1
|
Turab A, Nescolarde-Selva JA, Ullah F, Montoyo A, Alfiniyah C, Sintunavarat W, Rizk D, Zaidi SA. Deep neural networks and stochastic methods for cognitive modeling of rat behavioral dynamics in T -mazes. Cogn Neurodyn 2025; 19:66. [PMID: 40290756 PMCID: PMC12031716 DOI: 10.1007/s11571-025-10247-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 03/23/2025] [Accepted: 03/26/2025] [Indexed: 04/30/2025] Open
Abstract
Modeling animal decision-making requires mathematical rigor and computational analysis to capture underlying cognitive mechanisms. This study presents a cognitive model for rat decision-making behavior in T -mazes by combining stochastic methods with deep neural architectures. The model adapts Wyckoff's stochastic framework, originally grounded in Bush's discrimination learning theory, to describe probabilistic transitions between directional choices under reinforcement contingencies. The existence and uniqueness of solutions are demonstrated via fixed-point theorems, ensuring the formulation is well-posed. The asymptotic properties of the system are examined under boundary conditions to understand the convergence behavior of decision probabilities across trials. Empirical validation is performed using Monte Carlo simulations to compare expected trajectories with the model's predictive output. The dataset comprises spatial trajectory recordings of rats navigating toward food rewards under controlled experimental protocols. Trajectories are preprocessed through statistical filtering, augmented to address data imbalance, and embedded using t-SNE to visualize separability across behavioral states. A hybrid convolutional-recurrent neural network (CNN-LSTM) is trained on these representations and achieves a classification accuracy of 82.24%, outperforming conventional machine learning models, including support vector machines and random forests. In addition to discrete choice prediction, the network reconstructs continuous paths, enabling full behavioral sequence modeling from partial observations. The integration of stochastic dynamics and deep learning develops a computational basis for analyzing spatial decision-making in animal behavior. The proposed approach contributes to computational models of cognition by linking observable behavior to internal processes in navigational tasks.
Collapse
Affiliation(s)
- Ali Turab
- School of Software, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi’an, 710072 China
- Department of Software and Computing Systems, University of Alicante, Alicante, Spain
- Department of Mathematics, Faculty of Science and Technology, Universitas Airlangga, 60115 Surabaya, Indonesia
| | | | - Farhan Ullah
- Cybersecurity Center, Prince Mohammad Bin Fahd University, 617, Al Jawharah, Khobar, Dhahran 34754 Saudi Arabia
| | - Andrés Montoyo
- Department of Software and Computing Systems, University of Alicante, Alicante, Spain
| | - Cicik Alfiniyah
- Department of Mathematics, Faculty of Science and Technology, Universitas Airlangga, 60115 Surabaya, Indonesia
| | - Wutiphol Sintunavarat
- Department of Mathematics and Statistics, Faculty of Science and Technology, Thammasat University Rangsit Center, 12120 Pathum Thani, Thailand
| | - Doaa Rizk
- Department of Mathematics, College of Science, Qassim University, 51452 Buraydah, Saudi Arabia
| | - Shujaat Ali Zaidi
- Department of Computer Science, Faculty of Science, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
2
|
Uvarajan D, Ravikumar M, Durairaj B. Mitigating Bisphenol-Induced Neurotoxicity: Exploring the Therapeutic Potential of Diosmin in Zebrafish Larvae. Food Chem Toxicol 2025; 200:115402. [PMID: 40122510 DOI: 10.1016/j.fct.2025.115402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 03/09/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025]
Abstract
Neurological disorders are commonly accompanied by inflammation of the brain, which can be triggered by oxidative stress and cell damage caused by hazardous environmental substances. The ubiquitous harmful chemical bisphenol A (BPA) has been linked to several neuropsychiatric disorders and is thought to contribute to oxidative damage. This study explored the mechanisms underlying the effects of BPA on neurological health. Diosmin (DM) is a natural flavonoid (C28H32O15) found in various plants, including citrus fruits and it possess various pharmacological activities. This study investigated the neuroprotective effects of DM on BPA-induced neuroinflammation in zebrafish larvae, suggesting its potential therapeutic uses. Developmental toxicity, including mortality, hatching rate, and heart rate, was evaluated to determine DM toxicity. Oxidative stress biomarkers such as reactive oxygen species (ROS), superoxide anions (O-2), lipid peroxidation (LPO), and nitric oxide (NO) were quantified using colorimetric assays in the head region of the larvae. Antioxidant enzyme activities were measured to assess the impact of DM on antioxidant defences. Neuroinflammation was evaluated by analysing pro-inflammatory markers using RT-qPCR, and motor neuron function was assessed using acetylcholinesterase (AChE) activity and behavioural assays. The findings indicate that exposure to DM prevents neurotoxicity induced by BPA by increasing antioxidant defence enzymes and reducing the levels of ROS, O2-, LPO, and NO in the head region of zebrafish larvae. Furthermore, DM enhanced motor neuron function by increasing AChE activity and decreasing neuroinflammation by reducing the levels of pro-inflammatory markers influenced by BPA. This study suggests that DM offers neuroprotection against BPA-induced oxidative damage and neuroinflammation, thereby paving the way for the development of new treatment options for neurological disorders.
Collapse
Affiliation(s)
- Deenathayalan Uvarajan
- Department of Biochemistry, PSG College of Arts & Science, Coimbatore, Tamil Nadu, India
| | - Manish Ravikumar
- Department of Biochemistry, PSG College of Arts & Science, Coimbatore, Tamil Nadu, India
| | - Brindha Durairaj
- Department of Biochemistry, PSG College of Arts & Science, Coimbatore, Tamil Nadu, India.
| |
Collapse
|
3
|
Mirehei M, Motamedi F, Maghsoudi N, Mansouri Z, Naderi S, Khodagholi F, Abbaszadeh F. Effects of Bufexamac, a class IIb HDAC inhibitor, on behavior and neuropathological features in an Aβ-induced rat model of Alzheimer's disease. Exp Gerontol 2025; 204:112746. [PMID: 40185252 DOI: 10.1016/j.exger.2025.112746] [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: 01/26/2025] [Revised: 03/24/2025] [Accepted: 03/31/2025] [Indexed: 04/07/2025]
Abstract
It has been suggested that Alzheimer's disease (AD), a progressive neurological condition, can potentially be treated through epigenetic means by targeting histone deacetylases (HDACs), enzymes that regulate gene expression. In this study, we investigated the molecular mechanisms of Bufexamac, in an animal model of AD. Bufexamac specifically targets Class IIb HDACs, which are particularly relevant in the context of neuroinflammation and neurodegeneration. This selectivity may reduce off-target effects commonly associated with broader-spectrum HDAC inhibitors, such as pan-HDAC inhibitors, which can affect multiple HDAC classes and potentially lead to undesirable side effects. Male rats injected with Aβ25-35 for AD-like symptoms were treated with 20 μg/rat Bufexamac for 8 days. Cognitive function, depression, and anxiety were assessed through behavioral tests, while Western blotting, H&E staining, and ELISA were used to detect protein expression, morphological changes, and enzyme activity. Bufexamac treatment markedly improved cognitive and behavioral impairments in Aβ-injected rats and regulated the key proteins related to neuroinflammation (GFAP, Iba1), histone, and α-tubulin acetylation. Simultaneously, it decreased the expression of proteins in the stromal interaction molecule (STIM) pathway. Furthermore, Bufexamac lowered the activity of monoamine oxidase enzymes, elevated the count of healthy neurons, and ameliorated neuronal structure in the hippocampus. Overall, these findings suggest that Bufexamac could be a more targeted therapy for AD than other non-selective HDAC inhibitors, which often have diverse functions and potential side effects. Bufexamac enhances cognitive function and alleviates depression and anxiety by regulating proteins related to neuroinflammation, histone, and α-tubulin acetylation, as well as modulating STIM levels and MAO activity.
Collapse
Affiliation(s)
- Monireh Mirehei
- Neuroscience Research Center, Institute of Neuroscience and Cognition, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fereshteh Motamedi
- Neuroscience Research Center, Institute of Neuroscience and Cognition, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nader Maghsoudi
- Neuroscience Research Center, Institute of Neuroscience and Cognition, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Mansouri
- Neuroscience Research Center, Institute of Neuroscience and Cognition, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soudabeh Naderi
- Neuroscience Research Center, Institute of Neuroscience and Cognition, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Institute of Neuroscience and Cognition, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Fatemeh Abbaszadeh
- Neurobiology Research Center, Institute of Neuroscience and Cognition, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Shao X, He L, Liu Y. The effects of exercise interventions on brain-derived neurotrophic factor levels in children and adolescents: a meta-analysis. Neural Regen Res 2025; 20:1513-1520. [PMID: 39075917 PMCID: PMC11624860 DOI: 10.4103/nrr.nrr-d-23-01296] [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: 08/03/2023] [Revised: 12/01/2023] [Accepted: 01/27/2024] [Indexed: 07/31/2024] Open
Abstract
Brain-derived neurotrophic factor is a crucial neurotrophic factor that plays a significant role in brain health. Although the vast majority of meta-analyses have confirmed that exercise interventions can increase brain-derived neurotrophic factor levels in children and adolescents, the effects of specific types of exercise on brain-derived neurotrophic factor levels are still controversial. To address this issue, we used meta-analytic methods to quantitatively evaluate, analyze, and integrate relevant studies. Our goals were to formulate general conclusions regarding the use of exercise interventions, explore the physiological mechanisms by which exercise improves brain health and cognitive ability in children and adolescents, and provide a reliable foundation for follow-up research. We used the PubMed, Web of Science, Science Direct, Springer, Wiley Online Library, Weipu, Wanfang, and China National Knowledge Infrastructure databases to search for randomized controlled trials examining the influences of exercise interventions on brain-derived neurotrophic factor levels in children and adolescents. The extracted data were analyzed using ReviewManager 5.3. According to the inclusion criteria, we assessed randomized controlled trials in which the samples were mainly children and adolescents, and the outcome indicators were measured before and after the intervention. We excluded animal experiments, studies that lacked a control group, and those that did not report quantitative results. The mean difference (MD; before versus after intervention) was used to evaluate the effect of exercise on brain-derived neurotrophic factor levels in children and adolescents. Overall, 531 participants (60 children and 471 adolescents, 10.9-16.1 years) were included from 13 randomized controlled trials. Heterogeneity was evaluated using the Q statistic and I2 test provided by ReviewManager software. The meta-analysis showed that there was no heterogeneity among the studies (P = 0.67, I2 = 0.00%). The combined effect of the interventions was significant (MD = 2.88, 95% CI: 1.53-4.22, P < 0.0001), indicating that the brain-derived neurotrophic factor levels of the children and adolescents in the exercise group were significantly higher than those in the control group. In conclusion, different types of exercise interventions significantly increased brain-derived neurotrophic factor levels in children and adolescents. However, because of the small sample size of this meta-analysis, more high-quality research is needed to verify our conclusions. This meta-analysis was registered at PROSPERO (registration ID: CRD42023439408).
Collapse
Affiliation(s)
- Xueyun Shao
- Physical Education School, Shenzhen University, Shenzhen, Guangdong Province, China
- Shenzhen Institute of Neuroscience, Shenzhen, Guangdong Province, China
| | - Longfei He
- Shenzhen Institute of Neuroscience, Shenzhen, Guangdong Province, China
| | - Yangyang Liu
- Shenzhen Institute of Neuroscience, Shenzhen, Guangdong Province, China
| |
Collapse
|
5
|
Shi JX, Wang ZY, Wang SW, Shen Q, Tan X. Exercise-mediated muscle-hypothalamus crosstalk: Improvement for cognitive dysfunction caused by disrupted circadian rhythm. Life Sci 2025; 373:123657. [PMID: 40306358 DOI: 10.1016/j.lfs.2025.123657] [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: 02/18/2025] [Revised: 04/14/2025] [Accepted: 04/20/2025] [Indexed: 05/02/2025]
Abstract
In contemporary societal evolution, the increasing disruption of the natural sleep-wake cycle, attributable to factors such as shift work and overexposure to artificial light, has been paralleled by a marked escalation in the incidence of cognitive impairments and the prevalence of neurodegenerative diseases. Current management strategies for cognitive impairments include pharmacological and non-pharmacological interventions. Pharmacological interventions for cognitive impairments typically involve medications to manage cognitive symptoms and improve neurological functions. However, these drugs show limited long-term efficacy in slowing disease progression and may cause side effects. Given the widespread occurrence of cognitive dysfunction, it is crucial to develop accessible non-pharmacological interventions. Physical activity and exercise have emerged as pivotal lifestyle determinants known to exert a modulatory effect on the risk profile for cognitive dysfunction caused by disrupted circadian rhythms. The skeletal muscle, a dynamic tissue, undergoes a profound morphological and metabolic reconfiguration in response to physical exertion, along with the secretion of myokines. Additionally, the hypothalamus, particularly the ventromedial nuclei, arcuate nuclei, and the suprachiasmatic nucleus, have crucial functions in regulating physical activity, influencing energy metabolism, and managing circadian cycles. Nevertheless, the communication between the hypothalamus and skeletal muscle during exercise is not fully understood. This narrative review integrates current knowledge on the interaction between the hypothalamus and skeletal muscle during exercise, emphasizing its neuroendocrine effects and potential therapeutic implications for alleviating cognitive dysfunction associated with disrupted circadian rhythms.
Collapse
Affiliation(s)
- Jun-Xiao Shi
- School of Basic Medical Science, Naval Medical University, Shanghai 200433, China
| | - Zi-Yuan Wang
- School of Basic Medical Science, Naval Medical University, Shanghai 200433, China
| | - Sheng-Wen Wang
- School of Basic Medical Science, Naval Medical University, Shanghai 200433, China
| | - Qi Shen
- Naval Medical Center, Naval Medical University (Second Military Medical University), Shanghai 200433, China.
| | - Xing Tan
- Naval Medical Center, Naval Medical University (Second Military Medical University), Shanghai 200433, China.
| |
Collapse
|
6
|
Rajavand H, Zalouli V, Nematollahi Z, Fathy-Karkaragh F, Karimigharighi E, Jafarizadeh F, Rabiei Rad A. The Cooperation of Neurogranin with Calmodulin Promotes the Treatment of Aging-Related Diseases via Regular Exercise. Mol Neurobiol 2025:10.1007/s12035-025-04959-6. [PMID: 40285939 DOI: 10.1007/s12035-025-04959-6] [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: 08/02/2024] [Accepted: 04/14/2025] [Indexed: 04/29/2025]
Abstract
Research has demonstrated that engaging in regular exercise has the potential to enhance cognitive function, promote neuroplasticity, and mitigate the likelihood of experiencing cognitive decline. The underlying mechanisms responsible for these effects are intricate and encompass various pathways, including the interaction between neurogranin and calmodulin. The activation of calcium signaling pathways is a significant mechanism through which regular exercise facilitates the treatment of age-related diseases. The activation of neurogranin and calmodulin induced by exercise can provide protection against neurodegeneration by promoting neuronal survival, mitigating oxidative stress, and improving mitochondrial function through the regulation of calcium homeostasis and energy metabolism. In addition, there is evidence suggesting that engaging in regular exercise can lead to an upregulation of neurotrophic factors, specifically brain-derived neurotrophic factor (BDNF). These factors are crucial for the survival of neurons, the plasticity of synapses, and overall cognitive function. Researchers have discovered the involvement of neurogranin in the regulation of BDNF signaling, underscoring its significance in exercise-induced neuroprotection and cognitive enhancement. The current work offers valuable insights into how neurogranin/calmodulin cooperation, facilitated by regular exercise, promotes the treatment of aging-related diseases. The results suggest that regular exercise could enhance memory, learning, synaptic plasticity, and resilience to neurological damage; promote recovery after brain injury; and treat aging-related disorders such as Alzheimer's disease.
