Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 15.748
Filter
1.
Cell Mol Biol (Noisy-le-grand) ; 70(7): 161-167, 2024 Jul 28.
Article in English | MEDLINE | ID: mdl-39097880

ABSTRACT

Alzheimer's disease (AD) is a degenerative dementia illness that causes atrophy of the temporal and frontal lobes of the cerebral cortex. Linggui Zhugan (LGZG), a classic Chinese herbal formula, was initially recognized as a safe and effective treatment of cardiovascular diseases for long history. This study intended to assess the effects and the molecular mechanism of LGZG on AD progress. C57BL/6 mice were divided into six groups: normal mice, amyloid precursor protein/presenilin 1 (APP/PS1) mice (model group), positive control group (model mice treated with donepezil), high, medium and low LGZG group (model mice treated with 7g/kg/d, 3.5g/kg/d or 1.75g/kg/d LGZG respectively). Water maze results showed that the escape latency and path length of high and medium LGZG groups declined compared to the model mice, the decline degree was dose-dependent. The hippocampal slices of six groups were analyzed by Nissl-staining, Perls' iron staining and immunofluorescence assay. The results indicated LGZG could restore morphological anomalies and alleviate iron deposition of AD mice, and the GXP4 positive cells increased significantly. The MDA, Fe2+ and GSH were measured by biochemical testing, whose results illustrated that LGZG could normalize MDA, Fe2+ and GSH levels in AD model compared to un-treated APP/PS1 model. The higher dose of LGZG the mice received, the more intensive effects on those levels of molecules. Western blot results showed that LGZG could affect NeuN, AMPK, p53, SLC7A11 and GPX4 levels in the hippocampus of AD model, which was all proteins related to AMPK pathway. In conclusion, LGZG has a neuroprotective effect on AD through AMPK pathway by alleviating oxidative stress and ferroptosis.


Subject(s)
Alzheimer Disease , Disease Models, Animal , Drugs, Chinese Herbal , Hippocampus , Mice, Inbred C57BL , Neuroprotective Agents , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Mice , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , AMP-Activated Protein Kinases/metabolism , Signal Transduction/drug effects , Male , Mice, Transgenic , Oxidative Stress/drug effects , Neuroprotection/drug effects , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/genetics , Maze Learning/drug effects
2.
Front Immunol ; 15: 1433590, 2024.
Article in English | MEDLINE | ID: mdl-39139557

ABSTRACT

Salvianolic acid A (SalA), a bioactive compound extracted from Salvia miltiorrhiza, has garnered considerable interest for its potential in ameliorating the post-stroke neuroinflammation. This review delineates the possible molecular underpinnings of anti-inflammatory and neuroprotective roles of SalA, offering a comprehensive analysis of its therapeutic efficacy in preclinical studies of ischemic stroke. We explore the intricate interplay between post-stroke neuroinflammation and the modulatory effects of SalA on pro-inflammatory cytokines, inflammatory signaling pathways, the peripheral immune cell infiltration through blood-brain barrier disruption, and endothelial cell function. The pharmacokinetic profiles of SalA in the context of stroke, characterized by enhanced cerebral penetration post-ischemia, makes it particularly suitable as a therapeutic agent. Preliminary clinical findings have demonstrated that salvianolic acids (SA) has a positive impact on cerebral perfusion and neurological deficits in stroke patients, warranting further investigation. This review emphasizes SalA as a potential anti-inflammatory agent for the advancement of innovative therapeutic approaches in the treatment of ischemic stroke.


Subject(s)
Anti-Inflammatory Agents , Caffeic Acids , Neuroinflammatory Diseases , Stroke , Humans , Animals , Caffeic Acids/therapeutic use , Caffeic Acids/pharmacology , Stroke/drug therapy , Stroke/metabolism , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/etiology , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/pharmacology , Lactates/therapeutic use , Lactates/pharmacology , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects
3.
Neuromolecular Med ; 26(1): 32, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39090268

ABSTRACT

Parkinson's disease is a progressive neurodegenerative disorder marked by the death of dopaminergic neurons in the substantia nigra region of the brain. Aggregation of alpha-synuclein (α-synuclein) is a contributing factor to Parkinson's disease pathogenesis. The objective of this study is to investigate the neuroprotective effects of gut microbes on α-synuclein aggregation using both in silico and in vivo approaches. We focussed on the interaction between α-synuclein and metabolites released by gut bacteria that protect from PD. We employed three probiotic microbe strains against α-synuclein protein: Lactobacillus casei, Escherichia coli, and Bacillus subtilis, with their chosen PDB IDs being Dihydrofolate reductase (3DFR), methionine synthetase (6BM5), and tryptophanyl-tRNA synthetase (3PRH), respectively. Using HEX Dock 6.0 software, we examined the interactions between these proteins. Among the various metabolites, methionine synthetase produced by E. coli showed potential interactions with α-synuclein. To further evaluate the neuroprotective benefits of E. coli, an in vivo investigation was performed using a rotenone-induced Parkinsonian mouse model. The motor function of the animals was assessed through behavioural tests, and oxidative stress and neurotransmitter levels were also examined. The results demonstrated that, compared to the rotenone-induced PD mouse model, the rate of neurodegeneration was considerably reduced in mice treated with E. coli. Additionally, histopathological studies provided evidence of the neuroprotective effects of E. coli. In conclusion, this study lays the groundwork for future research, suggesting that gut bacteria may serve as potential therapeutic agents in the development of medications to treat Parkinson's disease. fig. 1.


