1
|
Agadagba SK, Yau SY, Liang Y, Dalton K, Thompson B. Bidirectional causality of physical exercise in retinal neuroprotection. Neural Regen Res 2025; 20:3400-3415. [PMID: 39688575 PMCID: PMC11974656 DOI: 10.4103/nrr.nrr-d-24-00942] [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: 08/16/2024] [Revised: 10/21/2024] [Accepted: 11/16/2024] [Indexed: 12/18/2024] Open
Abstract
Physical exercise is recognized as an effective intervention to improve mood, physical performance, and general well-being. It achieves these benefits through cellular and molecular mechanisms that promote the release of neuroprotective factors. Interestingly, reduced levels of physical exercise have been implicated in several central nervous system diseases, including ocular disorders. Emerging evidence has suggested that physical exercise levels are significantly lower in individuals with ocular diseases such as glaucoma, age-related macular degeneration, retinitis pigmentosa, and diabetic retinopathy. Physical exercise may have a neuroprotective effect on the retina. Therefore, the association between reduced physical exercise and ocular diseases may involve a bidirectional causal relationship whereby visual impairment leads to reduced physical exercise and decreased exercise exacerbates the development of ocular disease. In this review, we summarize the evidence linking physical exercise to eye disease and identify potential mediators of physical exercise-induced retinal neuroprotection. Finally, we discuss future directions for preclinical and clinical research in exercise and eye health.
Collapse
Affiliation(s)
- Stephen K. Agadagba
- Center for Eye and Vision Research Limited, 17W, Hong Kong Science Park, Hong Kong Special Administrative Region, China
| | - Suk-yu Yau
- Center for Eye and Vision Research Limited, 17W, Hong Kong Science Park, Hong Kong Special Administrative Region, China
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| | - Ying Liang
- Center for Eye and Vision Research Limited, 17W, Hong Kong Science Park, Hong Kong Special Administrative Region, China
| | - Kristine Dalton
- Center for Eye and Vision Research Limited, 17W, Hong Kong Science Park, Hong Kong Special Administrative Region, China
- School of Optometry and Vision Science, University of Waterloo, Waterloo, ON, Canada
| | - Benjamin Thompson
- Center for Eye and Vision Research Limited, 17W, Hong Kong Science Park, Hong Kong Special Administrative Region, China
- School of Optometry and Vision Science, University of Waterloo, Waterloo, ON, Canada
| |
Collapse
|
2
|
Medeiros EB, da Silva S, Mina F, Lidio AV, Boaventura A, de Jesus LC, Garcez ML, Zabot GC, Fenilli GP, Rodrigues MS, de Oliveira J, Valvassori SS, Budni J. Deregulation of Neuroinflammatory and Neurotrophic Factors as Biological Events Triggered by D-Galactose Chronic Administration in Wistar Rats. Neuromolecular Med 2025; 27:41. [PMID: 40397192 DOI: 10.1007/s12017-025-08846-0] [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: 10/01/2024] [Accepted: 03/22/2025] [Indexed: 05/22/2025]
Abstract
The chronic administration of D-galactose (D-gal) is widely used to model brain senescence in rodents. However, the effects of prolonged oral exposure of D-gal on the neuroinflammatory cytokines in rats remain poorly characterized. Therefore, we administered D-gal (100 mg/kg) in male Wistar rats aged 3-4 months, via oral gavage once a day for 1, 2, 4, 6, or 8 weeks. Cytokine and neurotrophin levels were analyzed using the ELISA method. D-gal administrations for 4, 6, and 8 weeks significantly increased interleukin -1β (IL-1β), tumor necrosis factor-alpha (TNF-α), and interleukin-4 (IL-4) levels in the frontal cortex and hippocampus. In addition, 4, 6, and 8 weeks of D-gal administration significantly increased interleukin-10 (IL-10) levels in the frontal cortex; however, in the hippocampus, only 6 and 8 weeks of D-gal administration significantly increased the IL-10 levels. In terms of neurotrophin levels, our results demonstrated that 1 week of D-gal administration significantly increased Brain-derived Neurotrophic Factor (BDNF) and Nerve Growth Factor (NGF) in the hippocampus. In the frontal cortex, D-gal increased BDNF levels when administered for 1 and 2 weeks and increased NGF levels when administered for only 2 weeks. However, we observed a reduction of BDNF, NGF, and Glial cell line-derived Neurotrophic Factor (GDNF) levels after 6 and 8 weeks of D-gal treatment in the frontal cortex. Moreover, GDNF levels also were reduced after 4 weeks of D-gal administration. These findings suggest that oral D-gal exposure disrupts the balance of cytokines and neurotrophins, which may be an essential mechanism in brain aging and neurodegenerative processes.
Collapse
Affiliation(s)
- Eduarda Behenck Medeiros
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences (PPGCS), University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Sabrina da Silva
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences (PPGCS), University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Francielle Mina
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences (PPGCS), University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Adrielly Vargas Lidio
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences (PPGCS), University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Amanda Boaventura
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences (PPGCS), University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Laura Ceolin de Jesus
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences (PPGCS), University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Michelle Lima Garcez
- Department of Clinical Analysis, Postgraduate Program in Pharmacy, Federal University of Santa Catarina -UFSC, Florianópolis, SC, Brazil
| | - Gabriel Casagrande Zabot
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences (PPGCS), University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Gabriela Piovesan Fenilli
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences (PPGCS), University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Matheus Scarpatto Rodrigues
- Department of Biochemistry, Postgraduate Program in Biological Sciences: Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande Do Sul, Porto Alegre, Brazil
| | - Jade de Oliveira
- Department of Biochemistry, Postgraduate Program in Biological Sciences: Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande Do Sul, Porto Alegre, Brazil
| | - Samira S Valvassori
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences (PPGCS), University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Josiane Budni
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences (PPGCS), University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil.
| |
Collapse
|
3
|
Li H, Zheng C, Wen K, Zhang T, Zhang Y. Neurotrophic and Neurotoxic Effects of Aβ42 and Its Oligomers on Neuronal Survival: Revealed by Their Opposite Influence on the Potency of Extracellular BDNF. Int J Mol Sci 2025; 26:4501. [PMID: 40429646 PMCID: PMC12111036 DOI: 10.3390/ijms26104501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 05/02/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is critical for neuronal survival. Amyloid-β monomers (Aβ42M) and oligomers (Aβ42O) have trophic and toxic effects on neuronal survival, respectively. Branched oligosaccharides (BOs) and catechins (CAs) can specifically bind to Aβ42M/Aβ42O, influencing both effects. However, whether and how Aβ42M/Aβ42O influences BDNF remains unknown. This study investigated the interaction between Aβ42M/Aβ42O and BDNF, the effects of Aβ42M and Aβ42O on BDNF binding to the TrkB/p75 receptor and their impact on BDNF-supported cell survival, and the roles of BOs and CAs in these processes. BDNF exhibited stronger binding affinity for Aβ42M and Aβ42O than BOs/CAs. Aβ42M increased neuronal viability by synergistically enhancing BDNF binding to TrkB and p75, whereas Aβ42O decreased neuronal viability by inactivating/consuming BDNF, thereby reducing its binding to these receptors. BDNF-Aβ42O binding appeared to mutually neutralize/counteract each other's biological effects; therefore, increasing BDNF levels might reduce Aβ42O's neurotoxicity. By competitively targeting Aβ42M/Aβ42O rather than BDNF or its receptors, BOs and CAs enhanced these effects. These findings suggest that Aβ42M's neurotrophicity was directly linked to its synergistic enhancement of BDNF activity, whereas Aβ42O's neurotoxicity was primarily due to its inactivation or consumption of BDNF. This study provided valuable insights for developing BOs/CAs-based neuroprotective therapeutics or nanomaterials against AD.
Collapse
Affiliation(s)
| | | | | | | | - Yingjiu Zhang
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China; (H.L.); (C.Z.); (K.W.); (T.Z.)
| |
Collapse
|
4
|
Tchekalarova J, Atanasova D, Krushovlieva D, Barbutska D, Atanasova M, Rashev P, Nenchovska Z, Mourdjeva M, Koeva Y. Age-related memory decline is accelerated by pinealectomy in young adult and middle-aged rats via BDNF / ERK / CREB signalling. Neurochem Int 2025; 185:105960. [PMID: 40043851 DOI: 10.1016/j.neuint.2025.105960] [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: 11/27/2024] [Revised: 02/24/2025] [Accepted: 03/02/2025] [Indexed: 03/10/2025]
Abstract
Memory decline is considered a normal part of aging, while the relationship between melatonin deficiency and cognitive function is complex and not fully understood. The present study investigated the role of melatonin deficiency at different ages on working and short-term recognition and spatial memory in rats. An age-related decline in memory function was tested using the Y-maze, the object recognition test, and the radial arm maze. The brain-derived neurotrophic factor (BDNF), TrkB receptor, the extracellular signal-regulated kinase (ERK)1/2 and pERK1/2 expression in the hippocampus was assessed by immunohistochemistry. The pCREB/CREB ratio in the frontal cortex (FC) and hippocampus was evaluated by ELISA. Young adult and middle-aged rats with pinealectomy had memory impairment whereas old melatonin-deficient rats were unaffected. Aging was associated with reduced expression of BDNF and its receptor throughout the hippocampus and reduced ratio of pCREB/CREB in the FC and hippocampus, whereas pinealectomy exacerbated this process in 3- and 14-month-old rats. The region-specific reduced expression of the ERK1/2 and pERK1/2 was observed in young adult rats with pinealectomy. However, in middle-aged rats, the expression of these signaling molecules was either downregulated or upregulated in different regions of the hippocampus. Our study provides insights into the molecular pathways involved in age-related memory changes associated with melatonin deficiency, highlighting the importance of the BDNF/ERK1/2/CREB pathway in the hippocampus and suggesting a critical period for intervention.
Collapse
Affiliation(s)
- Jana Tchekalarova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, 1113, Bulgaria; Department of Organic Chemistry, University of Chemical Technology and Metallurgy, 1756, Sofia, Bulgaria.
| | - Dimitrinka Atanasova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, 1113, Bulgaria; Department of Anatomy, Faculty of Medicine, Trakia University, Stara Zagora, 6003, Bulgaria
| | | | - Darina Barbutska
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Medical University - Plovdiv, 4002, Bulgaria
| | - Milena Atanasova
- Medical University - Pleven, 1 Kliment Ochridski Str., Pleven, 5800, Bulgaria
| | - Pavel Rashev
- Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, 1113, Bulgaria; Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Zlatina Nenchovska
- Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, 1113, Bulgaria
| | - Milena Mourdjeva
- Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Yvetta Koeva
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Medical University - Plovdiv, 4002, Bulgaria
| |
Collapse
|
5
|
Zheng X, Zhou F, Zhang Q, Zheng W, Shi F, Li R, Lv J, Li Q. Exploring the Effects of Changes in Dietary Protein Content on Naturally Aging Mice Based on Comprehensive Quantitative Scoring and Metabolomic Analysis. Nutrients 2025; 17:1542. [PMID: 40362850 PMCID: PMC12073357 DOI: 10.3390/nu17091542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Revised: 04/28/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND During aging, protein nutrition has a bidirectional role in regulating healthy lifespan by modulating body metabolism and neurological function. However, the current "low-high" hypothesis on the dynamics of protein requirements is mainly based on male animal models, and its applicability to female physiology (e.g., estrogen fluctuations) is unclear. The present study aims to fill the gap in the study of protein demand dynamics in female naturally aging mice and to investigate the effects of different protein levels on the health status of female C57BL/6J mice at different stages of aging. METHODS In this study, four dietary interventions (high protein, HP; low protein, LP; model test, MT; and control, C) were evaluated by constructing a C57BL/6J female mouse model at three ages, 9 M (9 months), 16 M (16 months), and 20 M (20 months), which are approximately equivalent to 34, 65, and 78 years of age in humans, respectively, to determine the effects on naturally aging mice. The effects of the interventions were quantitatively described by behavioral, neuropathological, oxidative, and inflammatory indices and NMR metabolomics using Principal Component Analysis to construct a comprehensive quantitative scoring method. RESULTS The comprehensive quantitative scores Fsum was highest in the HP group, lowest in the LP group, and in between in the MT group. The HP intervention showed the most significant improvement in the aged group (20 M) mice, with a 35.2% reduction in avoidance latency (p < 0.01) and a 32.9% increase in pyramidal cell density in the hippocampal CA1 region (p < 0.05), while the LP intervention led to a cognitive decline in the mice, with an avoidance latency that was prolonged by 15.2% (p < 0.05). Metabolomics analysis revealed that mouse samples of all ages showed age-dependent metabolic re-adaptation: the 9 M group may reflect gut microbial metabolism rather than direct host TCA cycle activity, suggesting an indirect association with energy metabolism; an enhanced degradation of branched-chain amino acids (BCAAs) was seen in the middle-aged group (16 M); and amino acid biosynthesis was predominant in the old group (20 M). CONCLUSIONS Female mice have sustained neuromotor benefits to high-protein diets at different aging stages, and the dynamics of their protein requirements differ significantly from those of males. The study reveals the critical role of gender factors in protein nutritional strategies and provides an experimental basis for precise protein supplementation in older women.
Collapse
Affiliation(s)
- Xiaohua Zheng
- College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China; (X.Z.)
| | - Fan Zhou
- College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China; (X.Z.)
| | - Qinren Zhang
- College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China; (X.Z.)
| | - Wenxuan Zheng
- College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China; (X.Z.)
| | - Fengcui Shi
- College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China; (X.Z.)
| | - Ruiding Li
- College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China; (X.Z.)
| | - Jingwen Lv
- College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China; (X.Z.)
| | - Quanyang Li
- College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China; (X.Z.)
- Guangxi Key Laboratory of Longevity Science and Technology, Nanning 530200, China
| |
Collapse
|
6
|
Pennington KR, Debs L, Chung S, Bava J, Garin CM, Vale FL, Bick SK, Englot DJ, Terry AV, Constantinidis C, Blake DT. Basal forebrain activation improves working memory in senescent monkeys. Brain Stimul 2025; 18:185-194. [PMID: 39924100 PMCID: PMC12076211 DOI: 10.1016/j.brs.2025.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/13/2025] [Accepted: 02/01/2025] [Indexed: 02/11/2025] Open
Abstract
Brain aging contributes to cognitive decline and risk of dementia. Degeneration of the basal forebrain cholinergic system parallels these changes in aging, Alzheimer's dementia, Parkinson's dementia, and Lewy body dementia, and thus is a common element linked to executive function across the lifespan and in disease states. Here, we tested the potential of one-hour daily intermittent basal forebrain stimulation to improve cognition in senescent Rhesus monkeys, and its mechanisms of action. Stimulation in five animals improved working memory duration in each animal over 8-12 weeks, with peak improvements observed in the first four weeks. In an ensuing three month period without stimulation, improvements were retained. With additional stimulation, performance remained above baseline throughout the 15 months of the study. Studies with a cholinesterase inhibitor in five animals produced inconsistent improvements in behavior. One of five animals improved significantly. Manipulating the stimulation pattern demonstrated selectivity for both stimulation and recovery period duration in two animals. Brain stimulation led to acute increases in cerebrospinal fluid levels of tissue plasminogen activator, which is an activating element for two brain neurotrophins, Nerve Growth Factor (NGF) and Brain-Derived Growth Factor (BDNF), in four animals. Stimulation also led to improved glucose utilization in stimulated hemispheres relative to contralateral in three animals. Glucose utilization also consistently declines with aging and some dementias. Together, these findings suggest that intermittent stimulation of the nucleus basalis of Meynert improves executive function and reverses some aspects of brain aging.