Collapse
Affiliation(s)
- Hosniyeh Rajavand
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Vahideh Zalouli
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Zeinab Nematollahi
- 4UCL Department of Nanotechnology, Division of Surgery and Interventional Science, University College London, London, UK
| | - Farshid Fathy-Karkaragh
- Department of Psychology, Faculty of Educational Sciences and Psychology, University of Tehran, Tehran, Iran
| | - Elham Karimigharighi
- Diagnostic Radiology and Nuclear Medicine, University of Maryland Baltimore, Baltimore, MD, USA
| | - Farzad Jafarizadeh
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - Amirhossein Rabiei Rad
- Department of Physiotherapy and Rehabilitation, Faculty of Health Sciences, Tokat Gaziosmanpaşa University, Tokat, Turkey.
| |
Collapse
|
7
|
Li J, Dan Y, Su W, Zhao M, Chen Z, Zhao Z. Immune-related biomarkers in the neuromyelitis optica spectrum disorder; pathogenesis and therapeutic approaches. Exp Eye Res 2025:110395. [PMID: 40274184 DOI: 10.1016/j.exer.2025.110395] [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: 02/21/2025] [Revised: 04/06/2025] [Accepted: 04/21/2025] [Indexed: 04/26/2025]
Abstract
Neuromyelitis optica spectrum disorder (NMOSD) is an autoimmune inflammatory disease of the central nervous system that mostly affects the optic nerves and spinal cord. About eighty percent of patients have antibodies that are directed against the water channel aquaporin-4 (AQP4)-IgG, which is expressed on astrocytes. This protein was shown to be both a biomarker and a pathogenic cause of NMOSD. Researchers have discovered that antibodies against myelin oligodendrocyte glycoprotein (MOG) IgG can serve as a biomarker for a distinct condition known as MOG antibody-associated disease (MOGAD). This condition shares some similarities with AQP4-IgG-positive NMOSD, but it has distinct differences in terms of its underlying causes, clinical characteristics, response to treatment, and prognosis. Identifying AQP4 antibodies in the blood serum confirms the diagnosis of seropositive NMOSD. Nevertheless, it remains uncertain if there is a correlation between AQP4-IgG levels and disease activity, severity, responsiveness to medication, or long-term effects. Furthermore, there is still a need to establish and confirm biomarkers specifically for patients diagnosed with seronegative NMOSD. This study primarily examines the immunological aspects of NMOSD, which might have significant consequences for clinical practice. These implications include the possible use of new biomarkers to aid in the early and correct diagnosis of NMOSD, as well as the development of current treatment options to enhance the long-term prognosis of NMOSD patients.
Collapse
Affiliation(s)
- Jingyong Li
- Department of Laboratory Pathology and Blood Transfusion, People's Liberation Army Joint Logistic Support Force 945th Hospital, Yaan, 625000, China
| | - Ya Dan
- Department of Laboratory Pathology and Blood Transfusion, People's Liberation Army Joint Logistic Support Force 945th Hospital, Yaan, 625000, China
| | - Wei Su
- Department of Medical Imaging, the Second People's Hospital of Yaan, 625000, China
| | - Mingjun Zhao
- Department of Laboratory Pathology and Blood Transfusion, People's Liberation Army Joint Logistic Support Force 945th Hospital, Yaan, 625000, China
| | - Zhiguo Chen
- People's Liberation Army Joint Logistic Support Force rehabilitation center, Da Lian, Yaan, 625000, China
| | - Zhuyang Zhao
- Department of Laboratory Pathology and Blood Transfusion, People's Liberation Army Joint Logistic Support Force 945th Hospital, Yaan, 625000, China.
| |
Collapse
|
8
|
Rani P, Chahal S, Ranolia A, Kiran, Kumar D, Kataria R, Kumar P, Singh D, Duhan A, Jha V, Wahajuddin M, Joshi G, Sindhu J. Design and development of sulfenylated 5-aminopyrazoles as inhibitors of acetylcholinesterase and butyrylcholinesterase: exploring the implication for Aβ 1-42 aggregation inhibition in Alzheimer's disease. RSC Med Chem 2025:d5md00069f. [PMID: 40256309 PMCID: PMC12005478 DOI: 10.1039/d5md00069f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 03/16/2025] [Indexed: 04/22/2025] Open
Abstract
Current therapeutic regimens approved to treat Alzheimer's disease (AD) provide symptomatic relief by replenishing the acetylcholine levels in the brain by inhibiting AChE. However, these drugs don't halt or slow down the progression of Alzheimer's disease, which remains a major challenge. Evidence suggests a significant increase in BuChE activity with a decrease in AChE activity as the AD progresses along with the Aβ1-42 aggregation. To address this unmet need, we rationally developed sulfenylated 5-aminopyrazoles (3a-3o) via electro-organic synthesis in good to excellent yields (68-89%) and duly characterized them using spectrophotometric techniques. The compounds were tested for acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE) inhibition, with 3b (4-NO2) showing the highest potency. It exhibited IC50 values of 1.634 ± 0.066 μM against AChE and 0.0285 ± 0.019 μM against BuChE, outperforming donepezil and tacrine. Admittedly, 3b effectively inhibited Aβ1-42 aggregation and enhanced working memory, as indicated by the Y-maze test, besides portraying no cytotoxicity. The outcome was further corroborated using in silico techniques, leading to the elucidation of plausible inhibition and metabolism mechanisms.
Collapse
Affiliation(s)
- Payal Rani
- Department of Chemistry, COBS&H, CCS Haryana Agricultural University Hisar 125004 India
| | - Sandhya Chahal
- Department of Chemistry, Chaudhary Ranbir Singh University Jind 126102 India
| | - Anju Ranolia
- Department of Chemistry, COBS&H, CCS Haryana Agricultural University Hisar 125004 India
| | - Kiran
- Department of Chemistry, COBS&H, CCS Haryana Agricultural University Hisar 125004 India
| | - Devendra Kumar
- School of Pharmacy, Narsee Monjee Institute of Management Studies (NMIMS) Dist. Dhule Maharashtra-42400 India
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University) Varanasi 221005 India
| | - Ramesh Kataria
- Department of Chemistry at Panjab University Chandigarh-160014 India
| | - Parvin Kumar
- Department of Chemistry, Kurukshetra University Kurukshetra-136119 India
| | - Devender Singh
- Department of Chemistry, Maharshi Dayanand University Rohtak-124001 India
| | - Anil Duhan
- Department of Chemistry, COBS&H, CCS Haryana Agricultural University Hisar 125004 India
| | - Vibhu Jha
- Institute of Cancer Therapeutics School of Pharmacy and Medical Sciences, University of Bradford UK
| | - Muhammad Wahajuddin
- Institute of Cancer Therapeutics School of Pharmacy and Medical Sciences, University of Bradford UK
| | - Gaurav Joshi
- Institute of Cancer Therapeutics School of Pharmacy and Medical Sciences, University of Bradford UK
- Department of Pharmaceutical Sciences, Chauras Campus, HNB Garhwal University (A Central University) Srinagar Uttarakhand 246174 India
| | - Jayant Sindhu
- Department of Chemistry, COBS&H, CCS Haryana Agricultural University Hisar 125004 India
| |
Collapse
|
9
|
Kömür M, Kıyan HT, Öztürk AA. Development of donepezil hydrochloride-loaded PLGA-based nanoparticles for Alzheimer's disease treatment. Sci Rep 2025; 15:13184. [PMID: 40240764 PMCID: PMC12003670 DOI: 10.1038/s41598-025-95792-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
In recent years, nanoparticle (NP) systems have demonstrated significant promise in pharmaceutical applications. This study focused on the development of donepezil hydrochloride-loaded PLGA-NPs, prepared using the 'Double Emulsion Solvent Evaporation' method. The impact of varying concentrations of polyvinyl alcohol-(PVA) in the aqueous phase and sonication time on NP characteristics was comprehensively examined. Results showed that increasing PVA concentration and sonication time resulted in a reduction in NP size, with an optimal formulation (I-DNP) achieving a particle size of 136.37 nm ± 0.93 and a PDI of 0.122 ± 0.011, indicating uniformity. The zeta potential was measured at - 24.17mV ± 1.21, confirming the electrostatic stability of the formulation, essential for long-term stability. Trehalose was incorporated to enhance stability, and gastrointestinal stability testing revealed that I-DNP degraded faster in acidic environments. The encapsulation efficiency reached 69.22 ± 4.84%, suggesting effective drug loading, and release studies exhibited a sustained release profile, with a Fickian and non-Fickian release mechanism. DSC, FT-IR, and 1H-NMR analyses confirmed the encapsulation and structural integrity of the formulation. In biological activity studies, I-DNP exhibited potent anti-AChE and anti-BuChE activities, with Chorioallantoic Membrane (CAM) assays showing significant inhibition of angiogenesis. These findings highlight the potential of I-DNP as a promising therapeutic strategy for Alzheimer's disease, demonstrating its ability to enhance drug stability, controlled release, and potential blood-brain barrier (BBB) penetration. Future studies will focus on long-term stability testing and in vivo Alzheimer's models to further validate its clinical applicability. This research contributes to the advancement of nanoparticle-based drug delivery systems for neurodegenerative diseases, paving the way for innovative therapeutic approaches.
Collapse
Affiliation(s)
- Merve Kömür
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Anadolu University, Eskişehir, Türkiye
| | - Hülya Tuba Kıyan
- Department of Pharmacognosy, Faculty of Pharmacy, Anadolu University, Eskişehir, Türkiye
| | - A Alper Öztürk
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Anadolu University, Eskişehir, Türkiye.
| |
Collapse
|
10
|
Eslami M, Adampour Z, Fadaee Dowlat B, Yaghmayee S, Motallebi Tabaei F, Oksenych V, Naderian R. A Novel Frontier in Gut-Brain Axis Research: The Transplantation of Fecal Microbiota in Neurodegenerative Disorders. Biomedicines 2025; 13:915. [PMID: 40299512 PMCID: PMC12025253 DOI: 10.3390/biomedicines13040915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 04/02/2025] [Accepted: 04/07/2025] [Indexed: 04/30/2025] Open
Abstract
The gut-brain axis (GBA) represents a sophisticated bidirectional communication system connecting the central nervous system (CNS) and the gastrointestinal (GI) tract. This interplay occurs primarily through neuronal, immune, and metabolic pathways. Dysbiosis in gut microbiota has been associated with multiple neurodegenerative diseases, such as Parkinson's disease (PD), Alzheimer's disease (AD), multiple sclerosis (MS), and amyotrophic lateral sclerosis (ALS). In recent years, fecal microbiota transplantation (FMT) has gained attention as an innovative therapeutic approach, aiming to restore microbial balance in the gut while influencing neuroinflammatory and neurodegenerative pathways. This review explores the mechanisms by which FMT impacts the gut-brain axis. Key areas of focus include its ability to reduce neuroinflammation, strengthen gut barrier integrity, regulate neurotransmitter production, and reinstate microbial diversity. Both preclinical and clinical studies indicate that FMT can alleviate motor and cognitive deficits in PD and AD, lower neuroinflammatory markers in MS, and enhance respiratory and neuromuscular functions in ALS. Despite these findings, several challenges remain, including donor selection complexities, uncertainties about long-term safety, and inconsistencies in clinical outcomes. Innovations such as synthetic microbial communities, engineered probiotics, and AI-driven analysis of the microbiome hold the potential to improve the precision and effectiveness of FMT in managing neurodegenerative conditions. Although FMT presents considerable promise as a therapeutic development, its widespread application for neurodegenerative diseases requires thorough validation through well-designed, large-scale clinical trials. It is essential to establish standardized protocols, refine donor selection processes, and deepen our understanding of the molecular mechanisms behind its efficacy.
Collapse
Affiliation(s)
- Majid Eslami
- Cancer Research Center, Semnan University of Medical Sciences, Semnan 35147-99442, Iran;
- Department of Bacteriology and Virology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan 35147-99442, Iran
| | - Zarifeh Adampour
- Institute of Science, Biotechnology and Biosafety Department, Eskishehir Osmangazi University, Eskishehir 26040, Türkiye;
| | - Bahram Fadaee Dowlat
- School of Medicine, Iran University of Medical Sciences, Tehran 14496-14535, Iran
| | - Shayan Yaghmayee
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan 35147-99442, Iran
| | - Faezeh Motallebi Tabaei
- Department of Medical Microbiology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan 49189-36316, Iran
| | | | - Ramtin Naderian
- Clinical Research Development Unit, Kowsar Educational, Research and Therapeutic Hospital, Semnan University of Medical Sciences, Semnan 35147-99442, Iran
| |
Collapse
|
11
|
Razavi ZS, Razavi FS, Alizadeh SS. Inorganic nanoparticles and blood-brain barrier modulation: Advancing targeted neurological therapies. Eur J Med Chem 2025; 287:117357. [PMID: 39947054 DOI: 10.1016/j.ejmech.2025.117357] [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: 12/09/2024] [Revised: 01/19/2025] [Accepted: 01/31/2025] [Indexed: 02/24/2025]
Abstract
The blood-brain barrier (BBB) is a protective barrier that complicates the treatment of neurological disorders. Pharmaceutical compounds encounter significant challenges in crossing the central nervous system (CNS). Nanoparticles (NPs) are promising candidates for treating neurological conditions as they help facilitate drug delivery. This review explores the diverse characteristics and mechanisms of inorganic NPs (INPs), including metal-based, ferric-oxide, and carbon-based nanoparticles, which facilitate their passage through the BBB. Emphasis is placed on the physicochemical properties of NPs such as size, shape, surface charge, and surface modifications and their role in enhancing drug delivery efficacy, reducing immune clearance, and improving BBB permeability. Specific synthesis approaches are demonstrated, with an emphasis on the influence of each one on NP property, biological activity and the capability of an NP for its intended application. As for the advances in the field, the review emphasizes those characterized the NP formulation and surface chemistry that conquered the BBB and tested the need for its alteration. Current findings indicate that NP therapy can in the future enable effective targeting of specific brain disorders and eventually evolve this drug delivery system, which would allow for lower doses with less side effects.
Collapse
Affiliation(s)
- Zahra Sadat Razavi
- Physiology Research Center, Iran University Medical Sciences, Tehran, Iran; Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran.
| | - Fateme Sadat Razavi
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | | |
Collapse
|
12
|
Alikhanzade M, Khosravi M, Hosseini M, Rajabian A. Gallic acid enhances memory, learning and reduces neuroinflammation in a rat model of scopolamine-induced cholinergic dysfunction. Inflammopharmacology 2025; 33:2095-2108. [PMID: 40090981 DOI: 10.1007/s10787-025-01699-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 09/09/2024] [Indexed: 03/19/2025]
Abstract
Gallic acid (GA), a potent polyphenol antioxidant, has demonstrated beneficial effects on the nervous system. This study aimed to investigate the neuroprotective potential of GA on learning and memory in a rat model of scopolamine-induced cholinergic dysfunction. Additionally, the roles of oxidative stress and neuroinflammation were examined. Rats were divided into six groups: Control, scopolamine (2 mg/kg/day), scopolamine plus 25, 50, or 100 mg/kg of GA, and scopolamine plus 2 mg/kg of donepezil (DN, administered once daily). Behavioral performance was evaluated using the Morris Water Maze (MWM) and Passive Avoidance Test. Biochemical parameters were assessed to determine oxidative stress, and gene expression analyses were conducted to explore neuroinflammation in the hippocampus. The behavioral tests revealed that both GA and DN treatments improved the rats' performance in the MWM, as evidenced by their ability to locate the platform and spend more time in the target area. Additionally, GA administration increased the latency of entering the dark compartment and extended the time spent in the light compartment while reducing the frequency of dark compartment entries in the Passive Avoidance Test. Furthermore, GA exhibited antioxidant, anti-acetylcholinesterase, and anti-inflammatory effects, as indicated by the modulation of malondialdehyde levels, thiol content, superoxide dismutase activity, acetylcholinesterase activity, and the expression of inflammatory cytokines, including tumor necrosis factor-alpha (TNF-α), interleukin (IL)-1β, and IL-6. In conclusion, this study provides evidence for the potential therapeutic benefits of GA in Alzheimer's disease, highlighting its ability to enhance memory function and mitigate oxidative stress, acetylcholinesterase activity, and inflammation.
Collapse
Affiliation(s)
- Mahbobe Alikhanzade
- Department of Biology, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Maryam Khosravi
- Department of Biology, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Mahmoud Hosseini
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, 9177948564, Iran.
- Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, 9177948564, Iran.
| | - Arezoo Rajabian
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, 9177948564, Iran
- Department of Neurosciences, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, 9177948564, Iran
| |
Collapse
|
13
|
Khan S, Iqbal T, Zahoor T, Hussain R, Islam MS, Dahlous KA. Insight the confirmation of benzothiazolidinone-derived thiadiazole scaffolds as promising antiurease and anti-Alzheimer agents: synthesis, in vitro, and in silico investigations. Z NATURFORSCH C 2025:znc-2024-0243. [PMID: 40114449 DOI: 10.1515/znc-2024-0243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 02/26/2025] [Indexed: 03/22/2025]
Abstract
Alzheimer's disease is a serious neurological disorder, and traditional therapies for Alzheimer's, like radiation and surgical procedures, as well as chemotherapeutics, are usually linked with multiple negative consequences. Finding a novel therapeutic anti-Alzheimer agent with high efficacy and minimal side effects, we have designed and synthesized benzothiazolidinone-derived thiadiazole-based Schiff base derivatives (1-15). Biological assessment of these compounds was carried out against acetylcholinesterase and butyrylcholinesterase, and all the derivatives showed varying degrees of inhibitory activity. Analog 8 (IC50 = 3.60 ± 0.20 and 4.10 ± 0.20 μM for acetylcholinesterase and butyrylcholinesterase, respectively) demonstrated spellbinding efficacy in contrast to standard donepezil (IC50 = 50 ± 0.10 and 8.10 ± 0.20 μM). The surpassing inhibition of analog 8 is due to highly reactive CF3 moiety at the para-position, inhibiting the enzymes via strong hydrogen bond. Analog 7 with IC50 value of 5.70 ± 0.10 and 6.20 ± 0.40 μM was also found with strong therapeutic potential than standard drug. The strong inhibition potential of lead compounds was also evaluated under enzyme kinetics and spellbinding potential was observed. Biological effectiveness of potent compounds was validated by visualizing the binding interactions via in silico molecular docking study and prediction of drug-likeness via ADME analysis. All the synthesized compounds were analyzed for their structural confirmation via 1HNMR, 13CNMR, and HREI-MS.
Collapse
Affiliation(s)
- Shoaib Khan
- Department of Chemistry, 469633 Abbottabad University of Science and Technology , Abbottabad 22500, Pakistan
| | - Tayyiaba Iqbal
- Department of Chemistry, 469633 Abbottabad University of Science and Technology , Abbottabad 22500, Pakistan
| | - Tayyaba Zahoor
- Department of Chemistry, 469633 Abbottabad University of Science and Technology , Abbottabad 22500, Pakistan
| | - Rafaqat Hussain
- College of Biology, Hunan University Changsha, Hunan 410082, P.R. China
| | - Mohammad Shahidul Islam
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Kholood A Dahlous
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| |
Collapse
|
14
|
Rekha A, Afzal M, Babu MA, Menon SV, Nathiya D, Supriya S, Mishra SB, Gupta S, Goyal K, Rana M, Ali H, Imran M. GSK-3β dysregulation in aging: Implications for tau pathology and Alzheimer's disease progression. Mol Cell Neurosci 2025; 133:104005. [PMID: 40120784 DOI: 10.1016/j.mcn.2025.104005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/05/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025] Open
Abstract
The role of glycogen synthase kinase-3β (GSK-3β) in the pathogenesis of Alzheimer's disease (AD) is critical for linking amyloid-beta (Aβ) and Tau pathology. The activity of GSK-3β is dysregulated in the regulation of Tau hyperphosphorylation, formation of neurofibrillary tangles (NFTs), and production of Aβ by modulating amyloid precursor protein (APP) processing. This review discusses the mechanisms controlling GSK-3β dysregulation in aging and its influence on AD progression, focusing on the role of neuroinflammation, oxidative stress, and defective signaling pathways, including PI3K/Akt and Wnt. Critical analysis is presented for therapeutic strategies targeting GSK-3β using natural compounds (e.g., curcumin, geniposide) and emerging approaches such as TREM2 modulation and miRNA therapies. In preclinical models, these interventions promise to reduce Tau hyperphosphorylation and Aβ burden, along with associated neurodegeneration. Nevertheless, achieving selective GSK-3β inhibition and optimizing drug delivery are still critical barriers to clinical translation. This review underscores the central role of GSK-3β in AD pathogenesis to highlight its potential as a multifaceted therapeutic target of an innovative strategy for treating this complex neurodegenerative disease.
Collapse
Affiliation(s)
- A Rekha
- D.Y.Patil Medical College, Hospital and Research centre, Pimpri, Pune, India
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA University, Mathura, India
| | - Soumya V Menon
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Deepak Nathiya
- NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - S Supriya
- Department of CHEMISTRY, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Shakti Bedanta Mishra
- Department of Anaesthesiology, IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha 751003, India
| | - Sofia Gupta
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307, Punjab, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India.
| | - Mohit Rana
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia; Center for Health Research, Northern Border University, Arar 73213, Saudi Arabia
| |
Collapse
|
15
|
Nibrad D, Shiwal A, Tadas M, Katariya R, Kale M, Kotagale N, Umekar M, Taksande B. Therapeutic modulation of mitochondrial dynamics by agmatine in neurodegenerative disorders. Neuroscience 2025; 569:43-57. [PMID: 39890051 DOI: 10.1016/j.neuroscience.2025.01.061] [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: 12/10/2024] [Revised: 01/24/2025] [Accepted: 01/28/2025] [Indexed: 02/03/2025]
Abstract
Mitochondrial dysfunction is a pivotal factor in the pathogenesis of neurodegenerative disorders, driving neuronal degeneration through mechanisms involving oxidative stress, impaired energy production, and dysregulated calcium homeostasis. Agmatine, an endogenous polyamine derived from arginine, has garnered attention for its neuroprotective properties, including anti-inflammatory, anti-oxidative, and antiapoptotic effects. Recent studies have highlighted the potential of agmatine in preserving mitochondrial function and mitigating neurodegeneration, making it a promising candidate for therapeutic intervention. One of the key mechanisms by which agmatine exerts its neuroprotective effects is through the maintenance of mitochondrial homeostasis. Agmatine has been shown to modulate mitochondrial dynamics, promoting mitochondrial fusion and fission balance essential for cellular energy metabolism and signaling. Moreover, agmatine acts as a regulator of mitochondrial permeability transition pore (mPTP) opening, preventing excessive calcium influx and subsequent mitochondrial dysfunction. Despite promising findings, challenges such as optimizing agmatine's pharmacokinetics, determining optimal dosing regimens, and elucidating its precise molecular targets within mitochondria remain to be addressed. Future research directions should focus on developing targeted delivery systems for agmatine, investigating its interactions with mitochondrial proteins, and conducting well-designed clinical trials to evaluate its therapeutic efficacy and safety profile in neurodegenerative disorders. Overall, agmatine emerges as a novel therapeutic agent with the potential to modulate mitochondrial homeostasis and alleviate neurodegenerative pathology, offering new avenues for treating these debilitating conditions.
Collapse
Affiliation(s)
- Dhanshree Nibrad
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, (M.S.) 441 002, India
| | - Amit Shiwal
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, (M.S.) 441 002, India
| | - Manasi Tadas
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, (M.S.) 441 002, India
| | - Raj Katariya
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, (M.S.) 441 002, India
| | - Mayur Kale
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, (M.S.) 441 002, India
| | - Nandkishor Kotagale
- Government College of Pharmacy, Kathora Naka, VMV Road, Amravati, (M.S.) 444604, India
| | - Milind Umekar
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, (M.S.) 441 002, India
| | - Brijesh Taksande
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, (M.S.) 441 002, India.
| |
Collapse
|
16
|
Kulkarni AS, Ramana SR, Nuthakki VK, Bhatt S, Jamwal A, Nandawadekar LD, Jotshi A, Kumar A, Nandi U, Bharate SB, Reddy DS. Silicon incorporated tacrine: design, synthesis, and evaluation of biological and pharmacokinetic parameters. RSC Med Chem 2025:d5md00019j. [PMID: 40177641 PMCID: PMC11959489 DOI: 10.1039/d5md00019j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 02/19/2025] [Indexed: 04/05/2025] Open
Abstract
Tacrine, an orally bioavailable cholinesterase inhibitor, was previously used to treat Alzheimer's disease but was withdrawn due to hepatotoxicity. The unique structural features of tacrine have once again captured the interest of medicinal chemists. However, the blood-brain barrier (BBB) permeability hampered the development of the majority of its new analogs. Herein, we employed a silicon switch approach for improving the BBB permeability of CNS drugs with tacrine as a tool compound. The replacement of C2 methylene of tacrine with dimethyl silicon yielded 'sila-tacrine' that inhibits acetylcholinesterase as well as butyrylcholinesterase with IC50 values of 3.18 and 6.09 μM, respectively. Sila-tacrine competitively inhibits acetylcholinesterase while it is a non-competitive inhibitor of butyrylcholinesterase. The molecular docking results corroborated with the in vitro cholinesterase inhibition activity of tacrine vs. sila-tacrine. Sila-tacrine demonstrated metabolic stability in HLM and MLM and exhibited superior plasma exposure in an oral pharmacokinetic study in Swiss albino mice. However, tissue distribution studies revealed lower-than-expected brain levels due to efflux pump-mediated transport. This study offers a proof-of-concept for the silicon switch approach in improving the BBB permeability of CNS-active compounds.
Collapse
Affiliation(s)
- Akshay S Kulkarni
- Organic Chemistry Division, CSIR-National Chemical Laboratory Dr. Homi Bhabha Road Pune 411008 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
| | - Sreenivasa Rao Ramana
- Organic Chemistry Division, CSIR-National Chemical Laboratory Dr. Homi Bhabha Road Pune 411008 India
| | - Vijay K Nuthakki
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
- Natural Products & Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine Canal Road Jammu 180001 India
| | - Shipra Bhatt
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine Canal Road Jammu 180001 India
| | - Ashiya Jamwal
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine Canal Road Jammu 180001 India
| | - Laxman D Nandawadekar
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
- Department of Organic Synthesis & Process Chemistry, CSIR-Indian Institute of Chemical Technology Tarnaka Hyderabad-500007 India
| | - Anshika Jotshi
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine Canal Road Jammu 180001 India
| | - Ajay Kumar
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine Canal Road Jammu 180001 India
| | - Utpal Nandi
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine Canal Road Jammu 180001 India
- Bose Institute Unified Academic Campus Kolkata 700091 India
| | - Sandip B Bharate
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
- Natural Products & Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine Canal Road Jammu 180001 India
- Department of Natural Products & Medicinal Chemistry, CSIR-Indian Institute of Chemical Technology Tarnaka Hyderabad-500007 India
| | - D Srinivasa Reddy
- Organic Chemistry Division, CSIR-National Chemical Laboratory Dr. Homi Bhabha Road Pune 411008 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
- Natural Products & Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine Canal Road Jammu 180001 India
- Department of Organic Synthesis & Process Chemistry, CSIR-Indian Institute of Chemical Technology Tarnaka Hyderabad-500007 India
| |
Collapse
|
17
|
Liu J, Zhao W, Wang Y. Lights and shadows of clozapine on the immune system in schizophrenia: a narrative literature review. Metab Brain Dis 2025; 40:128. [PMID: 39954151 DOI: 10.1007/s11011-025-01558-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 02/12/2025] [Indexed: 02/17/2025]
Abstract
Schizophrenia is a chronic mental disorder and one of the main causes of disability in the world. Approximately 1% of the general population suffers from this disorder, and almost 30% of cases are unresponsive to antipsychotic therapies. Clozapine is a Food and Drug Administration (FDA)-approved antipsychotic drug for treatment-resistant schizophrenia (TRS). Clozapine is also approved for the prevention of suicide associated with schizophrenia. However, clozapine is not the preferred first-line medication because of its potential AEs, including agranulocytosis, metabolic syndromes, and myocarditis. Clozapine prescription requires weekly absolute neutrophil count (ANC) monitoring for the first six months, followed by biweekly monitoring until the patient has finished one year of treatment. Several psychiatric disorders have been reported to be associated with inflammatory biomarkers. Dysregulation of the immune system and the elevation of pro-inflammatory cytokines were also reported to be associated with schizophrenia, highlighting the necessity of further research into the etiology of the disease and the relationship between the immune system and clozapine-responsiveness to support better management of symptoms and potential AEs. In this framework, we searched PubMed using the medical subject headings (MeSH) terms "clozapine", "antipsychotics", "schizophrenia", "treatment-resistant schizophrenia", "immune system", "inflammation", "neuroinflammation", "biomarker", "cytokine", and "chemokine" with the aim of overview the impact of clozapine on the immune system in individuals with treatment-responsive and treatment-resistant schizophrenia.
Collapse
Affiliation(s)
- Jian Liu
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| | - Weimin Zhao
- Department of Preventive Medicine, The Affiliated Hospital of Changchun University of Chinese Medicine, Jilin, China.
| | - Yitong Wang
- Dermatology Department of Changchun Traditional Chinese Medicine Hospital, Jilin, China
| |
Collapse
|
18
|
Tepebaşı MY, Aşcı H, Koşar PA, Dinçer EN, Selçuk E, Kolay Ö, Hüseynov İ. Potential Ameliorating Effects of Fluvoxamine in a Rat Model of Endotoxin-Induced Neuroinflammation: Molecular Aspects Through SIRT-1/GPX-4 and HMGB-1 Signaling. Mol Neurobiol 2025:10.1007/s12035-025-04764-1. [PMID: 39954165 DOI: 10.1007/s12035-025-04764-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 02/08/2025] [Indexed: 02/17/2025]
Abstract
Research on the tissue-protective effects of fluvoxamine (FLV), a selective serotonin reuptake inhibitor, rapidly expands. This study explores FLV's potential to protect against lipopolysaccharide (LPS)-induced neuroinflammation, a key factor in systemic inflammation-related neuronal damage. Four equal groups of thirty-two female Wistar Albino rats were created: FLV, LPS-FLV (50 mg/kg intraperitoneal), LPS (5 mg/kg intraperitoneal), and control. Both drugs were given in one dose on the same day. Tissues from the brain cortex, cerebellum, and hippocampus were taken for histopathology, immunohistochemistry, biochemistry, and genetic analysis. In the LPS group, histological examinations revealed hyperemia, edema, mild degeneration, neuronal death, and modest gliosis. Additionally, while apelin and total antioxidant status levels were reduced, greater levels of oxidative stress index, glial fibrillary acidic protein (GFAP), mammalian target of rapamycin (mTOR), and total oxidant status were noted. FLV treatment reversed all these findings. Genetic analyses revealed that LPS decreased sirtuin-1 (SIRT-1) and glutathione peroxidase 4 (GPX-4) while increasing high mobility group box protein 1 (HMGB-1). FLV treatment reversed all these parameters, and a significant result was obtained only with GPX-4. In this study, FLV treatment was shown to have anti-inflammatory and neuroprotective effects through various mechanisms on the brain cortex, cerebellum, and hippocampus tissues in addition to its antidepressant effects.
