083-862
| | - Nathane B.S. Rezende
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil, 22281-100
| | - Thaís L. Pinheiro
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil, 22281-100
- Institute of Medical Biochemistry Leopoldo De Meis (IBqM), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil, 21941-902
| | - Guilherme Reis-de-Oliveira
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil, 13083-862
| | - Mauro J. Cabral-Castro
- Institute of Microbiology Paulo de Goés, UFRJ, Rio de Janeiro, Brazil, 21941-902
- Department of Pathology, Faculty of Medicine, Universidade Federal Fluminense, Niterói, RJ, Brazil, 24210-346
| | - Daniel C. Queiroz
- Department of Genetics, Ecology, and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil, 31270-901
| | - Paula L.C. Fonseca
- Department of Genetics, Ecology, and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil, 31270-901
| | - Alessandro L. Gonçalves
- Department of Genetics, Ecology, and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil, 31270-901
| | | | - Felipe K. Sudo
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil, 22281-100
| | - Paulo Mattos
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil, 22281-100
| | - Fernando A. Bozza
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil, 22281-100
| | - Erika C. Rodrigues
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil, 22281-100
| | - Renato S. Aguiar
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil, 22281-100
- Department of Genetics, Ecology, and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil, 31270-901
| | - Rosana S. Rodrigues
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil, 22281-100
| | | | - Andrea S. Souza
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil, 22281-100
| | - Daniel Martins-de-Souza
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil, 22281-100
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil, 13083-862
| | - Fernanda G. De Felice
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil, 22281-100
- Institute of Medical Biochemistry Leopoldo De Meis (IBqM), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil, 21941-902
- Centre for Neuroscience Studies, Department of Biomedical and Molecular Sciences & Department of Psychiatry, Queen's University, Kingston, Ontario, Canada, K7L 3N6
| | - Fernanda Tovar-Moll
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil, 22281-100
| |
Collapse
|
31
|
Ribeiro FC, Cozachenco D, Argyrousi EK, Staniszewski A, Wiebe S, Calixtro JD, Soares‐Neto R, Al‐Chami A, Sayegh FE, Bermudez S, Arsenault E, Cossenza M, Lacaille J, Nader K, Sun H, De Felice FG, Lourenco MV, Arancio O, Aguilar‐Valles A, Sonenberg N, Ferreira ST. The ketamine metabolite (2R,6R)-hydroxynorketamine rescues hippocampal mRNA translation, synaptic plasticity and memory in mouse models of Alzheimer's disease. Alzheimers Dement 2024; 20:5398-5410. [PMID: 38934107 PMCID: PMC11350050 DOI: 10.1002/alz.14034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 04/16/2024] [Accepted: 05/06/2024] [Indexed: 06/28/2024]
Abstract
INTRODUCTION Impaired brain protein synthesis, synaptic plasticity, and memory are major hallmarks of Alzheimer's disease (AD). The ketamine metabolite (2R,6R)-hydroxynorketamine (HNK) has been shown to modulate protein synthesis, but its effects on memory in AD models remain elusive. METHODS We investigated the effects of HNK on hippocampal protein synthesis, long-term potentiation (LTP), and memory in AD mouse models. RESULTS HNK activated extracellular signal-regulated kinase 1/2 (ERK1/2), mechanistic target of rapamycin (mTOR), and p70S6 kinase 1 (S6K1)/ribosomal protein S6 signaling pathways. Treatment with HNK rescued hippocampal LTP and memory deficits in amyloid-β oligomers (AβO)-infused mice in an ERK1/2-dependent manner. Treatment with HNK further corrected aberrant transcription, LTP and memory in aged APP/PS1 mice. DISCUSSION Our findings demonstrate that HNK induces signaling and transcriptional responses that correct synaptic and memory deficits in AD mice. These results raise the prospect that HNK could serve as a therapeutic approach in AD. HIGHLIGHTS The ketamine metabolite HNK activates hippocampal ERK/mTOR/S6 signaling pathways. HNK corrects hippocampal synaptic and memory defects in two mouse models of AD. Rescue of synaptic and memory impairments by HNK depends on ERK signaling. HNK corrects aberrant transcriptional signatures in APP/PS1 mice.
Collapse
Affiliation(s)
- Felipe C. Ribeiro
- Institute of Medical Biochemistry Leopoldo de MeisFederal University of Rio de JaneiroRio de JaneiroRio de JaneiroBrazil
| | - Danielle Cozachenco
- Institute of Medical Biochemistry Leopoldo de MeisFederal University of Rio de JaneiroRio de JaneiroRio de JaneiroBrazil
| | - Elentina K. Argyrousi
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
| | - Agnieszka Staniszewski
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
| | - Shane Wiebe
- Department of BiochemistryMcGill UniversityMontrealQuebecCanada
| | - Joao D. Calixtro
- Institute of Medical Biochemistry Leopoldo de MeisFederal University of Rio de JaneiroRio de JaneiroRio de JaneiroBrazil
| | - Rubens Soares‐Neto
- Institute of Medical Biochemistry Leopoldo de MeisFederal University of Rio de JaneiroRio de JaneiroRio de JaneiroBrazil
| | - Aycheh Al‐Chami
- Department of NeuroscienceCarleton UniversityOttawaOntarioCanada
| | - Fatema El Sayegh
- Department of NeuroscienceCarleton UniversityOttawaOntarioCanada
| | - Sara Bermudez
- Department of BiochemistryMcGill UniversityMontrealQuebecCanada
| | - Emily Arsenault
- Department of NeuroscienceCarleton UniversityOttawaOntarioCanada
| | - Marcelo Cossenza
- Department of Physiology and Pharmacology, Fluminense Federal UniversityBiomedical InstituteNiteróiRio de JaneiroBrazil
| | - Jean‐Claude Lacaille
- Department of Neurosciences, Université de MontréalCentre for Interdisciplinary Research on Brain and Learning and Research Group on Neural Signaling and CircuitsMontrealQuebecCanada
| | - Karim Nader
- Department of PsychologyMcGill UniversityMontrealQuebecCanada
| | - Hongyu Sun
- Department of NeuroscienceCarleton UniversityOttawaOntarioCanada
| | - Fernanda G. De Felice
- Institute of Medical Biochemistry Leopoldo de MeisFederal University of Rio de JaneiroRio de JaneiroRio de JaneiroBrazil
- Department of Biomedical and Molecular Sciences, Centre for Neuroscience StudiesQueen's UniversityKingstonOntarioCanada
- Department of PsychiatryQueen's UniversityKingstonOntarioCanada
- D'Or Institute for Research and EducationRio de JaneiroRio de JaneiroBrazil
| | - Mychael V. Lourenco
- Institute of Medical Biochemistry Leopoldo de MeisFederal University of Rio de JaneiroRio de JaneiroRio de JaneiroBrazil
| | - Ottavio Arancio
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
| | | | - Nahum Sonenberg
- Department of BiochemistryMcGill UniversityMontrealQuebecCanada
| | - Sergio T. Ferreira
- Institute of Medical Biochemistry Leopoldo de MeisFederal University of Rio de JaneiroRio de JaneiroRio de JaneiroBrazil
- D'Or Institute for Research and EducationRio de JaneiroRio de JaneiroBrazil
- Institute of Biophysics Carlos Chagas FilhoFederal University of Rio de JaneiroRio de JaneiroRio de JaneiroBrazil
| |
Collapse
|
32
|
Zhao W, Zhang W, Hu Y, Zhou Y, Zhao J, Li Y, Xu Z. AdipoRon Ameliorates Synaptic Dysfunction and Inhibits tau Hyperphosphorylation through the AdipoR/AMPK/mTOR Pathway in T2DM Mice. Neurochem Res 2024; 49:2075-2086. [PMID: 38819697 DOI: 10.1007/s11064-024-04162-4] [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: 10/07/2023] [Revised: 02/20/2024] [Accepted: 05/22/2024] [Indexed: 06/01/2024]
Abstract
There is growing evidence showing that adiponectin (APN) can improve Alzheimer's disease(AD)-like pathological changes by improving insulin resistance. However, the role of AdipoRon (an Adiponectin receptor agonist) on synaptic plasticity and cognitive dysfunction in the early stages of type 2 diabetes mellitus(T2DM) remains unknown. In this study, we investigated the neuroprotective effect and the molecular mechanism underlying the effect of AdipoRon in T2DM mice. We found that AdipoRon significantly restored the cognitive deficits in T2DM mice, including shorter escape latency, more crossing times, increased distances, and percentage of time in the target quadrant. In addition, AdipoRon treatment up-regulated synaptic proteins (PSD95, SYN, GAP43, and SYP), increased the number of hippocampal synapses and attenuated synaptic damage, including the length, the number and the density of dendritic spines in CA1 and DG regions. Furthermore, AdipoRon attenuated Tau phosphorylation at multiple AD-related sites (p-tau 205, p-tau 396, p-tau 404) by promoting AdipoR expression and activating the AMPK/mTOR pathway. Our data suggests that AdipoRon exerts neuroprotective effects on the T2DM mice, which may be mediated by the activation of the AdipoR/AMPK/mTOR signaling pathway.