Collapse
Affiliation(s)
- Kendyl R Pennington
- Dept Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Luca Debs
- Dept Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Sophia Chung
- Neuroscience Program, Vanderbilt University, Nashville, TN, 37235, USA
| | - Janki Bava
- Dept Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Clément M Garin
- Dept Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Fernando L Vale
- Dept Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Sarah K Bick
- Dept Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA; Dept Neurosurgery, Vanderbilt University, Nashville, TN, USA
| | - Dario J Englot
- Dept Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA; Dept Neurosurgery, Vanderbilt University, Nashville, TN, USA
| | - Alvin V Terry
- Dept Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Christos Constantinidis
- Neuroscience Program, Vanderbilt University, Nashville, TN, 37235, USA; Dept Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA; Dept Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
| | - David T Blake
- Dept Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA; Dept Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, USA.
| |
Collapse
|
7
|
Xu M, Chen Y, Lin Y, Wang D, Zheng X. Serum-derived exosomal microRNAs as biomarkers for postoperative delirium. Front Neurosci 2025; 19:1525230. [PMID: 40092071 PMCID: PMC11906430 DOI: 10.3389/fnins.2025.1525230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 02/11/2025] [Indexed: 03/19/2025] Open
Abstract
Introduction Postoperative delirium (POD) is a frequent and challenging complication in elderly surgical patients, marked by abrupt cognitive and attentional disturbances. Current POD diagnosis depends on clinical assessments that are time-intensive and lack predictive accuracy before surgery. Although previous research has explored biomarkers such as neuroinflammatory factors and Alzheimer's-related proteins to enhance POD prediction, single molecular markers have proven insufficient for reliable prognosis. Methods This study investigated serum exosomal miRNA expression profiles in postoperative patients to assess their association with POD. We compared miRNA expression between POD and non-POD groups through cognitive assessments and serum analyses. Additionally, enrichment analysis was conducted to determine the biological pathways regulated by differentially expressed miRNAs. Results Our analysis identified 57 miRNAs with significantly altered expression between POD and non-POD patients, including 16 upregulated and 41 downregulated miRNAs in the POD group. Enrichment analysis revealed that these miRNAs are involved in genes regulating neurotrophin signaling, neuroactive ligand-receptor interactions, and pathways that influence neuronal plasticity and cell viability. Discussion This study highlights specific miRNAs as potential biomarkers for POD and suggests their involvement in the underlying mechanisms of cognitive decline following surgery. By enhancing diagnostic capabilities and identifying potential therapeutic targets, our findings could lead to more effective POD management strategies for elderly patients. Further research is recommended to validate these miRNAs and evaluate their clinical utility for predictive screening and therapeutic interventions.
Collapse
Affiliation(s)
- Maokai Xu
- Department of Anesthesiology, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
| | - Yingjie Chen
- Department of Anesthesiology, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
| | - Yujun Lin
- Department of Critical Care Medicine, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
| | - Danfeng Wang
- Department of Anesthesiology, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
| | - Xiaochun Zheng
- Department of Anesthesiology, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
| |
Collapse
|
8
|
Safi A, Giunti E, Melikechi O, Xia W, Melikechi N. Identification of blood plasma protein ratios for distinguishing Alzheimer's disease from healthy controls using machine learning. Heliyon 2025; 11:e42349. [PMID: 39981365 PMCID: PMC11840181 DOI: 10.1016/j.heliyon.2025.e42349] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 01/23/2025] [Accepted: 01/28/2025] [Indexed: 02/22/2025] Open
Abstract
Early detection of Alzheimer's disease is essential for effective treatment and the development of therapies that modify disease progression. Developing sensitive and specific noninvasive diagnostic tools is crucial for improving clinical outcomes and advancing our understanding of this condition. Liquid biopsy techniques, especially those involving plasma biomarkers, provide a promising noninvasive method for early diagnosis and disease monitoring. In this study, we analyzed the plasma proteomic profiles of 38 healthy individuals, with an average age of 66.5 years, and 22 patients with Alzheimer's disease, with an average age of 79.7 years. Proteins in the plasma were quantified using specialized panels designed for proteomic extension assays. Through computational analysis using a linear support vector machine algorithm, we identified 82 differentially expressed proteins between the two groups. From these, we calculated 6642 possible protein ratios and identified specific combinations of these ratios as significant features for distinguishing between individuals with Alzheimer's disease and healthy individuals. Notably, the protein ratios kynureninase to macrophage scavenger receptor type 1, Neurocan to protogenin, and interleukin-5 receptor alpha to glial cell line-derived neurotrophic factor receptor alpha 1 achieving accuracy up to 98 % in differentiating between the two groups. This study underscores the potential of leveraging protein relationships, expressed as ratios, in advancing Alzheimer's disease diagnostics. Furthermore, our findings highlight the promise of liquid biopsy techniques as a noninvasive and accurate approach for early detection and monitoring of Alzheimer's disease using blood plasma.
Collapse
Affiliation(s)
- Ali Safi
- Kennedy College of Sciences, University of Massachusetts Lowell, Lowell, MA, 01854, USA
| | - Elisa Giunti
- Bedford VA Healthcare System, Bedford, MA, 01730, USA
- Boston University Chobanian & Avedisian School of Medicine, Boston, MA, 02118, USA
| | - Omar Melikechi
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Weiming Xia
- Kennedy College of Sciences, University of Massachusetts Lowell, Lowell, MA, 01854, USA
- Bedford VA Healthcare System, Bedford, MA, 01730, USA
- Boston University Chobanian & Avedisian School of Medicine, Boston, MA, 02118, USA
| | - Noureddine Melikechi
- Kennedy College of Sciences, University of Massachusetts Lowell, Lowell, MA, 01854, USA
| |
Collapse
|
9
|
Puoyan‐Majd S, Parnow A, Rashno M, Heidarimoghadam R, komaki A. Effects of Pretreatment With Coenzyme Q10 (CoQ10) and High-Intensity Interval Training (HIIT) on FNDC5, Irisin, and BDNF Levels, and Amyloid-Beta (Aβ) Plaque Formation in the Hippocampus of Aβ-Induced Alzheimer's Disease Rats. CNS Neurosci Ther 2025; 31:e70221. [PMID: 39957598 PMCID: PMC11831071 DOI: 10.1111/cns.70221] [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: 12/10/2023] [Revised: 12/23/2024] [Accepted: 01/06/2025] [Indexed: 02/18/2025] Open
Abstract
AIMS Physical exercise has been shown to protect against cognitive decline in Alzheimer's disease (AD), likely through the upregulation of brain-derived neurotrophic factor (BDNF). Recent studies have reported that exercise activates the FNDC5/irisin pathway in the hippocampus of mice, triggering a neuroprotective gene program that includes BDNF. This study aimed to investigate the effects of 8 weeks of pretreatment with coenzyme Q10 (CoQ10) and high-intensity interval training (HIIT), both individually and in combination, on FNDC5, irisin, BDNF, and amyloid-beta (Aβ) plaque formation in the hippocampus of Aβ-related AD rats. METHODS In this study, 72 male Wistar rats were randomly assigned to one of the following groups: control, sham, HIIT (low intensity: 3 min running at 50%-60% VO2max; high intensity: 4 min running at 85%-90% VO2max), Q10 (50 mg/kg, orally administered), Q10 + HIIT, AD, AD + HIIT, AD + Q10, and AD + Q10 + HIIT. RESULTS Aβ injection resulted in a trend toward decreased levels of FNDC5, irisin, and BDNF, alongside increased Aβ plaque formation in the hippocampus of Aβ-induced AD rats. However, pretreatment with CoQ10, HIIT, or their combination significantly restored hippocampal levels of FNDC5, irisin, and BDNF, while also inhibiting Aβ plaque accumulation in these rats. CONCLUSION Pretreatment with CoQ10 and HIIT improved the Aβ-induced reduction in BDNF levels probably through the FNDC5/irisin pathway and preventing Aβ plaque formation.
Collapse
Affiliation(s)
- Samira Puoyan‐Majd
- Bio‐Sciences Department, Physical Education and Sport Sciences FacultyRazi UniversityKermanshahIran
- Neurophysiology Research CenterHamadan University of Medical SciencesHamadanIran
| | - Abdolhossein Parnow
- Bio‐Sciences Department, Physical Education and Sport Sciences FacultyRazi UniversityKermanshahIran
| | - Masome Rashno
- Asadabad School of Medical SciencesAsadabadIran
- Student Research Committee, Asadabad School of Medical SciencesAsadabadIran
| | - Rashid Heidarimoghadam
- Department of ErgonomicsSchool of Health, Hamadan University of Medical SciencesHamadanIran
| | - Alireza komaki
- Neurophysiology Research CenterHamadan University of Medical SciencesHamadanIran
- Department of NeuroscienceSchool of Science and Advanced Technologies in Medicine, Hamadan University of Medical SciencesHamadanIran
| |
Collapse
|
10
|
Dewi MM, Imron A, Risan NA, Mediana G, Judistiani RTD, Setiabudiawan B. The Association of Vitamin D, Nerve Growth Factor (NGF), Brain-Derived Neurotrophic Factor (BDNF), and Glial Cell-Derived Neurotrophic Factor (GDNF) with Development in Children. CHILDREN (BASEL, SWITZERLAND) 2025; 12:60. [PMID: 39857891 PMCID: PMC11763926 DOI: 10.3390/children12010060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 12/26/2024] [Accepted: 12/30/2024] [Indexed: 01/27/2025]
Abstract
BACKGROUND Short stature remains a global problem and is associated with vitamin D status. Vitamin D is also a neurosteroid with regard to neurotrophic factors but its role in development is unclear. Therefore, this study analyzed the relationships between vitamin D, NGF, GDNF, and BDNF and developmental status in children with a history of short stature (<2 years). METHODS This research is a prospective cross-sectional study conducted in March 2022. The vitamin D, NGF, GDNF, and BDNF levels were measured in stored biological materials from children aged 2-4 years, and their Ages and Stages Questionnaire (ASQ-3) scores were also assessed. The results were analyzed via the chi-square test, Fisher's exact test, Mann-Whitney test for NGF, unpaired t-test, and Spearman rank correlation. RESULTS Among the 85 study subjects, 41.2% were short in stature, with 37% having developmental deviation. Male sex (p = 0.038) and low maternal education (p = 0.024) were associated with short stature. The mean vitamin D level was lower (p = 0.041) in children with short stature (27.65 ng/dL). The risk factors associated with short stature were vitamin D levels ≤ 32.7 ng/dL, GDNF levels ≤ 12.99 ng/mL, male sex, and low maternal education. Children with short stature (<2 years old) also demonstrated impaired problem-solving as assessed by the ASQ-3 (p = 0.005). Vitamin D was also associated with gross motor skills (p = 0.035) and personal social development (p = 0.038). CONCLUSIONS There was no association of vitamin D with NGF, GDNF, or BDNF levels. Vitamin D levels are associated with short stature and development in children, especially gross motor skills, personal social development, and problem solving.
Collapse
Affiliation(s)
- Mia Milanti Dewi
- Child Health Department, Faculty of Medicine, Universitas Padjadjaran, Hasan Sadikin Hospital, Bandung 40161, West Java, Indonesia; (N.A.R.); (B.S.)
| | - Akhmad Imron
- Neurosurgery Department, Faculty of Medicine, Universitas Padjadjaran, Hasan Sadikin Hospital, Bandung 40161, West Java, Indonesia;
| | - Nelly Amalia Risan
- Child Health Department, Faculty of Medicine, Universitas Padjadjaran, Hasan Sadikin Hospital, Bandung 40161, West Java, Indonesia; (N.A.R.); (B.S.)
| | - Grace Mediana
- Bandung City Health Service, Bandung 40161, West Java, Indonesia;
| | - Raden Tina Dewi Judistiani
- Public Health Department, Faculty of Medicine, Universitas Padjadjaran, Bandung 40161, West Java, Indonesia;
| | - Budi Setiabudiawan
- Child Health Department, Faculty of Medicine, Universitas Padjadjaran, Hasan Sadikin Hospital, Bandung 40161, West Java, Indonesia; (N.A.R.); (B.S.)
| |
Collapse
|
11
|
Mitra S, Gera R, Eriksdotter M. NGF-based cholinergic therapies in Alzheimer disease. HANDBOOK OF CLINICAL NEUROLOGY 2025; 211:123-135. [PMID: 40340057 DOI: 10.1016/b978-0-443-19088-9.00007-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Abstract
The cholinergic system is part of the parasympathetic nervous system, which works in tandem with the sympathetic and enteric nervous systems to maintain the physiologic functioning of our body. The neurotransmitter acetylcholine (ACh) facilitates cholinergic signaling pathways by activating specific cell surface receptors (nicotinic and muscarinic receptors). Altered cholinergic signaling has been implicated in several pathologic conditions. In this chapter, conditions where cholinergic modulation in the central nervous system occurs through the neurotrophin nerve growth factor (NGF) are addressed. NGF is the master regulator of several pathways, ultimately leading to cell survival, ACh production, regenerative signaling, and anti-inflammatory tone. NGF and cholinergic-related pathways have been reported to be severely affected in the case of Alzheimer disease (AD), the most common dementia disorder. In AD, the cholinergic nuclei of the basal forebrain are affected early during the AD continuum, resulting in cholinergic cell loss and hampered ACh production, which overall affects the propagation of cholinergic signals in other brain regions. Since the 1990s clinically relevant strategies to treat AD patients have been the drugs that enhance cholinergic signaling-termed cholinesterase inhibitors (ChEIs), however, other strategies in AD have been and are presently being assessed for clinical efficacy. Delivery of NGF to the basal forebrain is considered crucial to revive the cholinergic cell bodies, restore ACh production, and sustain cognitive function. This chapter provides a description of the relevance of NGF-based therapies targeted for AD treatment, technical approaches for NGF delivery to the brain, and the status of ongoing clinical studies are provided.
Collapse
Affiliation(s)
- Sumonto Mitra
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Ruchi Gera
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Maria Eriksdotter
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Theme Inflammation and Aging, Medical Unit Aging, Karolinska University Hospital, Huddinge, Sweden.
| |
Collapse
|
12
|
Gong JH, Kim CS, Park J, Kang S, Jang Y, Kim MS, Chung HT, Joe Y, Yu R. Filbertone-Induced Nrf2 Activation Ameliorates Neuronal Damage via Increasing BDNF Expression. Neurochem Res 2024; 50:44. [PMID: 39636503 PMCID: PMC11621137 DOI: 10.1007/s11064-024-04290-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 12/07/2024]
Abstract
Neurotrophic factors are endogenous proteins that promote the survival of various neuronal cells. Increasing evidence has suggested a key role for brain-derived neurotrophic factor (BDNF) in the dopaminergic neurotoxicity associated with Parkinson's Disease (PD). This study explores the therapeutic potential of filbertone, a bioactive compound found in hazelnuts, in neurodegeneration, focusing on its effects on neurotrophic factors and the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway. In our study, filbertone markedly elevated the expression of neurotrophic factors, including BDNF, Glial cell line-Derived Neurotrophic Factor (GDNF), and Nerve Growth Factor (NGF), in human neuroblastoma SH-SY5Y cells, mouse astrocyte C8-D1A cells, and mouse hypothalamus mHypoE-N1 cells. Moreover, filbertone effectively countered neuroinflammation and reversed the decline in neurotrophic factors and Nrf2 activation induced by a high-fat diet (HFD) in neurodegeneration models. The neuroprotective effects of filbertone were further validated in models of neurotoxicity induced by palmitic acid (PA) and the neurotoxin MPTP/MPP+, where it was observed to counteract PA and MPTP/MPP+-induced decreases in cell viability and neuroinflammation, primarily through the activation of Nrf2 and the subsequent upregulation of BDNF and heme oxygenase-1 expression. Nrf2 deficiency negated the neuroprotective effects of filbertone in MPTP-treated mice. Consequently, our finding suggests that filbertone is a novel therapeutic agent for neurodegenerative diseases, enhancing neuronal resilience through the Nrf2 signaling pathway and upregulation of neurotrophic factors.
Collapse
Affiliation(s)
- Jeong Heon Gong
- College of Korean Medicine, Daegu Haany University, Gyeongsan, 38610, Republic of Korea
| | - Chu-Sook Kim
- Department of Biological Sciences, College of Information and Biotechnology, Ulsan National Institute of Science and Technology, Ulsan, 44919, Republic of Korea
| | - Jeongmin Park
- College of Korean Medicine, Daegu Haany University, Gyeongsan, 38610, Republic of Korea
| | - Soeun Kang
- Department of Food and Nutrition, University of Ulsan, Ulsan, 44610, Republic of Korea
| | - Yumi Jang
- Department of Food and Nutrition, University of Ulsan, Ulsan, 44610, Republic of Korea
| | - Min-Seon Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Diabetes Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Korea
| | - Hun Taeg Chung
- College of Korean Medicine, Daegu Haany University, Gyeongsan, 38610, Republic of Korea
| | - Yeonsoo Joe
- College of Korean Medicine, Daegu Haany University, Gyeongsan, 38610, Republic of Korea.
| | - Rina Yu
- Department of Food and Nutrition, University of Ulsan, Ulsan, 44610, Republic of Korea.
| |
Collapse
|
13
|
Ozgür-Gunes Y, Le Stunff C, Bougnères P. Oligodendrocytes, the Forgotten Target of Gene Therapy. Cells 2024; 13:1973. [PMID: 39682723 PMCID: PMC11640421 DOI: 10.3390/cells13231973] [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: 10/29/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
If the billions of oligodendrocytes (OLs) populating the central nervous system (CNS) of patients could express their feelings, they would undoubtedly tell gene therapists about their frustration with the other neural cell populations, neurons, microglia, or astrocytes, which have been the favorite targets of gene transfer experiments. This review questions why OLs have been left out of most gene therapy attempts. The first explanation is that the pathogenic role of OLs is still discussed in most CNS diseases. Another reason is that the so-called ubiquitous CAG, CBA, CBh, or CMV promoters-widely used in gene therapy studies-are unable or poorly able to activate the transcription of episomal transgene copies brought by adeno-associated virus (AAV) vectors in OLs. Accordingly, transgene expression in OLs has either not been found or not been evaluated in most gene therapy studies in rodents or non-human primates. The aims of the current review are to give OLs their rightful place among the neural cells that future gene therapy could target and to encourage researchers to test the effect of OL transduction in various CNS diseases.