Subject(s)
Bacillus subtilis , Escherichia coli , Gastrointestinal Microbiome , Molecular Docking Simulation , Oxidative Stress , Probiotics , Rotenone , alpha-Synuclein , Animals , Mice , Gastrointestinal Microbiome/physiology , Probiotics/therapeutic use , Probiotics/pharmacology , alpha-Synuclein/metabolism , Oxidative Stress/drug effects , Rotenone/toxicity , Lacticaseibacillus casei/physiology , Methionine-tRNA Ligase , Tryptophan-tRNA Ligase/physiology , Male , Tetrahydrofolate Dehydrogenase/metabolism , Computer Simulation , Parkinsonian Disorders/microbiology , Humans , Neuroprotective Agents/therapeutic use , Mice, Inbred C57BL , Disease Models, Animal , Parkinson Disease, Secondary/chemically induced , Dopaminergic Neurons/drug effects , Parkinson Disease/microbiology
4.
Cell Physiol Biochem ; 58(4): 361-381, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39092504

ABSTRACT

BACKGROUND/AIMS: Traumatic brain injury is a significant public problem with an incidence of 10 million people per year, causing the largest deaths and disabilities worldwide. Head injuries can be classified into primary and secondary head injuries. Secondary head injuries can be caused by several factors such as ischemia, cerebral edema, and neuroinflammation. AIF and MMP-9 are two parameters that can be indicators in measuring the effect of Oleuropein on traumatic brain injury in rats. Oleuropein itself has many activities such as antioxidant, anti-apoptotic, antimicrobial, anti-inflammatory, and neuroprotective. METHODS: Adult male Sprague-Dawley rats (250-350 grams) were exposed to head injury, with or without intraperitoneal administration of Oleuropein. Within 24-72 hours brain tissue was isolated for immunohistochemical analysis, ELISA, and TUNEL. AIF, GFAP, MMP-9, and HMGB-1 levels were determined using immunohistochemistry in both the control and treatment groups. Statistical analysis was made using the One-Way Analysis of Variance (ANOVA) and paired t-test. RESULTS: The results showed that Oleuropein was able to reduce AIF and MMP-9 levels in rats with traumatic brain injury. This indicates that Oleuropein has a neuroprotective effect by reducing inflammation and apoptosis. CONCLUSION: Oleuropein has a potential neuroprotective effect in traumatic brain injury by reducing inflammation and apoptosis. Therefore, Oleuropein can be considered as a potential therapeutic agent for traumatic brain injury in the future.


Subject(s)
Apoptosis Inducing Factor , Brain Injuries, Traumatic , Disease Models, Animal , Iridoid Glucosides , Iridoids , Matrix Metalloproteinase 9 , Rats, Sprague-Dawley , Animals , Iridoid Glucosides/pharmacology , Iridoid Glucosides/therapeutic use , Matrix Metalloproteinase 9/metabolism , Male , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/pathology , Iridoids/pharmacology , Iridoids/therapeutic use , Rats , Apoptosis Inducing Factor/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , HMGB1 Protein/metabolism , Apoptosis/drug effects , Glial Fibrillary Acidic Protein/metabolism , Brain/metabolism , Brain/pathology , Brain/drug effects
5.
Molecules ; 29(15)2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39124919

ABSTRACT

The main feature of neurodegenerative diseases, including Alzheimer's disease, is the network of complex and not fully recognized neuronal pathways and targets involved in their onset and progression. The therapeutic treatment, at present mainly symptomatic, could benefit from a polypharmacological approach based on the development of a single molecular entity designed to simultaneously modulate different validated biological targets. This strategy is principally based on molecular hybridization, obtained by linking or merging different chemical moieties acting with synergistic and/or complementary mechanisms. The coumarin core, widely found in nature, endowed with a recognized broad spectrum of pharmacological activities, large synthetic accessibility and favourable pharmacokinetic properties, appears as a valuable, privileged scaffold to be properly modified in order to obtain compounds able to engage different selected targets. The scientific literature has long been interested in the multifaceted profiles of coumarin derivatives, and in this review, a survey of the most important results of the last four years, on both natural and synthetic coumarin-based compounds, regarding the development of anti-Alzheimer's compounds is reported.