Collapse
Affiliation(s)
- Wenyan Zhao
- Department of Neuropsychology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Wei Zhang
- Department of Neuropsychology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yingying Hu
- Department of Neuropsychology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yuliang Zhou
- Department of Neuropsychology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jinying Zhao
- Department of Neuropsychology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yahong Li
- Department of Applied Psychology, South-Central Minzu University, Wuhan, Hubei, China.
| | - Zhipeng Xu
- Department of Neuropsychology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
33
|
Abdalla MMI. Insulin resistance as the molecular link between diabetes and Alzheimer's disease. World J Diabetes 2024; 15:1430-1447. [PMID: 39099819 PMCID: PMC11292327 DOI: 10.4239/wjd.v15.i7.1430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/08/2024] [Accepted: 05/06/2024] [Indexed: 07/08/2024] Open
Abstract
Diabetes mellitus (DM) and Alzheimer's disease (AD) are two major health concerns that have seen a rising prevalence worldwide. Recent studies have indicated a possible link between DM and an increased risk of developing AD. Insulin, while primarily known for its role in regulating blood sugar, also plays a vital role in protecting brain functions. Insulin resistance (IR), especially prevalent in type 2 diabetes, is believed to play a significant role in AD's development. When insulin signalling becomes dysfunctional, it can negatively affect various brain functions, making individuals more susceptible to AD's defining features, such as the buildup of beta-amyloid plaques and tau protein tangles. Emerging research suggests that addressing insulin-related issues might help reduce or even reverse the brain changes linked to AD. This review aims to explore the rela-tionship between DM and AD, with a focus on the role of IR. It also explores the molecular mechanisms by which IR might lead to brain changes and assesses current treatments that target IR. Understanding IR's role in the connection between DM and AD offers new possibilities for treatments and highlights the importance of continued research in this interdisciplinary field.
Collapse
Affiliation(s)
- Mona Mohamed Ibrahim Abdalla
- Department of Human Biology, School of Medicine, International Medical University, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| |
Collapse
|
34
|
Nguyen DPQ, Jallow AW, Lin YF, Lin YF. Exploring the Potential Role of Oligodendrocyte-Associated PIP4K2A in Alzheimer's Disease Complicated with Type 2 Diabetes Mellitus via Multi-Omic Analysis. Int J Mol Sci 2024; 25:6640. [PMID: 38928345 PMCID: PMC11204139 DOI: 10.3390/ijms25126640] [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: 05/17/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Alzheimer's disease (AD) and type 2 diabetes mellitus (T2DM) are two common diseases that affect the elderly population worldwide. The identification of common genes associated with AD and T2DM holds promise for potential biomarkers and intriguing pathogenesis of these two complicated diseases. This study utilized a comprehensive approach by integrating transcriptome data from multiple cohorts, encompassing both AD and T2DM. The analysis incorporated various data types, including blood and tissue samples as well as single-cell datasets, allowing for a detailed assessment of gene expression patterns. From the brain region-specific single-cell analysis, PIP4K2A, which encodes phosphatidylinositol-5-phosphate 4-kinase type 2 alpha, was found to be expressed mainly in oligodendrocytes compared to other cell types. Elevated levels of PIP4K2A in AD and T2DM patients' blood were found to be associated with key cellular processes such as vesicle-mediated transport, negative regulation of autophagosome assembly, and cytosolic transport. The identification of PIP4K2A's potential roles in the cellular processes of AD and T2DM offers valuable insights into the development of biomarkers for diagnosis and therapy, especially in the complication of these two diseases.
Collapse
Affiliation(s)
- Doan Phuong Quy Nguyen
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, New Taipei City 235, Taiwan; (D.P.Q.N.); (A.W.J.)
- Institute of Biomedicine, Hue University of Medicine and Pharmacy, Hue University, Hue City 49120, Vietnam
- Department of Medical Genetics, Hue University of Medicine and Pharmacy, Hue University, Hue City 49120, Vietnam
| | - Amadou Wurry Jallow
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, New Taipei City 235, Taiwan; (D.P.Q.N.); (A.W.J.)
| | - Yi-Fang Lin
- Department of Laboratory Medicine, Taipei Medical University—Shuang Ho Hospital, New Taipei City 235, Taiwan;
| | - Yung-Feng Lin
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, New Taipei City 235, Taiwan; (D.P.Q.N.); (A.W.J.)
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, New Taipei City 235, Taiwan
- Department of Laboratory Medicine, Taipei Medical University Hospital, Taipei City 110, Taiwan
| |
Collapse
|
35
|
Yang Y, Qiu L. Research Progress on the Pathogenesis, Diagnosis, and Drug Therapy of Alzheimer's Disease. Brain Sci 2024; 14:590. [PMID: 38928590 PMCID: PMC11201671 DOI: 10.3390/brainsci14060590] [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: 05/15/2024] [Revised: 06/03/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
As the population ages worldwide, Alzheimer's disease (AD), the most prevalent kind of neurodegenerative disorder among older people, has become a significant factor affecting quality of life, public health, and economies. However, the exact pathogenesis of Alzheimer's remains elusive, and existing highly recognized pathogenesis includes the amyloid cascade hypothesis, Tau neurofibrillary tangles hypothesis, and neuroinflammation hypothesis. The major diagnoses of Alzheimer's disease include neuroimaging positron emission computed tomography, magnetic resonance imaging, and cerebrospinal fluid molecular diagnosis. The therapy of Alzheimer's disease primarily relies on drugs, and the approved drugs on the market include acetylcholinesterase drugs, glutamate receptor antagonists, and amyloid-β monoclonal antibodies. Still, the existing drugs can only alleviate the symptoms of the disease and cannot completely reverse it. This review aims to summarize existing research results on Alzheimer's disease pathogenesis, diagnosis, and drug therapy, with the objective of facilitating future research in this area.