Collapse
Affiliation(s)
- Yasemin Ozgür-Gunes
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA;
| | - Catherine Le Stunff
- MIRCen Institute, Laboratoire des Maladies Neurodégénératives, Commissariat à l’Energie Atomique, 92260 Fontenay-aux-Roses, France;
- NEURATRIS at MIRCen, 92260 Fontenay-aux-Roses, France
- UMR1195 Inserm and University Paris Saclay, 94270 Le Kremlin-Bicêtre, France
| | - Pierre Bougnères
- MIRCen Institute, Laboratoire des Maladies Neurodégénératives, Commissariat à l’Energie Atomique, 92260 Fontenay-aux-Roses, France;
- NEURATRIS at MIRCen, 92260 Fontenay-aux-Roses, France
- Therapy Design Consulting, 94300 Vincennes, France
| |
Collapse
|
14
|
Pennington KR, Debs L, Chung S, Bava J, Garin CM, Vale FL, Bick SK, Englot DJ, Terry AV, Constantinidis C, Blake DT. Basal forebrain activation improves working memory in senescent monkeys. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.01.582925. [PMID: 39574741 PMCID: PMC11580932 DOI: 10.1101/2024.03.01.582925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/28/2024]
Abstract
Brain aging contributes to cognitive decline and risk of dementia. Degeneration of the basal forebrain cholinergic system parallels these changes in aging, Alzheimer's dementia, Parkinson's dementia, and Lewy body dementia, and thus is a common element linked to executive function across the lifespan and in disease states. Here, we tested the potential of one-hour daily intermittent basal forebrain stimulation to improve cognition in senescent monkeys, and its mechanisms of action. Stimulation in five animals improved working memory duration in 8-12 weeks across all animals, with peak improvements observed in the first four weeks. In an ensuing three month period without stimulation, improvements were retained. With additional stimulation, performance remained above baseline throughout the 15 months of the study. Studies with a cholinesterase inhibitor produced inconsistent improvements in behavior. One of five animals improved significantly. Manipulating the stimulation pattern demonstrated selectivity for both stimulation and recovery period duration. Brain stimulation led to acute increases in cerebrospinal levels of tissue plasminogen activator, which is an activating element for two brain neurotrophins, Nerve Growth Factor (NGF) and Brain-Derived Growth Factor (BDNF). Stimulation also led to improved glucose utilization in stimulated hemispheres relative to contralateral. Glucose utilization also consistently declines with aging and some dementias. Together, these findings suggest that intermittent stimulation of the nucleus basalis of Meynert improves executive function and reverses some aspects of brain aging.
Collapse
Affiliation(s)
- Kendyl R Pennington
- Dept Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA
| | - Luca Debs
- Dept Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA
| | - Sophia Chung
- Neuroscience Program, Vanderbilt University, Nashville, TN 37235
| | - Janki Bava
- Dept Biomedical Engineering, Vanderbilt University, Nashville, TN 37235
| | - Clément M Garin
- Dept Biomedical Engineering, Vanderbilt University, Nashville, TN 37235
| | - Fernando L Vale
- Dept Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA
| | - Sarah K Bick
- Dept Biomedical Engineering, Vanderbilt University, Nashville, TN 37235
- Dept Neurosurgery, Vanderbilt University, Nashville TN
| | - Dario J Englot
- Dept Biomedical Engineering, Vanderbilt University, Nashville, TN 37235
- Dept Neurosurgery, Vanderbilt University, Nashville TN
| | - Alvin V Terry
- Dept Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA
| | - Christos Constantinidis
- Neuroscience Program, Vanderbilt University, Nashville, TN 37235
- Dept Biomedical Engineering, Vanderbilt University, Nashville, TN 37235
- Dept Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville, TN 37232
| | - David T Blake
- Dept Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA
| |
Collapse
|
15
|
Hassan NF, El-Ansary MR, El-Ansary AR, El-Saied MA, Zaki OS. Unveiling the protective potential of mirabegron against thioacetamide-induced hepatic encephalopathy in rats: Insights into cAMP/PPAR-γ/p-ERK1/2/p S536 NF-κB p 65 and p-CREB/BDNF/TrkB in parallel with oxidative and apoptotic trajectories. Biochem Pharmacol 2024; 229:116504. [PMID: 39179118 DOI: 10.1016/j.bcp.2024.116504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 08/12/2024] [Accepted: 08/20/2024] [Indexed: 08/26/2024]
Abstract
Hepatic encephalopathy (HE) is one of the most prevalent and severe hepatic and brain disorders in which escalation of the oxidative, inflammatory and apoptotic trajectories pathologically connects acute liver injury with neurological impairment. Mirabegron (Mira) is a beta3 adrenergic receptor agonist with proven antioxidant and anti-inflammatory activities. The current research pointed to exploring Mira's hepato-and neuroprotective impacts against thioacetamide (TAA)-induced HE in rats. Rats were distributed into three experimental groups: the normal control group, the TAA group, received TAA (200 mg/kg/day for three consecutive days) and the Mira-treated group received Mira (10 mg/kg/day; oral gavage) for 15 consecutive days and intoxicated with TAA from the 13th to the 15th day of the experimental period. Mira counteracted hyperammonemia, enhanced rats' locomotor capability and motor coordination. It attenuated hepatic/neurological injuries by its antioxidant, anti-apoptotic as well as anti-inflammatory potentials. Mira predominantly targeted cyclic adenosine monophosphate (cAMP)/phosphorylated extracellular signal-regulated kinase (p-Erk1/2)/peroxisome proliferator-activated receptor gamma (PPARγ) dependent pathways via downregulation of p S536-nuclear factor kappa B p65 (p S536 NF-κB p 65)/tumor necrosis alpha (TNF-α) axis. Meanwhile, it attenuated nuclear factor erythroid 2-related factor (Nrf2) depletion in parallel with restoring of the neuroprotective defensive pathway by upregulation of cerebral cAMP/PPAR-γ/p-ERK1/2 and p-CREB/BDNF/TrkB besides reduction of GFAP immunoreactivity. Mira showed anti-apoptotic activity through inhibition of Bax immunoreactivity and elevation of Bcl2. To summarize, Mira exhibited a hepato-and neuroprotective effect against TAA-induced HE in rats via shielding antioxidant defense and mitigation of the pathological inflammatory and apoptotic axis besides upregulation of neuroprotective signaling pathways.
Collapse
Affiliation(s)
- Noha F Hassan
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Modern University for Technology and Information, Cairo, Egypt
| | - Mona R El-Ansary
- Department of Biochemistry, Faculty of Pharmacy, Modern University for Technology and Information, Cairo, Egypt
| | - Amira R El-Ansary
- Department of Internal Medicine, Faculty of Medicine, Misr University for Science and Technology, Giza, Egypt
| | - Mohamed A El-Saied
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Omnia S Zaki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Modern University for Technology and Information, Cairo, Egypt.
| |
Collapse
|
16
|
Svenningsson AL, Bocancea DI, Stomrud E, van Loenhoud A, Barkhof F, Mattsson-Carlgren N, Palmqvist S, Hansson O, Ossenkoppele R. Biological mechanisms of resilience to tau pathology in Alzheimer's disease. Alzheimers Res Ther 2024; 16:221. [PMID: 39396028 PMCID: PMC11470552 DOI: 10.1186/s13195-024-01591-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 09/29/2024] [Indexed: 10/14/2024]
Abstract
BACKGROUND In Alzheimer's disease (AD), the associations between tau pathology and brain atrophy and cognitive decline are well established, but imperfect. We investigate whether cerebrospinal fluid (CSF) biomarkers of biological processes (vascular, synaptic, and axonal integrity, neuroinflammation, neurotrophic factors) explain the disconnection between tau pathology and brain atrophy (brain resilience), and tau pathology and cognitive decline (cognitive resilience). METHODS We included 428 amyloid positive participants (134 cognitively unimpaired (CU), 128 with mild cognitive impairment (MCI), 166 with AD dementia) from the BioFINDER-2 study. At baseline, participants underwent tau positron emission tomography (tau-PET), magnetic resonance imaging (MRI), cognitive testing, and lumbar puncture. Longitudinal data were available for MRI (mean (standard deviation) follow-up 26.4 (10.7) months) and cognition (25.2 (11.4) months). We analysed 18 pre-selected CSF proteins, reflecting vascular, synaptic, and axonal integrity, neuroinflammation, and neurotrophic factors. Stratifying by cognitive status, we performed linear mixed-effects models with cortical thickness (brain resilience) and global cognition (cognitive resilience) as dependent variables to assess whether the CSF biomarkers interacted with tau-PET levels in its effect on cortical atrophy and cognitive decline. RESULTS Regarding brain resilience, interaction effects were observed in AD dementia, with vascular integrity biomarkers (VEGF-A (βinteraction = -0.009, pFDR = 0.047) and VEGF-B (βinteraction = -0.010, pFDR = 0.037)) negatively moderating the association between tau-PET signal and atrophy. In MCI, higher NfL levels were associated with more longitudinal cortical atrophy (β = -0.109, pFDR = 0.033) and lower baseline cortical thickness (β = -0.708, pFDR = 0.033) controlling for tau-PET signal. Cognitive resilience analyses in CU revealed interactions with tau-PET signal for inflammatory (GFAP, IL-15; βinteraction -0.073--0.069, pFDR 0.001-0.045), vascular (VEGF-A, VEGF-D, PGF; βinteraction -0.099--0.063, pFDR < 0.001-0.046), synaptic (14-3-3ζ/δ; βinteraction = -0.092, pFDR = 0.041), axonal (NfL; βinteraction = -0.079, pFDR < 0.001), and neurotrophic (NGF; βinteraction = 0.091, pFDR < 0.001) biomarkers. In MCI higher NfL levels (βmain = -0.690, pFDR = 0.025) were associated with faster cognitive decline independent of tau-PET signal. CONCLUSIONS Biomarkers of co-existing pathological processes, in particular vascular pathology and axonal degeneration, interact with levels of tau pathology on its association with the downstream effects of AD pathology (i.e. brain atrophy and cognitive decline). This indicates that vascular pathology and axonal degeneration could impact brain and cognitive resilience.
Collapse
Affiliation(s)
- Anna L Svenningsson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, 211 46, Lund, Sweden.
- Memory Clinic, Skåne University Hospital, 214 28, Malmö, Sweden.
| | - Diana I Bocancea
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, 1081 HZ, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, 1081, Amsterdam, The Netherlands
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, 211 46, Lund, Sweden
- Memory Clinic, Skåne University Hospital, 214 28, Malmö, Sweden
| | - Anita van Loenhoud
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, 1081 HZ, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, 1081, Amsterdam, The Netherlands
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081 HV, Amsterdam, The Netherlands
- Queen Square Institute of Neurology and Center for Medical Image Computing, University College London, London, WC1N 3BG, UK
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, 211 46, Lund, Sweden
- Department of Neurology, Skåne University Hospital, 211 84, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, 211 46, Lund, Sweden
- Memory Clinic, Skåne University Hospital, 214 28, Malmö, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, 211 46, Lund, Sweden
- Memory Clinic, Skåne University Hospital, 214 28, Malmö, Sweden
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, 211 46, Lund, Sweden
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, 1081 HZ, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, 1081, Amsterdam, The Netherlands
| |
Collapse
|
17
|
Li D, Jia J, Zeng H, Zhong X, Chen H, Yi C. Efficacy of exercise rehabilitation for managing patients with Alzheimer's disease. Neural Regen Res 2024; 19:2175-2188. [PMID: 38488551 PMCID: PMC11034587 DOI: 10.4103/1673-5374.391308] [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: 06/26/2023] [Revised: 10/16/2023] [Accepted: 11/25/2023] [Indexed: 04/24/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive and degenerative neurological disease characterized by the deterioration of cognitive functions. While a definitive cure and optimal medication to impede disease progression are currently unavailable, a plethora of studies have highlighted the potential advantages of exercise rehabilitation for managing this condition. Those studies show that exercise rehabilitation can enhance cognitive function and improve the quality of life for individuals affected by AD. Therefore, exercise rehabilitation has been regarded as one of the most important strategies for managing patients with AD. Herein, we provide a comprehensive analysis of the currently available findings on exercise rehabilitation in patients with AD, with a focus on the exercise types which have shown efficacy when implemented alone or combined with other treatment methods, as well as the potential mechanisms underlying these positive effects. Specifically, we explain how exercise may improve the brain microenvironment and neuronal plasticity. In conclusion, exercise is a cost-effective intervention to enhance cognitive performance and improve quality of life in patients with mild to moderate cognitive dysfunction. Therefore, it can potentially become both a physical activity and a tailored intervention. This review may aid the development of more effective and individualized treatment strategies to address the challenges imposed by this debilitating disease, especially in low- and middle-income countries.
Collapse
Affiliation(s)
- Dan Li
- Department of Pathology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi Province, China
| | - Jinning Jia
- Department of Pathology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi Province, China
| | - Haibo Zeng
- Department of Pathology, Huichang County People’s Hospital, Ganzhou, Jiangxi Province, China
| | - Xiaoyan Zhong
- Department of Pathology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi Province, China
| | - Hui Chen
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
| | - Chenju Yi
- Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong Province, China
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Shenzhen, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou, Guangdong Province, China
| |
Collapse
|
18
|
Xu L, Yao S, Ding YE, Xie M, Feng D, Sha P, Tan L, Bei F, Yao Y. Designing and optimizing AAV-mediated gene therapy for neurodegenerative diseases: from bench to bedside. J Transl Med 2024; 22:866. [PMID: 39334366 PMCID: PMC11429861 DOI: 10.1186/s12967-024-05661-2] [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: 05/21/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
Recombinant adeno-associated viruses (rAAVs) have emerged as an attractive tool for gene delivery, and demonstrated tremendous promise in gene therapy and gene editing-therapeutic modalities with potential "one-and-done" treatment benefits compared to conventional drugs. Given their tropisms for the central nervous system (CNS) across various species including humans, rAAVs have been extensively investigated in both pre-clinical and clinical studies targeting neurodegenerative disease. However, major challenges remain in the application of rAAVs for CNS gene therapy, such as suboptimal vector design, low CNS transduction efficiency and specificity, and therapy-induced immunotoxicity. Therefore, continuing efforts are being made to optimize the rAAV vectors from their "core" genetic payloads to their "coat" or capsid structure. In this review, we describe current approaches for rAAV vector design tailored for transgene expression in the CNS, summarize the development of CNS-targeting AAV serotypes, and highlight recent advancements in AAV capsid engineering, aimed at generating a new generation of rAAVs with improved CNS tropism. Additionally, we discuss various administration routes for delivering rAAVs to the CNS and provide an overview of AAV-mediated gene therapies currently under investigation in clinical trials for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Liang Xu
- Clinical Research Center of Neurological Disease, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Shun Yao
- Department of Neurosurgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Yifan Evan Ding
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mengxiao Xie
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Dingqi Feng
- Center of Clinical Laboratory, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, 215123, China
| | - Pengfei Sha
- Clinical Research Center of Neurological Disease, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
| | - Lu Tan
- Clinical Research Center of Neurological Disease, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Fengfeng Bei
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Yizheng Yao
- Clinical Research Center of Neurological Disease, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Soochow University, Suzhou, China.
| |
Collapse
|
19
|
Magrì A, Tomasello B, Naletova I, Tabbì G, Cairns WRL, Greco V, Sciuto S, La Mendola D, Rizzarelli E. New BDNF and NT-3 Cyclic Mimetics Concur with Copper to Activate Trophic Signaling Pathways as Potential Molecular Entities to Protect Old Brains from Neurodegeneration. Biomolecules 2024; 14:1104. [PMID: 39334869 PMCID: PMC11430436 DOI: 10.3390/biom14091104] [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: 08/05/2024] [Accepted: 08/30/2024] [Indexed: 09/30/2024] Open
Abstract
A low level of Neurotrophins (NTs), their Tyrosine Kinase Receptors (Trks), Vascular Endothelial Growth Factors (VEGFs) and their receptors, mainly VEGFR1 and VEGFR2, characterizes AD brains. The use of NTs and VEGFs as drugs presents different issues due to their low permeability of the blood-brain barrier, the poor pharmacokinetic profile, and the relevant side effects. To overcome these issues, different functional and structural NT mimics have been employed. Being aware that the N-terminus domain as the key domain of NTs for the binding selectivity and activation of Trks and the need to avoid or delay proteolysis, we herein report on the mimicking ability of two cyclic peptide encompassing the N-terminus of Brain Derived Growth Factor (BDNF), (c-[HSDPARRGELSV-]), cBDNF(1-12) and of Neurotrophin3 (NT3), (c-[YAEHKSHRGEYSV-]), cNT3(1-13). The two cyclic peptide features were characterized by a combined thermodynamic and spectroscopic approach (potentiometry, NMR, UV-vis and CD) that was extended to their copper(II) ion complexes. SH-SY5Y cell assays show that the Cu2+ present at the sub-micromolar level in the complete culture media affects the treatments with the two peptides. cBDNF(1-12) and cNT3(1-13) act as ionophores, induce neuronal differentiation and promote Trks and CREB phosphorylation in a copper dependent manner. Consistently, both peptide and Cu2+ stimulate BDNF and VEGF expression as well as VEGF release; cBDNF(1-12) and cNT3(1-13) induce the expression of Trks and VEGFRs.