Subject(s)
Alzheimer Disease , Coumarins , Drug Discovery , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Coumarins/chemistry , Coumarins/therapeutic use , Coumarins/pharmacology , Humans , Animals , Biological Products/chemistry , Biological Products/therapeutic use , Biological Products/pharmacology , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/pharmacology , Neuroprotective Agents/chemistry
6.
Gen Physiol Biophys ; 43(5): 385-397, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39140683

ABSTRACT

Stroke is one of the major causes of disability and death worldwide. The lack of effective medical treatment for stroke heightens the need for new therapeutic targets. In this study, we obtained two microarray data sets from the Gene Expression Omnibus (GEO) database and identified differential genes (DEGs) between MCAO and control groups. Then, enrichment analysis of the DEGs was performed using DAVID and Metascape. The results show 27 DEGs shared between the two datasets. The functional enrichment analysis showed that these genes are mainly enriched in immune response, complement and coagulation cascades, apoptotic processes. The four hub genes (C1qc, Fcgr2b, C1qb, and Cd14) were screened out using the Cytoscape. Next, real-time PCR and Western blot analysis showed that expression of C1q and CD14 increased at 14 days after tMCAO. Furthermore, we took eight small molecule compounds with the lowest score using Cmap and studied their background characteristics. These results are built on a meta-analysis of data, which are generally accessible from the online space. Finally, we evaluated the protective effect of the rolipram through behavior tests after tMCAO, and results showed that the rolipram significantly attenuated neurobehavioral dysfunction at 14 days after brain ischemia. The present results provide novel insights into the biological process and potential therapeutic drugs involved in stroke.


Subject(s)
Computational Biology , Ischemic Stroke , Ischemic Stroke/genetics , Ischemic Stroke/drug therapy , Animals , Male , Mice , Gene Expression Profiling , Lipopolysaccharide Receptors/genetics , Lipopolysaccharide Receptors/metabolism , Neuroprotective Agents/therapeutic use , Complement C1q/genetics , Complement C1q/metabolism , Brain Ischemia/genetics , Brain Ischemia/drug therapy
7.
Neurosurg Rev ; 47(1): 430, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39141183

ABSTRACT

This letter commends the article by Luzzi et al. on alternative neuroprotection strategies for aneurysmal subarachnoid hemorrhage (SAH). It highlights the pharmacological advantages of nicardipine, cilostazol, and clazosentan over nimodipine in managing cerebral vasospasm and delayed cerebral ischemia. Emphasizing the need for personalized medicine, it advocates for integrating genetic screening and advanced monitoring techniques to tailor treatments to individual patient profiles. This approach could significantly improve clinical outcomes by optimizing drug efficacy and minimizing adverse effects.


Subject(s)
Brain Ischemia , Neuroprotective Agents , Nimodipine , Subarachnoid Hemorrhage , Vasospasm, Intracranial , Humans , Subarachnoid Hemorrhage/complications , Subarachnoid Hemorrhage/drug therapy , Vasospasm, Intracranial/drug therapy , Vasospasm, Intracranial/prevention & control , Vasospasm, Intracranial/etiology , Nimodipine/therapeutic use , Brain Ischemia/drug therapy , Brain Ischemia/prevention & control , Neuroprotective Agents/therapeutic use , Nicardipine/therapeutic use , Neuroprotection/drug effects , Cilostazol/therapeutic use , Dioxanes/therapeutic use , Vasodilator Agents/therapeutic use , Pyridines/therapeutic use , Pyrimidines , Sulfonamides , Tetrazoles
8.
Mol Biol Rep ; 51(1): 908, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39141244

ABSTRACT

BACKGROUND: Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by oxidative stress and neuroinflammation. Sofalcone (SFC), a chalcone derivative known for its antioxidative and anti-inflammatory properties, is widely used clinically as a gastric mucosa protective agent. However, its therapeutic potential in PD remains to be fully explored. In this study, we investigated the neuroprotective effects of SFC in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mouse model. METHODS AND RESULTS: We found that SFC ameliorated MPTP-induced motor impairments in mice, as assessed by the rotarod and wire tests. Moreover, SFC administration prevented the loss of dopaminergic neurons and striatal degeneration induced by MPTP. Subsequent investigations revealed that SFC reversed MPTP-induced downregulation of NRF2, reduced elevated levels of reactive oxygen species (ROS) and malondialdehyde (MDA), and increased total antioxidant capacity (TAOC). Furthermore, SFC suppressed MPTP-induced activation of microglia and astrocytes, downregulated the pro-inflammatory cytokine TNF-α, and upregulated the anti-inflammatory cytokine IL-4. Additionally, SFC ameliorated the MPTP-induced downregulation of phosphorylation of Akt at Ser473. CONCLUSIONS: This study provides evidence for the neuroprotective effects of SFC, highlighting its antioxidative and anti-inflammatory properties and its role in Akt activation in the PD model. These findings underscore SFC's potential as a promising therapeutic candidate for PD, warranting further clinical investigation.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Chalcones , Disease Models, Animal , Neuroprotective Agents , Oxidative Stress , Animals , Oxidative Stress/drug effects , Mice , Male , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Chalcones/pharmacology , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Antioxidants/pharmacology , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Microglia/drug effects , Microglia/metabolism , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , NF-E2-Related Factor 2/metabolism , Anti-Inflammatory Agents/pharmacology
9.
J Nanobiotechnology ; 22(1): 463, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39095888