Collapse
Affiliation(s)
- Yixuan Yang
- College of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China;
| | - Lina Qiu
- College of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China;
- Beijing Key Laboratory for Science and Application of Functional Molecular and Crystalline Materials, University of Science and Technology Beijing, Beijing 100083, China
| |
Collapse
|
36
|
Bennici G, Almahasheer H, Alghrably M, Valensin D, Kola A, Kokotidou C, Lachowicz J, Jaremko M. Mitigating diabetes associated with reactive oxygen species (ROS) and protein aggregation through pharmacological interventions. RSC Adv 2024; 14:17448-17460. [PMID: 38813124 PMCID: PMC11135279 DOI: 10.1039/d4ra02349h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 05/22/2024] [Indexed: 05/31/2024] Open
Abstract
Diabetes mellitus, a complex metabolic disorder, presents a growing global health challenge. In 2021, there were 529 million diabetics worldwide. At the super-regional level, Oceania, the Middle East, and North Africa had the highest age-standardized rates. The majority of cases of diabetes in 2021 (>90.0%) were type 2 diabetes, which is largely indicative of the prevalence of diabetes in general, particularly in older adults (K. L. Ong, et al., Global, regional, and national burden of diabetes from 1990 to 2021, with projections of prevalence to 2050: a systematic analysis for the Global Burden of Disease Study 2021, Lancet, 2023, 402(10397), 203-234). Nowadays, slowing the progression of diabetic complications is the only effective way to manage diabetes with the available therapeutic options. However, novel biomarkers and treatments are urgently needed to control cytokine secretion, advanced glycation end products (AGEs) production, vascular inflammatory effects, and cellular death. Emerging research has highlighted the intricate interplay between reactive oxygen species (ROS) and protein aggregation in the pathogenesis of diabetes. In this scenario, the main aim of this paper is to provide a comprehensive review of the current understanding of the molecular mechanisms underlying ROS-induced cellular damage and protein aggregation, specifically focusing on their contribution to diabetes development. The role of ROS as key mediators of oxidative stress in diabetes is discussed, emphasizing their impact on cellular components and signaling. Additionally, the involvement of protein aggregation in impairing cellular function and insulin signaling is explored. The synergistic effects of ROS and protein aggregation in promoting β-cell dysfunction and insulin resistance are examined, shedding light on potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Giulia Bennici
- Division of Biological and Environmental Sciences and Engineering (BESE), King Abdullah University of Science and Technology (KAUST) Thuwal 23955-6900 Saudi Arabia
| | - Hanan Almahasheer
- Department of Biology, College of Science, Imam Abdulrahman Bin Faisal University (IAU) Dammam 31441-1982 Saudi Arabia
| | - Mawadda Alghrably
- Division of Biological and Environmental Sciences and Engineering (BESE), King Abdullah University of Science and Technology (KAUST) Thuwal 23955-6900 Saudi Arabia
| | - Daniela Valensin
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena Via Aldo Moro 2 53100 Siena Italy
| | - Arian Kola
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena Via Aldo Moro 2 53100 Siena Italy
| | - Chrysoula Kokotidou
- Department of Materials Science and Technology, University of Crete 70013 Heraklion Crete Greece
- Institute of Electronic Structure and Laser (IESL) FORTH 70013 Heraklion Crete Greece
| | - Joanna Lachowicz
- Department of Population Health, Division of Environmental Health and Occupational Medicine, Wroclaw Medical University Mikulicza-Radeckiego 7 Wroclaw PL 50-368 Poland
| | - Mariusz Jaremko
- Division of Biological and Environmental Sciences and Engineering (BESE), King Abdullah University of Science and Technology (KAUST) Thuwal 23955-6900 Saudi Arabia
| |
Collapse
|
37
|
Kula B, Antal B, Weistuch C, Gackière F, Barre A, Velado V, Hubbard JM, Kukley M, Mujica-Parodi LR, Smith NA. D-β-hydroxybutyrate stabilizes hippocampal CA3-CA1 circuit during acute insulin resistance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.23.554428. [PMID: 37662316 PMCID: PMC10473684 DOI: 10.1101/2023.08.23.554428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
The brain primarily relies on glycolysis for mitochondrial respiration but switches to alternative fuels such as ketone bodies (KBs) when less glucose is available. Neuronal KB uptake, which does not rely on glucose transporter 4 (GLUT4) or insulin, has shown promising clinical applicability in alleviating the neurological and cognitive effects of disorders with hypometabolic components. However, the specific mechanisms by which such interventions affect neuronal functions are poorly understood. In this study, we pharmacologically blocked GLUT4 to investigate the effects of exogenous KB D-β-hydroxybutyrate (D-βHb) on mouse brain metabolism during acute insulin resistance (AIR). We found that both AIR and D-βHb had distinct impacts across neuronal compartments: AIR decreased synaptic activity and long-term potentiation (LTP) and impaired axonal conduction, synchronization, and action potential (AP) properties, while D-βHb rescued neuronal functions associated with axonal conduction, synchronization, and LTP.
Collapse
Affiliation(s)
- Bartosz Kula
- Del Monte Institute for Neuroscience, Department of Neuroscience, University of Rochester, School of Medicine and Dentistry, Rochester, USA
| | - Botond Antal
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, USA
| | - Corey Weistuch
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Florian Gackière
- Neuroservices Alliance, Les Jardins de l’Entreprise, Quartier de le Confrérie, Le Puy Ste Réparade, France
| | - Alexander Barre
- Neuroservices Alliance, Les Jardins de l’Entreprise, Quartier de le Confrérie, Le Puy Ste Réparade, France
| | - Victor Velado
- Center for Neuroscience Research, Children’s National Research Institute, Children’s National Hospital, Washington D.C., USA
| | - Jeffrey M Hubbard
- Neuroservices Alliance, Les Jardins de l’Entreprise, Quartier de le Confrérie, Le Puy Ste Réparade, France
| | - Maria Kukley
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- Ikerbasque - Basque Foundation for Science, Bilbao, Spain
| | - Lilianne R Mujica-Parodi
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, USA
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, USA
| | - Nathan A Smith
- Del Monte Institute for Neuroscience, Department of Neuroscience, University of Rochester, School of Medicine and Dentistry, Rochester, USA
- Center for Neuroscience Research, Children’s National Research Institute, Children’s National Hospital, Washington D.C., USA
- George Washington University School of Medicine and Health Sciences, Washington D.C., USA
| |
Collapse
|
38
|
Kula B, Antal B, Weistuch C, Gackière F, Barre A, Velado V, Hubbard JM, Kukley M, Mujica-Parodi LR, Smith NA. D-ꞵ-hydroxybutyrate stabilizes hippocampal CA3-CA1 circuit during acute insulin resistance. PNAS NEXUS 2024; 3:pgae196. [PMID: 38818236 PMCID: PMC11138115 DOI: 10.1093/pnasnexus/pgae196] [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: 02/02/2024] [Accepted: 05/06/2024] [Indexed: 06/01/2024]
Abstract
The brain primarily relies on glycolysis for mitochondrial respiration but switches to alternative fuels such as ketone bodies (KBs) when less glucose is available. Neuronal KB uptake, which does not rely on glucose transporter 4 (GLUT4) or insulin, has shown promising clinical applicability in alleviating the neurological and cognitive effects of disorders with hypometabolic components. However, the specific mechanisms by which such interventions affect neuronal functions are poorly understood. In this study, we pharmacologically blocked GLUT4 to investigate the effects of exogenous KB D-ꞵ-hydroxybutyrate (D-ꞵHb) on mouse brain metabolism during acute insulin resistance (AIR). We found that both AIR and D-ꞵHb had distinct impacts across neuronal compartments: AIR decreased synaptic activity and long-term potentiation (LTP) and impaired axonal conduction, synchronization, and action potential properties, while D-ꞵHb rescued neuronal functions associated with axonal conduction, synchronization, and LTP.
Collapse
Affiliation(s)
- Bartosz Kula
- Del Monte Institute for Neuroscience, Department of Neuroscience, University of Rochester, School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Botond Antal
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
| | - Corey Weistuch
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Florian Gackière
- Neuroservices Alliance, Les Jardins de l’Entreprise, Quartier de le Confrérie, 13610 Le Puy-Sainte-Réparade, France
| | - Alexander Barre
- Neuroservices Alliance, Les Jardins de l’Entreprise, Quartier de le Confrérie, 13610 Le Puy-Sainte-Réparade, France
| | - Victor Velado
- Center for Neuroscience Research, Children’s National Research Institute, Children’s National Hospital, Washington, DC 20012, USA
| | - Jeffrey M Hubbard
- Neuroservices Alliance, Les Jardins de l’Entreprise, Quartier de le Confrérie, 13610 Le Puy-Sainte-Réparade, France
| | - Maria Kukley
- Achucarro Basque Center for Neuroscience, 48940 Leioa, Bizkaia, Spain
- Ikerbasque—Basque Foundation for Science, 48009 Bilbao, Spain
| | - Lilianne R Mujica-Parodi
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Nathan A Smith
- Del Monte Institute for Neuroscience, Department of Neuroscience, University of Rochester, School of Medicine and Dentistry, Rochester, NY 14642, USA
- Center for Neuroscience Research, Children’s National Research Institute, Children’s National Hospital, Washington, DC 20012, USA
- School of Medicine and Health Sciences, George Washington University, Washington, DC 20052, USA
| |
Collapse
|
39
|
Albar NY, Hassaballa H, Shikh H, Albar Y, Ibrahim AS, Mousa AH, Alshanberi AM, Elgebaly A, Bahbah EI. The interaction between insulin resistance and Alzheimer's disease: a review article. Postgrad Med 2024; 136:377-395. [PMID: 38804907 DOI: 10.1080/00325481.2024.2360887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024]
Abstract
Insulin serves multiple functions as a growth-promoting hormone in peripheral tissues. It manages glucose metabolism by promoting glucose uptake into cells and curbing the production of glucose in the liver. Beyond this, insulin fosters cell growth, drives differentiation, aids protein synthesis, and deters degradative processes like glycolysis, lipolysis, and proteolysis. Receptors for insulin and insulin-like growth factor-1 are widely expressed in the central nervous system. Their widespread presence in the brain underscores the varied and critical functions of insulin signaling there. Insulin aids in bolstering cognition, promoting neuron extension, adjusting the release and absorption of catecholamines, and controlling the expression and positioning of gamma-aminobutyric acid (GABA). Importantly, insulin can effortlessly traverse the blood-brain barrier. Furthermore, insulin resistance (IR)-induced alterations in insulin signaling might hasten brain aging, impacting its plasticity and potentially leading to neurodegeneration. Two primary pathways are responsible for insulin signal transmission: the phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) pathway, which oversees metabolic responses, and the mitogen-activated protein kinase (MAPK) pathway, which guides cell growth, survival, and gene transcription. This review aimed to explore the potential shared metabolic traits between Alzheimer's disease (AD) and IR disorders. It delves into the relationship between AD and IR disorders, their overlapping genetic markers, and shared metabolic indicators. Additionally, it addresses existing therapeutic interventions targeting these intersecting pathways.