Collapse
Affiliation(s)
- Antonio Magrì
- Institute of Crystallography, National Council of Research (CNR), P. Gaifami 18, 95126 Catania, Italy; (A.M.); (I.N.); (G.T.)
| | - Barbara Tomasello
- Department of Drug and Health Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy;
| | - Irina Naletova
- Institute of Crystallography, National Council of Research (CNR), P. Gaifami 18, 95126 Catania, Italy; (A.M.); (I.N.); (G.T.)
| | - Giovanni Tabbì
- Institute of Crystallography, National Council of Research (CNR), P. Gaifami 18, 95126 Catania, Italy; (A.M.); (I.N.); (G.T.)
| | - Warren R. L. Cairns
- CNR-Institute of Polar Sciences (CNR-ISP), 155 Via Torino, 30172 Venice, Italy;
| | - Valentina Greco
- Department of Chemical Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy; (V.G.); (S.S.)
| | - Sebastiano Sciuto
- Department of Chemical Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy; (V.G.); (S.S.)
| | - Diego La Mendola
- Department of Pharmacy, University of Pisa, via Bonanno Pisano 6, 56126 Pisa, Italy;
| | - Enrico Rizzarelli
- Institute of Crystallography, National Council of Research (CNR), P. Gaifami 18, 95126 Catania, Italy; (A.M.); (I.N.); (G.T.)
- Department of Chemical Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy; (V.G.); (S.S.)
| |
Collapse
|
20
|
Li M, Xu X, Jia Y, Yuan Y, Na G, Zhu L, Xiao X, Zhang Y, Ye H. Transformation of mulberry polyphenols by Lactobacillus plantarum SC-5: Increasing phenolic acids and enhancement of anti-aging effect. Food Res Int 2024; 192:114778. [PMID: 39147466 DOI: 10.1016/j.foodres.2024.114778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 07/02/2024] [Accepted: 07/14/2024] [Indexed: 08/17/2024]
Abstract
Fermentation can transform bioactive compounds in food and improve their biological activity. This study aims to explore the transformation of polyphenols in mulberry juice and the improvement of its anti-aging effect. The results demonstrated that Lactobacillus plantarum SC-5 transformed anthocyanin in mulberry juice into more phenolic acids, especially improved 2-hydroxy-3-(4-hydroxyphenyl) propanoic acid from 4.16 ± 0.06 to 10.07 ± 0.03. In the D-gal-induced mouse model, fermented mulberry juice significantly raised the abundance of Bifidobacteriaceae (303.7 %) and Lactobacillaceae (237.2 %) and Short-chain fatty acids (SCFAs) in intestine, further reducing the level of oxidative stress (12.3 %). Meanwhile, the expression of Sirtuin 1 (SIRT1) and Brain-derived neurotrophic factor (BDNF) increased, which protected the integrity of hippocampal tissue. Morris water maze results approved that fermented mulberry juice improved cognitive ability in aging mice (30.3 %). This study provides theoretical support for the view that fermentation is an effective means of developing functional foods.
Collapse
Affiliation(s)
- Mengyao Li
- College of Food Science and Engineering, Jilin University, 5333 Xi'an Road, Changchun 130062, China
| | - Xiangxiu Xu
- Changchun City Market Supervision Comprehensive Administrative Law Enforcement Detachment, 1150a-1 Wanfu Road, Changchun 130062, China
| | - Yifan Jia
- College of Food Science and Engineering, Jilin University, 5333 Xi'an Road, Changchun 130062, China
| | - Yuan Yuan
- College of Food Science and Engineering, Jilin University, 5333 Xi'an Road, Changchun 130062, China
| | - Guo Na
- College of Food Science and Engineering, Jilin University, 5333 Xi'an Road, Changchun 130062, China
| | - Ling Zhu
- College of Food Science and Engineering, Jilin University, 5333 Xi'an Road, Changchun 130062, China
| | - Xiaowei Xiao
- College of Food Science and Engineering, Jilin University, 5333 Xi'an Road, Changchun 130062, China
| | - Yamin Zhang
- College of Food Science and Engineering, Jilin University, 5333 Xi'an Road, Changchun 130062, China
| | - Haiqing Ye
- College of Food Science and Engineering, Jilin University, 5333 Xi'an Road, Changchun 130062, China.
| |
Collapse
|
21
|
Zhang L, Jiang Z, Hu S, Ni H, Zhao Y, Tan X, Lang Y, Na R, Li Y, Du Q, Li QX, Dong Y. GSK3β Substrate-competitive Inhibitors Regulate the gut Homeostasis and Barrier Function to Inhibit Neuroinflammation in Scopolamine-induced Alzheimer's Disease Model Mice. Inflammation 2024:10.1007/s10753-024-02133-z. [PMID: 39180577 DOI: 10.1007/s10753-024-02133-z] [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: 05/03/2024] [Revised: 08/18/2024] [Accepted: 08/19/2024] [Indexed: 08/26/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease mainly characterized by cognitive impairment. Glycogen synthase kinase 3 (GSK3β) is a potential therapeutic target against AD. Isoorientin (ISO), a GSK3β substrate competitive inhibitor, plays anti-AD effects in in vitro and in vivo AD model. TFGF-18 is an ISO synthetic analog with improved potency, but its neuroprotective effect in vivo remains to be elucidated, and the underlying mechanisms of GSK3β inhibitor against AD need to be clarified. This study investigated the TFGF-18 and ISO effects on gut homeostasis and neuroinflammation in scopolamine (SCOP)-induced AD mice. And the protection on barrier function was observed in in vitro blood-brain barrier (BBB) model of mouse brain microvascular endothelial cells (bEnd.3). The results show that TFGF-18 and ISO improved cognitive function in SCOP-induced mice, and inhibited cholinergic system disorders and inflammation in the brain and intestine, decreased the level of lipopolysaccharides (LPS) in serum and intestine, protected the diversity and balance of intestinal microbiome, increased the expressions of tight junction protein (ZO-1, occludin), brain derived neurotrophic factor (BDNF) and glial cell-derived neurotrophic factor (GDNF) in the mouse brain and intestine. In addition, TFGF-18 and ISO protected against barrier damage in LPS-stimulated BBB model of bEnd.3 cells in vitro. TFGF-18 and ISO increased the ratio of p-GSK3β/GSK3β, suppressed toll-like receptors 4 (TLR-4) expression and nuclear factor kappa-B (NF-κB) activation in vivo and in vitro, and increased the expressions of β-catenin, nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) in vitro. In conclusion, The GSK3β inhibitors TFGF-18 and ISO modulate the gut homeostasis and barrier function to inhibit neuroinflammation and attenuate cognitive impairment by regulating NF-κB, β-catenin and Nrf2/HO-1 pathways.
Collapse
Affiliation(s)
- Lingyu Zhang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Zhihao Jiang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Shaozhen Hu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Haojie Ni
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Yijing Zhao
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Xiaoqin Tan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
- Medical Department, Wuhan City College, Wuhan, 430083, China
| | - Yi Lang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Risong Na
- College of Plant Protection, Henan Agricultural University, Wenhua Road No. 95, Zhengzhou, 450002, China
| | - Yanwu Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Qun Du
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Qing X Li
- Department of Molecular Bioscience and Bioengineering, University of Hawaii at Manoa, 1955 East-West Road, Honolulu, HI, 96822, USA.
| | - Yan Dong
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China.
| |
Collapse
|
22
|
Askari R, NasrAbadi M, Haghighi AH, Mahin MJ, Somayeh R, Pusceddu M. Effect of combined training in water on hippocampal neuronal Plasticity and memory function in healthy elderly rats. AIMS Neurosci 2024; 11:260-274. [PMID: 39431271 PMCID: PMC11486616 DOI: 10.3934/neuroscience.2024017] [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: 06/29/2024] [Revised: 08/03/2024] [Accepted: 08/15/2024] [Indexed: 10/22/2024] Open
Abstract
Purpose The cyclic AMP response element-binding protein (CREB) and nerve growth factor (NGF) have been proposed as key modulators of brain health and are involved in synaptic plasticity. The study investigates how combined water-based training affects hippocampal neuron plasticity and memory function in old rats. Methods 16 Wistar male rats 24-month-old were randomly divided into two groups: combined training (n = 8) and control (n = 8). Four sessions were performed per week for 10 weeks, and consisted of resistance and endurance training in water. The control group was placed in a water container during training for 30 minutes to be homogenized in terms of the stress conditions. The.NGF and CREB genes in the hippocampus were evaluated and the working memory was measured using real-time PCR and Y-maze tests. The SPSS 26 software was utilized in which independent t-tests were used to analyze the genes and the Mann-Whitney U test was used to analyze functional memory with a significant level of (P < 0.05). Results The combined training resulted in a significant rise in NGF and CREB gene expression in the hippocampus tissue of elderly rats compared to the control group (P < 0.05); however, there was no notable difference in the Y maze performance test between the two groups (P < 0.05). Conclusions These findings suggest that water-based combined training has beneficial effects on gene expression of NGF and CREB; however, it is necessary to conduct more studies to comprehend the effects of combined training on memory function.
Collapse
Affiliation(s)
- Roya. Askari
- Department of Exercise Physiology, Faculty of Sport Science, Hakim Sabzevari University, Sabzevar, Iran
| | - Mohadeseh. NasrAbadi
- Department of Exercise Physiology, Faculty of Sport Science, Hakim Sabzevari University, Sabzevar, Iran
| | - Amir Hossein. Haghighi
- Department of Exercise Physiology, Faculty of Sport Science, Hakim Sabzevari University, Sabzevar, Iran
| | - Mohammad Jahan Mahin
- Department of Exercise Physiology, Faculty of Sport Science, Hakim Sabzevari University, Sabzevar, Iran
| | - Rajabi Somayeh
- Department of Exercise Physiology, Faculty of Human Sciences, Shahrood Branch, Islamic Azad University, Shahrood, Iran
| | | |
Collapse
|
23
|
Yang M, Yan C, Ospondpant D, Wang L, Lin S, Tang WL, Dong TT, Tong P, Xu Q, Tsim KWK. Unveiling the therapeutic potential of Lobaria extract and its depsides/depsidones in combatting A β42 peptides aggregation and neurotoxicity in Alzheimer's disease. Front Pharmacol 2024; 15:1426569. [PMID: 39193345 PMCID: PMC11347406 DOI: 10.3389/fphar.2024.1426569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 07/29/2024] [Indexed: 08/29/2024] Open
Abstract
Background: The development of effective inhibitors that can inhibit amyloid β (Aβ) peptides aggregation and promote neurite outgrowth is crucial for the possible treatment of Alzheimer's disease (AD). Lobaria (Schreb.) Hoffm., a traditional Chinese medicine used in Himalaya region for inflammatory diseases, contains depsides/depsidones (DEPs) such as gyrophoric acid, norstictic acid, and stictic acid known for their anti-cancer and anti-inflammation properties. Methods: Lobaria extracts were analyzed using HPLC to identify DEPs and establish standards. The inhibitory effects of Lobaria on Aβ42 fibrillization and depolymerization were assessed using various approaches with biophysical and cellular methods. The neuroprotective activity of Lobaria extracts and its DEPs aganist Aβ-mediated cytotoxicity was also evaluated. Results: Norstictic and stictic acid were found in the water extract, while norstictic, stictic, and gyrophoric acid were detected in the ethanol extract of Lobaria. Both extracts, and their DEPs effectively inhibited Aβ42 fibrillation and disaggregate mature Aβ42 fibrils. Notably, the ethanol extract showed superior inhibitory effect compared to the water extract, with gyrophoric acid being the most effective DEPs. Additionally, herbal extract-treated Aβ42 aggregation species significantly protected neuronal cells from Aβ42-induced cell damage and promoted neurite outgrowth. Conclusion: This study is the first to investigate the effect of Lobaria on Aβ42 and neuronal cell in AD. Given that Lobaria is commonly used in ethnic medicine and food with good safety records, our findings propose that Lobaria extracts and DEPs have potential as neuroprotective and therapeutic agents for AD patients.
Collapse
Affiliation(s)
- Meixia Yang
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Hong Kong SAR, China
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, HKUST Shenzhen Research Institute, Shenzhen, China
| | - Caishan Yan
- Department of Physics, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Dusadee Ospondpant
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Hong Kong SAR, China
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, HKUST Shenzhen Research Institute, Shenzhen, China
| | - Lisong Wang
- Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Shengying Lin
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Hong Kong SAR, China
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, HKUST Shenzhen Research Institute, Shenzhen, China
| | - Wai Lun Tang
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Hong Kong SAR, China
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, HKUST Shenzhen Research Institute, Shenzhen, China
| | - Tina Tingxia Dong
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Hong Kong SAR, China
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, HKUST Shenzhen Research Institute, Shenzhen, China
| | - Penger Tong
- Department of Physics, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Qin Xu
- Department of Physics, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Karl Wah Keung Tsim
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Hong Kong SAR, China
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, HKUST Shenzhen Research Institute, Shenzhen, China
| |
Collapse
|
24
|
Wei W, Wu Q, Wang S, Dong C, Shao S, Zhang Z, Zhang X, Zhang X, Kan J, Liu F. Treatment with walnut peptide ameliorates memory impairment in zebrafish and rats: promoting the expression of neurotrophic factors and suppressing oxidative stress. Food Funct 2024; 15:8043-8052. [PMID: 38988249 DOI: 10.1039/d4fo00074a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
Walnut peptide, a low molecular weight peptide separated from walnuts by enzymatic hydrolysis, is considered as a potential nutraceutical with a variety of biological activities. In this study, we characterized the walnut peptide prepared by alkaline protease hydrolysis and evaluated its neuroprotective effect in zebrafish and rat models of memory disorders. Series of concentrations of the walnut peptide were orally administered to zebrafish and rats to examine its impact on the behavior and biochemical indicators. The results showed that the oral administration of walnut peptide significantly ameliorated the behavioral performance in zebrafish exposed to bisphenol AF (1 μg mL-1) and rats exposed to alcohol (30% ethanol, 10 mL kg-1). Furthermore, the walnut peptide upregulated the expression of neurotrophic-related molecules in zebrafish, such as the brain-derived neurotrophic factor (BDNF) and the glial cell-derived neurotrophic factor (GDNF). In the rat brain, the walnut peptide increased the activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px), while dramatically reduced malondialdehyde (MDA) level. Together, these findings elucidated that the walnut peptide might partially offset the declarative memory deficits via regulation of neurotrophic-related molecule expression and promotion of the antioxidant defense ability. Therefore, walnut peptide holds the potential for development into functional foods as a nutritional supplement for the management of certain neurodegenerative disorders.
Collapse
Affiliation(s)
- Wei Wei
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, PR China.