ABSTRACT

Neurodegenerative disorders are complex, progressive, and life-threatening. They cause mortality and disability for millions of people worldwide. Appropriate treatment for neurodegenerative diseases (NDs) is still clinically lacking due to the presence of the blood-brain barrier (BBB). Developing an effective transport system that can cross the BBB and enhance the therapeutic effect of neuroprotective agents has been a major challenge for NDs. Exosomes are endogenous nano-sized vesicles that naturally carry biomolecular cargoes. Many studies have indicated that exosome content, particularly microRNAs (miRNAs), possess biological activities by targeting several signaling pathways involved in apoptosis, inflammation, autophagy, and oxidative stress. Exosome content can influence cellular function in healthy or pathological ways. Furthermore, since exosomes reflect the features of the parental cells, their cargoes offer opportunities for early diagnosis and therapeutic intervention of diseases. Exosomes have unique characteristics that make them ideal for delivering drugs directly to the brain. These characteristics include the ability to pass through the BBB, biocompatibility, stability, and innate targeting properties. This review emphasizes the role of exosomes in alleviating NDs and discusses the associated signaling pathways and molecular mechanisms. Furthermore, the unique biological features of exosomes, making them a promising natural transporter for delivering various medications to the brain to combat several NDs, are also discussed.


Subject(s)
Blood-Brain Barrier , Drug Delivery Systems , Exosomes , Neurodegenerative Diseases , Exosomes/metabolism , Humans , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Animals , Drug Delivery Systems/methods , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/pharmacology , MicroRNAs/metabolism , Signal Transduction/drug effects , Drug Carriers/chemistry
10.
Cells ; 13(15)2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39120318

ABSTRACT

The first objective is to highlight the lack of tools to measure whether a given intervention affords neuroprotection in patients with Alzheimer's or Parkinson's diseases. A second aim is to present the primary outcome measures used in clinical trials in cohorts of patients with neurodegenerative diseases. The final aim is to discuss whether metabolomics using body fluids may lead to the discovery of biomarkers of neuroprotection. Information on the primary outcome measures in clinical trials related to Alzheimer's and Parkinson's disease registered since 2018 was collected. We analysed the type of measures selected to assess efficacy, not in terms of neuroprotection since, as stated in the aims, there is not yet any marker of neuroprotection. Proteomic approaches using plasma or CSF have been proposed. PET could estimate the extent of lesions, but disease progression does not necessarily correlate with a change in tracer uptake. We propose some alternatives based on considering the metabolome. A new opportunity opens with metabolomics because there have been impressive technological advances that allow the detection, among others, of metabolites related to mitochondrial function and mitochondrial structure in serum and/or cerebrospinal fluid; some of the differentially concentrated metabolites can become reliable biomarkers of neuroprotection.


Subject(s)
Alzheimer Disease , Biomarkers , Disease Progression , Metabolomics , Neuroprotective Agents , Parkinson Disease , Humans , Metabolomics/methods , Biomarkers/metabolism , Biomarkers/cerebrospinal fluid , Biomarkers/blood , Parkinson Disease/cerebrospinal fluid , Parkinson Disease/metabolism , Parkinson Disease/drug therapy , Parkinson Disease/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/pathology , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/pharmacology , Neuroprotection/drug effects
11.
J Mol Neurosci ; 74(3): 75, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39112893

ABSTRACT

Alzheimer's disease is a neurodegenerative disorder with polygenic etiology. Genetic risk variants for Alzheimer's disease differ among populations. Thus, discovering them in each population is clinically important. A total of 118 patients and 97 controls for VDR rs11568820 and 88 patients and 100 healthy controls for MTHFR C677T polymorphism were genotyped to evaluate the association of these polymorphisms with late-onset Alzheimer's disease in the Iranian population, along with their impacts on the response to Rivastigmine treatment. The VDR C allele was significantly associated with Alzheimer's disease and provided protection against it (P = 0.003, RR = 1.14, 95% CI 1.04-1.24), while the T allele increased susceptibility (P = 0.003, RR = 1.93, 95% CI 1.23-3.02). These results were also considerable upon excluding the effect of APOE ε4 allele. The Prevalence-corrected Positive Predictive Value was 1.71% for the VDR CC genotype and 4% for the VDR CT genotype, indicating lower and almost twofold higher chances of developing Alzheimer's disease, respectively. No significant correlation was observed between MTHFR C677T and Alzheimer's disease. Based on our pharmacogenetic study, MTHFR T allele carriers lacking APOE ε4 allele showed a better response to Rivastigmine treatment after a 2-year follow-up. Moreover, patients with VDR CC genotype displayed milder Alzheimer's disease, particularly when coincided with the APOE ε4 allele. The VDR rs11568820 polymorphism affects both Alzheimer's disease risk and the response to Rivastigmine in Iranian patients. Also, MTHFR C677T polymorphism may play a role in the response to Rivastigmine, through a pathway that needs to be elucidated in future studies.