Collapse
Affiliation(s)
| | - Halil Aşcı
- Department of Pharmacology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - Pınar Aslan Koşar
- Department of Medical Biology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - Emine Nur Dinçer
- Department of Pathology, Faculty of Veterinary Medicine, Mehmet Akif Ersoy University, Burdur, Turkey
| | - Esma Selçuk
- Department of Medical Biology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - Öznur Kolay
- Institute of Health Sciences, Department of Pharmacology, Suleyman Demirel University, Isparta, Turkey
| | - İbrahim Hüseynov
- Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| |
Collapse
|
19
|
Sadat Razavi Z, Sina Alizadeh S, Sadat Razavi F, Souri M, Soltani M. Advancing neurological disorders therapies: Organic nanoparticles as a key to blood-brain barrier penetration. Int J Pharm 2025; 670:125186. [PMID: 39788400 DOI: 10.1016/j.ijpharm.2025.125186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 01/03/2025] [Accepted: 01/05/2025] [Indexed: 01/12/2025]
Abstract
The blood-brain barrier (BBB) plays a vital role in protecting the central nervous system (CNS) by preventing the entry of harmful pathogens from the bloodstream. However, this barrier also presents a significant obstacle when it comes to delivering drugs for the treatment of neurodegenerative diseases and brain cancer. Recent breakthroughs in nanotechnology have paved the way for the creation of a wide range of nanoparticles (NPs) that can serve as carriers for diagnosis and therapy. Regarding their promising properties, organic NPs have the potential to be used as effective carriers for drug delivery across the BBB based on recent advancements. These remarkable NPs have the ability to penetrate the BBB using various mechanisms. This review offers a comprehensive examination of the intricate structure and distinct properties of the BBB, emphasizing its crucial function in preserving brain balance and regulating the transport of ions and molecules. The disruption of the BBB in conditions such as stroke, Alzheimer's disease, and Parkinson's disease highlights the importance of developing creative approaches for delivering drugs. Through the encapsulation of therapeutic molecules and the precise targeting of transport processes in the brain vasculature, organic NP formulations present a hopeful strategy to improve drug transport across the BBB. We explore the changes in properties of the BBB in various pathological conditions and investigate the factors that affect the successful delivery of organic NPs into the brain. In addition, we explore the most promising delivery systems associated with NPs that have shown positive results in treating neurodegenerative and ischemic disorders. This review opens up new possibilities for nanotechnology-based therapies in cerebral diseases.
Collapse
Affiliation(s)
- Zahra Sadat Razavi
- Physiology Research Center, Iran University Medical Sciences, Tehran, Iran; Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | | | - Fateme Sadat Razavi
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - Mohammad Souri
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - M Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran; Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, Canada; Centre for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, Canada; Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, Canada; Centre for Sustainable Business, International Business University, Toronto, Canada.
| |
Collapse
|
20
|
Singh A, Maheshwari S, Yadav JP, Kumar R, Verma A, Singh S, Prajapati BG. Bioactive Compound-Fortified Nanocarriers in the Management of Neurodegenerative Disease: A Review. Chem Biodivers 2025:e202402018. [PMID: 39928755 DOI: 10.1002/cbdv.202402018] [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: 08/21/2024] [Revised: 02/09/2025] [Accepted: 02/10/2025] [Indexed: 02/12/2025]
Abstract
Individual around the globe faces enormous problems from illnesses of the neurological system and the cerebrum, including neurodegenerative conditions and brain tumors. There are still no demonstrated viable treatments for neurological conditions, despite advances in drug delivery technologies such as solid lipid nanoparticles, nanostructured lipid carriers, and nano-liposomes. To address this, there is growing interest in leveraging naturally occurring bioactive substances for their therapeutic potential. However, challenges such as limited bioavailability and metabolism hinder their efficacy, particularly in the brain. Although various pharmaceutical interventions exist for neurodegenerative diseases, they often come with significant side effects, and there is currently no specific treatment to cure or slow down disease progression. Challenges such as the blood-brain barrier and blood-cerebrospinal fluid barrier present significant obstacles to deliver drugs into the brain. Strategies to improve drug penetration across these barriers include targeting specific transport systems and developing innovative drug delivery approaches. Hence, the development of nanocarriers capable of targeting bioactive compounds to the brain represents a promising approach for neurodegenerative disease therapy. This review explores the potential of bioactive compound-fortified nano-delivery systems for treating neurodegenerative diseases, with various compounds offering unique avenues for investigating neurodegeneration pathways and strategies in overcoming associated challenges.
Collapse
Affiliation(s)
- Aditya Singh
- Department of Pharmacy, Faculty of Pharmacy, Integral University, Lucknow, Uttar Pradesh, India
| | - Shubhrat Maheshwari
- Faculty of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj, India
| | - Jagat Pal Yadav
- Faculty of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj, India
| | - Ravi Kumar
- Institute of pharmaceutical sciences, J.S. University, Shikohabad, Uttar Pradesh, India
| | - Amita Verma
- Faculty of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj, India
- School of Pharmacy, Graphic Era Hill University, Dehradun, Uttrakhand, India
| | - Sudarshan Singh
- Office of Research Administration, Chiang Mai University, Chiang Mai, Thailand
- Faculty of Pharmacy, Chiang Mai University, Chiang Mai, Thailand
| | - Bhupendra G Prajapati
- Shree S. K. Patel College of Pharmaceutical Education and Research, Ganpat University, Gujarat, India
- Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand
| |
Collapse
|
21
|
Huang Z, Liu Q, Guo Q, Gao J, Zhang L, Li L. Effects and mechanisms of Apelin in treating central nervous system diseases. Neuroscience 2025; 566:177-189. [PMID: 39681256 DOI: 10.1016/j.neuroscience.2024.12.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 12/18/2024]
Abstract
Apelin, an endogenous ligand of G protein-coupled receptor APJ, is widely distributed in the central nervous system (CNS). It can be divided into such subtypes as Apelin-13, Apelin-17, and Apelin-36 as they have different amino acid structures. All Apelin is widely studied as an adipokine, showing a significant protective effect through regulating apoptosis, autophagy, oxidative stress, angiogenesis, inflammation, and other pathophysiological processes. As an adipokine, Apelin has been found to play a crucial role in cardiovascular disease development. In this paper, we reviewed the effects and mechanisms of Apelin in treating CNS diseases, such as neurotrauma, stroke, spinal cord injury, primary tumors, neurodegenerative diseases, psychiatric diseases, epilepsy, and pain.
Collapse
Affiliation(s)
- Zimeng Huang
- Medicine School, Qingdao University, 308 Ningxia Road, Shinan District, Qingdao 266071, China
| | - Qing Liu
- Department of Anatomy, School of Basic Medicine, Shandong University, Jinan, Shandong, 250021, China; School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266071, China
| | - Qixuan Guo
- Department of Human Anatomy, Binzhou Medical University, Yantai, Shandong, 264003, China
| | - Jianqing Gao
- College of Pharmaceutical Sciences and Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China.
| | - Luping Zhang
- Department of Human Anatomy, Binzhou Medical University, Yantai, Shandong, 264003, China.
| | - Liming Li
- Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao 266071, China.
| |
Collapse
|
22
|
Divya, Faruq M, Nazir SS, Kaushik P, Parvez S, Vohora D. Ganaxolone Reverses the Effect of Amyloid β-Induced Neurotoxicity by Regulating the Liver X Receptor Expression in APP Transfected SH-SY5Y Cells and Murine Model of Alzheimer's Disease. J Neurochem 2025; 169:e70007. [PMID: 39936324 DOI: 10.1111/jnc.70007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/06/2024] [Accepted: 01/01/2025] [Indexed: 02/13/2025]
Abstract
Inhibiting β-amyloid aggregation and enhancing its clearance are the key strategies in Alzheimer's disease (AD) treatment. Liver X receptors (LXRs) plays a crucial role in cholesterol homeostasis and inflammation, and their activation can clear Aβ aggregates in AD. Allopregnanolone, a neurosteroid, positively influences AD through LXR regulation, while ganaxolone, its synthetic analog, is known for its neuroprotective properties. This study explores the effect of ganaxolone on LXR activation and regulation of genes involved in mitigating Aβ toxicity and tauopathy in SH-SY5Y cells transfected with APP695 Swe/Ind plasmid and an Aβ1-42 induced AD mouse model. Molecular docking stimulations indicated ganaxolone's binding and interaction with LXRβ. Subsequently, transfected neuronal cells exhibited increased mRNA levels of APP, TNF-α and IL-1β, decreased cell viability, reduced MMP and altered protein expression of Aβ, LXR, BCL-2, APOE, ABCA1, along with increased levels of mROS, Bax, and caspase 3 activity. Ganaxolone treatment significantly abrogated Aβ-induced effect in transfected neuronal cells by enhancing LXRβ expression, inducing LXR:RXR colocalization, thereby increasing APOE and ABCA1 expression. It also decreased tau mRNA levels in transfected cells. Importantly, in AD mice, ganaxolone ameliorated cognitive impairment, reduced Aβ toxicity, tau levels, and neuroinflammatory markers, restored mitochondrial function, and decreased neuronal apoptosis. Taken together, these novel results highlight the central role of LXR in mediating Aβ-induced toxicity and provide preclinical evidence for ganaxolone as a potential agent to reduce toxicity in an LXR-dependent manner. This may serve as a promising treatment strategy to slow or prevent neurodegeneration in AD patients.
Collapse
Affiliation(s)
- Divya
- Department of Pharmacology, School of Pharmaceutical Education & Research (SPER), Jamia Hamdard, New Delhi, India
| | - Mohammed Faruq
- Division of Genomics and Molecular Medicine, CSIR-Institute of Genomics and Integrative Biology (IGIB), New Delhi, India
| | - Sheikh Sana Nazir
- Department of Pharmacology, School of Pharmaceutical Education & Research (SPER), Jamia Hamdard, New Delhi, India
| | - Pooja Kaushik
- Department of Toxicology, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi, India
| | - Suhel Parvez
- Department of Toxicology, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi, India
| | - Divya Vohora
- Department of Pharmacology, School of Pharmaceutical Education & Research (SPER), Jamia Hamdard, New Delhi, India
| |
Collapse
|
23
|
Dhariwal R, Jain M, Mir YR, Singh A, Jain B, Kumar P, Tariq M, Verma D, Deshmukh K, Yadav VK, Malik T. Targeted drug delivery in neurodegenerative diseases: the role of nanotechnology. Front Med (Lausanne) 2025; 12:1522223. [PMID: 39963432 PMCID: PMC11831571 DOI: 10.3389/fmed.2025.1522223] [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: 11/04/2024] [Accepted: 01/06/2025] [Indexed: 02/20/2025] Open
Abstract
Neurodegenerative diseases, characterized by progressive neuronal loss and cognitive impairments, pose a significant global health challenge. This study explores the potential of nanotherapeutics as a promising approach to enhance drug delivery across physiological barriers, particularly the blood-brain barrier (BBB) and blood-cerebrospinal fluid barrier (B-CSFB). By employing nanoparticles, this research aims to address critical challenges in the diagnosis and treatment of conditions such as Alzheimer's, Parkinson's, and Huntington's diseases. The multifactorial nature of these disorders necessitates innovative solutions that leverage nanomedicine to improve drug solubility, circulation time, and targeted delivery while minimizing off-target effects. The findings underscore the importance of advancing nanomedicine applications to develop effective therapeutic strategies that can alleviate the burden of neurodegenerative diseases on individuals and healthcare systems.
Collapse
Affiliation(s)
- Rupal Dhariwal
- Research and Development Cell, Parul University, Vadodara, India
- Department of Life Sciences, Parul Institute of Applied Sciences, Parul University, Vadodara, India
| | - Mukul Jain
- Research and Development Cell, Parul University, Vadodara, India
- Department of Life Sciences, Parul Institute of Applied Sciences, Parul University, Vadodara, India
| | - Yaser Rafiq Mir
- Department of Biotechnology, Baba Ghulam Shah Badshah University, Rajouri, India
| | - Abhayveer Singh
- Centre for Research Impact and Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, India
| | - Bhavik Jain
- Chitkara Centre for Research and Development, Chitkara University, Baddi, India
| | - Pankaj Kumar
- Department of Environmental Science, Parul Institute of Applied Sciences, Parul University, Vadodara, India
| | - Mohd Tariq
- Department of Life Sciences, Parul Institute of Applied Sciences, Parul University, Vadodara, India
- Department of Biotechnology, Graphic Era (Deemed to be University), Dehradun, India
| | - Devvret Verma
- Department of Biotechnology, Graphic Era (Deemed to be University), Dehradun, India
| | - Khemraj Deshmukh
- Department of Biomedical Engineering, Parul Institute of Technology, Parul University, Vadodara, India
| | | | - Tabarak Malik
- Department of Biomedical Sciences, Institute of Health, Jimma University, Jimma, Ethiopia
- Division of Research & Development, Lovely Professional University, Phagwara, Punjab, India
| |
Collapse
|
24
|
Saleh SR, Khamiss SE, Aly Madhy S, Khattab SN, Sheta E, Elnozahy FY, Thabet EH, Ghareeb DA, Awad D, El-Bessoumy AA. Biochemical investigation and in silico analysis of the therapeutic efficacy of Ipriflavone through Tet-1 Surface-Modified-PLGA nanoparticles in Streptozotocin-Induced Alzheimer's like Disease: Reduced oxidative damage and etiological Descriptors. Int J Pharm 2025; 669:125021. [PMID: 39631714 DOI: 10.1016/j.ijpharm.2024.125021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 11/29/2024] [Accepted: 11/30/2024] [Indexed: 12/07/2024]
Abstract
Ipriflavone (IPRI), an isoflavone derivative, is clinically used to prevent postmenopausal bone loss in addition to its antioxidant and cognitive benefits. However, its poor aqueous solubility retained its bioavailability. New strategies have been developed to improve the bioavailability and solubility of neurological medications to enhance their potency and limit adverse effects. This study aimed to prepare targeted IPRI-poly-lactic-co-glycolic acid (PLGA) nanoparticles coupled with Tet-1 peptide to increase the therapeutic potency of IPRI in a rat model of Alzheimer's disease (AD). Streptozotocin (STZ) exacerbates Alzheimer-related alterations by promoting central insulin resistance resulted from defective signaling pathways related to neuroinflammation and neurotoxicity. Bilateral intracerebroventricular (icv) injection of STZ was used to introduce the AD model. Icv-STZ injection significantly affected brain insulin, oxidative stress, inflammatory, and apoptotic indicators and caused behavioral abnormalities. STZ promoted the formation of amyloid β42 (Aβ42) by increasing BACE1 and reducing ADAM10 and ADAM17 expression levels. STZ also triggered the accumulation of neurofibrillary tangles and synaptic dysfunction, which are crucial for neurological impairments. Icv-STZ injection showed evident degenerative changes in the pyramidal cell layer and significantly reduced the count of viable cells in both CA1 and prefrontal cortex, indicating increased neuronal cell death. IPRI successfully ameliorated cognitive dysfunction by improving the phosphorylated forms of cAMP-response element-binding protein (pCREB) and extracellular signal-regulated kinase 1/2 (pERK1/2) related to synaptic plasticity. Targeted IPRI nanoparticles exceeded free IPRI potential in reducing oxidative stress, acetylcholinesterase/monoamine oxidase activities, Tau phosphorylation, and Aβ42 levels revealing less degenerative changes and increased viable neuron counts. IPRI-targeted nanoparticles improved the neuroprotective potential of free IPRI, making this strategy applicable to treat many neurodegenerative diseases. Finally, the in silico study predicted its ability to cross the BBB and to bind various protein targets in the brain.