- Zhong Shi Du Qing (Shandong) Biotechnology Company, Heze, Shandong, 274108, PR China
| | - Qiming Wu
- Amway (Shanghai) Innovation & Science Co., Ltd, Shanghai, 201203, PR China.
| | - Shuai Wang
- School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Chuanmin Dong
- Institute of Scientific and Technical Information of Heze, Heze, Shandong, 274005, PR China
| | - Shujuan Shao
- Heze Administrative Examination and Approval Service Bureau, Heze, Shandong, 274000, PR China
| | - Zhao Zhang
- Zhong Shi Du Qing (Shandong) Biotechnology Company, Heze, Shandong, 274108, PR China
| | - Xiping Zhang
- Zhong Shi Du Qing (Shandong) Biotechnology Company, Heze, Shandong, 274108, PR China
| | - Xuejun Zhang
- Zhong Shi Du Qing (Shandong) Biotechnology Company, Heze, Shandong, 274108, PR China
| | - Juntao Kan
- Amway (Shanghai) Innovation & Science Co., Ltd, Shanghai, 201203, PR China.
| | - Fuguo Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, PR China.
| |
Collapse
|
25
|
Lin X, Ren P, Xue Z, Liu X, Cao Y, Li T, Miao H. Astrocytic GDNF ameliorates anesthesia and surgery-induced cognitive impairment by promoting hippocampal synaptic plasticity in aged mice. Neurochem Int 2024; 177:105765. [PMID: 38750960 DOI: 10.1016/j.neuint.2024.105765] [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: 01/06/2024] [Revised: 05/08/2024] [Accepted: 05/12/2024] [Indexed: 05/21/2024]
Abstract
BACKGROUND Perioperative neurocognitive disorders (PND) are common complications after surgery in older patients. However, the specific mechanism of this condition remains unclear. Glial cell line-derived neurotrophic factor (GDNF) is an important neurotrophin that abundantly expressed throughout the brain. It can enhance synaptic plasticity and alleviate learning and memory impairments. Thus, the purpose of this study was to investigate the role of GDNF in PND and the mechanisms involved. METHODS The PND animal model was established by performing left tibial fracture surgery on 18-month-old C57BL/6 mice under sevoflurane anesthesia. Recombinant adeno-associated virus (rAAV)-GDNF or empty vectors were injected bilaterally into the hippocampal CA1 region of aged mice 3 weeks before anesthesia/surgery. The open field and fear conditioning test were used to assess the behavior changes. Golgi staining and electrophysiology were utilized to evaluate the morphological and functional alterations of neuronal synaptic plasticity. Western blot analysis was carried out to measure the proteins expression levels and immunofluorescence staining was performed to probe the cellular localization of GDNF. RESULTS Mice with surgery and anesthesia showed a significant decrease in hippocampus-dependent learning and memory, accompanied by a decline in hippocampal synaptic plasticity. Anesthesia/surgery induced a reduction of GDNF, which was colocalized with astrocytes. Overexpression of GDNF in astrocytes could ameliorate the decline in cognitive function by improving hippocampal synaptic plasticity, meanwhile astrocytic GDNF rescued the anesthesia/surgery-induced decrease in GFRα1 and NCAM. CONCLUSION The study concludes that astrocytic GDNF may improve anesthesia/surgery-induced cognitive impairment by promoting hippocampal synaptic plasticity in aged mice via the GFRα1/NCAM pathway.
Collapse
Affiliation(s)
- Xiaowan Lin
- Department of Anesthesiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Department of Anesthesiology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Peng Ren
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Ziyi Xue
- Department of Anesthesiology, Peking University First Hospital, Beijing, China
| | - Xiao Liu
- Department of Anesthesiology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Ying Cao
- Department of Anesthesiology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Tianzuo Li
- Department of Anesthesiology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.
| | - Huihui Miao
- Department of Anesthesiology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
26
|
Sushma, Sahu MR, Murugan NA, Mondal AC. Amelioration of Amyloid-β Induced Alzheimer's Disease by Bacopa monnieri through Modulation of Mitochondrial Dysfunction and GSK-3β/Wnt/β-Catenin Signaling. Mol Nutr Food Res 2024; 68:e2300245. [PMID: 38143280 DOI: 10.1002/mnfr.202300245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 09/21/2023] [Indexed: 12/26/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is the most prevalent dementia, affecting a large number of populations. Despite being under scrutiny for decades, an effective therapeutic option is still not available. METHODS AND RESULTS This study explores the therapeutic role of a nootropic herb Bacopa monnieri (BM) in AD-like pathological conditions produced by injecting preformed amyloid-β42 (Aβ42) fibril bilaterally into hippocampus of Wistar rats, and ethanolic extract of BM is orally administered for 4 weeks. Assessment of behavioral changes reveals that BM treatment ameliorates Aβ42-induced cognitive impairment and compromised explorative behavior. Supplementation of BM also reduces oxidative stress biomarkers, proinflammatory cytokines, and cholinesterase activity in the AD rats. Additionally, BM treatment restores Bcl-2-associated X protein (Bax)/ B-cell lymphoma 2 (Bcl-2) imbalance, increases neurotrophic factors expression, and prevents neurodegeneration validated by quantifying Nissl-positive hippocampal neurons. Interestingly, BM administration eliminates amyloid plaques in the hippocampal region and normalizes the Aβ42-induced increase in phospho-tau and total tau expression. Mechanistic investigations reveal that BM interacts with glycogen synthase kinase (GSK-3β) and restores Wnt/β-catenin signaling. CONCLUSION BM has been used in diet as a nootropic herb for several centuries. This study highlights the anti-Alzheimer activity of BM from the behavioral to the molecular level by modulating mitochondrial dysfunction, and GSK-3β mediates the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Sushma
- Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Manas Ranjan Sahu
- Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Natarajan Arul Murugan
- Department of Computational Biology, Indraprastha Institute of Information Technology, New Delhi, 110067, India
| | - Amal Chandra Mondal
- Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| |
Collapse
|
27
|
Niazi SK, Magoola M, Mariam Z. Innovative Therapeutic Strategies in Alzheimer's Disease: A Synergistic Approach to Neurodegenerative Disorders. Pharmaceuticals (Basel) 2024; 17:741. [PMID: 38931409 PMCID: PMC11206655 DOI: 10.3390/ph17060741] [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: 02/29/2024] [Revised: 05/31/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Alzheimer's disease (AD) remains a significant challenge in the field of neurodegenerative disorders, even nearly a century after its discovery, due to the elusive nature of its causes. The development of drugs that target multiple aspects of the disease has emerged as a promising strategy to address the complexities of AD and related conditions. The immune system's role, particularly in AD, has gained considerable interest, with nanobodies representing a new frontier in biomedical research. Advances in targeting antibodies against amyloid-β (Aβ) and using messenger RNA for genetic translation have revolutionized the production of antibodies and drug development, opening new possibilities for treatment. Despite these advancements, conventional therapies for AD, such as Cognex, Exelon, Razadyne, and Aricept, often have limited long-term effectiveness, underscoring the need for innovative solutions. This necessity has led to the incorporation advanced technologies like artificial intelligence and machine learning into the drug discovery process for neurodegenerative diseases. These technologies help identify therapeutic targets and optimize lead compounds, offering a more effective approach to addressing the challenges of AD and similar conditions.
Collapse
Affiliation(s)
| | | | - Zamara Mariam
- Centre for Health and Life Sciences, Coventry University, Coventry CV1 5FB, UK
| |
Collapse
|
28
|
Liu WY, Yu Y, Zang J, Liu Y, Li FR, Zhang L, Guo RB, Kong L, Ma LY, Li XT. Menthol-Modified Quercetin Liposomes with Brain-Targeting Function for the Treatment of Senescent Alzheimer's Disease. ACS Chem Neurosci 2024; 15:2283-2295. [PMID: 38780450 DOI: 10.1021/acschemneuro.4c00109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024] Open
Abstract
Oxidative stress and neuroinflammation in the aging brain are correlated with the development of neurodegenerative diseases, such as Alzheimer's disease (AD). The blood-brain barrier (BBB) poses a significant challenge to the effective delivery of therapeutics for AD. Prior research has demonstrated that menthol (Men) can augment the permeability of the BBB. Consequently, in the current study, we modified Men on the surface of liposomes to construct menthol-modified quercetin liposomes (Men-Qu-Lips), designed to cross the BBB and enhance quercetin (Qu) concentration in the brain for improved therapeutic efficacy. The experimental findings indicate that Men-Qu-Lips exhibited good encapsulation efficiency and stability, successfully crossed the BBB, improved oxidative stress and neuroinflammation in the brains of aged mice, protected neurons, and enhanced their learning and memory abilities.
Collapse
Affiliation(s)
- Wan-Ying Liu
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China
| | - Yang Yu
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China
| | - Juan Zang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China
| | - Yang Liu
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China
| | - Feng-Rui Li
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China
| | - Lu Zhang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China
| | - Rui-Bo Guo
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China
| | - Liang Kong
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China
| | - Ling-Yue Ma
- Department of Pharmacy, Peking University First Hospital, Beijing 100034, China
| | - Xue-Tao Li
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China
| |
Collapse
|
29
|
Zhou F, Zhang Q, Zheng X, Shi F, Ma K, Ji F, Meng N, Li R, Lv J, Li Q. Antiaging Effects of Human Fecal Transplants with Different Combinations of Bifidobacterium bifidum LTBB21J1 and Lactobacillus casei LTL1361 in d-Galactose-Induced Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:9818-9827. [PMID: 38647087 DOI: 10.1021/acs.jafc.3c09815] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
The feces of healthy middle-aged and old people were first transplanted into d-galactose-induced aging mice to construct humanized aging mice with gut microbiota (FMTC) to confirm the antiaging effect of probiotics produced from centenarians. The mouse model was then treated with centenarian-derived Bifidobacterium bifidum (FMTL), Lactobacillus casei (FMTB), and their mixtures (FMTM), and young mice were used as the control. Compared with the FMTC group, the results demonstrated that the probiotics and their combinations alleviated neuronal damage, increased antioxidant capacity, decreased inflammation, and enhanced cognitive and memory functions in aging mice. In the gut microbiota, the relative abundance of Lactobacillus, Ligilactobacillus, and Akkermansia increased and that of Desulfovibrio and Colidextribacter decreased in the FMTM group compared with that in the FMTC group. The three probiotic groups displayed significant changes in 15 metabolites compared with the FMTC group, with 4 metabolites showing increased expression and 11 metabolites showing decreased expression. The groups were graded as Control > FMTM > FMTB > FMTL > FMTC using a newly developed comprehensive quantitative scoring system that thoroughly analyzed the various indicators of this study. The beneficial antiaging effects of probiotics derived from centenarians were quantitatively described using a novel perspective in this study; it is confirmed that both probiotics and their combinations exert antiaging effects, with the probiotic complex group exhibiting a larger effect.
Collapse
Affiliation(s)
- Fan Zhou
- College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| | - Qinren Zhang
- College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| | - Xiaohua Zheng
- College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| | - Fengcui Shi
- School of Chemical and Biological Engineering, Qilu Institute of Technology, Shandong 250200, China
| | - Kai Ma
- Jiangsu New-Bio Biotechnology Co.,Ltd, Jiangsu 214400, China
| | - Feng Ji
- Jiangsu New-Bio Biotechnology Co.,Ltd, Jiangsu 214400, China
| | - Ning Meng
- College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| | - Ruiding Li
- College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| | - Jingwen Lv
- College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| | - Quanyang Li
- College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| |
Collapse
|
30
|
Liu X, Shen L, Wan M, Xie H, Wang Z. Peripheral extracellular vesicles in neurodegeneration: pathogenic influencers and therapeutic vehicles. J Nanobiotechnology 2024; 22:170. [PMID: 38610012 PMCID: PMC11015679 DOI: 10.1186/s12951-024-02428-1] [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: 11/17/2023] [Accepted: 03/19/2024] [Indexed: 04/14/2024] Open
Abstract
Neurodegenerative diseases (NDDs) such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis epitomize a class of insidious and relentless neurological conditions that are difficult to cure. Conventional therapeutic regimens often fail due to the late onset of symptoms, which occurs well after irreversible neurodegeneration has begun. The integrity of the blood-brain barrier (BBB) further impedes efficacious drug delivery to the central nervous system, presenting a formidable challenge in the pharmacological treatment of NDDs. Recent scientific inquiries have shifted focus toward the peripheral biological systems, investigating their influence on central neuropathology through the lens of extracellular vesicles (EVs). These vesicles, distinguished by their ability to breach the BBB, are emerging as dual operatives in the context of NDDs, both as conveyors of pathogenic entities and as prospective vectors for therapeutic agents. This review critically summarizes the burgeoning evidence on the role of extracerebral EVs, particularly those originating from bone, adipose tissue, and gut microbiota, in modulating brain pathophysiology. It underscores the duplicity potential of peripheral EVs as modulators of disease progression and suggests their potential as novel vehicles for targeted therapeutic delivery, positing a transformative impact on the future landscape of NDD treatment strategies. Search strategy A comprehensive literature search was conducted using PubMed, Web of Science, and Scopus from January 2000 to December 2023. The search combined the following terms using Boolean operators: "neurodegenerative disease" OR "Alzheimer's disease" OR "Parkinson's disease" OR "Amyotrophic lateral sclerosis" AND "extracellular vesicles" OR "exosomes" OR "outer membrane vesicles" AND "drug delivery systems" AND "blood-brain barrier". MeSH terms were employed when searching PubMed to refine the results. Studies were included if they were published in English, involved human subjects, and focused on the peripheral origins of EVs, specifically from bone, adipose tissue, and gut microbiota, and their association with related diseases such as osteoporosis, metabolic syndrome, and gut dysbiosis. Articles were excluded if they did not address the role of EVs in the context of NDDs or did not discuss therapeutic applications. The titles and abstracts of retrieved articles were screened using a dual-review process to ensure relevance and accuracy. The reference lists of selected articles were also examined to identify additional relevant studies.
Collapse
Affiliation(s)
- Xixi Liu
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Lu Shen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, Hunan, 410008, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Changsha, Hunan, 410008, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, Hunan, 410008, China
| | - Meidan Wan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Hui Xie
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, Hunan, 410008, China.
| | - Zhenxing Wang
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, Hunan, 410008, China.
| |
Collapse
|
31
|
Chen M, Liu M, Chen J, Liu X, Tang L, Wang C, Yu Z, Zhang Y, Tian J. Potential Function of 3,5-Dihydroxy-4-Methoxybenzyl Alcohol from Pacific Oyster (Crassostrea gigas) in Brain of Old Mice. Mol Nutr Food Res 2024; 68:e2300469. [PMID: 38522025 DOI: 10.1002/mnfr.202300469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 10/25/2023] [Indexed: 03/25/2024]
Abstract
SCOPE 3,5-Dihydroxy-4-methoxybenzyl alcohol (DHMBA) is found in oyster extracts in recent years and is reported to have antioxidant activity. Although it has been reported to be protective in various models of oxidative stress, the therapeutic effect of DHMBA on neurological damage caused by aging remains to be demonstrated. METHODS AND RESULTS The present study investigates the potential functions of DHMBA in brain of old C57BL/6J mice and aging cell model. Administration of DHMBA improves working memory, reduces anxiety behavior, decreases the expression levels of cell cycle proteins, cycin-dependent kinase inhibitor 1(P21) and peptidase inhibitor 16(P16) and inhibits neuronal loss in old mice. The data obtained from the aging cell model are consistent with those from the old mice. The interaction between DHMBA and Kelch-like ECH-associated protein 1 (Keap1) is predicted by molecular docking assay, and then it is verified by co-immunopricipitation (CoIP) that factor red lineage 2-related factor 2 (Nrf2)-Keap1 protein-protein interaction is inhibited by DHMBA. Protein levels of Nrf2 and its target genes, such as glutathione peroxidase 4(GPX4) and heme oxygenase 1 (HO-1), are detected in old mice and aging cell model. CONCLUSION This study provides new evidence that explores the antioxidant mechanism of DHMBA and implies a potential role of DHMBA on antiaging in brain.
Collapse
Affiliation(s)
- MinYu Chen
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life, Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, 518055, China
| | - Min Liu
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life, Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, 518055, China
| | - JingHong Chen
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life, Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, 518055, China
| | - Xinwei Liu
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life, Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, 518055, China
| | - LiWei Tang
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life, Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, 518055, China
| | - Chao Wang
- Chemical Analysis & Physical Testing Institute, Shenzhen Center for Disease Control and Prevention, Shenzhen, Guangdong, 518055, China
| | - Ziniu Yu
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Key Laboratory of Tropical Marine Bio-resources and Ecology and Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, 510301, China
| | - Yang Zhang
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Key Laboratory of Tropical Marine Bio-resources and Ecology and Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, 510301, China
| | - Jing Tian
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life, Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, 518055, China
| |
Collapse
|
32
|
Valipour B, Simorgh S, Mirsalehi M, Moradi S, Taghizadeh-Hesary F, Seidkhani E, Akbarnejad Z, Alizadeh R. Improvement of spatial learning and memory deficits by intranasal administration of human olfactory ecto-mesenchymal stem cells in an Alzheimer's disease rat model. Brain Res 2024; 1828:148764. [PMID: 38242524 DOI: 10.1016/j.brainres.2024.148764] [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: 10/09/2023] [Revised: 12/14/2023] [Accepted: 01/09/2024] [Indexed: 01/21/2024]
Abstract
Mesenchymal stem cells therapy provides a new perspective of therapeutic approaches in the treatment of neurodegenerative diseases. The present study aimed to investigate the effects of intranasally transplanted human "olfactory ecto-mesenchymal stem cells" (OE-MSCs) in Alzheimer's disease (AD) rats. In this study, we isolated OE-MSCs from human olfactory lamina propria and phenotypically characterized them using immunocytochemistry and flow cytometry. The undifferentiated OE-MSCs were transplanted either by intranasal (IN) or intrahippocampal (IH) injection to rat models of AD, which were induced by injecting amyloid-beta (Aβ) intrahippocampally. Behavioral, histological, and molecular assessments were performed after a three-month recovery period. Based on the results, intranasal administration of OE-MSCs significantly reduced Aβ accumulation and neuronal loss, improved learning and memory impairments, and increased levels of BDNF (brain-derived neurotrophic factor) and NMDAR (N-methyl-D-Aspartate receptors) in the AD rat model. These changes were more significant in animals who received OE-MSCs by intranasal injection. The results of this study suggest that OE-MSCs have the potential to enhance cognitive function in AD, possibly mediated by BDNF and the NMDA receptors.