Subject(s)
Alzheimer Disease , Methylenetetrahydrofolate Reductase (NADPH2) , Polymorphism, Single Nucleotide , Receptors, Calcitriol , Rivastigmine , Humans , Alzheimer Disease/genetics , Alzheimer Disease/drug therapy , Rivastigmine/therapeutic use , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Male , Female , Aged , Iran , Receptors, Calcitriol/genetics , Aged, 80 and over , Cholinesterase Inhibitors/therapeutic use , Cholinesterase Inhibitors/pharmacology , Middle Aged , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/pharmacology
12.
Article in Russian | MEDLINE | ID: mdl-39113450

ABSTRACT

OBJECTIVE: To evaluate the efficacy of Cortexin in the treatment of patients with post-Covid impairment. MATERIAL AND METHODS: Treatment results of 109 patients with post-Covid cognitive impairment aged from 42 to 65 years were analyzed. The main group (MG, n=52), 29 (55.8%) men and 23 (44.2%) women, average age 58.1±4.9 years, who received Cortexin continuously for 20 days at a dose of 10 mg i.m., after 3, 6 months. The comparison group included 57 people, 31 (54.4%) men and 26 (45.6%) women (average age 59.3±3.8 years). The effectiveness of therapy was assessed using neuropsychological testing with international scales. RESULTS: During treatment, statistically significant differences were obtained in MG patients in the form of improved concentration (p<0.05), increased control of exutative functions (p<0.05), and auditory-verbal memory (p=0.002). There were no adverse events in the MG. CONCLUSION: Cortexin is highly effective and safe, and can be recommended as part of a combined staged therapy for post-Covid cognitive impairment. Thus, the study confirms the feasibility of using Cortexin, which has a neurocytoprotective effect.


Subject(s)
COVID-19 , Cognitive Dysfunction , Humans , Female , Male , Middle Aged , Cognitive Dysfunction/etiology , Cognitive Dysfunction/drug therapy , COVID-19/complications , Aged , Adult , SARS-CoV-2 , Treatment Outcome , Neuropsychological Tests , COVID-19 Drug Treatment , Post-Acute COVID-19 Syndrome , Neuroprotective Agents/therapeutic use , Intercellular Signaling Peptides and Proteins
13.
Sci Rep ; 14(1): 17949, 2024 08 02.
Article in English | MEDLINE | ID: mdl-39095453

ABSTRACT

Stroke is a leading cause of permanent disability worldwide. Despite intensive research over the last decades, key anti-inflammatory strategies that have proven beneficial in pre-clinical animal models have often failed in translation. The importance of neutrophils as pro- and anti-inflammatory peripheral immune cells has often been overlooked in ischemic stroke. However, neutrophils rapidly infiltrate into the brain parenchyma after stroke and secrete an array of pro-inflammatory factors including reactive oxygen species, proteases, cytokines, and chemokines exacerbating damage. In this study, we demonstrate the neuroprotective and anti-inflammatory effect of benserazide, a clinically used DOPA decarboxylase inhibitor, using both in vitro models of inflammation and in vivo mouse models of focal cerebral ischemia. Benserazide significantly attenuated PMA-induced NETosis in isolated human neutrophils. Furthermore, benserazide was able to protect both SH-SY5Y and iPSC-derived human cortical neurons when challenged with activated neutrophils demonstrating the clinical relevance of this study. Additional in vitro data suggest the ability of benserazide to polarize macrophages towards M2-phenotypes following LPS stimulation. Neuroprotective effects of benserazide are further demonstrated by in vivo studies where peripheral administration of benserazide significantly attenuated neutrophil infiltration into the brain, altered microglia/macrophage phenotypes, and improved the behavioral outcome post-stroke. Overall, our data suggest that benserazide could serve as a drug candidate for the treatment of ischemic stroke. The importance of our results for future clinical trials is further underlined as benserazide has been approved by the European Medicines Agency as a safe and effective treatment in Parkinson's disease when combined with levodopa.


Subject(s)
Benserazide , Ischemic Stroke , Neuroprotective Agents , Neutrophils , Benserazide/pharmacology , Animals , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Humans , Ischemic Stroke/drug therapy , Ischemic Stroke/immunology , Ischemic Stroke/metabolism , Mice , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/metabolism , Disease Models, Animal , Recovery of Function/drug effects , Male , Mice, Inbred C57BL , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Microglia/drug effects , Microglia/metabolism , Neurons/drug effects , Neurons/metabolism
14.
J Transl Med ; 22(1): 727, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39103918