Collapse
Affiliation(s)
- Nezar Y Albar
- Internal Medicine Department, Dr. Samir Abbas Hospital, Jeddah, Saudi Arabia
| | | | - Hamza Shikh
- Ibn Sina National College for Medical Studies, Jeddah, Saudi Arabia
| | - Yassin Albar
- Fakeeh College of Medical Sciences, Jeddah, Saudi Arabia
| | | | - Ahmed Hafez Mousa
- Department of Neurosurgery, Postgraduate Medical Education, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
- Department of Neurosurgery, Rashid Hospital, Dubai Academic Health Cooperation, Dubai, United Arab Emirates
| | - Asim Muhammed Alshanberi
- Department of Community Medicine and Pilgrims Health Care, Umm Alqura University, Makkah, Saudi Arabia
- Medicine Program, Batterjee Medical College, Jeddah, Saudi Arabia
| | - Ahmed Elgebaly
- Smart Health Academic Unit, University of East London, London, UK
| | - Eshak I Bahbah
- Faculty of Medicine, Al-Azhar University, Damietta, Egypt
| |
Collapse
|
40
|
Das-Earl P, Schreihofer DA, Sumien N, Schreihofer AM. Temporal and region-specific tau hyperphosphorylation in the medulla and forebrain coincides with development of functional changes in male obese Zucker rats. J Neurophysiol 2024; 131:689-708. [PMID: 38416718 PMCID: PMC11305650 DOI: 10.1152/jn.00409.2023] [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: 11/07/2023] [Revised: 02/14/2024] [Accepted: 02/26/2024] [Indexed: 03/01/2024] Open
Abstract
Metabolic syndrome (MetS) is associated with development of tauopathies that contribute to cognitive decline. Without functional leptin receptors, male obese Zucker rats (OZRs) develop MetS, and they have increased phosphorylated tau (ptau) with impaired cognitive function. In addition to regulating energy balance, leptin enhances activation of the hippocampus, which is essential for spatial learning and memory. Whether spatial learning and memory are always impaired in OZRs or develop with MetS is unknown. We hypothesized that male OZRs develop MetS traits that promote regional increases in ptau and functional deficits associated with those brain regions. In the medulla and cortex, tau-pSer199,202 and tau-pSer396 were comparable in juvenile (7-8 wk old) lean Zucker rats (LZRs) and OZRs but increased in 18- to 19-wk-old OZRs. Elevated tau-pSer396 was concentrated in the dorsal vagal complex of the medulla, and by this age OZRs had hypertension with increased arterial pressure variability. In the hippocampus, tau-pSer199,202 and tau-pSer396 were still comparable in 18- to 19-wk-old OZRs and LZRs but elevated in 28- to 29-wk-old OZRs, with emergence of deficits in Morris water maze performance. Comparable escape latencies observed during acquisition in 18- to 19-wk-old OZRs and LZRs were increased in 28- to 29-wk-old OZRs, with greater use of nonspatial search strategies. Increased ptau developed with changes in the insulin/phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway in the hippocampus and cortex but not medulla, suggesting different underlying mechanisms. These data demonstrate that leptin is not required for spatial learning and memory in male OZRs. Furthermore, early development of MetS-associated autonomic dysfunction by the medulla may be predictive of later hippocampal dysfunction and cognitive impairment.NEW & NOTEWORTHY Male obese Zucker rats (OZRs) lack functional leptin receptors and develop metabolic syndrome (MetS). At 16-19 wk, OZRs are insulin resistant, with increased ptau in dorsal medulla and impaired autonomic regulation of AP. At 28-29 wk OZRs develop increased ptau in hippocampus with deficits in spatial learning and memory. Juvenile OZRs lack elevated ptau and these deficits, demonstrating that leptin is not essential for normal function. Elevated ptau and deficits emerge before the onset of diabetes in insulin-resistant OZRs.
Collapse
Affiliation(s)
- Paromita Das-Earl
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Derek A Schreihofer
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Nathalie Sumien
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Ann M Schreihofer
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| |
Collapse
|
41
|
Yu F, Pituch KA, Maxfield M, Baena E, Geda YE, Pruzin JJ, Coon DW, Shaibi GQ. The associations between type 2 diabetes and plasma biomarkers of Alzheimer's disease in the Health and Aging Brain Study: Health Disparities (HABS-HD). PLoS One 2024; 19:e0295749. [PMID: 38558059 PMCID: PMC10984470 DOI: 10.1371/journal.pone.0295749] [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: 03/02/2023] [Accepted: 11/28/2023] [Indexed: 04/04/2024] Open
Abstract
Alzheimer's disease (AD) affects Latinos disproportionately. One of the reasons underlying this disparity may be type 2 diabetes (T2D) that is a risk factor for AD. The purpose of this study was to examine the associations of T2D and AD blood biomarkers and the differences in these associations between Mexican Americans and non-Hispanic Whites. This study was a secondary analysis of baseline data from the observational Health and Aging Brain Study: Health Disparities (HABS-HD) that investigated factors underlying health disparities in AD in Mexican Americans in comparison to non-Hispanic Whites. HABS-HD participants were excluded if they had missing data or were large outliers (z-scores >|4|) on a given AD biomarker. Fasting blood glucose and glycosylated hemoglobin (HbA1c) levels were measured from clinical labs. T2D was diagnosed by licensed clinicians. Plasma amyloid-beta 42 and 40 (Aβ42/42) ratio, total tau (t-tau), and neurofilament light (NfL) were measured via ultra-sensitive Simoa assays. The sample sizes were 1,552 for Aβ42/40 ratio, 1,570 for t-tau, and 1,553 for NfL. Mexican Americans were younger (66.6±8.7 vs. 69.5±8.6) and had more female (64.9% female vs. 55.1%) and fewer years of schooling (9.5±4.6 vs. 15.6±2.5) than non-Hispanic Whites. Mexican Americans differed significantly from non-Hispanic Whites in blood glucose (113.5±36.6 vs. 99.2±17.0) and HbA1c (6.33±1.4 vs. 5.51±0.6) levels, T2D diagnosis (35.3% vs. 11.1%), as well as blood Aβ42/40 ratio (.051±.012 vs. .047±.011), t-tau (2.56±.95 vs. 2.33±.90), and NfL levels (16.3±9.5 vs. 20.3±10.3). Blood glucose, blood HbA1c, and T2D diagnosis were not related to Aβ42/40 ratio and t-tau but explained 3.7% of the variation in NfL (p < .001). Blood glucose and T2D diagnosis were not, while HbA1c was positively (b = 2.31, p < .001, β = 0.26), associated with NfL among Mexican Americans. In contrast, blood glucose, HbA1c, and T2D diagnosis were negatively (b = -0.09, p < .01, β = -0.26), not (b = 0.34, p = .71, β = 0.04), and positively (b = 3.32, p < .01, β = 0.33) associated with NfL, respectively in non-Hispanic Whites. To conclude, blood glucose and HbA1c levels and T2D diagnosis are associated with plasma NfL levels, but not plasma Aβ and t-tau levels. These associations differ in an ethnicity-specific manner and need to be further studied as a potential mechanism underlying AD disparities.