Collapse
Affiliation(s)
- Behnaz Valipour
- Department of Anatomical Sciences, Sarab Faculty of Medical Sciences, Sarab, Iran; Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sara Simorgh
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Marjan Mirsalehi
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Salah Moradi
- Department of Life Science Engineering, Faculty of New Science and Technology, University of Tehran, Tehran, Iran
| | - Farzad Taghizadeh-Hesary
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Elham Seidkhani
- Department of Anatomical Sciences, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zeinab Akbarnejad
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Rafieh Alizadeh
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
33
|
Abdelaziz M, Mohamed AF, Zaki HF, Gad SS. Agomelatine improves memory and learning impairments in a rat model of LPS-induced neurotoxicity by modulating the ERK/SorLA/BDNF/TrkB pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:1701-1714. [PMID: 37712973 PMCID: PMC10858839 DOI: 10.1007/s00210-023-02717-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 09/07/2023] [Indexed: 09/16/2023]
Abstract
The mutual interplay between neuroinflammation, synaptic plasticity, and autophagy has piqued researchers' interest, particularly when it comes to linking their impact and relationship to cognitive deficits. Being able to reduce inflammation and apoptosis, melatonin has shown to have positive neuroprotective effects; that is why we thought to check the possible role of agomelatine (AGO) as a promising candidate that could have a positive impact on cognitive deficits. In the current study, AGO (40 mg/kg/day, p.o., 7 days) successfully ameliorated the cognitive and learning disabilities caused by lipopolysaccharide (LPS) in rats (250 μg/kg/day, i.p., 7 days). This positive impact was supported by improved histopathological findings and improved spatial memory as assessed using Morris water maze. AGO showed a strong ability to control BACE1 activity and to rein in the hippocampal amyloid beta (Aβ) deposition. Also, it improved neuronal survival, neuroplasticity, and neurogenesis by boosting BDNF levels and promoting its advantageous effects and by reinforcing the pTrkB expression. In addition, it upregulated the pre- and postsynaptic neuroplasticity biomarkers resembled in synapsin I, synaptophysin, and PSD-95. Furthermore, AGO showed a modulatory action on Sortilin-related receptor with A-type repeats (SorLA) pathway and adjusted autophagy. It is noteworthy that all of these actions were abolished by administering PD98059 a MEK/ERK pathway inhibitor (0.3 mg/kg/day, i.p., 7 days). In conclusion, AGO administration significantly improves memory and learning disabilities associated with LPS administration by modulating the ERK/SorLA/BDNF/TrkB signaling pathway parallel to its capacity to adjust the autophagic process.
Collapse
Affiliation(s)
- Mahmoud Abdelaziz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA University), Giza, Egypt
| | - Ahmed F Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini St, Cairo, 11562, Egypt.
- Faculty of Pharmacy, King Salman International University (KSIU), 46612, Ras Sedr, South Sinai, Egypt.
| | - Hala F Zaki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini St, Cairo, 11562, Egypt
| | - Sameh S Gad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA University), Giza, Egypt
| |
Collapse
|
34
|
Nguyen QTN, Park J, Kim DY, Tran DT, Han IO. Forskolin rescues hypoxia-induced cognitive dysfunction in zebrafish with potential involvement of O-GlcNAc cycling regulation. Biochem Pharmacol 2024; 221:116032. [PMID: 38281601 DOI: 10.1016/j.bcp.2024.116032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 12/28/2023] [Accepted: 01/23/2024] [Indexed: 01/30/2024]
Abstract
Repeated sublethal hypoxia exposure induces brain inflammation and affects the initiation and progression of cognitive dysfunction. Experiments from the current study showed that hypoxic exposure downregulates PKA/CREB signaling, which is restored by forskolin (FSK), an adenylate cyclase activator, in both Neuro2a (N2a) cells and zebrafish brain. FSK significantly protected N2a cells from hypoxia-induced cell death and neurite shrinkage. Intraperitoneal administration of FSK for 5 days on zebrafish additionally led to significant recovery from hypoxia-induced social interaction impairment and learning and memory (L/M) deficit. FSK suppressed hypoxia-induced neuroinflammation, as indicated by the observed decrease in NF-κB activation and GFAP expression. We further investigated the potential effect of FSK on O-GlcNAcylation changes induced by hypoxia. Intriguingly FSK induced marked upregulation of the protein level of O-GlcNAc transferase catalyzing addition of the GlcNAc group to target proteins, accompanied by elevated O-GlcNAcylation of nucleocytoplasmic proteins. The hypoxia-induced O-GlcNAcylation decrease in the brain of zebrafish was considerably restored following FSK treatment. Based on the collective results, we propose that FSK rescues hypoxia-induced cognitive dysfunction, potentially through regulation of HBP/O-GlcNAc cycling.
Collapse
Affiliation(s)
- Quynh T N Nguyen
- Department of Biomedical Science, Program in Biomedical Science and Engineering, College of Medicine, Inha University, Incheon, South Korea
| | - Jiwon Park
- Department of Biomedical Science, Program in Biomedical Science and Engineering, College of Medicine, Inha University, Incheon, South Korea
| | - Dong Yeol Kim
- Department of Biomedical Science, Program in Biomedical Science and Engineering, College of Medicine, Inha University, Incheon, South Korea
| | - Duong T Tran
- Department of Biomedical Science, Program in Biomedical Science and Engineering, College of Medicine, Inha University, Incheon, South Korea
| | - Inn Oc Han
- Department of Biomedical Science, Program in Biomedical Science and Engineering, College of Medicine, Inha University, Incheon, South Korea.
| |
Collapse
|
35
|
Zhan Y, Wen Y, Zheng F, Du LJ, Chen TY, Shen XL, Wu R, Tang XG. MiR-26b-3p Promotes Intestinal Motility Disorder by Targeting FZD10 to Inhibit GSK3β/β-Catenin Signaling and Induce Enteric Glial Cell Apoptosis. Mol Neurobiol 2024; 61:1543-1561. [PMID: 37728849 DOI: 10.1007/s12035-023-03600-8] [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: 02/08/2023] [Accepted: 08/20/2023] [Indexed: 09/21/2023]
Abstract
Enteric glial cells (EGCs) are the major component of the enteric nervous system and affect the pathophysiological process of intestinal motility dysfunction. MicroRNAs (miRNAs) play an important role in regulating gastrointestinal homeostasis. However, the mechanism of miRNA-mediated regulation of EGCs in intestinal dysmotility remains unclear. In this study, we investigated the effect of EGC apoptosis on intestinal dysmotility, and the effect of miR-26b-3p on EGC proliferation and apoptosis in vivo and in vitro. A loperamide hydrochloride (Lop)-induced constipated mouse model and an in vitro culture system of rat EGCs were established. The transcriptome was used to predict the differentially expressed gene miR-26b-3p and the target gene Frizzled 10 (FZD10), and their targeting binding relationship was verified by luciferase. EGCs were transfected with miR-26b-3p mimic or antagomir, and the FZD10 expression was down-regulated by siRNA. Immunofluorescence and flow cytometry were used to detect EGC apoptosis. MiR-26b-3p and FZD10 expressions were examined using quantitative real-time PCR (qRT-PCR). The CCK-8 assay was used to detect EGC proliferation. The protein levels were detected by Western blotting and enzyme-linked immunosorbent assay (ELISA). The results showed that miR-26b-3p was up-regulated in the Lop group, whereas FZD10 was down-regulated, and EGC apoptosis was increased in the colon of intestinal dysmotility mice. FZD10 down-regulation and miR-26b-3p mimic significantly increased glycogen synthase kinase-3β phosphorylation (p-GSK3β) levels, decreased β-catenin expression, and promoted EGC apoptosis. MiR-26b-3p antagomir alleviated intestinal dysmotility, promoted EGC increased activity of EGCs, and reduced EGC apoptosis in vivo. In conclusion, this study indicated that miR-26b-3p promotes intestinal motility disorders by targeting FZD10 to block GSK3β/β-catenin signaling and induces apoptosis in EGCs. Our results provide a new research target for the treatment and intervention of intestinal dysmotility.
Collapse
Affiliation(s)
- Yu Zhan
- Hospital of Chengdu University of TCM, Chengdu, China
- Anorectal Department, Affiliated Hospital of Integrative Chinese Medicine and Western Medicine of Chengdu University of TCM, Chengdu, China
- Chengdu First People's Hospital, Chengdu, China
| | - Yong Wen
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Fan Zheng
- Anorectal Department, People's Hospital of Deyang City, Deyang, China
- Universiti Sains Malaysia, George Town, Pulau Pinang, Malaysia
| | - Li-Juan Du
- The Third People's Hospital of Chengdu, Chengdu, China
- Southwest Jiaotong University College of Medicine, Chengdu, China
| | - Tai-Yu Chen
- Department of Integrated Traditional and Western Medicine Anorectal, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xu-Long Shen
- Anorectal Department, Luzhou People's Hospital, Luzhou, China
| | - Rong Wu
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xue-Gui Tang
- Department of Integrated Traditional and Western Medicine Anorectal, Affiliated Hospital of North Sichuan Medical College, Nanchong, China.
| |
Collapse
|
36
|
Angelucci F, Veverova K, Katonová A, Vyhnalek M, Hort J. Plasminogen activator inhibitor-1 serum levels in frontotemporal lobar degeneration. J Cell Mol Med 2024; 28:e18013. [PMID: 38386354 PMCID: PMC10902304 DOI: 10.1111/jcmm.18013] [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: 05/02/2023] [Revised: 09/25/2023] [Accepted: 10/17/2023] [Indexed: 02/23/2024] Open
Abstract
Plasminogen activator inhibitor-1 (PAI-1) impedes brain plasmin synthesis. Reduced plasmin activity facilitates cumulation of amyloid beta (Aβ) in Alzheimer's disease (AD). Since plasmin also regulates the synaptic activity, it is possible that altered PAI-1 is present in other neurodegenerative disorders. We investigated whether PAI-1 and its counter-regulatory tissue plasminogen activator (tPA) are altered in serum of patients with dementia due to frontotemporal lobar degeneration (FTLD). Thirty five FTLD patients (21 in mild cognitive impairment stage (MCI) and 14 in dementia stage) and 10 cognitively healthy controls were recruited. Serum tPA and PAI-1 protein levels were measured by anova. Correlation between biochemical and demographic data were explored by measuring Pearson correlation coefficient. Serum PAI-1 levels were elevated in the FTLD dementia group as compared to FTLD MCI and controls. tPA serum levels and PAI-1/tPA ratio did not significantly differ among groups. There was a negative correlation between PAI-1 serum levels and disease severity measured by MMSE score. No correlations of tPA serum levels and PAI-1/tPA ratio with MMSE were found. Increased PAI-1 serum levels may serve as a marker of dementia in FTLD, suggesting that, besides Aβ pathway, the plasmin system may affect cognition through synaptic activity.
Collapse
Affiliation(s)
- Francesco Angelucci
- Memory ClinicDepartment of NeurologySecond Faculty of MedicineCharles University and Motol University HospitalPragueCzech Republic
- International Clinical Research CentreSt. Anne's University HospitalBrnoCzech Republic
| | - Katerina Veverova
- Memory ClinicDepartment of NeurologySecond Faculty of MedicineCharles University and Motol University HospitalPragueCzech Republic
| | - Alžbeta Katonová
- Memory ClinicDepartment of NeurologySecond Faculty of MedicineCharles University and Motol University HospitalPragueCzech Republic
| | - Martin Vyhnalek
- Memory ClinicDepartment of NeurologySecond Faculty of MedicineCharles University and Motol University HospitalPragueCzech Republic
| | - Jakub Hort
- Memory ClinicDepartment of NeurologySecond Faculty of MedicineCharles University and Motol University HospitalPragueCzech Republic
- International Clinical Research CentreSt. Anne's University HospitalBrnoCzech Republic
| |
Collapse
|
37
|
Bu L, Wang C, Bai J, Song J, Zhang Y, Chen H, Suo H. Gut microbiome-based therapies for alleviating cognitive impairment: state of the field, limitations, and future perspectives. Food Funct 2024; 15:1116-1134. [PMID: 38224464 DOI: 10.1039/d3fo02307a] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
Cognitive impairment (CI) is a multifaceted neurological condition that can trigger negative emotions and a range of concurrent symptoms, imposing significant public health and economic burdens on society. Therefore, it is imperative to discover a remedy for CI. Nevertheless, the mechanisms behind the onset of this disease are multifactorial, which makes the search for effective amelioration difficult and complex, hindering the search for effective measures. Intriguingly, preclinical research indicates that gut microbiota by influencing brain function, plays an important role in the progression of CI. Furthermore, numerous preclinical studies have highlighted the potential of probiotics, prebiotics, fecal microbiota transplantation (FMT), and diet in modulating the gut microbiota, thereby ameliorating CI symptoms. This review provides a comprehensive evaluation of CI pathogenesis, emphasizing the contribution of gut microbiota disorders to CI development. It also summarizes and discusses current strategies and mechanisms centered on the synergistic role of gut microbiota modulation in the microbiota-gut-brain axis in CI development. Finally, problems with existing approaches are contemplated and the development of microbial modulation strategies as therapeutic approaches to promote and restore brain cognition is discussed. Further research considerations and directions are highlighted to provide ideas for future CI prevention and treatment strategies.
Collapse
Affiliation(s)
- Linli Bu
- College of Food Science, Southwest University, Chongqing 400715, China.
- Modern "Chuan Cai Yu Wei" Food Industry Innovation Research Institute, Chongqing 400715, China
| | - Chen Wang
- College of Food Science, Southwest University, Chongqing 400715, China.
- Modern "Chuan Cai Yu Wei" Food Industry Innovation Research Institute, Chongqing 400715, China
| | - Junying Bai
- Citrus Research Institute, Southwest University, Chongqing 400715, China
| | - Jiajia Song
- College of Food Science, Southwest University, Chongqing 400715, China.
- Modern "Chuan Cai Yu Wei" Food Industry Innovation Research Institute, Chongqing 400715, China
| | - Yuhong Zhang
- Institute of Food Sciences and Technology, Tibet Academy of Agricultural and Animal Husbandry Sciences, Xizang 850000, China
| | - Hongyu Chen
- College of Food Science, Southwest University, Chongqing 400715, China.
- Modern "Chuan Cai Yu Wei" Food Industry Innovation Research Institute, Chongqing 400715, China
| | - Huayi Suo
- College of Food Science, Southwest University, Chongqing 400715, China.
- Modern "Chuan Cai Yu Wei" Food Industry Innovation Research Institute, Chongqing 400715, China
| |
Collapse
|
38
|
Syed RA, Hayat M, Qaiser H, Uzair M, Al-Regaiey K, Khallaf R, Kaleem I, Bashir S. Aging-Related Protein Alterations in the Brain. J Alzheimers Dis 2024; 99:S5-S22. [PMID: 38339930 DOI: 10.3233/jad-230801] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2024]
Abstract
Aging is an intrinsic aspect of an organism's life cycle and is characterized by progressive physiological decline and increased susceptibility to mortality. Many age-associated disorders, including neurological disorders, are most commonly linked with the aging process, such as Alzheimer's disease (AD). This review aims to provide a comprehensive overview of the effects of aging and AD on the molecular pathways and levels of different proteins in the brain, including metalloproteins, neurotrophic factors, amyloid proteins, and tau proteins. AD is caused by the aggregation of amyloid proteins in the brain. Factors such as metal ions, protein ligands, and the oligomerization state of amyloid precursor protein significantly influence the proteolytic processing of amyloid-β protein precursor (AβPP). Tau, a disordered cytosolic protein, serves as the principal microtubule-associated protein in mature neurons. AD patients exhibit decreased levels of nerve growth factor within their nervous systems and cerebrospinal fluid. Furthermore, a significant increase in brain-derived neurotrophic factor resulting from the neuroprotective effect of glial cell line-derived neurotrophic factor suggests that the synergistic action of these proteins plays a role in inhibiting neuronal degeneration and atrophy. The mechanism through which Aβ and AβPP govern Cu2+ transport and their influence on Cu2+ and other metal ion pools requires elucidation in future studies. A comprehensive understanding of the influence of aging and AD on molecular pathways and varying protein levels may hold the potential for the development of novel diagnostic and therapeutic methods for the treatment of AD.