ABSTRACT

BACKGROUND: Vascular dysregulation is one of the major risk factors of glaucoma, and endothelin-1 (ET-1) may have a role in the pathogenesis of vascular-related glaucoma. Fruit extract from Lycium Barbarum (LB) exhibits anti-ageing and multitarget mechanisms in protecting retinal ganglion cells (RGC) in various animal models. To investigate the therapeutic efficacy of LB glycoproteins (LbGP) in ET-1 induced RGC degeneration, LbGP was applied under pre- and posttreatment conditions to an ET-1 mouse model. Retina structural and functional outcomes were characterised using clinical-based techniques. METHODS: Adult C57BL/6 mice were randomly allocated into four experimental groups, namely vehicle control (n = 9), LbGP-Pretreatment (n = 8), LbGP-Posttreatment (day 1) (n = 8) and LbGP-Posttreatment (day 5) (n = 7). Oral administration of LbGP 1 mg/Kg or PBS for vehicle control was given once daily. Pre- and posttreatment (day 1 or 5) were commenced at 1 week before and 1 or 5 days after intravitreal injections, respectively, and were continued until postinjection day 28. Effects of treatment on retinal structure and functions were evaluated using optical coherence tomography (OCT), doppler OCT and electroretinogram measurements at baseline, post-injection days 10 and 28. RGC survival was evaluated by using RBPMS immunostaining on retinal wholemounts. RESULTS: ET-1 injection in vehicle control induced transient reductions in arterial flow and retinal functions, leading to significant RNFL thinning and RGC loss at day 28. Although ET-1 induced a transient loss in blood flow or retinal functions in all LbGP groups, LbGP treatments facilitated better restoration of retinal flow and retinal functions as compared with the vehicle control. Also, all three LbGP treatment groups (i.e. pre- and posttreatments from days 1 or 5) significantly preserved thRNFL thickness and RGC densities. No significant difference in protective effects was observed among the three LbGP treatment groups. CONCLUSION: LbGP demonstrated neuroprotective effects in a mouse model of ET-1 induced RGC degeneration, with treatment applied either as a pretreatment, immediate or delayed posttreatment. LbGP treatment promoted a better restoration of retinal blood flow, and protected the RNFL, RGC density and retinal functions. This study showed the translational potential of LB as complementary treatment for glaucoma management.


Subject(s)
Endothelin-1 , Mice, Inbred C57BL , Neuroprotection , Retinal Ganglion Cells , Animals , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , Retinal Ganglion Cells/metabolism , Endothelin-1/metabolism , Neuroprotection/drug effects , Electroretinography , Lycium/chemistry , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Retinal Degeneration/drug therapy , Retinal Degeneration/pathology , Tomography, Optical Coherence , Male , Mice , Nerve Degeneration/pathology , Nerve Degeneration/drug therapy
15.
Acta Cir Bras ; 39: e393124, 2024.
Article in English | MEDLINE | ID: mdl-39109777

ABSTRACT

PURPOSE: To investigate the neuroprotective effects of the SOD2 gene in cerebral ischemia reperfusion injury function and the underlying mechanisms in a mice model of middle cerebral artery ischemia reperfusion. METHODS: SOD2 transgenic mice were engineered using transcription activator-like effector nucleases, and the genotype was identified using PCR after every three generations. Transgenic and C57BL/6J wild type mice were simultaneously subjected to the middle cerebral artery occlusion model. RESULTS: SOD2 expression in the brain, heart, kidney, and skeletal muscle of transgenic mice was significantly higher than that in the wild type. Following ischemia reperfusion, the infarct volume of wild type mice decreased after treatment with fenofibrate compared to the CMC group. Infarction volume in SOD2 transgenic mice after CMC and fenofibrate treatment was significantly reduced. The recovery of cerebral blood flow in wild type mice treated with fenofibrate was significantly enhanced compared with that in the CMC group. CONCLUSIONS: The expression of SOD2 in transgenic mice was significantly higher than that in wild type mice, the neuroprotective role of fenofibrate depends on an increase in SOD2 expression.


Subject(s)
Disease Models, Animal , Fenofibrate , Mice, Inbred C57BL , Mice, Transgenic , Reperfusion Injury , Superoxide Dismutase , Animals , Reperfusion Injury/genetics , Superoxide Dismutase/genetics , Fenofibrate/pharmacology , Fenofibrate/therapeutic use , Brain Ischemia/genetics , Humans , Male , Mice , Infarction, Middle Cerebral Artery/genetics , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use
16.
Acta Cir Bras ; 39: e395329, 2024.
Article in English | MEDLINE | ID: mdl-39109783