Collapse
Affiliation(s)
- Fang Yu
- Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, Arizona, United States of America
| | - Keenan A. Pituch
- Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, Arizona, United States of America
| | - Molly Maxfield
- Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, Arizona, United States of America
| | - Elsa Baena
- Clinical Neuropsychology Department, Barrow Neurological Institute, Phoenix, Arizona, United States of America
| | - Yonas E. Geda
- Department of Neurology and the Franke Neursciene Education Center, Barrow Neurological Institute, Phoenix, Arizona, United States of America
| | - Jeremy J. Pruzin
- Department of Neurology, Banner Alzheimer’s Institute, Phoenix, Arizona, United States of America
| | - David W. Coon
- Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, Arizona, United States of America
| | - Gabriel Q. Shaibi
- Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, Arizona, United States of America
| | | |
Collapse
|
42
|
Boyd ED, Zhang L, Ding G, Li L, Lu M, Li Q, Huang R, Kaur J, Hu J, Chopp M, Zhang Z, Jiang Q. The Glymphatic Response to the Development of Type 2 Diabetes. Biomedicines 2024; 12:401. [PMID: 38398003 PMCID: PMC10886551 DOI: 10.3390/biomedicines12020401] [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: 01/05/2024] [Revised: 01/21/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
The glymphatic system has recently been shown to be important in neurological diseases, including diabetes. However, little is known about how the progressive onset of diabetes affects the glymphatic system. The aim of this study is to investigate the glymphatic system response to the progressive onset of diabetes in a rat model of type 2 diabetic mellitus. Male Wistar rats (n = 45) with and without diabetes were evaluated using MRI glymphatic tracer kinetics, functional tests, and brain tissue immunohistochemistry. Our data demonstrated that the contrast agent clearance impairment gradually progressed with the diabetic duration. The MRI data showed that an impairment in contrast clearance occurred prior to the cognitive deficits detected using functional tests and permitted the detection of an early DM stage compared to the immuno-histopathology and cognitive tests. Additionally, the quantitative MRI markers of brain waste clearance demonstrated region-dependent sensitivity in glymphatic impairment. The improved sensitivity of MRI markers in the olfactory bulb and the whole brain at an early DM stage may be attributed to the important role of the olfactory bulb in the parenchymal efflux pathway. MRI can provide sensitive quantitative markers of glymphatic impairment during the progression of DM and can be used as a valuable tool for the early diagnosis of DM with a potential for clinical application.
Collapse
Affiliation(s)
- Edward D. Boyd
- Department of Neurology, Henry Ford Health System, E&R B126, 2799 West Grand Boulevard, Detroit, MI 48202, USA; (L.Z.); (G.D.); (L.L.); (Q.L.); (J.K.); (M.C.); (Z.Z.); (Q.J.)
- Department of Radiology, Michigan State University, East Lansing, MI 48824, USA
| | - Li Zhang
- Department of Neurology, Henry Ford Health System, E&R B126, 2799 West Grand Boulevard, Detroit, MI 48202, USA; (L.Z.); (G.D.); (L.L.); (Q.L.); (J.K.); (M.C.); (Z.Z.); (Q.J.)
| | - Guangliang Ding
- Department of Neurology, Henry Ford Health System, E&R B126, 2799 West Grand Boulevard, Detroit, MI 48202, USA; (L.Z.); (G.D.); (L.L.); (Q.L.); (J.K.); (M.C.); (Z.Z.); (Q.J.)
- Department of Radiology, Michigan State University, East Lansing, MI 48824, USA
| | - Lian Li
- Department of Neurology, Henry Ford Health System, E&R B126, 2799 West Grand Boulevard, Detroit, MI 48202, USA; (L.Z.); (G.D.); (L.L.); (Q.L.); (J.K.); (M.C.); (Z.Z.); (Q.J.)
| | - Mei Lu
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI 48202, USA;
| | - Qingjiang Li
- Department of Neurology, Henry Ford Health System, E&R B126, 2799 West Grand Boulevard, Detroit, MI 48202, USA; (L.Z.); (G.D.); (L.L.); (Q.L.); (J.K.); (M.C.); (Z.Z.); (Q.J.)
| | - Rui Huang
- Department of Neurology, Henry Ford Health System, E&R B126, 2799 West Grand Boulevard, Detroit, MI 48202, USA; (L.Z.); (G.D.); (L.L.); (Q.L.); (J.K.); (M.C.); (Z.Z.); (Q.J.)
| | - Jasleen Kaur
- Department of Neurology, Henry Ford Health System, E&R B126, 2799 West Grand Boulevard, Detroit, MI 48202, USA; (L.Z.); (G.D.); (L.L.); (Q.L.); (J.K.); (M.C.); (Z.Z.); (Q.J.)
- Department of Physics, Oakland University, Rochester, MI 48309, USA
| | - Jiani Hu
- Department of Radiology, Wayne State University, Detroit, MI 48202, USA;
| | - Michael Chopp
- Department of Neurology, Henry Ford Health System, E&R B126, 2799 West Grand Boulevard, Detroit, MI 48202, USA; (L.Z.); (G.D.); (L.L.); (Q.L.); (J.K.); (M.C.); (Z.Z.); (Q.J.)
- Department of Physics, Oakland University, Rochester, MI 48309, USA
- Department of Neurology, Wayne State University, Detroit, MI 28202, USA
| | - Zhenggang Zhang
- Department of Neurology, Henry Ford Health System, E&R B126, 2799 West Grand Boulevard, Detroit, MI 48202, USA; (L.Z.); (G.D.); (L.L.); (Q.L.); (J.K.); (M.C.); (Z.Z.); (Q.J.)
- Department of Neurology, Wayne State University, Detroit, MI 28202, USA
| | - Quan Jiang
- Department of Neurology, Henry Ford Health System, E&R B126, 2799 West Grand Boulevard, Detroit, MI 48202, USA; (L.Z.); (G.D.); (L.L.); (Q.L.); (J.K.); (M.C.); (Z.Z.); (Q.J.)
- Department of Radiology, Michigan State University, East Lansing, MI 48824, USA
- Department of Physics, Oakland University, Rochester, MI 48309, USA
- Department of Neurology, Wayne State University, Detroit, MI 28202, USA
| |
Collapse
|
43
|
Wang K, Zhao S, Lee EKP, Yau SZM, Wu Y, Hung CT, Yeoh EK. Risk of Dementia Among Patients With Diabetes in a Multidisciplinary, Primary Care Management Program. JAMA Netw Open 2024; 7:e2355733. [PMID: 38345817 PMCID: PMC10862158 DOI: 10.1001/jamanetworkopen.2023.55733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 12/19/2023] [Indexed: 02/15/2024] Open
Abstract
Importance Although poorly controlled diabetes is associated with a higher incidence of dementia, few studies have examined the association of diabetes management interventions with dementia incidence. Objective To examine the association of receiving a multidisciplinary diabetes management program (the Risk Assessment and Management Program-Diabetes Mellitus [RAMP-DM]) that enables better glycemic control with subsequent risk of dementia incidence and the association of dementia with glycemic control. Design, Setting, and Participants This territory-wide, retrospective, matched cohort study with more than 8 years of follow-up was conducted using electronic health care records from all the patients who used public health care services in Hong Kong from 2011 to 2019. Eligible participants included all patients with type 2 diabetes (T2D) who were managed in primary care settings. Patients who received RAMP-DM were matched in a 1:1 ratio with patients who received usual care only. Data analysis occurred from April 2023 to July 2023. Exposures Diagnosis of T2D, hemoglobin A1C (HbA1C) level, and attendance at a general outpatient clinic or family medicine clinic. Patients received either RAMP-DM or usual care. Main Outcomes and Measures Incidence of all-cause dementia and subtypes of dementia were compared between the RAMP-DM and usual care participants using a Cox proportional hazard model with other baseline characteristics, biomarkers, and medication history adjusted. HbA1C levels were measured as a secondary outcome. Results Among the 55 618 matched participants (mean [SD] age, 62.28 [11.90] years; 28 561 female [51.4%]; 27 057 male [48.6%]), including the 27 809 patients in the RAMP-DM group and 27 809 patients in the usual care group, patients had been diagnosed with T2D for a mean (SD) of 5.90 (4.20) years. During a median (IQR) follow-up period of 8.4 (6.8-8.8) years, 1938 patients in the RAMP-DM group (6.97%) and 2728 patients in the usual care group (9.81%) received a diagnosis of dementia. Compared with those receiving usual care, RAMP-DM participants had a lower risk of developing all-cause dementia (adjusted hazard ratio [aHR], 0.72; 95% CI, 0.68-0.77; P < .001), Alzheimer disease (aHR, 0.85; 95% CI, 0.76-0.96; P = .009), vascular dementia (aHR, 0.61; 95% CI, 0.51-0.73; P < .001), and other or unspecified dementia (aHR, 0.71; 95% CI, 0.66-0.77; P < .001). Compared with having a mean HbA1C level during the first 3 years after cohort entry between 6.5% and 7.5%, a higher risk of dementia incidence was detected for patients with a 3-year mean HbA1C level greater than 8.5% (aHR, 1.54; 95% CI, 1.31-1.80]), between 7.5% and 8.5% (aHR, 1.33; 95% CI, 1.19-1.48), between 6% and 6.5% (aHR, 1.17; 95% CI, 1.07-1.29), and 6% or less (aHR, 1.39; 95% CI, 1.24-1.57). Conclusions and Relevance In this cohort study of patients with T2D, the findings strengthened evidence of an association of glycemic control with dementia incidence, and revealed that a multidisciplinary primary care diabetes management program was associated with beneficial outcomes for T2D patients against dementia and its major subtypes. A moderate glycemic control target of HbA1C between 6.5% and 7.5% was associated with lower dementia incidence.