Collapse
Affiliation(s)
- Samar R Saleh
- Biochemistry Department, Faculty of Science, Alexandria University, Alexandria 21511, Egypt; Bio-Screening and Preclinical Trial Lab, Biochemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt.
| | - Salma E Khamiss
- Biochemistry Department, Faculty of Science, Alexandria University, Alexandria 21511, Egypt; Bio-Screening and Preclinical Trial Lab, Biochemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt.
| | - Somaya Aly Madhy
- Chemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt.
| | - Sherine N Khattab
- Chemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt.
| | - Eman Sheta
- Pathology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt.
| | - Fatma Y Elnozahy
- Medical Physiology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt.
| | - Eman H Thabet
- Medical Physiology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt; Center of Excellence for Research in Regenerative Medicine and Applications (CERRMA), Faculty of Medicine, Alexandria University, Alexandria, Egypt.
| | - Doaa A Ghareeb
- Biochemistry Department, Faculty of Science, Alexandria University, Alexandria 21511, Egypt; Bio-Screening and Preclinical Trial Lab, Biochemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt.
| | - Doaa Awad
- Biochemistry Department, Faculty of Science, Alexandria University, Alexandria 21511, Egypt.
| | - Ashraf A El-Bessoumy
- Biochemistry Department, Faculty of Science, Alexandria University, Alexandria 21511, Egypt.
| |
Collapse
|
25
|
Li Z, Zhao Q, Zhou J, Li Y, Zheng Y, Chen L. A reactive oxygen species-responsive hydrogel loaded with Apelin-13 promotes the repair of spinal cord injury by regulating macrophage M1/M2 polarization and neuroinflammation. J Nanobiotechnology 2025; 23:12. [PMID: 39794784 PMCID: PMC11724542 DOI: 10.1186/s12951-024-02978-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 11/04/2024] [Indexed: 01/13/2025] Open
Abstract
Spinal cord injury (SCI) is a chronic condition whereby persistent aberrant macrophage activation hinders the repair process. During acute trauma, dominant M1 macrophages produce high levels of reactive oxygen species (ROS), leading to increased apoptosis in neurons, glial cells, and oligodendrocytes. This study investigated the specific effects of a ROS-responsive hydrogel loaded with Apelin-13 (Apelin-13@ROS-hydrogel) on macrophage polarization and neuroinflammation, thereby exploring its role in boosting SCI repair. Apelin-13@ROS-hydrogel was prepared, and its ROS-scavenging capacities were evaluated using DPPH, H2O2, and ·O2- assays. The effects of Apelin-13@ROS-hydrogel on macrophage polarization, inflammatory mediators and oxidative stress were assessed in LPS-pre-treated microglia BV2 cells and an SCI rat model. Apelin-13 was downregulated in SCI rats. Treatment with Apelin-13 improved functional recovery and reduced inflammatory factors and M1 markers but increased the M2 marker Arg-1. Apelin-13@ROS-hydrogel showed significantly higher ROS-scavenging capacities compared to the control hydrogel. Apelin-13@ROS-hydrogel decreased pro-inflammatory mediators and increased anti-inflammatory mediators in BV2 cells. Apelin-13@ROS-hydrogel enhanced the healing process and neurological functions, reducing inflammatory factors and M1 markers while increasing Arg-1 levels by day 28 in SCI rats. Collectively, Apelin-13 enhances SCI repair through macrophage regulation, M1/M2 polarization, and neuroinflammation. The ROS-responsive hydrogel further amplifies these effects, offering a promising therapeutic strategy for SCI.
Collapse
Affiliation(s)
- Zhiyue Li
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Qun Zhao
- Health Management Medicine Center, The Third Xiangya Hospital, Central South University, Changsha, 410013, China.
| | - Jiahui Zhou
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, 410013, China.
| | - Yuyan Li
- NanChang University Queen Mary School, Nanchang, 330038, China
| | - Yifan Zheng
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Linxi Chen
- School of Pharmaceutical Science, University of South China, Hengyang, 421001, China
| |
Collapse
|
26
|
Nabizadeh F, Sheykhlou S, Mahmoodi S, Khalili E, Zafari R, Hosseini H. Neuroimaging Findings of Psychosis in Alzheimer's Disease: A Systematic Review. Brain Behav 2025; 15:e70205. [PMID: 39740792 DOI: 10.1002/brb3.70205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 11/15/2024] [Accepted: 11/23/2024] [Indexed: 01/02/2025] Open
Abstract
BACKGROUND Previous studies on neuroimaging findings in Alzheimer's disease (AD) patients with hallucinations and delusions have yielded inconsistent results. We aimed to systematically review neuroimaging findings of delusions and hallucinations in AD patients to describe the most prominent neuroimaging features. METHODS We performed a comprehensive search in three online databases, including PubMed, Scopus, and Web of Science in June 2023. We included studies that reported neuroimaging features of AD patients with delusion, hallucination, or psychosis. RESULTS After the screening, 34 studies with 2241 AD patients were eligible to be included in our qualitative synthesis. On the basis of the included studies, there are significant changes in the volume and perfusion levels of broad brain areas, including the hippocampus, amygdala, insula, cingulate, occipital, frontal, prefrontal, orbitofrontal, temporal, and parietal cortices in these patients. Moreover, AD patients with psychosis, hallucinations, or delusions reflected different EEG waves compared to AD patients without these disorders. CONCLUSION The results of our review provided evidence about the neuroimaging alterations in AD patients suffering from psychosis, hallucinations, and delusions using different imaging methods. AD patients with psychosis, hallucinations, or delusions have significant differences in the volume and perfusion levels of various brain regions along with alterations in EEG waves and biological molecules compared to patients with only AD.
Collapse
Affiliation(s)
- Fardin Nabizadeh
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Alzheimer's Disease Institute, Tehran, Iran
| | - Shadi Sheykhlou
- Medical Laboratory Department, Iran University of Medical Sciences, Tehran, Iran
| | - Sara Mahmoodi
- Medical Laboratory Department, Iran University of Medical Sciences, Tehran, Iran
| | - Elham Khalili
- Universal Scientific Education and Research Network (USERN), Bandar Abbas, Hormozgan, Iran
- Cardiovascular Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Rasa Zafari
- School of Medicine, Tehran University of Medical Science, Tehran, Iran
| | - Helia Hosseini
- Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
27
|
Oruç KY, Ağtürk G, Oruç A, Yanar K, Seymen HO. Protective effect of Apelin-13 on D-glutamic acid-induced excitotoxicity in SH-SY5Y cell line: An in-vitro study. Neuropeptides 2025; 109:102483. [PMID: 39547009 DOI: 10.1016/j.npep.2024.102483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/08/2024] [Accepted: 11/03/2024] [Indexed: 11/17/2024]
Abstract
Excitotoxicity, resulting from excessive accumulation of glutamate in the extracellular space, leads to neuronal cell death. This study investigates the protective effects of Apelin-13 on D-Glutamic acid-induced excitotoxicity in SH-SY5Y human neuroblastoma cells, an in-vitro model for neurodegenerative diseases. Unlike the commonly studied L-glutamic acid, this research focuses on D-Glutamic acid to understand its specific impacts. SH-SY5Y cells were treated with varying concentrations of D-Glutamic acid and Apelin-13, followed by analyses at 12 and 24 h to evaluate cell viability, oxidative stress markers, and inflammatory cytokine levels. Cell viability assays revealed significant cytotoxic effects of D-Glutamic acid at doses of 10 mM and 20 mM, reducing viability by over 50 %. However, Apelin-13 treatment mitigated these effects, especially at 2 μg/ml, enhancing cell viability and reducing inflammatory cytokine levels (IL-1β and TNF-α). Apelin-13 also increased anti-inflammatory cytokine levels (IL-10 and TGF-β1) and brain-derived neurotrophic factor (BDNF), indicating its neuroprotective role. Oxidative stress markers, including ROS, AGE, AOPP, DT, T-SH, were significantly elevated by D-Glutamic acid but effectively reduced by Apelin-13. The neuroprotective mechanisms of Apelin-13 involve modulation of cAMP/PKA and MAPK signaling pathways, enhancing BDNF synthesis and suppressing oxidative stress and inflammatory responses. This study is the first to demonstrate the effects of D-Glutamic acid on SH-SY5Y cells. It highlights Apelin-13's potential as a therapeutic agent against excitotoxicity-induced neuronal damage, emphasizing its ability to modulate key molecular pathways involved in inflammation and oxidative stress. Further in-vivo studies are warranted to explore the long-term neuroprotective effects of Apelin-13 in treating neurodegenerative diseases.
Collapse
Affiliation(s)
- Kadriye Yağmur Oruç
- Istanbul University-Cerrahpaşa, Cerrahpaşa Faculty of Medicine, Department of Physiology, Istanbul, Turkey; Istinye University, Faculty of Medicine, Department of Physiology, Istanbul, Turkey.
| | - Gökhan Ağtürk
- Haliç University, Faculty of Medicine, Department of Physiology, Istanbul, Turkey
| | - Aykut Oruç
- Istanbul University-Cerrahpaşa, Cerrahpaşa Faculty of Medicine, Department of Physiology, Istanbul, Turkey
| | - Karolin Yanar
- Istanbul University-Cerrahpaşa, Cerrahpaşa Faculty of Medicine, Department of Medical Biochemistry, Istanbul, Turkey
| | - Hakkı Oktay Seymen
- Istanbul University-Cerrahpaşa, Cerrahpaşa Faculty of Medicine, Department of Physiology, Istanbul, Turkey
| |
Collapse
|
28
|
Jászberényi M, Thurzó B, Jayakumar AR, Schally AV. The Aggravating Role of Failing Neuropeptide Networks in the Development of Sporadic Alzheimer's Disease. Int J Mol Sci 2024; 25:13086. [PMID: 39684795 DOI: 10.3390/ijms252313086] [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: 10/12/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
Alzheimer's disease imposes an increasing burden on aging Western societies. The disorder most frequently appears in its sporadic form, which can be caused by environmental and polygenic factors or monogenic conditions of incomplete penetrance. According to the authors, in the majority of cases, Alzheimer's disease represents an aggravated form of the natural aging of the central nervous system. It can be characterized by the decreased elimination of amyloid β1-42 and the concomitant accumulation of degradation-resistant amyloid plaques. In the present paper, the dysfunction of neuropeptide regulators, which contributes to the pathophysiologic acceleration of senile dementia, is reviewed. However, in the present review, exclusively those neuropeptides or neuropeptide families are scrutinized, and the authors' investigations into their physiologic and pathophysiologic activities have made significant contributions to the literature. Therefore, the pathophysiologic role of orexins, neuromedins, RFamides, corticotrope-releasing hormone family, growth hormone-releasing hormone, gonadotropin-releasing hormone, ghrelin, apelin, and natriuretic peptides are discussed in detail. Finally, the therapeutic potential of neuropeptide antagonists and agonists in the inhibition of disease progression is discussed here.
Collapse
Affiliation(s)
- Miklós Jászberényi
- Department of Pathophysiology, University of Szeged, P.O. Box 427, H-6701 Szeged, Hungary
| | - Balázs Thurzó
- Department of Pathophysiology, University of Szeged, P.O. Box 427, H-6701 Szeged, Hungary
- Emergency Patient Care Unit, Albert Szent-Györgyi Health Centre, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
| | - Arumugam R Jayakumar
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Andrew V Schally
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
29
|
Zhou Y, Meng Z, Han Y, Yang X, Kuai J, Bao H. The effects of apelin-13 in a mouse model of post-traumatic stress disorder. Neuroreport 2024; 35:1098-1106. [PMID: 39423326 DOI: 10.1097/wnr.0000000000002104] [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: 10/21/2024]
Abstract
The objective is to investigate the effects of apelin-13 in models of post-traumatic stress disorder (PTSD). Mature male CD1 mice were subjected to the single prolonged stress method to induce PTSD-related behaviors. These behaviors were then evaluated using the elevated plus maze test, Morris water maze test, and open field test. Hippocampal neural cell death was assessed using propidium iodide labeling. The expression of hippocampal autophagy pathway-associated proteins was determined through immunoblotting analysis, and LC3 levels were also measured via quantitative real-time reverse transcription-PCR. The results demonstrate that administration of apelin-13 suppressed PTSD-induced hippocampal neural cell death and alleviated PTSD-related behaviors in mice. Additionally, PTSD led to an up-regulation of LC3 and FoxO3a, and down-regulation of P62, p-PI3K, p-Akt, and p-FoxO3a in the hippocampus. However, these changes were reversed by apelin-13 treatment. These findings support the hypothesis that apelin-13 prevents the development of PTSD-like behavior and inhibits autophagy of neuronal cells in a mouse model of PTSD. Apelin-13 may hold potential as a therapeutic agent for PTSD in clinical applications.
Collapse
Affiliation(s)
- Yang Zhou
- Department of Forensic Medicine, Xuzhou Medical University, Xuzhou
| | - Zijun Meng
- Department of Forensic Medicine, Xuzhou Medical University, Xuzhou
| | - Yuqing Han
- Department of Forensic Medicine, Xuzhou Medical University, Xuzhou
| | - Xiaofang Yang
- Department of Histology and Embryology, Fenyang College, Shanxi Medical University, Fenyang
| | - Jinxia Kuai
- Department of Science and Technology, Public Experimental Research Center, Xuzhou Medical University, Xuzhou, China
| | - Haijun Bao
- Department of Forensic Medicine, Xuzhou Medical University, Xuzhou
| |
Collapse
|
30
|
Murugan R, Nayak SPRR, Haridevamuthu B, Priya D, Rajagopal R, Pasupuleti M, Guru A, Kumaradoss KM, Arockiaraj J. Multifaceted evaluation of pyrazole derivative (T4)-chitosan (CS) nanoparticles: Morphology, drug release, and anti-tumor efficacy in a rat model. Int J Biol Macromol 2024; 283:137702. [PMID: 39549794 DOI: 10.1016/j.ijbiomac.2024.137702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 11/08/2024] [Accepted: 11/13/2024] [Indexed: 11/18/2024]
Abstract
The development of targeted nanotherapeutics has emerged as a pivotal advancement in cancer treatment, aiming to enhance the efficacy and specificity of drug delivery while minimizing systemic toxicity. Due to their biocompatibility and modifiable surface properties, Chitosan-based nanoparticles have shown considerable promise in encapsulating and delivering therapeutic agents directly to tumor sites. This study investigates the potential of 1,5-diary pyrazole derivative (T4)-loaded chitosan (CS) nanoparticles as a novel anticancer agent, evaluating their physical characteristics, in vivo biodistribution, and therapeutic efficacy against cancerous cells. SEM morphological analysis confirmed chitosan-based nanoparticles' smooth, spherical structure, with aggregation patterns typical of high surface energy nanoparticle synthesis. UV-visible spectroscopy and XRD analysis validated the successful incorporation of T4, showing characteristic absorption peaks and indicating a reduction in crystallinity desirable for enhanced drug release. In vivo imaging demonstrated the rapid systemic distribution of T4-CS nanoparticles, essential for delivering therapeutic agents effectively. The cytotoxic potential of T4-CS nanoparticles was significantly higher against cancer cells compared to controls, confirmed by MTT and scratch assays, indicating enhanced anti-cancer activity and potential inhibition of cancer metastasis. Furthermore, histological and gene expression analyses supported the anti-tumor and pro-apoptotic capabilities of T4-CS nanoparticles, showing reduced proliferation markers and inflammatory pathways. Behavioral assessments in rats highlighted the neuroprotective effects of T4-CS nanoparticles against 7,12-dimethyl benzanthracene (DMBA) induced neurotoxicity, suggesting their utility as both anticancer and neuroprotective agents. This multifaceted evaluation underscores the versatility and therapeutic potential of T4-CS nanoparticles, warranting further investigation into their mechanistic effects and clinical applications.
Collapse
Affiliation(s)
- Raghul Murugan
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai 600077, Tamil Nadu, India
| | - S P Ramya Ranjan Nayak
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India
| | - B Haridevamuthu
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India
| | - D Priya
- Dr APJ Abdul Kalam Research Lab, Department of Pharmaceutical Chemistry, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India
| | - Rajakrishnan Rajagopal
- Department of Botany & Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mukesh Pasupuleti
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute (CDRI), Sitapur Road, Sector 10, Janakipuram Extension, Lucknow 226031, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Ajay Guru
- Department of Cariology, Saveetha Dental College and Hospitals, SIMATS, Chennai 600 077, Tamil Nadu, India.
| | - Kathiravan Muthu Kumaradoss
- Dr APJ Abdul Kalam Research Lab, Department of Pharmaceutical Chemistry, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India.
| | - Jesu Arockiaraj
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India.
| |
Collapse
|
31
|
Sepúlveda P, Ferreira AFF, Sandoval C, Bergoc G, Moreno ACR, Nunes MT, Torrão ADS. Thyroid Hormone Supplementation Restores Cognitive Deficit, Insulin Signaling, and Neuroinflammation in the Hippocampus of a Sporadic Alzheimer's-like Disease Rat Model. Cells 2024; 13:1793. [PMID: 39513900 PMCID: PMC11545223 DOI: 10.3390/cells13211793] [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: 05/27/2024] [Revised: 07/03/2024] [Accepted: 07/16/2024] [Indexed: 11/16/2024] Open
Abstract
Thyroid hormones play a crucial role in the development of the central nervous system and are considered pivotal to cognitive functions in the adult brain. Recently, thyroid dysfunction has been associated with Alzheimer's disease. The aim of this study was to assess the neuroprotective effects of triiodothyronine (T3) on insulin signaling, neuroinflammation, apoptosis, and cognitive function in a streptozotocin (STZ)-induced sporadic Alzheimer's disease-like model. Male Wistar rats underwent stereotaxic surgery for intracerebroventricular injections of streptozotocin (STZ; 2 mg/kg) or vehicle in the lateral ventricles to induce an AD-like model. The animals received a daily dose of 1.5 μg of T3/100 g body weight or the same volume of vehicle for 30 days and were subdivided into four experimental groups: (1) animals receiving citrate treated with saline (Control = CTL); (2) animals receiving citrate treated with T3 (T3); (3) animals receiving STZ treated with saline (STZ); and (4) animals receiving STZ treated with T3 (STZ + T3). The novel object recognition test was used to measure cognitive function. Serum analysis, real-time RT-PCR, immunohistochemistry, and immunoblotting analyses were also carried out. Our results demonstrated that T3 treatment reversed cognitive impairment and increased Akt and GSK3 phosphorylation in the treated group, while also reducing microglial activation (Iba-1) and GFAP expression (reactive astrocytes), along with TNF-α, IL-6, and IL-1β levels in the hippocampus. Additionally, T3 treatment increased levels of the anti-apoptotic protein Bcl-2 and reduced the expression of the pro-apoptotic protein BAX in the hippocampus. Our study demonstrated that T3 could potentially protect neurons in an AD model induced by STZ.