ABSTRACT

PURPOSE: To evaluate the neuroprotective effect of resveratrol, urapidil, and a combined administration of these drugs against middle cerebral artery occlusion (MCAO) induced ischemia/reperfusion (IR) injury model in rats. METHODS: Thirty-five rats were divided into five groups of seven animals each. Animals in IR, IR resveratrol (IRr), IR urapidil (IRu), and IR + combination of resveratrol and urapidil (IRc) were exposed to MCAO induced cerebral ischemia reperfusion injury model. Rats in IRr and IRu groups received 30-mg/kg resveratrol and 5-mg/kg urapidil respectively. Animals in IRc received a combined treatment of both drugs. At the end of the study, brain tissues were used for oxidative stress (malondialdehyde, glutathione, and superoxide dismutase), pro-apoptotic caspase-3, anti-apoptotic Bcl-2, and pro-inflammatory tumor necrosis factor-α cytokine level measurements. RESULTS: The MCAO model successfully replicated IR injury with significant histopathological changes, elevated tissue oxidative stress, and upregulated apoptotic and inflammatory protein expression in IR group compared to control group (p < 0.001). All parameters were significantly alleviated in IRr group compared to IR group (all p < 0.05). In IRu group, all parameters except for caspase-3 and Bcl-2 were also significantly different than IR group (all p < 0.05). The IRc group showed the biggest difference compared to IR group in all parameters (all p < 0.001). The IRc had higher superoxide dismutase and Bcl-2 levels, and lower caspase-3 levels compared to both IRr and IRu groups (all p < 0.05). Also, the IRc group had lower MDA and TNF-α levels compared to IRu group (all p < 0.05). CONCLUSIONS: The results indicate that combined treatment of resveratrol and urapidil may be a novel strategy to downregulate neurodegeneration in cerebral IR injury.


Subject(s)
Disease Models, Animal , Neuroprotective Agents , Oxidative Stress , Reperfusion Injury , Resveratrol , Stilbenes , Animals , Resveratrol/pharmacology , Resveratrol/therapeutic use , Reperfusion Injury/drug therapy , Reperfusion Injury/prevention & control , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/pharmacology , Male , Oxidative Stress/drug effects , Stilbenes/therapeutic use , Stilbenes/pharmacology , Drug Therapy, Combination , Rats, Wistar , Infarction, Middle Cerebral Artery/drug therapy , Treatment Outcome , Rats , Tumor Necrosis Factor-alpha/analysis , Superoxide Dismutase/analysis , Superoxide Dismutase/metabolism , Malondialdehyde/analysis , Malondialdehyde/metabolism , Reproducibility of Results , Apoptosis/drug effects , Random Allocation , Brain Ischemia/drug therapy , Antioxidants/therapeutic use , Antioxidants/pharmacology , Caspase 3/metabolism , Caspase 3/analysis
17.
Neuromolecular Med ; 26(1): 33, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39138706

ABSTRACT

The newly identified estrogen receptor, G protein-coupled receptor 30 (GPR30), is prevalent in the brain and has been shown to provide significant neuroprotection. Recent studies have linked ferroptosis, a newly characterized form of programmed cell death, closely with cerebral ischemia-reperfusion injury (CIRI), highlighting it as a major contributing factor. Consequently, our research aimed to explore the potential of GPR30 targeting in controlling neuronal ferroptosis and lessening CIRI impacts. Results indicated that GPR30 activation not only improved neurological outcomes and decreased infarct size in a mouse model but also lessened iron accumulation and malondialdehyde formation post-middle cerebral artery occlusion (MCAO). This protective effect extended to increased levels of Nrf2 and GPX4 proteins. Similar protective results were replicated in PC12 cells subjected to Oxygen Glucose Deprivation and Reoxygenation (OGD/R) using the GPR30-specific agonist G1. Importantly, inhibition of Nrf2 with ML385 curtailed the neuroprotective effects of GPR30 activation, suggesting that GPR30 mitigates CIRI primarily through inhibition of neuronal ferroptosis via upregulation of Nrf2 and GPX4.


Subject(s)
Ferroptosis , Infarction, Middle Cerebral Artery , Mice, Inbred C57BL , NF-E2-Related Factor 2 , Phospholipid Hydroperoxide Glutathione Peroxidase , Receptors, Estrogen , Receptors, G-Protein-Coupled , Reperfusion Injury , Signal Transduction , Animals , NF-E2-Related Factor 2/metabolism , Reperfusion Injury/prevention & control , Reperfusion Injury/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/physiology , Ferroptosis/drug effects , Ferroptosis/physiology , Signal Transduction/drug effects , Mice , PC12 Cells , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Rats , Male , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/pharmacology , Disease Models, Animal
18.
J Nanobiotechnology ; 22(1): 499, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39164747

ABSTRACT

Ischemic stroke poses significant challenges in terms of mortality and disability rates globally. A key obstacle to the successful treatment of ischemic stroke lies in the limited efficacy of administering therapeutic agents. Leveraging the unique properties of nanoparticles for brain targeting and crossing the blood-brain barrier, researchers have engineered diverse nanoparticle-based drug delivery systems to improve the therapeutic outcomes of ischemic stroke. This review provides a concise overview of the pathophysiological mechanisms implicated in ischemic stroke, encompassing oxidative stress, glutamate excitotoxicity, neuroinflammation, and cell death, to elucidate potential targets for nanoparticle-based drug delivery systems. Furthermore, the review outlines the classification of nanoparticle-based drug delivery systems according to these distinct physiological processes. This categorization aids in identifying the attributes and commonalities of nanoparticles that target specific pathophysiological pathways in ischemic stroke, thereby facilitating the advancement of nanomedicine development. The review discusses the potential benefits and existing challenges associated with employing nanoparticles in the treatment of ischemic stroke, offering new perspectives on designing efficacious nanoparticles to enhance ischemic stroke treatment outcomes.