Collapse
Affiliation(s)
- Kailu Wang
- Centre for Health Systems and Policy Research, Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Shi Zhao
- School of Public Health, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Environment, Nutrition, and Public Health, Tianjin Medical University, Tianjin, China
- Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Eric Kam-Pui Lee
- Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Susan Zi-May Yau
- Centre for Health Systems and Policy Research, Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yushan Wu
- Centre for Health Systems and Policy Research, Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Chi-Tim Hung
- Centre for Health Systems and Policy Research, Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Eng-Kiong Yeoh
- Centre for Health Systems and Policy Research, Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
44
|
Tian CY, Yang QH, Lv HZ, Yue F, Zhou FF. Combined untargeted and targeted lipidomics approaches reveal potential biomarkers in type 2 diabetes mellitus cynomolgus monkeys. J Med Primatol 2024; 53:e12688. [PMID: 38083989 DOI: 10.1111/jmp.12688] [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: 09/24/2023] [Revised: 11/14/2023] [Accepted: 12/01/2023] [Indexed: 02/13/2024]
Abstract
BACKGROUND The significantly increasing incidence of type 2 diabetes mellitus (T2DM) over the last few decades triggers the demands of T2DM animal models to explore the pathogenesis, prevention, and therapy of the disease. The altered lipid metabolism may play an important role in the pathogenesis and progression of T2DM. However, the characterization of molecular lipid species in fasting serum related to T2DM cynomolgus monkeys is still underrecognized. METHODS Untargeted and targeted LC-mass spectrometry (MS)/MS-based lipidomics approaches were applied to characterize and compare the fasting serum lipidomic profiles of T2DM cynomolgus monkeys and the healthy controls. RESULTS Multivariate analysis revealed that 196 and 64 lipid molecules differentially expressed in serum samples using untargeted and targeted lipidomics as the comparison between the disease group and healthy group, respectively. Furthermore, the comparative analysis of differential serum lipid metabolites obtained by untargeted and targeted lipidomics approaches, four common serum lipid species (phosphatidylcholine [18:0_22:4], lysophosphatidylcholine [14:0], phosphatidylethanolamine [PE] [16:1_18:2], and PE [18:0_22:4]) were identified as potential biomarkers and all of which were found to be downregulated. By analyzing the metabolic pathway, glycerophospholipid metabolism was associated with the pathogenesis of T2DM cynomolgus monkeys. CONCLUSION The study found that four downregulated serum lipid species could serve as novel potential biomarkers of T2DM cynomolgus monkeys. Glycerophospholipid metabolism was filtered out as the potential therapeutic target pathway of T2DM progression. Our results showed that the identified biomarkers may offer a novel tool for tracking disease progression and response to therapeutic interventions.
Collapse
Affiliation(s)
- Chao-Yang Tian
- Sanya Research Institute of Hainan University, School of Biomedical Engineering, Hainan University, Sanya, China
- Key Laboratory of Biomedical Engineering of Hainan Province, One Health Institute, Hainan University, Haikou, China
| | | | - Hai-Zhou Lv
- Hainan Jingang Biotech Co., Ltd, Haikou, China
| | - Feng Yue
- Sanya Research Institute of Hainan University, School of Biomedical Engineering, Hainan University, Sanya, China
- Key Laboratory of Biomedical Engineering of Hainan Province, One Health Institute, Hainan University, Haikou, China
| | - Fei-Fan Zhou
- Sanya Research Institute of Hainan University, School of Biomedical Engineering, Hainan University, Sanya, China
- Key Laboratory of Biomedical Engineering of Hainan Province, One Health Institute, Hainan University, Haikou, China
| |
Collapse
|
45
|
Gao Y, Yu H, Liu Y, Xu Z, He B, Liu H, Wang Y, Zhang Y, Liang Y, Yang Y, Zheng J, Wang J. GSK-3β activation mediates apolipoprotein E4-associated cognitive impairment in type 2 diabetes mellitus: A multicenter, cross-sectional study. J Diabetes 2024; 16:e13470. [PMID: 37700547 PMCID: PMC10809305 DOI: 10.1111/1753-0407.13470] [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: 05/05/2023] [Revised: 07/17/2023] [Accepted: 08/16/2023] [Indexed: 09/14/2023] Open
Abstract
AIM Both the activation of glycogen synthase kinase-3β (GSK-3β) and the presence of ApoE ε4 genotype have been found to respectively correlate with cognitive decline in patients with type 2 diabetes mellitus (T2DM), who further show a high incidence of developing Alzheimer's disease. However, the relationship between ApoE ε4 and GSK-3β in the cognitive impairment of T2DM patients remains unclear. METHODS ApoE genotypes and platelet GSK-3β level were measured in 1139 T2DM patients recruited from five medical centers in Wuhan, China. Cognitive functions were assessed by Mini-Mental State Examination (MMSE). The association and the relationships among apolipoprotein E (ApoE) genotypes, GSK-3β activity and cognitive function were analyzed by regression and mediating effect analyses, respectively. RESULTS T2DM patients with ApoE ε4 but not ApoE ε2 haplotype showed poorer cognitive function and elevated platelet GSK-3β activity, when using ApoE ε3 as reference. The elevation of GSK-3β activity was positively correlated the diabetes duration, as well as plasma glycated hemoglobin (HbA1c) and glucose levels. Moreover, correlation and regression analysis also revealed significant pairwise correlations among GSK-3β activity, ApoE gene polymorphism and cognitive function. Lastly, using Baron and Kenny modeling, we unveiled a mediative role of GSK-3β activity between ApoE ε4 and cognitive impairment. CONCLUSION We reported here that the upregulation of GSK-3β activity mediates the exacerbation of cognitive impairment by ApoE ε4-enhanced cognitive impairment in T2DM patients, suggesting GSK-3β inhibitors as promising drugs for preserving cognitive function in T2DM patients, especially to those with ApoE ε4 genotype.