Collapse
Affiliation(s)
- Paulina Sepúlveda
- Departamento de Ciencias Preclínicas, Facultad de Medicina, Universidad de La Frontera, Temuco 4811230, Chile
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil; (A.F.F.F.); (G.B.); (A.C.R.M.); (M.T.N.)
| | - Ana Flavia Fernandes Ferreira
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil; (A.F.F.F.); (G.B.); (A.C.R.M.); (M.T.N.)
| | - Cristian Sandoval
- Escuela de Tecnología Médica, Facultad de Salud, Universidad Santo Tomás, Los Carreras 753, Osorno 5310431, Chile;
- Departamento de Medicina Interna, Facultad de Medicina, Universidad de La Frontera, Temuco 4811230, Chile
| | - Giovanna Bergoc
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil; (A.F.F.F.); (G.B.); (A.C.R.M.); (M.T.N.)
| | - Ana Caroline Rippi Moreno
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil; (A.F.F.F.); (G.B.); (A.C.R.M.); (M.T.N.)
| | - Maria Tereza Nunes
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil; (A.F.F.F.); (G.B.); (A.C.R.M.); (M.T.N.)
| | - Andréa da Silva Torrão
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil; (A.F.F.F.); (G.B.); (A.C.R.M.); (M.T.N.)
| |
Collapse
|
32
|
Li M, Chen Z, Deng S, Wang L, Yu X. MOSDNET: A multi-omics classification framework using simplified multi-view deep discriminant representation learning and dynamic edge GCN with multi-task learning. Comput Biol Med 2024; 181:109040. [PMID: 39168014 DOI: 10.1016/j.compbiomed.2024.109040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 08/05/2024] [Accepted: 08/15/2024] [Indexed: 08/23/2024]
Abstract
The integration of multi-omics data offers a robust approach to understanding the complexity of diseases by combining information from various biological levels, such as genomics, transcriptomics, proteomics, and metabolomics. This integrated approach is essential for a comprehensive understanding of disease mechanisms and for developing more effective diagnostic and therapeutic strategies. Nevertheless, most current methodologies fail to effectively extract both shared and specific representations from omics data. This study introduces MOSDNET, a multi-omics classification framework that effectively extracts shared and specific representations. This framework leverages Simplified Multi-view Deep Discriminant Representation Learning (S-MDDR) and Dynamic Edge GCN (DEGCN) to enhance the accuracy and efficiency of disease classification. Initially, MOSDNET utilizes S-MDDR to establish similarity and orthogonal constraints for extracting these representations, which are then concatenated to integrate the multi-omics data. Subsequently, MOSDNET constructs a comprehensive view of the sample data by employing patient similarity networks. By incorporating similarity networks into DEGCN, MOSDNET learns intricate network structures and node representations, which enables superior classification outcomes. MOSDNET is trained through a multitask learning approach, effectively leveraging the complementary knowledge from both the data integration and classification components. After conducting extensive comparative experiments, we have conclusively demonstrated that MOSDNET outperforms leading state-of-the-art multi-omics classification models in terms of classification accuracy. Simultaneously, we employ MOSDNET to identify pivotal biomarkers within the multi-omics data, providing insights into disease etiology and progression.
Collapse
Affiliation(s)
- Min Li
- School of Information Engineering, Nanchang Institute of Technology, No. 289 Tianxiang Road, Nanchang, Jiangxi, PR China.
| | - Zihao Chen
- School of Information Engineering, Nanchang Institute of Technology, No. 289 Tianxiang Road, Nanchang, Jiangxi, PR China
| | - Shaobo Deng
- School of Information Engineering, Nanchang Institute of Technology, No. 289 Tianxiang Road, Nanchang, Jiangxi, PR China
| | - Lei Wang
- School of Information Engineering, Nanchang Institute of Technology, No. 289 Tianxiang Road, Nanchang, Jiangxi, PR China
| | - Xiang Yu
- School of Information Engineering, Nanchang Institute of Technology, No. 289 Tianxiang Road, Nanchang, Jiangxi, PR China
| |
Collapse
|
33
|
Yan YJ, Huang CQ. Cognitive impairment induced by circadian rhythm disorders involves hippocampal brain-derived neurotrophic factor reduction and amyloid-β deposition. Chronobiol Int 2024; 41:1299-1306. [PMID: 39311588 DOI: 10.1080/07420528.2024.2406545] [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: 01/25/2024] [Revised: 08/14/2024] [Accepted: 09/13/2024] [Indexed: 10/30/2024]
Abstract
Circadian rhythm disruptions have been implicated in numerous health issues, including cognitive decline and the exacerbation of neurodegenerative diseases, like Alzheimer disease (AD). Brain-derived neurotrophic factor (BDNF), vital for neuronal plasticity and cognitive function, is regulated by the circadian clock and exerts protective effects against AD. Thus, we investigated the impact of circadian rhythm disorders (CRDs) on cognitive impairment and explored the underlying neurobiological mechanisms by assessing BDNF and amyloid-β (Aβ) levels. We divided male C57BL/6 mice into three groups (n = 30): a control group (normal 12/12 hour light-dark cycle) and two CRD model groups (3/3 and 22/22 hour cycles, respectively). After 12 weeks, we assessed cognitive functions using the Morris water maze. Following behavioral tests, hippocampal levels of BDNF and Aβ were quantified using enzyme-linked immunosorbent assays. CRDs significantly impaired learning and memory, as evidenced by longer times to reach and find the platform in the CRD groups (p < 0.01). Furthermore, BDNF levels were notably decreased and Aβ levels increased in the CRD groups compared with the control group (p < 0.01). Thus, CRDs elicit cognitive impairment by reducing BDNF levels and increasing Aβ deposition in the hippocampus.
Collapse
Affiliation(s)
- Yue-Jia Yan
- Department of Geriatrics, The Third Hospital of Mianyang (Sichuan Mental Health Center), Mianyang, Sichuan, China
| | - Chang-Quan Huang
- Department of Geriatrics, The Third Hospital of Mianyang (Sichuan Mental Health Center), Mianyang, Sichuan, China
- Department of General Medicine, Chengdu Second People's Hospital, Chengdu, Sichuan, China
| |
Collapse
|
34
|
Wang Y, Chen J, Wang C, Chen T, He L. GW9508 ameliorates cognitive dysfunction via autophagy pathway in streptozotocin-induced mouse model of Alzheimer's disease. Fundam Clin Pharmacol 2024; 38:906-923. [PMID: 38486405 DOI: 10.1111/fcp.13002] [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: 02/22/2023] [Revised: 12/18/2023] [Accepted: 02/19/2024] [Indexed: 11/21/2024]
Abstract
BACKGROUND G protein-coupled receptor 40 (GPR40) is a potential drug target for Alzheimer's disease (AD), and its agonist GW9508 ameliorates cognitive impairment by intravenous administration. OBJECTIVES The present study was conducted to investigate the efficacy of GW9508 administered peripherally on cognitive dysfunction in streptozotocin (STZ)-induced AD mice. METHODS Seventy male ICR mice were randomly divided into seven groups: vehicle sham group, model, Donepezil, GW9508-L, GW9508-M, GW9508-H, and GW1100 + GW9508-H groups, and administered either vehicle (artificial cerebrospinal fluid [aCSF]) or STZ (3 mg/kg in the vehicle) once a day (9:00 a.m.) by intracerebroventricular injection bilaterally on day 1 and day 3, respectively. After 2 weeks of recovery, all mice were given drug treatment. Behavioral experiments were applied to test the recognition and spatial memory of mice, while molecular biology experiments such as Western blot, ELISA, and Nissl staining were used to detect the corresponding changes of signaling pathways. RESULTS Intraperitoneal administration of GW9508 prevented STZ-induced cognitive impairment as well as decreased the level of p-tau and Aβ1-42 in plasma and brain. GW9508 upregulated the expression of gut-brain peptides like PYY, CCK, IGF-1, and GLP-1 both in blood circulation and brain and downregulated the expression level of autophagy-related proteins through activating Akt/mTOR signaling pathway. Meanwhile, the treatment effect of GW9508 was reversed by GPR40 antagonist GW1100 significantly. CONCLUSION Peripheral administration of GW9508 exhibits neuroprotective effects, and it could be a promising therapy for AD. The neuroprotective mechanism of GW9508 was based on promoting gut-brain peptide secretion, activating Akt/mTOR signal pathway, and regulating neuronal autophagy.
Collapse
Affiliation(s)
- Yanan Wang
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Jingjing Chen
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Chen Wang
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Tong Chen
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Ling He
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
35
|
Mitchell AK, Bliss RR, Church FC. Exercise, Neuroprotective Exerkines, and Parkinson's Disease: A Narrative Review. Biomolecules 2024; 14:1241. [PMID: 39456173 PMCID: PMC11506540 DOI: 10.3390/biom14101241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/23/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024] Open
Abstract
Parkinson's disease (PD) is a prevalent neurodegenerative disease in which treatment often includes an exercise regimen. Exercise is neuroprotective in animal models of PD, and, more recently, human clinical studies have verified exercise's disease-modifying effect. Aerobic exercise and resistance training improve many of PD's motor and non-motor symptoms, while neuromotor therapy and stretching/flexibility exercises positively contribute to the quality of life in people with PD. Therefore, understanding the role of exercise in managing this complex disorder is crucial. Exerkines are bioactive substances that are synthesized and released during exercise and have been implicated in several positive health outcomes, including neuroprotection. Exerkines protect neuronal cells in vitro and rodent PD models in vivo. Aerobic exercise and resistance training both increase exerkine levels in the blood, suggesting a role for exerkines in the neuroprotective theory. Many exerkines demonstrate the potential for protecting the brain against pathological missteps caused by PD. Every person (people) with Parkinson's (PwP) needs a comprehensive exercise plan tailored to their unique needs and abilities. Here, we provide an exercise template to help PwP understand the importance of exercise for treating PD, describe barriers confronting many PwP in their attempt to exercise, provide suggestions for overcoming these barriers, and explore the role of exerkines in managing PD. In conclusion, exercise and exerkines together create a powerful neuroprotective system that should contribute to slowing the chronic progression of PD.
Collapse
Affiliation(s)
- Alexandra K. Mitchell
- Department of Health Sciences, Division of Physical Therapy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | | | - Frank C. Church
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
36
|
Cao Q, Zeng W, Nie J, Ye Y, Chen Y. The protective effects of apelin-13 in HIV-1 tat- induced macrophage infiltration and BBB impairment. Tissue Barriers 2024:2392361. [PMID: 39264117 DOI: 10.1080/21688370.2024.2392361] [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: 06/04/2024] [Revised: 08/05/2024] [Accepted: 08/11/2024] [Indexed: 09/13/2024] Open
Abstract
Impairment of the blood - brain barrier (BBB) and subsequent inflammatory responses contribute to the development of human immunodeficiency virus (HIV)-1-associated neurocognitive disorders (HAND). Apelin-13, the most abundant member of the apelin family, acts as the ligand of the angiotensin receptor-like 1 (APJ). However, its pharmacological function in HAND and its underlying mechanism are unknown. In the current study, we report that the presence of HIV-1 Tat reduced the levels of Apelin-13 and APJ in the cortex tissue of mice. Importantly, Apelin-13 preserved BBB integrity against HIV-1 Tat in mice by increasing the expression of the tight junction protein zonula occludens-1 (ZO-1) and occludin. Interestingly, increased macrophage infiltration, indicated by elevated CD68-positive staining was observed in the cortex after stimulation with HIV-1, which was mitigated by the administration of Apelin-13. Correspondingly, Apelin-13 reduced the expression of monocyte chemoattractant protein-1; (MCP-1). An in vitro two-chamber and two-cell trans-well assay demonstrated that HIV-1 Tat challenge significantly promoted macrophage migration, which was notably attenuated by the introduction of Apelin-13. Accordingly, treatment with Apelin-13 restored the HIV-1 Tat-induced reduction of occludin and ZO-1, while preventing the upregulation of MCP-1 in human brain microvascular endothelial cells (HBMVECs). Our results suggest that Apelin-13 may reduce macrophage infiltration into brain tissues and mitigate BBB dysfunction in patients with HAND.
Collapse
Affiliation(s)
- Qi Cao
- First Department of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
| | - Wei Zeng
- Department of Emergency, Chongqing Public Health Medical Center, Chongqing, China
| | - Jingmin Nie
- First Department of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
| | - Yongjun Ye
- Department of General Surgery, Chongqing Public Health Medical Center, Chongqing, China
| | - Yanchao Chen
- Department of General Internal Medicine, Chongqing Public Health Medical Center, Chongqing, China
| |
Collapse
|
37
|
Liu X, Ding Y, Jiang C, Xin Y, Ma X, Xu M, Wang Q, Hou B, Li Y, Zhang S, Shao B. Astragaloside IV mediates radiation-induced neuronal damage through activation of BDNF-TrkB signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155803. [PMID: 38876008 DOI: 10.1016/j.phymed.2024.155803] [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: 03/21/2024] [Revised: 05/06/2024] [Accepted: 06/02/2024] [Indexed: 06/16/2024]
Abstract
BACKGROUND Electromagnetic radiation is relevant to human life, and radiation can trigger neurodegenerative diseases by altering the function of the central nervous system through oxidative stress, mitochondrial dysfunction, and protein degradation. Astragaloside IV (AS-IV) is anti-oxidative, anti-apoptotic, activates the BDNF-TrkB pathway and enhances synaptic plasticity in radiated mice, which can exert its neuroprotection. However, the exact molecular mechanisms are still unclear. PURPOSE This study investigated whether AS-IV could play a neuroprotective role by regulating BDNF-TrkB pathway in radiation damage and its underlying molecular mechanisms. METHODS Transgenic mice (Thy1-YFP line H) were injected with AS-IV (40 mg/kg/day body weight) by intraperitoneal injection daily for 4 weeks, followed by X-rays. PC12 cells and primary cortical neurons were also exposed to UVA after 24 h of AS-IV treatment (25 μg/ml and 50 μg/ml) in vitro. The impact of radiation on learning and cognitive functions was visualized in the Morris water maze assay. Subsequently, Immunofluorescence and Golgi-Cox staining analyses were utilized to investigate the structural damage of neuronal dendrites and the density of dendritic spines. Transmission electron microscopy was performed to examine how the radiation affected the ultrastructure of neurons. Finally, western blotting analysis and Quantitative RT-PCR were used to evaluate the expression levels and locations of proteins in vitro and in vivo. RESULTS Radiation induced BDNF-TrkB signaling dysregulation and decreased the levels of neuron-related functional genes (Ngf, Bdnf, Gap-43, Ras, Psd-95, Arc, Creb, c-Fos), PSD-95 and F-actin, which subsequently led to damage of neuronal ultrastructure and dendrites, loss of dendritic spines, and decreased dendritic complexity index, contributing to spatial learning and memory deficits. These abnormalities were prevented by AS-IV treatment. In addition, TrkB receptor antagonists antagonized these neuroprotective actions of AS-IV. 7,8-dihydroxyflavone and AS-IV had neuroprotective effects after radiation. CONCLUSION AS-IV inhibits morphological damage of neurons and cognitive dysfunction in mice after radiation exposure, resulting in a neuroprotective effect, which were mediated by activating the BDNF-TrkB pathway.