Collapse
Affiliation(s)
- Rafay Ali Syed
- Department of Biological Sciences, Faculty of Basic & Applied Sciences, International Islamic University Islamabad, Pakistan
- Department of Biotechnology, Quaid-i-Azam University, Islamabad, Pakistan
| | - Mahnoor Hayat
- Department of Biological Sciences, Faculty of Basic & Applied Sciences, International Islamic University Islamabad, Pakistan
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Hammad Qaiser
- Department of Biological Sciences, Faculty of Basic & Applied Sciences, International Islamic University Islamabad, Pakistan
| | - Mohammad Uzair
- Department of Biological Sciences, Faculty of Basic & Applied Sciences, International Islamic University Islamabad, Pakistan
| | - Khalid Al-Regaiey
- Department of Physiology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Roaa Khallaf
- Department of Neurology, Neuroscience Center, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | - Imdad Kaleem
- Department of Biosciences, COMSATS University, Islamabad, Pakistan
| | - Shahid Bashir
- Neuroscience Center, King Fahad Specialist Hospital Dammam, Dammam, Saudi Arabia
| |
Collapse
|
39
|
Wang T, Chen S, Mao Z, Shang Y, Brinton RD. Allopregnanolone pleiotropic action in neurons and astrocytes: calcium signaling as a unifying mechanism. Front Endocrinol (Lausanne) 2023; 14:1286931. [PMID: 38189047 PMCID: PMC10771836 DOI: 10.3389/fendo.2023.1286931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 11/13/2023] [Indexed: 01/09/2024] Open
Abstract
Objective Allopregnanolone (Allo) is a neurosteroid with pleiotropic action in the brain that includes neurogenesis, oligogenesis, human and rodent neural stem cell regeneration, increased glucose metabolism, mitochondrial respiration and biogenesis, improved cognitive function, and reduction of both inflammation and Alzheimer's disease (AD) pathology. Because the breadth of Allo-induced responses requires activation of multiple systems of biology in the absence of an Allo-specific nuclear receptor, analyses were conducted in both neurons and astrocytes to identify unifying systems and signaling pathways. Methods Mechanisms of Allo action were investigated in embryonic hippocampal neurons and astrocytes cultured in an Aging Model (AM) media. Cellular morphology, mitochondrial function, and transcriptomics were investigated followed by mechanistic pathway analyses. Results In hippocampal neurons, Allo significantly increased neurite outgrowth and synaptic protein expression, which were paralleled by upregulated synaptogenesis and long-term potentiation gene expression profiles. Mechanistically, Allo induced Ca2+/CREB signaling cascades. In parallel, Allo significantly increased maximal mitochondrial respiration, mitochondrial membrane potential, and Complex IV activity while reducing oxidative stress, which required both the GABAA and L-type Ca2+ channels. In astrocytes, Allo increased ATP generation, mitochondrial function and dynamics while reducing oxidative stress, inflammasome indicators, and apoptotic signaling. Mechanistically, Allo regulation of astrocytic mitochondrial function required both the GABAA and L-type Ca2+ channels. Furthermore, Allo activated NRF1-TFAM signaling and increased the DRP1/OPA1 protein ratio, which led to increased mitochondrial biogenesis and dynamics. Conclusion Collectively, the cellular, mitochondrial, transcriptional, and pharmacological profiles provide evidence in support of calcium signaling as a unifying mechanism for Allo pleiotropic actions in the brain.
Collapse
Affiliation(s)
- Tian Wang
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ, United States
- Department of Neurology, College of Medicine Tucson, University of Arizona, Tucson, AZ, United States
| | - Shuhua Chen
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ, United States
| | - Zisu Mao
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ, United States
| | - Yuan Shang
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ, United States
| | - Roberta Diaz Brinton
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ, United States
- Department of Neurology, College of Medicine Tucson, University of Arizona, Tucson, AZ, United States
- Department of Pharmacology, College of Medicine Tucson, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
40
|
Tuon L, Tramontin NS, Custódio I, Comim VH, Costa B, Tietbohl LTW, Muller AP. Serum Biomarkers to Mild Cognitive Deficits in Children and Adolescents. Mol Neurobiol 2023; 60:7080-7087. [PMID: 37526895 DOI: 10.1007/s12035-023-03536-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 07/22/2023] [Indexed: 08/02/2023]
Abstract
Intellectual disability (ID) is a condition characterized by significant limitations in both cognitive development and adaptive behavior. The diagnosis is made through clinical assessment, standardized tests, and intelligence quotient (IQ). Genetic, inflammation, oxidative stress, and diet have been suggested to contribute to ID, and biomarkers could potentially aid in diagnosis and treatment. Study included children and adolescents aged 6-16 years. The ID group (n = 16) and the control group (n = 18) underwent the Wechsler Intelligence Scale for Children (WISC-IV) test, and blood samples were collected. Correlations between biomarker levels and WISC-IV test scores were analyzed. The ID group had an IQ score below 75, and the values of four domains (IQ, IOP, IMO, and IVP) were lower compared to the control group. Serum levels of FKN, NGF-β, and vitamin B12 were decreased in the ID group, while DCFH and nitrite levels were increased. Positive correlations were found between FKN and the QIT and IOP domains, NGF and the QIT and IMO domains, and vitamin B12 and the ICV domain. TNF-α showed a negative correlation with the ICV domain. Our study identified FKN, NGF-β, and vitamin B12 as potential biomarkers specific to ID, which could aid in the diagnosis and treatment of ID. TNF-α and oxidative stress biomarkers suggest that ID has a complex etiology, and further research is needed to better understand this condition and develop effective treatments. Future studies could explore the potential implications of these biomarkers and develop targeted interventions based on their findings.
Collapse
Affiliation(s)
- Lisiane Tuon
- Pós-Graduação em Saúde Coletiva, Universidade Do Extremo Sul Catarinense, Criciuma, SC, Brazil
| | | | - Isis Custódio
- Pós-Graduação em Saúde Coletiva, Universidade Do Extremo Sul Catarinense, Criciuma, SC, Brazil
| | - Vitor Hugo Comim
- Pós-Graduação em Saúde Coletiva, Universidade Do Extremo Sul Catarinense, Criciuma, SC, Brazil
| | - Barbara Costa
- Pós-Graduação em Saúde Coletiva, Universidade Do Extremo Sul Catarinense, Criciuma, SC, Brazil
| | | | - Alexandre Pastoris Muller
- Department of Pharmacology, Federal University of Santa Catarina, Florianópolis, SC, Brazil.
- Department of Biochemistry, Federal University of Santa Catarina, Florianópolis, SC, Brazil.
- Post-Graduate Program in Biochemistry, Federal University of Santa Catarina, Florianópolis, SC, Brazil.
| |
Collapse
|
41
|
Purcell J, Wiley R, Won J, Callow D, Weiss L, Alfini A, Wei Y, Carson Smith J. Increased neural differentiation after a single session of aerobic exercise in older adults. Neurobiol Aging 2023; 132:67-84. [PMID: 37742442 DOI: 10.1016/j.neurobiolaging.2023.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 08/19/2023] [Accepted: 08/24/2023] [Indexed: 09/26/2023]
Abstract
Aging is associated with decreased cognitive function. One theory posits that this decline is in part due to multiple neural systems becoming dedifferentiated in older adults. Exercise is known to improve cognition in older adults, even after only a single session. We hypothesized that one mechanism of improvement is a redifferentiation of neural systems. We used a within-participant, cross-over design involving 2 sessions: either 30 minutes of aerobic exercise or 30 minutes of seated rest (n = 32; ages 55-81 years). Both functional Magnetic Resonance Imaging (fMRI) and Stroop performance were acquired soon after exercise and rest. We quantified neural differentiation via general heterogeneity regression. There were 3 prominent findings following the exercise. First, participants were better at reducing Stroop interference. Second, while there was greater neural differentiation within the hippocampal formation and cerebellum, there was lower neural differentiation within frontal cortices. Third, this greater neural differentiation in the cerebellum and temporal lobe was more pronounced in the older ages. These data suggest that exercise can induce greater neural differentiation in healthy aging.
Collapse
Affiliation(s)
- Jeremy Purcell
- Department of Kinesiology, University of Maryland, College Park, MD, USA; Maryland Neuroimaging Center, University of Maryland, College Park, MD, USA.
| | - Robert Wiley
- Department of Psychology, University of North Carolina at Greensboro, Greensboro, NC, USA
| | - Junyeon Won
- Department of Kinesiology, University of Maryland, College Park, MD, USA; Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Dallas, Dallas, TX, USA
| | - Daniel Callow
- Department of Kinesiology, University of Maryland, College Park, MD, USA; Program in Neuroscience and Cognitive Science, University of Maryland, College Park, MD, USA
| | - Lauren Weiss
- Department of Kinesiology, University of Maryland, College Park, MD, USA; Program in Neuroscience and Cognitive Science, University of Maryland, College Park, MD, USA
| | - Alfonso Alfini
- National Center on Sleep Disorders Research, Division of Lung Diseases, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Yi Wei
- Maryland Neuroimaging Center, University of Maryland, College Park, MD, USA
| | - J Carson Smith
- Department of Kinesiology, University of Maryland, College Park, MD, USA; Program in Neuroscience and Cognitive Science, University of Maryland, College Park, MD, USA.
| |
Collapse
|
42
|
Xiao Z, Li Q, Wang Z, Zhang H. Single- and combined-phthalate exposures are associated with biological ageing among adults. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 268:115715. [PMID: 37992641 DOI: 10.1016/j.ecoenv.2023.115715] [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: 09/10/2023] [Revised: 11/09/2023] [Accepted: 11/17/2023] [Indexed: 11/24/2023]
Abstract
BACKGROUND Previous research has emphasized the effects of lifestyle and genetics on ageing. However, the association between exposure to phthalates, which are extensively used in cosmetics and personal care products, and ageing is still unclear. METHOD Data for 4711 subjects from the National Health and Nutrition Examination Survey (NHANES) from 2005 to 2010 were incorporated in the present study. The acceleration of the Klemera-Doubal method-biological age (KDM-BA) and phenotypic Age (PhenoAge) were measured by the composite of 13 biomarkers. Multiple-linear and weighted-quantile sum (WQS) regression models were constructed to explore the relationships of single- and combined-phthalate exposures, as indicated by urinary phthalate metabolites, with KDM-BA and PhenoAge. A generalized additive model (GAM) was fitted to explore the potential nonlinear relationships among the above variables. RESULTS Except for mono-(carboxynonyl), all urinary phthalate metabolites were associated with biological ageing, with correlation coefficients ranging from 0.241 to 0.526; however, mono-ethyl presented a negative correlation. The WQS models revealed mixed effects of combined urinary phthalate metabolites on ageing, with a 0.22-year ((95 % CI) 0.09, 0.32) increase in KDM-BA acceleration and a 0.27-year ((95 % CI) 0.13, 0.37) increase in PhenoAge acceleration for each decile increase in urinary phthalate metabolites. Moreover, MCPP, MEOHP, and MBzP seemed to be the top three phthalates in terms of biological ageing, with weights of 33.3 % and 32.2 %, 29.2 % and 17.2 %, and 21.5 % and 30.1 % in KDM-BA and PhenoAge acceleration, respectively. CONCLUSION Single-phthalate exposure was mostly associated with the ageing process, and combined-phthalate exposure presented mixed effects on biological ageing, emphasizing phthalate exposure as a significant risk factor for ageing.
Collapse
Affiliation(s)
- Zhihao Xiao
- School of Public Health, Nanjing Medical University, China
| | - Qian Li
- School of Public Health, Nanjing Medical University, China
| | - Zhiqi Wang
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing 210008, China.
| | - Hongmei Zhang
- Department of Rehabilitation, Children's Hospital of Nanjing Medical University, Nanjing 210008, China.
| |
Collapse
|
43
|
Tancheva L, Kalfin R, Minchev B, Uzunova D, Tasheva K, Tsvetanova E, Georgieva A, Alexandrova A, Stefanova M, Solak A, Lazarova M, Hodzhev Y, Grigorova V, Yarkov D, Petkova-Kirova P. Memory Recovery Effect of a New Bioactive Innovative Combination in Rats with Experimental Dementia. Antioxidants (Basel) 2023; 12:2050. [PMID: 38136170 PMCID: PMC10740861 DOI: 10.3390/antiox12122050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 12/24/2023] Open
Abstract
Alzheimer's disease manifests as a complex pathological condition, with neuroinflammation, oxidative stress and cholinergic dysfunction being a few of the many pathological changes. Due to the complexity of the disease, current therapeutic strategies aim at a multitargeted approach, often relying on a combination of substances with versatile and complementary effects. In the present study, a unique combination of α-lipoic acid, citicoline, extracts of leaves from olive tree and green tea, vitamin D3, selenium and an immune-supporting complex was tested in scopolamine-induced dementia in rats. Using behavioral and biochemical methods, we assessed the effects of the combination on learning and memory, and elucidated the mechanisms of these effects. Our results showed that, compared to its components, the experimental combination was most efficient in improving short- and long-term memory as assessed by the step-through method as well as spatial memory as assessed by T-maze and Barnes maze underlined by decreases in AChE activity (p < 0.05) and LPO (p < 0.001), increases in SOD activity in the cortex (p < 0.05) and increases in catalase (p < 0.05) and GPx (p < 0.01) activities and BDNF (p < 0.001) and pCREB (p < 0.05) levels in the hippocampus. No significant histopathological changes or blood parameter changes were detected, making the experimental combination an effective and safe candidate in a multitargeted treatment of AD.
Collapse
Affiliation(s)
- Lyubka Tancheva
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str. 23, 1113 Sofia, Bulgaria; (L.T.); (B.M.); (D.U.); (E.T.); (A.G.); (A.A.); (M.S.); (A.S.); (M.L.)
| | - Reni Kalfin
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str. 23, 1113 Sofia, Bulgaria; (L.T.); (B.M.); (D.U.); (E.T.); (A.G.); (A.A.); (M.S.); (A.S.); (M.L.)
- Department of Healthcare, South-West University “Neofit Rilski”, Ivan Mihailov Str. 66, 2700 Blagoevgrad, Bulgaria
| | - Borislav Minchev
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str. 23, 1113 Sofia, Bulgaria; (L.T.); (B.M.); (D.U.); (E.T.); (A.G.); (A.A.); (M.S.); (A.S.); (M.L.)
| | - Diamara Uzunova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str. 23, 1113 Sofia, Bulgaria; (L.T.); (B.M.); (D.U.); (E.T.); (A.G.); (A.A.); (M.S.); (A.S.); (M.L.)
| | - Krasimira Tasheva
- Institute of Plant Physiology and Genetics, Bulgarian Academy of Sciences, Acad. G. Bonchev Str. 21, 1113 Sofia, Bulgaria;
| | - Elina Tsvetanova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str. 23, 1113 Sofia, Bulgaria; (L.T.); (B.M.); (D.U.); (E.T.); (A.G.); (A.A.); (M.S.); (A.S.); (M.L.)
| | - Almira Georgieva
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str. 23, 1113 Sofia, Bulgaria; (L.T.); (B.M.); (D.U.); (E.T.); (A.G.); (A.A.); (M.S.); (A.S.); (M.L.)
| | - Albena Alexandrova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str. 23, 1113 Sofia, Bulgaria; (L.T.); (B.M.); (D.U.); (E.T.); (A.G.); (A.A.); (M.S.); (A.S.); (M.L.)
- National Sports Academy, Department of Physiology and Biochemistry, Acad. S. Mladenov Str. 21, 1700 Sofia, Bulgaria
| | - Miroslava Stefanova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str. 23, 1113 Sofia, Bulgaria; (L.T.); (B.M.); (D.U.); (E.T.); (A.G.); (A.A.); (M.S.); (A.S.); (M.L.)
| | - Ayten Solak
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str. 23, 1113 Sofia, Bulgaria; (L.T.); (B.M.); (D.U.); (E.T.); (A.G.); (A.A.); (M.S.); (A.S.); (M.L.)