Collapse
Affiliation(s)
- Yang Gao
- Department of Pathophysiology, School of Basic Medicine, Ministry of Education Key Laboratory for Neurological Disorders, Hubei Key Laboratory for Neurological DisordersTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
- Department of RadiologyWuhan Brain HospitalWuhanChina
| | - Haitao Yu
- Department of Fundamental Medicine, Wuxi School of MedicineJiangnan UniversityWuxiChina
| | - Yanchao Liu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Zhipeng Xu
- Department of NeurologyZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Benrong He
- Department of Pathophysiology, School of Basic Medicine, Ministry of Education Key Laboratory for Neurological Disorders, Hubei Key Laboratory for Neurological DisordersTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Honghai Liu
- School of Medicine and Health Management, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yuying Wang
- Department of Pathophysiology, School of Basic Medicine, Ministry of Education Key Laboratory for Neurological Disorders, Hubei Key Laboratory for Neurological DisordersTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Yao Zhang
- Li‐Yuan Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yi Liang
- Department of RadiologyWuhan Brain HospitalWuhanChina
| | - Ying Yang
- Department of Pathophysiology, School of Basic Medicine, Ministry of Education Key Laboratory for Neurological Disorders, Hubei Key Laboratory for Neurological DisordersTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Jie Zheng
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical SciencesPeking University; Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking UniversityBeijingChina
| | - Jian‐Zhi Wang
- Department of Pathophysiology, School of Basic Medicine, Ministry of Education Key Laboratory for Neurological Disorders, Hubei Key Laboratory for Neurological DisordersTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
- Co‐innovation Center of NeuroregenerationNantong UniversityNantongChina
| |
Collapse
|
46
|
Abubakar M, Nama L, Ansari MA, Ansari MM, Bhardwaj S, Daksh R, Syamala KLV, Jamadade MS, Chhabra V, Kumar D, Kumar N. GLP-1/GIP Agonist as an Intriguing and Ultimate Remedy for Combating Alzheimer's Disease through its Supporting DPP4 Inhibitors: A Review. Curr Top Med Chem 2024; 24:1635-1664. [PMID: 38803170 DOI: 10.2174/0115680266293416240515075450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 04/14/2024] [Accepted: 04/22/2024] [Indexed: 05/29/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) is a widespread neurological illness in the elderly, which impacted about 50 million people globally in 2020. Type 2 diabetes has been identified as a risk factor. Insulin and incretins are substances that have various impacts on neurodegenerative processes. Preclinical research has shown that GLP-1 receptor agonists decrease neuroinflammation, tau phosphorylation, amyloid deposition, synaptic function, and memory formation. Phase 2 and 3 studies are now occurring in Alzheimer's disease populations. In this article, we present a detailed assessment of the therapeutic potential of GLP-1 analogues and DPP4 inhibitors in Alzheimer's disease. AIM This study aimed to gain insight into how GLP-1 analogues and associated antagonists of DPP4 safeguard against AD. METHODS This study uses terms from search engines, such as Scopus, PubMed, and Google Scholar, to explore the role, function, and treatment options of the GLP-1 analogue for AD. RESULTS The review suggested that GLP-1 analogues may be useful for treating AD because they have been linked to anti-inflammatory, neurotrophic, and neuroprotective characteristics. Throughout this review, we discuss the underlying causes of AD and how GLP signaling functions. CONCLUSION With a focus on AD, the molecular and pharmacological effects of a few GLP-1/GIP analogs, both synthetic and natural, as well as DPP4 inhibitors, have been mentioned, which are in the preclinical and clinical studies. This has been demonstrated to improve cognitive function in Alzheimer's patients.
Collapse
Affiliation(s)
- Mohammad Abubakar
- Department of Pharmacology and Toxicology, National Institution of Pharmaceutical Education and Research, Hajipur, Vaishali, 844102, Bihar, India
| | - Lokesh Nama
- Department of Pharmacology and Toxicology, National Institution of Pharmaceutical Education and Research, Hajipur, Vaishali, 844102, Bihar, India
| | - Mohammad Arif Ansari
- Department of Pharmacology and Toxicology, National Institution of Pharmaceutical Education and Research, Hajipur, Vaishali, 844102, Bihar, India
| | - Mohammad Mazharuddin Ansari
- Department of Pharmacology and Toxicology, National Institution of Pharmaceutical Education and Research, Hajipur, Vaishali, 844102, Bihar, India
| | - Shivani Bhardwaj
- Department of Pharmacology and Toxicology, National Institution of Pharmaceutical Education and Research, Hajipur, Vaishali, 844102, Bihar, India
| | - Rajni Daksh
- Department of Pharmacology and Toxicology, National Institution of Pharmaceutical Education and Research, Hajipur, Vaishali, 844102, Bihar, India
| | - Katta Leela Venkata Syamala
- Department of Regulatory and Toxicology, National Institution of Pharmaceutical Education and Research, Hajipur, Vaishali, 844102, Bihar, India
| | - Mohini Santosh Jamadade
- Department of Pharmacology and Toxicology, National Institution of Pharmaceutical Education and Research, Hajipur, Vaishali, 844102, Bihar, India
| | - Vishal Chhabra
- Department of Pharmacology and Toxicology, National Institution of Pharmaceutical Education and Research, Hajipur, Vaishali, 844102, Bihar, India
| | - Dileep Kumar
- Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune, Maharashtra, 411038, India
- Department of Entomology, University of California, Davis, One Shields Ave, Davis, CA, 95616, USA
| | - Nitesh Kumar
- Department of Pharmacology and Toxicology, National Institution of Pharmaceutical Education and Research, Hajipur, Vaishali, 844102, Bihar, India
| |
Collapse
|
47
|
Sakib MN, Ramezan R, Hall PA. Diabetes status and cognitive function in middle-aged and older adults in the Canadian longitudinal study on aging. Front Endocrinol (Lausanne) 2023; 14:1293988. [PMID: 38107512 PMCID: PMC10722407 DOI: 10.3389/fendo.2023.1293988] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 11/07/2023] [Indexed: 12/19/2023] Open
Abstract
Objectives Diabetes is recognized as a significant risk factor for cognitive impairment. However, this association has not been thoroughly examined using large-scale population-based datasets in the Canadian context. The objective of this study was to investigate the potential association between cognitive function and diabetes in a large population-based sample of middle-aged and older Canadians. Methods We utilized baseline data from the Canadian Longitudinal Study on Aging (N=30,097) to test our hypotheses, using five indicators of cognitive function (animal fluency, Stroop interference, reaction time, immediate and delayed memory recall). We conducted multivariate multivariable linear regression and subsequently performed tests for moderation analysis with lifestyle factors and health status. Results The analysis revealed that type 2 diabetes (T2DM) was associated with lower performance on most cognitive tasks, including those assessing executive function (b=0.60, 95% CI 0.31 to 0.90), reaction time (b=16.94, 95% CI 9.18 to 24.70), immediate memory recall (b=-0.10, 95% CI -0.18 to -0.02), and delayed memory recall (b=-0.12, 95% CI -0.21 to -0.02). However, no significant association was observed between other types of diabetes and cognitive performance. Moderation effects were largely null for T2DM, with the exception of alcohol intake for reaction time, and physical activity for animal fluency. Conclusions The study showed that individuals with T2DM exhibit poor performance on tasks that assess executive function, reaction time, and memory. Therefore, optimizing cognitive health among individuals with T2DM should be a priority in primary care. Additionally, further studies should examine this association using longitudinal data.
Collapse
Affiliation(s)
- Mohammad Nazmus Sakib
- Department of Health Research Methods, Evidence, and Impact, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
- School of Public Health Sciences, Faculty of Health, University of Waterloo, Waterloo, ON, Canada
| | - Reza Ramezan
- Department of Statistics and Actuarial Science, University of Waterloo, Waterloo, ON, Canada
| | - Peter A. Hall
- School of Public Health Sciences, Faculty of Health, University of Waterloo, Waterloo, ON, Canada
- Centre for Bioengineering and Biotechnology, University of Waterloo, Waterloo, ON, Canada
- Department of Psychology, University of Waterloo, Waterloo, ON, Canada
| |
Collapse
|
48
|
Ruegsegger GN, Ekholm ER, Monroe CE, Rappaport CI, Huppert RD, Anton CR, Ferguson MJ. Glucose tolerance status associates with improvements in cognitive function following high-intensity exercise in adults with obesity. Physiol Behav 2023; 272:114389. [PMID: 37890604 DOI: 10.1016/j.physbeh.2023.114389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/28/2023] [Accepted: 10/24/2023] [Indexed: 10/29/2023]
Abstract
PURPOSE Obesity, insulin resistance (IR), and proinflammatory cytokines associate with cognitive decline. Numerous studies document cognitive benefits of acute exercise bouts in lean individuals. However, how co-morbidities such as obesity and IR influence cognitive changes induced by acute exercise is unclear. We examined the effects of acute high-intensity aerobic exercise on cognitive function in age-matched and BMI-matched obese adults with normal glucose tolerance (NGT) or impaired glucose tolerance (IGT) and in lean, NGT adults. METHODS 49 adults (15 Lean, 18 Obese-NGT, 16 Obese-IGT) performed one session of high-intensity interval exercise (four cycles of 4-min at 75% Wmax with 3-min rest). Cognitive function testing and blood sampling were performed pre- and post-exercise. RESULTS Following exercise, measurements of executive function and working memory were improved in Lean and Obese-NGT (p < 0.05), but not Obese-IGT. Changes in cognitive function following exercise negatively correlated with 2-hr glucose during an OGTT after controlling for body weight and body composition (rp = -0.40, p = 0.007). Serum levels of inflammatory cytokines IL-6 and CRP remained increased 60-minutes post-exercise in Obese-IGT, but not in Lean or Obese-NGT, which positively associated with 2-hr glucose during an OGTT (p < 0.01) and negatively with changes in cognitive function following exercise (p < 0.01). Greater insulin levels in Obese-IGT post-exercise also negatively correlated with changes in cognitive function following exercise (p < 0.01). CONCLUSION Improvements in cognition following acute high-intensity exercise positively associate with glucose tolerance, independent of body weight and body composition. Further, poorer changes in cognitive performance following exercise associate with persistent peripheral inflammation.