Collapse
Affiliation(s)
- Xin Liu
- School of Life Sciences, Lanzhou University, Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou 730000, Gansu Province, PR China
| | - Yanping Ding
- School of Life Sciences, Northwest Normal University, Lanzhou 730070, Gansu Province, PR China
| | - Chenxin Jiang
- School of Life Sciences, Lanzhou University, Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou 730000, Gansu Province, PR China
| | - Yuanyuan Xin
- School of Life Sciences, Lanzhou University, Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou 730000, Gansu Province, PR China
| | - Xin Ma
- School of Life Sciences, Lanzhou University, Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou 730000, Gansu Province, PR China
| | - Min Xu
- School of Life Sciences, Lanzhou University, Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou 730000, Gansu Province, PR China
| | - Qianhao Wang
- School of Life Sciences, Lanzhou University, Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou 730000, Gansu Province, PR China
| | - Boru Hou
- Department of Neurosurgery, Second Hospital of Lanzhou University, Lanzhou 730030, Gansu Province, PR China
| | - Yingdong Li
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, PR China
| | - Shengxiang Zhang
- School of Life Sciences, Lanzhou University, Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou 730000, Gansu Province, PR China
| | - Baoping Shao
- School of Life Sciences, Lanzhou University, Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou 730000, Gansu Province, PR China.
| |
Collapse
|
38
|
Tomruk C, Şirin Tomruk C, Denizlioğlu B, Olukman M, Ercan G, Duman S, Köse T, Çetin Uyanıkgil EÖ, Uyanıkgil Y, Uysal A. Effects of apelin on neonatal brain neurogenesis in L-NAME-induced maternal preeclampsia. Sci Rep 2024; 14:19347. [PMID: 39164321 PMCID: PMC11335761 DOI: 10.1038/s41598-024-69326-2] [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: 02/18/2024] [Accepted: 08/02/2024] [Indexed: 08/22/2024] Open
Abstract
The aim of this study was to investigate the possible protective effects of apelin, which is known to have antioxidant and anti-inflammatory effects, on changes in neurogenesis in newborns of pregnant rats with L-NAME-induced preeclampsia. Wistar albino female rats were divided into four experimental groups: Control, Apelin, Preeclampsia and Preeclampsia + Apelin. Blood pressure was measured on the 5th, 11th and 17th days of gestation, urine protein was analyzed from urine samples collected for 24 h on the 6th, 12th and 18th days and serum creatinine was analyzed from serum samples. Maternal kidney and placenta tissues were obtained to establish the preeclampsia model, and neonatal brain tissues including the cortex, hippocampus and cerebellum regions were obtained to investigate neurogenesis and examined by histological and immunohistochemical methods. The number of newborns, body weight and brain weight of the newborns were measured. eNOS, IL-10, nNOS and NO levels in the brain analyzed via ELISA. Mean arterial pressure, urine protein and serum creatinine increased in the preeclampsia. Newborn weight decreased in the Preeclampsia group, the values in the Preeclampsia + Apelin group were closer to the Control and Apelin groups. In the Preeclampsia group, edema and dilatation in the proximal and distal tubules of kidneys, perivillous fibrin deposition and increase in syncytial nodules of placenta were observed. VEGF immunoreactivity decreased and iNOS immunoreactivity increased in both kidney and placenta. In neonatal brain tissue examinations, cytotoxic edema accompanied by thinning of cortex, delayed migration and lower cell counts in the hippocampus, and increase in intercellular spaces and EGL thickening in the cerebellum were observed in the preeclampsia. Expression of NeuN, GFAP, MBP, IL-10, eNOS, nNOS and NO levels decreased, whereas expression of Iba-1 increased in the preeclampsia. In the Preeclampsia + Apelin group, these findings were similar to the Control and Apelin groups. Apelin administration was found to be beneficial for preventing the adverse consequences of preeclampsia, but further experimental and clinical studies are needed to better understand these effects.
Collapse
Affiliation(s)
- Canberk Tomruk
- Department of Histology and Embryology, Faculty of Medicine, Ege University, Bornova, İzmir, Türkiye
- Histology and Embryology, Samsun Training and Research Hospital, İlkadim, Samsun, Türkiye
| | - Cansın Şirin Tomruk
- Department of Histology and Embryology, Faculty of Medicine, Ege University, Bornova, İzmir, Türkiye
| | - Burcu Denizlioğlu
- Department of Medical Biochemistry, Faculty of Medicine, Ege University, Bornova, İzmir, Türkiye
- Emergency Medicine, Aydın State Hospital, Efeler, Aydın, Türkiye
| | - Murat Olukman
- Department of Medical Pharmacology, Faculty of Medicine, Ege University, Bornova, İzmir, Türkiye
| | - Gülinnaz Ercan
- Department of Medical Biochemistry, Faculty of Medicine, Ege University, Bornova, İzmir, Türkiye
| | - Soner Duman
- Department of Internal Medicine, Faculty of Medicine, Ege University, Bornova, İzmir, Türkiye
| | - Timur Köse
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Ege University, Bornova, İzmir, Türkiye
| | - Emel Öykü Çetin Uyanıkgil
- Department of Pharmaceutical Technology, Department of Biopharmaceutics and Pharmacokinetics, Faculty of Pharmacy, Ege University, Bornova, İzmir, Türkiye
| | - Yiğit Uyanıkgil
- Department of Histology and Embryology, Faculty of Medicine, Ege University, Bornova, İzmir, Türkiye.
| | - Ayşegül Uysal
- Department of Histology and Embryology, Faculty of Medicine, Ege University, Bornova, İzmir, Türkiye
| |
Collapse
|
39
|
Baghaee P, Yoonesi M, Esfahani DE, Beirami E, Dargahi L, Rashidi FS, Valian N. Yttrium oxide nanoparticles alleviate cognitive deficits, neuroinflammation, and mitochondrial biogenesis impairment induced by streptozotocin. Neurosci Lett 2024; 837:137895. [PMID: 39025434 DOI: 10.1016/j.neulet.2024.137895] [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: 11/10/2023] [Revised: 06/17/2024] [Accepted: 07/09/2024] [Indexed: 07/20/2024]
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disorder characterized by progressive cognitive decline. Yttrium oxide nanoparticles (Y2O3NPs) have recently attracted much attention for their potential anti-inflammatory and antioxidant properties. However, the effects of Y2O3NPs in animal models of AD are less studied. This study aimed to investigate the potential therapeutic effects of Y2O3NPs in streptozotocin (STZ)-treated rats, a reliable animal model of AD, with special emphasis on cognitive function, neuroinflammation, and mitochondrial biogenesis in the hippocampus. Male Wistar rats were stereotaxically injected with STZ (3 mg/kg, 3 µl/ventricle). Three weeks after STZ injection, cognitive function was assessed using the Morris water maze, elevated plus maze, and passive avoidance tasks. Intraperitoneal treatment with Y2O3NPs (0.1, 0.3, or 0.5 mg/kg) was started 24 h after the STZ injection and continued for 21 days. The mRNA and protein levels of pro-inflammatory cytokines (TNF-α, IL-6, and IL-1β) and components involved in mitochondrial biogenesis (PGC-1α, NRF-1, and TFAM) were measured in the hippocampus. The results indicated that STZ induced cognitive impairment and led to neuroinflammation and mitochondrial biogenesis impairment in the hippocampus of rats. Interestingly, treatment with Y2O3NPs effectively reduced STZ-induced cognitive deficits in a dose-dependent manner, possibly by attenuating neuroinflammation and mitochondrial biogenesis impairment. These findings suggest that Y2O3NPs can be considered as a promising therapeutic agent for treating or ameliorating the neuropathological effects associated with AD.
Collapse
Affiliation(s)
- Pooya Baghaee
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Mohammad Yoonesi
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Delaram Eslimi Esfahani
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran.
| | - Elmira Beirami
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran.
| | - Leila Dargahi
- NeuroBiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Sadat Rashidi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Neda Valian
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
40
|
Weijie Z, Meng Z, Chunxiao W, Lingjie M, Anguo Z, Yan Z, Xinran C, Yanjiao X, Li S. Obesity-induced chronic low-grade inflammation in adipose tissue: A pathway to Alzheimer's disease. Ageing Res Rev 2024; 99:102402. [PMID: 38977081 DOI: 10.1016/j.arr.2024.102402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/19/2024] [Accepted: 06/30/2024] [Indexed: 07/10/2024]
Abstract
Alzheimer's disease (AD) is a leading cause of cognitive impairment worldwide. Overweight and obesity are strongly associated with comorbidities, such as hypertension, diabetes, and insulin resistance (IR), which contribute substantially to the development of AD and subsequent morbidity and mortality. Adipose tissue (AT) is a highly dynamic organ composed of a diverse array of cell types, which can be classified based on their anatomic localization or cellular composition. The expansion and remodeling of AT in the context of obesity involves immunometabolic and functional shifts steered by the intertwined actions of multiple immune cells and cytokine signaling within AT, which contribute to the development of metabolic disorders, IR, and systemic markers of chronic low-grade inflammation. Chronic low-grade inflammation, a prolonged, low-dose stimulation by specific immunogens that can progress from localized sites and affect multiple organs throughout the body, leads to neurodystrophy, increased apoptosis, and disruption of homeostasis, manifesting as brain atrophy and AD-related pathology. In this review, we sought to elucidate the mechanisms by which AT contributes to the onset and progression of AD in obesity through the mediation of chronic low-grade inflammation, particularly focusing on the roles of adipokines and AT-resident immune cells.
Collapse
Affiliation(s)
- Zhai Weijie
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China; Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Zhao Meng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China; Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Wei Chunxiao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China; Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Meng Lingjie
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China; Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Zhao Anguo
- Department of Urology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou 215000 China
| | - Zhang Yan
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China; Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Cui Xinran
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China; Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Xu Yanjiao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China; Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Sun Li
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China; Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China.
| |
Collapse
|
41
|
Birmpili D, Charmarké-Askar I, Spenlé C, Riché S, Pham-Van LD, Kuntzel T, Xhurxhi T, Riou A, Bonnet D, Bagnard D. Fluorinated apelin-13 mediates neuroprotective effects in multiple sclerosis models. Neurobiol Dis 2024; 198:106552. [PMID: 38844244 DOI: 10.1016/j.nbd.2024.106552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 05/31/2024] [Accepted: 06/01/2024] [Indexed: 06/11/2024] Open
Abstract
Multiple sclerosis (MS) is an autoimmune and neurodegenerative disease leading to demyelination and axonal loss. Current treatments are immunomodulatory or immunosuppressive drugs acting on the inflammatory component. However, these treatments do not adequately address the crucial aspect of neuroprotection. Recently, an association between an altered balance of adipokines and MS has been proposed as both a risk factor for developing MS and a chronic disease aggravating factor. Specifically, a decrease of apelin plasma levels in MS patients compared to controls correlates with the number of relapses and disease severity. Here we report a dramatic downregulation of apelin levels in the CNS of EAE mice which is also detected in MS patients brain samples compared to controls. Exploiting innovative design and synthesis techniques, we engineered a novel fluorinated apelin-13 peptide characterized by enhanced plasmatic stability compared to its native counterpart. With this peptide, we assessed the potential therapeutic benefits of apelin preventive supplementation in the EAE mouse model. We show that the fluorinated Apelin-13 peptide ameliorates EAE clinical score and preserves myelin content in the EAE MOG model recapitulating the progressive form of disease. These results combined with ex-vivo experiments in brain organotypic slices and in vitro studies in neurons and primary microglia and macrophages suggest that apelin has neuroprotective effects and influences the microglia/macrophages function.
Collapse
Affiliation(s)
- Dafni Birmpili
- Centre national de la Recherche Scientifique (CNRS) UMRS7242, Biotechnology and Cell Signaling, Therapeutic Peptides Team, Institut du Médicament de Strasbourg (IMS), ESBS 300 Boulevard S. Brant, 67400 Illkirch-Graffenstaden, France
| | - Imane Charmarké-Askar
- Centre national de la Recherche Scientifique (CNRS) UMRS7242, Biotechnology and Cell Signaling, Therapeutic Peptides Team, Institut du Médicament de Strasbourg (IMS), ESBS 300 Boulevard S. Brant, 67400 Illkirch-Graffenstaden, France
| | - Caroline Spenlé
- Centre national de la Recherche Scientifique (CNRS) UMRS7242, Biotechnology and Cell Signaling, Therapeutic Peptides Team, Institut du Médicament de Strasbourg (IMS), ESBS 300 Boulevard S. Brant, 67400 Illkirch-Graffenstaden, France
| | - Stéphanie Riché
- Laboratoire d'Innovation Thérapeutique, UMR7200 CNRS/Université de Strasbourg, Institut du Médicament de Strasbourg (IMS), Faculté de Pharmacie, Illkirch, France
| | - Lucas Dinh Pham-Van
- Centre national de la Recherche Scientifique (CNRS) UMRS7242, Biotechnology and Cell Signaling, Therapeutic Peptides Team, Institut du Médicament de Strasbourg (IMS), ESBS 300 Boulevard S. Brant, 67400 Illkirch-Graffenstaden, France
| | - Thomas Kuntzel
- Centre national de la Recherche Scientifique (CNRS) UMRS7242, Biotechnology and Cell Signaling, Therapeutic Peptides Team, Institut du Médicament de Strasbourg (IMS), ESBS 300 Boulevard S. Brant, 67400 Illkirch-Graffenstaden, France
| | - Thanos Xhurxhi
- Centre national de la Recherche Scientifique (CNRS) UMRS7242, Biotechnology and Cell Signaling, Therapeutic Peptides Team, Institut du Médicament de Strasbourg (IMS), ESBS 300 Boulevard S. Brant, 67400 Illkirch-Graffenstaden, France
| | - Aurélien Riou
- Centre national de la Recherche Scientifique (CNRS) UMRS7242, Biotechnology and Cell Signaling, Therapeutic Peptides Team, Institut du Médicament de Strasbourg (IMS), ESBS 300 Boulevard S. Brant, 67400 Illkirch-Graffenstaden, France
| | - Dominique Bonnet
- Laboratoire d'Innovation Thérapeutique, UMR7200 CNRS/Université de Strasbourg, Institut du Médicament de Strasbourg (IMS), Faculté de Pharmacie, Illkirch, France
| | - Dominique Bagnard
- Centre national de la Recherche Scientifique (CNRS) UMRS7242, Biotechnology and Cell Signaling, Therapeutic Peptides Team, Institut du Médicament de Strasbourg (IMS), ESBS 300 Boulevard S. Brant, 67400 Illkirch-Graffenstaden, France.
| |
Collapse
|
42
|
Khamies SM, El-Yamany MF, Ibrahim SM. Canagliflozin Mitigated Cognitive Impairment in Streptozotocin-Induced Sporadic Alzheimer's Disease in Mice: Role of AMPK/SIRT-1 Signaling Pathway in Modulating Neuroinflammation. J Neuroimmune Pharmacol 2024; 19:39. [PMID: 39073453 DOI: 10.1007/s11481-024-10140-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 07/14/2024] [Indexed: 07/30/2024]
Abstract
Sporadic Alzheimer's disease (SAD) represents a major health concern especially among elderly. Noteworthy, neuroinflammation and oxidative stress are highly implicated in AD pathogenesis resulting in enhanced disease progression. Moreover, most of the available anti-Alzheimer drugs have several adverse effects with variable efficacy, therefore new strategies, including agents with anti-inflammatory and antioxidant effects, are encouraged. Along these lines, canagliflozin (CAN), with its anti-inflammatory and anti-apoptotic activities, presents a promising candidate for AD treatment. Therefore, this study aimed to evaluate the therapeutic potential of CAN via regulation of AMPK/SIRT-1/BDNF/GSK-3β signaling pathway in SAD. SAD model was induced by intracerebroventricular streptozotocin injection (ICV-STZ;3 mg/kg, once), while CAN was administered (10 mg/kg/day, orally) to STZ-treated mice for 21 days. Behavioral tests, novel object recognition (NOR), Y-Maze, and Morris Water Maze (MWM) tests, histopathological examination, total adenosine monophosphate-activated protein kinase (T-AMPK) expression, p-AMPK, and silent information regulator-1 (SIRT-1) were evaluated. Furthermore, brain-derived neurotrophic factor (BDNF), glycogen synthase kinase-3β (GSK-3β), acetylcholinesterase (AChE), Tau protein, insulin-degrading enzyme (IDE), nuclear factor erythroid-2 (Nrf-2), interleukin-6 (IL-6), nuclear factor kappa-B-p65 (NFκB-p65), beta-site APP cleaving enzyme 1 (BACE-1), and amyloid beta (Aβ) plaque were assessed. CAN restored STZ-induced cognitive deficits, confirmed by improved behavioral tests and histopathological examination. Besides, CAN halted STZ-induced neurotoxicity through activation of p-AMPK/SIRT-1/BDNF pathway, subsequently reduction of GSK-3β, Tau protein, AChE, NFκB-p65, IL-6, BACE-1, and Aβ plaque associated with increased IDE and Nrf-2. Consequentially, our findings assumed that CAN, via targeting p-AMPK/SIRT-1 pathway, combated neuroinflammation and oxidative stress in STZ-induced AD. Thus, this study highlighted the promising effect of CAN for treating AD.