- Institute of Cryobiology and Food Technologies, Cherni Vrah Blvd 53, 1407 Sofia, Bulgaria
| | - Maria Lazarova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str. 23, 1113 Sofia, Bulgaria; (L.T.); (B.M.); (D.U.); (E.T.); (A.G.); (A.A.); (M.S.); (A.S.); (M.L.)
| | - Yordan Hodzhev
- National Center of Infectious and Parasitic Diseases, Yanko Sakazov Blvd 26, 1504 Sofia, Bulgaria;
| | - Valya Grigorova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str. 23, 1113 Sofia, Bulgaria; (L.T.); (B.M.); (D.U.); (E.T.); (A.G.); (A.A.); (M.S.); (A.S.); (M.L.)
| | - Dobri Yarkov
- Faculty of Veterinary Medicine, Trakia University, 6000 Stara Zagora, Bulgaria;
| | - Polina Petkova-Kirova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str. 23, 1113 Sofia, Bulgaria; (L.T.); (B.M.); (D.U.); (E.T.); (A.G.); (A.A.); (M.S.); (A.S.); (M.L.)
| |
Collapse
|
44
|
Wu Y, Angelova A. Recent Uses of Lipid Nanoparticles, Cell-Penetrating and Bioactive Peptides for the Development of Brain-Targeted Nanomedicines against Neurodegenerative Disorders. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:3004. [PMID: 38063700 PMCID: PMC10708303 DOI: 10.3390/nano13233004] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/17/2023] [Accepted: 11/21/2023] [Indexed: 12/19/2024]
Abstract
The lack of effective treatments for neurodegenerative diseases (NDs) is an important current concern. Lipid nanoparticles can deliver innovative combinations of active molecules to target the various mechanisms of neurodegeneration. A significant challenge in delivering drugs to the brain for ND treatment is associated with the blood-brain barrier, which limits the effectiveness of conventional drug administration. Current strategies utilizing lipid nanoparticles and cell-penetrating peptides, characterized by various uptake mechanisms, have the potential to extend the residence time and bioavailability of encapsulated drugs. Additionally, bioactive molecules with neurotropic or neuroprotective properties can be delivered to potentially mediate the ND targeting pathways, e.g., neurotrophin deficiency, impaired lipid metabolism, mitochondrial dysfunction, endoplasmic reticulum stress, accumulation of misfolded proteins or peptide fragments, toxic protein aggregates, oxidative stress damage, and neuroinflammation. This review discusses recent advancements in lipid nanoparticles and CPPs in view of the integration of these two approaches into nanomedicine development and dual-targeted nanoparticulate systems for brain delivery in neurodegenerative disorders.
Collapse
Affiliation(s)
| | - Angelina Angelova
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400 Orsay, France;
| |
Collapse
|
45
|
Chen L, Sun Y, Li J, Liu S, Ding H, Wang G, Li X. Assessing Cannabidiol as a Therapeutic Agent for Preventing and Alleviating Alzheimer's Disease Neurodegeneration. Cells 2023; 12:2672. [PMID: 38067101 PMCID: PMC10705747 DOI: 10.3390/cells12232672] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/03/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
Alzheimer's disease (AD) is a leading neurodegenerative condition causing cognitive and memory decline. With small-molecule drugs targeting Aβ proving ineffective, alternative targets are urgently needed. Neuroinflammation, which is central to AD's pathology, results in synaptic and neuronal damage, highlighting the importance of addressing inflammation and conserving neuronal integrity. Cannabidiol (CBD), derived from cannabis, is noted for its neuroprotective and anti-inflammatory properties, having shown efficacy in neuropathic pain management for epilepsy. To investigate the therapeutic efficacy of CBD in AD and to elucidate its underlying mechanisms, we aimed to contribute valuable insights for incorporating AD prevention recommendations into future CBD nutritional guidelines. Aβ1-42 was employed for in vivo or in vitro model establishment, CBD treatment was utilized to assess the therapeutic efficacy of CBD, and RNA-seq analysis was conducted to elucidate the underlying therapeutic mechanism. CBD mitigates Aβ-induced cognitive deficits by modulating microglial activity, promoting neurotrophic factor release, and regulating inflammatory genes. The administration of CBD demonstrated a protective effect against Aβ toxicity both in vitro and in vivo, along with an amelioration of cognitive impairment in mice. These findings support the potential inclusion of CBD in future nutritional guidelines for Alzheimer's disease prevention.
Collapse
Affiliation(s)
- Long Chen
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211166, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211166, China
| | - Yuan Sun
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211166, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211166, China
| | - Jinran Li
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211166, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211166, China
| | - Sai Liu
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211166, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211166, China
| | - Hancheng Ding
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211166, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211166, China
| | - Guangji Wang
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211166, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211166, China
| | - Xinuo Li
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211166, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211166, China
| |
Collapse
|
46
|
Angelucci F, Veverova K, Katonová A, Vyhnalek M, Hort J. Serum PAI-1/BDNF Ratio Is Increased in Alzheimer's Disease and Correlates with Disease Severity. ACS OMEGA 2023; 8:36025-36031. [PMID: 37810633 PMCID: PMC10552510 DOI: 10.1021/acsomega.3c04076] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 07/27/2023] [Indexed: 10/10/2023]
Abstract
We previously demonstrated that serum levels of plasminogen activator inhibitor-1 (PAI-1), which inhibits both the tissue plasminogen activator (tPA) and plasmin activity, are increased in patients with Alzheimer's disease. tPA/plasmin not only prevents the accumulation of β-amyloid in the brain but also is involved in the synthesis of the brain-derived neurotrophic factor (BDNF), a neurotrophin whose levels are reduced in Alzheimer. In the present study, we compared BDNF serum levels in Alzheimer patients with dementia to those in Alzheimer patients with amnestic mild cognitive impairment and to cognitively healthy controls. Moreover, we examined whether the PAI-1/BDNF ratio correlates with disease severity, as measured by Mini-Mental State Examination. Our results showed that BDNF serum levels are lower (13.7% less) and PAI-1 levels are higher in Alzheimer patients with dementia than in Alzheimer patients with amnestic mild cognitive impairment patients (23% more) or controls (36% more). Furthermore, the PAI-1/BDNF ratio was significantly increased in Alzheimer patients as compared to amnestic mild cognitive impairment (36.4% more) and controls (40% more). Lastly, the PAI-1/BDNF ratio negatively correlated with the Mini-Mental score. Our results suggest that increased PAI-1 levels in Alzheimer, by impairing the production of the BDNF, are implicated in disease progression. They also indicate that the PAI-1/BDNF ratio could be used as a marker of Alzheimer. In support of this hypothesis, a strong negative correlation between the PAI-1/BDNF ratio and the Mini-Mental score was observed.
Collapse
Affiliation(s)
- Francesco Angelucci
- Memory
Clinic, Department of Neurology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague 150 06, Czech Republic
- International
Clinical Research Centre, St. Anne’s
University Hospital, Brno 602 00,Czech Republic
| | - Katerina Veverova
- Memory
Clinic, Department of Neurology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague 150 06, Czech Republic
| | - Alžbeta Katonová
- Memory
Clinic, Department of Neurology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague 150 06, Czech Republic
| | - Martin Vyhnalek
- Memory
Clinic, Department of Neurology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague 150 06, Czech Republic
| | - Jakub Hort
- Memory
Clinic, Department of Neurology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague 150 06, Czech Republic
- International
Clinical Research Centre, St. Anne’s
University Hospital, Brno 602 00,Czech Republic
| |
Collapse
|
47
|
Ploydang T, Khovidhunkit W, Tanaka H, Suksom D. Nordic Walking in Water on Cerebrovascular Reactivity and Cognitive Function in Elderly Patients with Type 2 Diabetes. Med Sci Sports Exerc 2023; 55:1803-1811. [PMID: 37204017 DOI: 10.1249/mss.0000000000003216] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
INTRODUCTION Aquatic Nordic walking (ANW) is a novel whole-body low-impact exercise that can be practiced by a variety of older adults with chronic conditions. However, its efficacy on several aspects of health is largely unknown. PURPOSE This study aimed to determine the effects of regular ANW on glycemic control and vascular function in older adults with type 2 diabetes and mild cognitive impairment. METHODS Thirty-three older adults with type 2 diabetes age 60-75 yr were randomly allocated to nonexercising control ( n = 17) or ANW ( n = 17) groups. Nordic walking was performed in a pool at water temperature of 34°C-36°C, three times per week for 12 wk. RESULTS Measures of functional physical fitness including chair stand, timed up and go, chair sit and reach, reach and back scratch, and 6-min walk test scores were all improved after ANW (all P < 0.05). Plasma glucose, glycosylated hemoglobin, and homeostasis model assessment of insulin resistance decreased (all P < 0.05) in ANW. Vascular reactivity as assessed by brachial flow-mediated dilation increased, and arterial stiffness as assessed by brachial-ankle pulse wave velocity decreased in ANW (all P < 0.05). No significant changes were observed in the control group. Middle cerebral artery pulsatility index decreased with ANW under normocapnia condition ( P < 0.05). Cerebrovascular conductance increased with ANW under hypercapnia condition. Montreal Cognitive Assessment score increased in the ANW group ( P < 0.001). Changes in Montreal Cognitive Assessment scores were positively associated with corresponding changes in brain-derived neurotrophic factor ( r = 0.540, P = 0.031). CONCLUSIONS Nordic walking in water was a safe and effective innovative exercise modality to improve glycemic control, vascular function, physical fitness, cerebrovascular reactivity, and cognitive function in older adults with type 2 diabetes.
Collapse
Affiliation(s)
| | | | - Hirofumi Tanaka
- Department of Kinesiology and Health Education, The University of Texas at Austin, Austin, TX
| | | |
Collapse
|
48
|
Chen MH, Liu XZ, Qu XW, Guo RB, Zhang L, Kong L, Yu Y, Liu Y, Zang J, Li XY, Li XT. ApoE-modified liposomes encapsulating resveratrol and salidroside alleviate manifestations of Alzheimer's disease in APP/PS-1 mice. Drug Dev Ind Pharm 2023; 49:559-571. [PMID: 37649422 DOI: 10.1080/03639045.2023.2252062] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 08/18/2023] [Accepted: 08/21/2023] [Indexed: 09/01/2023]
Abstract
OBJECTIVE Alzheimer's disease (AD) is a neurodegenerative disease that is associated with aging and is influenced by both genetic and environmental factors. Several studies and clinical trials have demonstrated that resveratrol (Res) and salidroside (Sal) are not only biologically safe but also influence AD biomarker trajectories. However, their clinical applications have been quite limited due to poor specificity, low solubility, and insufficient blood-brain barrier (BBB) penetration. Therefore, we developed a nano-drug delivery system in which Res and Sal were encapsulated in liposomes, which were surface-modified with ApoE (ApoE-Res/Sal-Lips) to compensate for these deficiencies. METHOD In this study, ApoE-Res/Sal-Lips were prepared using a standard thin-film hydration method for liposomes. Then, cellular uptake of the loaded liposomes was assessed in vitro using fluorescent staining assays. A BBB model was constructed to investigate the capacity of the liposomes to cross the BBB in vitro, and the ability of liposomes to target the brain was observed by in vivo imaging. In addition, the neuroprotective effects of the different liposome formulations in APP/PS-1 mice were evaluated by measuring the changes in levels of oxidative, anti-inflammatory, and anti-apoptotic factors in the mice brains. RESULTS In vitro, ApoE-Res/Sal-Lips increased the uptake of Res and Sal by bEnd.3 and N2a cells, enhanced BBB penetration, and improved transport efficiency. In vivo, the ApoE-Res/Sal-Lips were found to alleviate AD pathological symptoms, reduce learning and memory impairments, and improve brain function. CONCLUSION ApoE-Res/Sal-Lips provide a new method for the treatment of AD.
Collapse
Affiliation(s)
- Mu-Han Chen
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, P.R. China
| | - Xin-Ze Liu
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, P.R. China
| | - Xiu-Wu Qu
- Shanxi Key Laboratory of Innovative Drug for the Treatment of Serious Diseases Basing on the Chronic Inflammation, Shanxi University of Chinese Medicine, Jinzhong, P.R. China
| | - Rui-Bo Guo
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, P.R. China
| | - Lu Zhang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, P.R. China
| | - Liang Kong
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, P.R. China
| | - Yang Yu
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, P.R. China
| | - Yang Liu
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, P.R. China
| | - Juan Zang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, P.R. China
| | - Xiu-Ying Li
- Shanxi Key Laboratory of Innovative Drug for the Treatment of Serious Diseases Basing on the Chronic Inflammation, Shanxi University of Chinese Medicine, Jinzhong, P.R. China
| | - Xue-Tao Li
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, P.R. China
| |
Collapse
|
49
|
Rejdak K, Sienkiewicz-Jarosz H, Bienkowski P, Alvarez A. Modulation of neurotrophic factors in the treatment of dementia, stroke and TBI: Effects of Cerebrolysin. Med Res Rev 2023; 43:1668-1700. [PMID: 37052231 DOI: 10.1002/med.21960] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/21/2023] [Accepted: 03/27/2023] [Indexed: 04/14/2023]
Abstract
Neurotrophic factors (NTFs) are involved in the pathophysiology of neurological disorders such as dementia, stroke and traumatic brain injury (TBI), and constitute molecular targets of high interest for the therapy of these pathologies. In this review we provide an overview of current knowledge of the definition, discovery and mode of action of five NTFs, nerve growth factor, insulin-like growth factor 1, brain derived NTF, vascular endothelial growth factor and tumor necrosis factor alpha; as well as on their contribution to brain pathology and potential therapeutic use in dementia, stroke and TBI. Within the concept of NTFs in the treatment of these pathologies, we also review the neuropeptide preparation Cerebrolysin, which has been shown to resemble the activities of NTFs and to modulate the expression level of endogenous NTFs. Cerebrolysin has demonstrated beneficial treatment capabilities in vitro and in clinical studies, which are discussed within the context of the biochemistry of NTFs. The review focuses on the interactions of different NTFs, rather than addressing a single NTF, by outlining their signaling network and by reviewing their effect on clinical outcome in prevalent brain pathologies. The effects of the interactions of these NTFs and Cerebrolysin on neuroplasticity, neurogenesis, angiogenesis and inflammation, and their relevance for the treatment of dementia, stroke and TBI are summarized.
Collapse
Affiliation(s)
- Konrad Rejdak
- Department of Neurology, Medical University of Lublin, Lublin, Poland
| | | | | | - Anton Alvarez
- Medinova Institute of Neurosciences, Clinica RehaSalud, Coruña, Spain
| |
Collapse
|
50
|
Ziegler T, Tsiountsioura M, Meixner-Goetz L, Cvirn G, Lamprecht M. Polyphenols' Impact on Selected Biomarkers of Brain Aging in Healthy Middle-Aged and Elderly Subjects: A Review of Clinical Trials. Nutrients 2023; 15:3770. [PMID: 37686802 PMCID: PMC10490411 DOI: 10.3390/nu15173770] [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: 07/20/2023] [Revised: 08/25/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
With a constantly growing elderly population, incidences of neurodegenerative diseases are also rising and are expected to further increase over the next years, while costing health systems across the world trillions of dollars. Therefore, biomarkers to detect manifestations of brain aging early and interventions to slow down its pace are of great interest. In the last years, the importance of the neurotrophins brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF) in the context of cognitive function and the aging brain has increased, besides the already well-established amyloid-beta (Aβ) and tau plaques. Due to their wide range of beneficial health effects as well as their antioxidant and anti-inflammatory properties, a class of secondary plant-metabolites, the so-called polyphenols, gained increasing attention. In this review, we discuss the roles of BDNF, Aβ, NGF, and tau proteins as biomarkers of brain aging and the effect of dietary polyphenol interventions on these biomarkers, assessed via blood analysis, magnetic resonance imaging (MRI), and positron emission tomography (PET).
Collapse
Affiliation(s)
- Tobias Ziegler
- Division of Medicinal Chemistry, Otto Loewi Research Center, Medical University of Graz, 8010 Graz, Austria; (T.Z.); (M.T.); (G.C.)
- Juice Plus+ Science Institute, Memphis, TN 38017, USA;
| | - Melina Tsiountsioura
- Division of Medicinal Chemistry, Otto Loewi Research Center, Medical University of Graz, 8010 Graz, Austria; (T.Z.); (M.T.); (G.C.)
- Juice Plus+ Science Institute, Memphis, TN 38017, USA;
| | | | - Gerhard Cvirn
- Division of Medicinal Chemistry, Otto Loewi Research Center, Medical University of Graz, 8010 Graz, Austria; (T.Z.); (M.T.); (G.C.)
| | - Manfred Lamprecht
- Juice Plus+ Science Institute, Memphis, TN 38017, USA;
- Green Beat Institute of Nutrient Research, 8010 Graz, Austria
| |
Collapse
|