Subject(s)
Blood-Brain Barrier , Drug Delivery Systems , Ischemic Stroke , Nanoparticles , Humans , Ischemic Stroke/drug therapy , Animals , Blood-Brain Barrier/metabolism , Drug Delivery Systems/methods , Nanoparticles/chemistry , Oxidative Stress/drug effects , Nanoparticle Drug Delivery System/chemistry , Brain Ischemia/drug therapy , Nanomedicine/methods , Brain/metabolism , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/chemistry
19.
BMC Pregnancy Childbirth ; 24(1): 519, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39090579

ABSTRACT

BACKGROUND: Intravenous administration of magnesium sulfate (MgSO4) to expectant individuals before childbirth, has been evaluated to reduce the likelihood of mortality and occurrence cerebral palsy in their offspring. Therefore, this systematic review and meta-analysis conducted to determine if were the prophylactic use of magnesium sulfate in women at risk for preterm delivery leads to decrease in the incidence of death or cerebral palsy. METHODS: A comprehensive search of electronic databases was done to identify relevant studies. Selection of eligible studies was based on predetermined inclusion criteria. Data extraction was performed, and the methodological quality of the selected studies was assessed using appropriate evaluative tools. A meta-analysis was carried out to estimate the overall effect of intravenous administration of magnesium sulfate on the incidence of death or cerebral palsy. RESULTS: A total of 7 studies met the inclusion criteria and were included in the final analysis. No significant publication bias was observed. The risk of fetal neurological impairment was significantly lower in the MgSO4 group compared to the control group relative risk (RR = 0.70, 95% CI: 0.56 to 0.87; I20%). However, neonatal mortality was not significantly associated with MgSO4 injection. (RR = 1.03, 95% CI: 0.88 to 1.21; I2 = 42%). Subgroup analysis was done based on the bolus dosage of MgSO4 and the duration of the trial follow-up. revealing a non-significant differences between-group. CONCLUSION: This study demonstrated that MgSO4 administration can improve fetal neurological impairment and cerebral palsy but is not linked to reducing mortality. Further studies are necessary to strengthen the evidence and clarify the underlying mechanisms.


Subject(s)
Cerebral Palsy , Magnesium Sulfate , Neuroprotective Agents , Randomized Controlled Trials as Topic , Female , Humans , Infant, Newborn , Pregnancy , Cerebral Palsy/etiology , Cerebral Palsy/prevention & control , Magnesium Sulfate/therapeutic use , Magnesium Sulfate/administration & dosage , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/administration & dosage , Premature Birth
20.
Arch Biochem Biophys ; 759: 110109, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39117070

ABSTRACT

Chronic inflammation is an important pathogenetic factor that leads to the progression of Alzheimer's disease (AD), and specialized pro-resolving lipid mediators (SPMs) play critical role in regulating inflammatory responses during AD pathogenesis. Maresin1 (MaR1) is the latest discovered SPMs, and it is found that MaR1 improves AD cognitive impairment by regulating neurotrophic pathways to protect AD synapses and reduce Aß production, which made MaR1 as candidate agent for AD treatment. Unfortunately, the underlying mechanisms are still largely known. In this study, the AD mice and cellular models were subjected to MaR1 treatment, and we found that MaR1 reduced Aß production to ameliorate AD-related symptoms and increased the expression levels of ADAM10/17, sAPPα and sAPPß to exert its anti-inflammatory role. In addition, as it was determined by Western Blot analysis, we observed that MaR1 could affected the neuroprotective signal pathways. Specifically, MaR1 downregulated p57NTR and upregulated TrkA to activate the p75NTR/TrkA signal pathway, and it could increase the expression levels of p-PI3K and p-Akt, and downregulated p-mTOR to activate the PI3K/AKT/ERK/mTOR pathway. Finally, we verified the role of ADAM10/17 in regulating AD progression, and we found that silencing of ADAM10/17 inactivated the above neuroprotective signal pathways to aggravate AD pathogenesis. In conclusion, MaR1 is verified as potential therapeutic agent for AD by eliminating Aß production, upregulating ADAM10/17, sAPPα and sAPPß, and activating the neuroprotective p75NTR/TrkA pathway and the PI3K/AKT/ERK/mTOR pathway.


Subject(s)
ADAM10 Protein , Alzheimer Disease , Amyloid Precursor Protein Secretases , Amyloid beta-Peptides , Docosahexaenoic Acids , Signal Transduction , Alzheimer Disease/metabolism , Alzheimer Disease/drug therapy , Animals , ADAM10 Protein/metabolism , ADAM10 Protein/genetics , Amyloid Precursor Protein Secretases/metabolism , Signal Transduction/drug effects , Docosahexaenoic Acids/pharmacology , Docosahexaenoic Acids/metabolism , Amyloid beta-Peptides/metabolism , Mice , Inflammation/metabolism , Pilot Projects , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Humans , Membrane Proteins/metabolism , Membrane Proteins/genetics , Male
SELECTION OF CITATIONS
SEARCH DETAIL