Collapse
Affiliation(s)
- Gregory N Ruegsegger
- Department of Health and Human Performance, University of Wisconsin-River Falls, A173 Falcon Center, 410 S. 3rd St., River Falls, WI, 54022, United States.
| | - Emily R Ekholm
- Department of Health and Human Performance, University of Wisconsin-River Falls, A173 Falcon Center, 410 S. 3rd St., River Falls, WI, 54022, United States
| | - Chandler E Monroe
- Department of Health and Human Performance, University of Wisconsin-River Falls, A173 Falcon Center, 410 S. 3rd St., River Falls, WI, 54022, United States
| | - Chapin I Rappaport
- Department of Health and Human Performance, University of Wisconsin-River Falls, A173 Falcon Center, 410 S. 3rd St., River Falls, WI, 54022, United States
| | - Rocco D Huppert
- Department of Health and Human Performance, University of Wisconsin-River Falls, A173 Falcon Center, 410 S. 3rd St., River Falls, WI, 54022, United States
| | - Caleb R Anton
- Department of Health and Human Performance, University of Wisconsin-River Falls, A173 Falcon Center, 410 S. 3rd St., River Falls, WI, 54022, United States
| | - Mia J Ferguson
- Department of Health and Human Performance, University of Wisconsin-River Falls, A173 Falcon Center, 410 S. 3rd St., River Falls, WI, 54022, United States
| |
Collapse
|
49
|
Zegarra-Valdivia JA, Pignatelli J, Nuñez A, Torres Aleman I. The Role of Insulin-like Growth Factor I in Mechanisms of Resilience and Vulnerability to Sporadic Alzheimer's Disease. Int J Mol Sci 2023; 24:16440. [PMID: 38003628 PMCID: PMC10671249 DOI: 10.3390/ijms242216440] [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: 10/11/2023] [Revised: 11/06/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Despite decades of intense research, disease-modifying therapeutic approaches for Alzheimer's disease (AD) are still very much needed. Apart from the extensively analyzed tau and amyloid pathological cascades, two promising avenues of research that may eventually identify new druggable targets for AD are based on a better understanding of the mechanisms of resilience and vulnerability to this condition. We argue that insulin-like growth factor I (IGF-I) activity in the brain provides a common substrate for the mechanisms of resilience and vulnerability to AD. We postulate that preserved brain IGF-I activity contributes to resilience to AD pathology as this growth factor intervenes in all the major pathological cascades considered to be involved in AD, including metabolic impairment, altered proteostasis, and inflammation, to name the three that are considered to be the most important ones. Conversely, disturbed IGF-I activity is found in many AD risk factors, such as old age, type 2 diabetes, imbalanced diet, sedentary life, sociality, stroke, stress, and low education, whereas the Apolipoprotein (Apo) E4 genotype and traumatic brain injury may also be influenced by brain IGF-I activity. Accordingly, IGF-I activity should be taken into consideration when analyzing these processes, while its preservation will predictably help prevent the progress of AD pathology. Thus, we need to define IGF-I activity in all these conditions and develop a means to preserve it. However, defining brain IGF-I activity cannot be solely based on humoral or tissue levels of this neurotrophic factor, and new functionally based assessments need to be developed.
Collapse
Affiliation(s)
- Jonathan A. Zegarra-Valdivia
- Achucarro Basque Center for Neuroscience, 48940 Leioa, Spain;
- Biomedical Research Networking Center on Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain;
- School of Medicine, Universidad Señor de Sipán, Chiclayo 14000, Peru
| | - Jaime Pignatelli
- Biomedical Research Networking Center on Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain;
- Cajal Institute (CSIC), 28002 Madrid, Spain
| | - Angel Nuñez
- Department of Anatomy, Histology and Neuroscience, Universidad Autónoma de Madrid, 28049 Madrid, Spain;
| | - Ignacio Torres Aleman
- Achucarro Basque Center for Neuroscience, 48940 Leioa, Spain;
- Biomedical Research Networking Center on Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain;
- Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
| |
Collapse
|
50
|
Li J, Xue C, Yang H, Zhang J, Li G, Li J, Kuang F, Chen J, Zhang S, Gao F, Kou Z, Zhang X, Dong L. Simulated weightlessness induces hippocampal insulin resistance and cognitive impairment. Life Sci 2023; 333:122112. [PMID: 37758017 DOI: 10.1016/j.lfs.2023.122112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 09/05/2023] [Accepted: 09/18/2023] [Indexed: 10/03/2023]
Abstract
Growing evidence highlights the potential consequences of long-term spaceflight, including gray matter volume reduction and cognitive dysfunction with subclinical manifestations of diabetes mellitus among astronauts, but the underlying mechanisms remain unknown. In this study, we found that long-term simulated weightlessness induced hippocampal insulin resistance and subsequent neuronal damage and cognitive impairment in rats. Rats subjected to 4-week tail suspension exhibited peripheral insulin resistance, evidenced by increased fasting blood glucose and abnormal glucose tolerance and insulin tolerance, alongside reduced spontaneous activity and impaired recognition memory. In addition, 4 weeks of simulated weightlessness induced neuronal apoptosis and degeneration in the hippocampus, as evidenced by increased TUNEL and Fluoro-Jade B staining-positive neurons. Mechanistically, insulin-stimulated hippocampal Akt phosphorylation was decreased, while PTEN, the negative regulator of insulin signaling, was increased in the hippocampus in tail-suspended rats. Interestingly, treatment with berberine, an insulin sensitizer, partly reversed the above-mentioned effects induced by simulated weightlessness. These data suggest that long-term simulated weightlessness induces cognitive impairment as well as neuronal apoptosis and neural degeneration, partially through hippocampal insulin resistance via PTEN up-regulation. Berberine treatment attenuates hippocampal insulin resistance and improves cognitive function.
Collapse
Affiliation(s)
- Jiahui Li
- Key Laboratory of Aerospace Medicine of the Ministry of Education, School of Aerospace Medicine, Air Force Medical University, Xi'an, China; Department of Psychology, Air Force Hospital, Western Theater Command, Chengdu, China
| | - Caiyan Xue
- Key Laboratory of Aerospace Medicine of the Ministry of Education, School of Aerospace Medicine, Air Force Medical University, Xi'an, China
| | - Hongyan Yang
- Key Laboratory of Aerospace Medicine of the Ministry of Education, School of Aerospace Medicine, Air Force Medical University, Xi'an, China
| | - Jiaxin Zhang
- Key Laboratory of Aerospace Medicine of the Ministry of Education, School of Aerospace Medicine, Air Force Medical University, Xi'an, China
| | - Guohua Li
- Key Laboratory of Aerospace Medicine of the Ministry of Education, School of Aerospace Medicine, Air Force Medical University, Xi'an, China
| | - Jijun Li
- Key Laboratory of Aerospace Medicine of the Ministry of Education, School of Aerospace Medicine, Air Force Medical University, Xi'an, China
| | - Fang Kuang
- Department of Neurobiology, Air Force Medical University, Xi'an, China
| | - Jing Chen
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Air Force Medical University, Xi'an, China
| | - Shu Zhang
- Key Laboratory of Aerospace Medicine of the Ministry of Education, School of Aerospace Medicine, Air Force Medical University, Xi'an, China
| | - Feng Gao
- Key Laboratory of Aerospace Medicine of the Ministry of Education, School of Aerospace Medicine, Air Force Medical University, Xi'an, China
| | - Zhenzhen Kou
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Air Force Medical University, Xi'an, China.
| | - Xing Zhang
- Key Laboratory of Aerospace Medicine of the Ministry of Education, School of Aerospace Medicine, Air Force Medical University, Xi'an, China.
| | - Ling Dong
- Key Laboratory of Aerospace Medicine of the Ministry of Education, School of Aerospace Medicine, Air Force Medical University, Xi'an, China.
| |
Collapse
|