Collapse
Affiliation(s)
- Sara M Khamies
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, Menoufia University, Menoufia, 32511, Egypt
| | - Mohammed F El-Yamany
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, Cairo University, ElKasr Elaini Street, Cairo, 11562, Egypt
| | - Sherehan M Ibrahim
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, Cairo University, ElKasr Elaini Street, Cairo, 11562, Egypt.
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, Modern University for Technology and Information, Cairo, Egypt.
| |
Collapse
|
43
|
Li T, Li S, Xiong Y, Li X, Ma C, Guan Z, Yang L. Binary Nano-inhalant Formulation of Icariin Enhances Cognitive Function in Vascular Dementia via BDNF/TrkB Signaling and Anti-inflammatory Effects. Neurochem Res 2024; 49:1720-1734. [PMID: 38520637 DOI: 10.1007/s11064-024-04129-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 02/12/2024] [Accepted: 02/14/2024] [Indexed: 03/25/2024]
Abstract
Vascular dementia (VaD) has a serious impact on the patients' quality of life. Icariin (Ica) possesses neuroprotective potential for treating VaD, yet its oral bioavailability and blood-brain barrier (BBB) permeability remain challenges. This research introduced a PEG-PLGA-loaded chitosan hydrogel-based binary formulation tailored for intranasal delivery, enhancing the intracerebral delivery efficacy of neuroprotective agents. The formulation underwent optimization to facilitate BBB crossing, with examinations conducted on its particle size, morphology, drug-loading capacity, in vitro release, and biodistribution. Using the bilateral common carotid artery occlusion (BCCAO) rat model, the therapeutic efficacy of this binary formulation was assessed against chitosan hydrogel and PEG-PLGA nanoparticles loaded with Ica. Post-intranasal administration, enhanced cognitive function was evident in chronic cerebral hypoperfusion (CCH) rats. Further mechanistic evaluations, utilizing immunohistochemistry (IHC), RT-PCR, and ELISA, revealed augmented transcription of synaptic plasticity-associated proteins like SYP and PSD-95, and a marked reduction in hippocampal inflammatory markers such as IL-1β and TNF-α, highlighting the formulation's promise in alleviating cognitive impairment. The brain-derived neurotrophic factor (BDNF)/tropomyosin related kinase B (TrkB) pathway was activated significantly in the binary formulation compared with the other two. Our study demonstrates that the intranasal application of chitosan hydrogel loaded with Ica-encapsulated PEG-PLGA could effectively deliver Ica into the brain and enhance its neuroprotective effect.
Collapse
Affiliation(s)
- Tieshu Li
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, People's Republic of China
| | - Shuling Li
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, People's Republic of China
| | - Yin Xiong
- School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, 130021, People's Republic of China
| | - Xinxin Li
- Affiliated Hospital of Yangzhou University, Yangzhou University, 88 South Daxue Road, Yangzhou, 225009, People's Republic of China
| | - Chun Ma
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, People's Republic of China
| | - Zhiying Guan
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, People's Republic of China
| | - Lihua Yang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, People's Republic of China.
| |
Collapse
|
44
|
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
|
45
|
Kamińska K, Borzuta H, Buczma K, Cudnoch-Jędrzejewska A. Neuroprotective effect of apelin-13 and other apelin forms-a review. Pharmacol Rep 2024; 76:439-451. [PMID: 38568371 DOI: 10.1007/s43440-024-00587-4] [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: 11/20/2023] [Revised: 03/05/2024] [Accepted: 03/05/2024] [Indexed: 05/25/2024]
Abstract
Neurodegenerative diseases, which occur when neurons begin to deteriorate, affect millions of people worldwide. These age-related disorders are becoming more common partly because the elderly population has increased in recent years. While no treatments are accessible, every year an increasing number of therapeutic and supportive options become available. Various substances that may have neuroprotective effects are currently being researched. One of them is apelin. This review aims to illustrate the results of research on the neuroprotective effect of apelin amino acid oligopeptide which binds to the apelin receptor and exhibits neuroprotective effects in the central nervous system. The collected data indicate that apelin can protect the central nervous system against injury by several mechanisms. More studies are needed to thoroughly investigate the potential neuroprotective effects of this peptide in neurodegenerative diseases and various other types of brain damage.
Collapse
Affiliation(s)
- Katarzyna Kamińska
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland.
| | - Hubert Borzuta
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland
| | - Kasper Buczma
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland
| | - Agnieszka Cudnoch-Jędrzejewska
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland
| |
Collapse
|
46
|
Lu H, Chen M, Zhu C. Intranasal Administration of Apelin-13 Ameliorates Cognitive Deficit in Streptozotocin-Induced Alzheimer's Disease Model via Enhancement of Nrf2-HO1 Pathways. Brain Sci 2024; 14:488. [PMID: 38790466 PMCID: PMC11118954 DOI: 10.3390/brainsci14050488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/01/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
BACKGROUND The discovery of novel diagnostic methods and therapies for Alzheimer's disease (AD) faces significant challenges. Previous research has shed light on the neuroprotective properties of Apelin-13 in neurodegenerative disorders. However, elucidating the mechanism underlying its efficacy in combating AD-related nerve injury is imperative. In this study, we aimed to investigate Apelin-13's mechanism of action in an in vivo model of AD induced by streptozocin (STZ). METHODS We utilized an STZ-induced nerve injury model of AD in mice to investigate the effects of Apelin-13 administration. Apelin-13 was administered intranasally, and cognitive impairment was assessed using standardized behavioral tests, primarily, behavioral assessment, histological analysis, and biochemical assays, in order to evaluate synaptic plasticity and oxidative stress signaling pathways. RESULTS Our findings indicate that intranasal administration of Apelin-13 ameliorated cognitive impairment in the STZ-induced AD model. Furthermore, we observed that this effect was potentially mediated by the enhancement of synaptic plasticity and the attenuation of oxidative stress signaling pathways. CONCLUSIONS The results of this study suggest that intranasal administration of Apelin-13 holds promise as a therapeutic strategy for preventing neurodegenerative diseases such as AD. By improving synaptic plasticity and mitigating oxidative stress, Apelin-13 may offer a novel approach to neuroprotection in AD and related conditions.
Collapse
Affiliation(s)
- Hai Lu
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Center for Brain Science, Fudan University, Shanghai 200032, China; (H.L.); (M.C.)
- College of Clinical Medicine, Jining Medical University, Jining 272067, China
| | - Ming Chen
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Center for Brain Science, Fudan University, Shanghai 200032, China; (H.L.); (M.C.)
| | - Cuiqing Zhu
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Center for Brain Science, Fudan University, Shanghai 200032, China; (H.L.); (M.C.)
| |
Collapse
|
47
|
Song J. BDNF Signaling in Vascular Dementia and Its Effects on Cerebrovascular Dysfunction, Synaptic Plasticity, and Cholinergic System Abnormality. J Lipid Atheroscler 2024; 13:122-138. [PMID: 38826183 PMCID: PMC11140249 DOI: 10.12997/jla.2024.13.2.122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/29/2023] [Accepted: 12/19/2023] [Indexed: 06/04/2024] Open
Abstract
Vascular dementia (VaD) is the second most common type of dementia and is characterized by memory impairment, blood-brain barrier disruption, neuronal cell loss, glia activation, impaired synaptic plasticity, and cholinergic system abnormalities. To effectively prevent and treat VaD a good understanding of the mechanisms underlying its neuropathology is needed. Brain-derived neurotrophic factor (BDNF) is an important neurotrophic factor with multiple functions in the systemic circulation and the central nervous system and is known to regulate neuronal cell survival, synaptic formation, glia activation, and cognitive decline. Recent studies indicate that when compared with normal subjects, patients with VaD have low serum BDNF levels and that BDNF deficiency in the serum and cerebrospinal fluid is an important indicator of VaD. Here, we review current knowledge on the role of BDNF signaling in the pathology of VaD, such as cerebrovascular dysfunction, synaptic dysfunction, and cholinergic system impairment.
Collapse
Affiliation(s)
- Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun, Korea
| |
Collapse
|
48
|
Zhang W, Mou M, Hu W, Lu M, Zhang H, Zhang H, Luo Y, Xu H, Tao L, Dai H, Gao J, Zhu F. MOINER: A Novel Multiomics Early Integration Framework for Biomedical Classification and Biomarker Discovery. J Chem Inf Model 2024; 64:2720-2732. [PMID: 38373720 DOI: 10.1021/acs.jcim.4c00013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2024]
Abstract
In the context of precision medicine, multiomics data integration provides a comprehensive understanding of underlying biological processes and is critical for disease diagnosis and biomarker discovery. One commonly used integration method is early integration through concatenation of multiple dimensionally reduced omics matrices due to its simplicity and ease of implementation. However, this approach is seriously limited by information loss and lack of latent feature interaction. Herein, a novel multiomics early integration framework (MOINER) based on information enhancement and image representation learning is thus presented to address the challenges. MOINER employs the self-attention mechanism to capture the intrinsic correlations of omics-features, which make it significantly outperform the existing state-of-the-art methods for multiomics data integration. Moreover, visualizing the attention embedding and identifying potential biomarkers offer interpretable insights into the prediction results. All source codes and model for MOINER are freely available https://github.com/idrblab/MOINER.
Collapse
Affiliation(s)
- Wei Zhang
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| | - Minjie Mou
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Wei Hu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Mingkun Lu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Hanyu Zhang
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Hongning Zhang
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yongchao Luo
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Hongquan Xu
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
| | - Lin Tao
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
| | - Haibin Dai
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jianqing Gao
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| |
Collapse
|
49
|
Kadian M, Saini N, Khera A, Kumar A. Neuroprotective mechanism of trans,trans-Farnesol in an ICV-STZ-induced rat model of Alzheimer's pathology. Inflammopharmacology 2024; 32:1545-1573. [PMID: 38308793 DOI: 10.1007/s10787-023-01413-8] [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: 04/02/2023] [Accepted: 12/13/2023] [Indexed: 02/05/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) is a prominent cause of dementia, resulting in neurodegeneration and memory impairment. This condition imposes a considerable public health burden on both patients and their families due to the patients' functional impairments as well as the psychological and financial constraints. It has been well demonstrated that its aetiology involves proteinopathy, mitochondriopathies, and enhanced reactive oxygen species (ROS) generation, which are some of the key features of AD brains that further result in oxidative stress, excitotoxicity, autophagy, and mitochondrial dysfunction. OBJECTIVE The current investigation was created with the aim of elucidating the neurological defence mechanism of trans,trans-Farnesol (TF) against intracerebroventricular-streptozotocin (ICV-STZ)-induced Alzheimer-like symptoms and related pathologies in rodents. MATERIALS AND METHODS The current investigation involved male SD rats receiving TF (25-100 mg/kg, per oral) consecutively for 21 days in ICV-STZ-treated animals. An in silico study was carried out to explore the possible interaction between TF and NADH dehydrogenase and succinate dehydrogenase. Further, various behavioural (Morris water maze and novel object recognition test), biochemical (oxidants and anti-oxidant markers), activities of mitochondrial enzyme complexes and acetylcholinesterase (AChE), pro-inflammatory (tumor necrosis factor-alpha; TNF-α) levels, and histopathological studies were evaluated in specific brain regions. RESULTS Rats administered ICV-STZ followed by treatment with TF (25, 50, and 100 mg/kg) for 21 days had significantly better mental performance (reduced escape latency to access platform, extended time spent in target quadrant, and improved differential index) in the Morris water maze test and new object recognition test models when compared to control (ICV-STZ)-treated groups. Further, TF treatment significantly restored redox proportion, anti-oxidant levels, regained mitochondrial capacities, attenuated altered AChE action, levels of TNF-α, and histopathological alterations in certain brain regions in comparison with control. In in silico analysis, TF caused greater interaction with NADH dehydrogenase and succinate dehydrogenase. CONCLUSION The current work demonstrates the neuroprotective ability of TF in an experimental model with AD-like pathologies. The study further suggests that the neuroprotective impacts of TF may be related to its effects on TNF-α levels, oxidative stress pathways, and mitochondrial complex capabilities.
Collapse
Affiliation(s)
- Monika Kadian
- Pharmacology Division, University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India
| | - Neetu Saini
- Pharmacology Division, University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India
| | - Alka Khera
- Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Anil Kumar
- Pharmacology Division, University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
50
|
Zhu XQ, Gao D. Naringenin alleviates cognitive dysfunction in rats with cerebral ischemia/reperfusion injury through up-regulating hippocampal BDNF-TrkB signaling: involving suppression in neuroinflammation and oxidative stress. Neuroreport 2024; 35:216-224. [PMID: 38141009 PMCID: PMC10852040 DOI: 10.1097/wnr.0000000000001989] [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: 10/29/2023] [Accepted: 12/03/2023] [Indexed: 12/24/2023]
Abstract
Cognitive dysfunction is one of the common complications of cerebral ischemia-reperfusion (CI/R) injury after ischemic stroke. Neuroinflammation and oxidative stress are the core pathological mechanism of CI/R injury. The activation of brain derived neurotrophic factor (BDNF)-tyrosine receptor kinase B (TrkB) signaling antagonize cognitive dysfunction in a series of neuropathy. Naringenin (NAR) improves cognitive function in many diseases, but the role of NAR in CI/R injury-induced cognitive dysfunction remains unexplored. The study aimed to explore the potential protective effects of NAR in CI/R injury-induced cognitive dysfunction and underlying mechanism. The rats were exposed to transient middle cerebral artery occlusion (MCAO) and then treated with distilled water or NAR (50 or 100 mg/kg/day, p.o.) for 30 days. The Y-maze test, Novel object recognition test and Morris water maze test were performed to assess cognitive function. The levels of oxidative stress and inflammatory cytokines were measured by ELISA. The expressions of BDNF/TrkB signaling were detected by Western blot. NAR prevented cognitive impairment in MCAO-induced CI/R injury rats. Moreover, NAR inhibited oxidative stress (reduced levels of malondialdehyde and 4-hydroxynonenal, increased activities of superoxide dismutase and Glutathione peroxidase) and inflammatory cytokines (reduced levels of tumor necrosis factor-α, Interleukin-1β and Interleukin-6), up-regulated the expressions of BDNF and p-TrkB in hippocampus of MCAO-induced CI/R rats. NAR ameliorated cognitive dysfunction of CI/R rats via inhibiting oxidative stress, reducing inflammatory response, and up-regulating BDNF/TrkB signaling pathways in the hippocampus.
Collapse
Affiliation(s)
- Xiao-Qin Zhu
- Health School of Nuclear Industry, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China
| | - Dong Gao
- The Affiliated Nanhua Hospital, Medical administration division, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|