Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 145
Filter
Add more filters

Publication year range
1.
Inflammopharmacology ; 31(3): 1449-1464, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36856916

ABSTRACT

Alzheimer's disease (AD) is one of the most prevalent neurodegenerative illnesses, and yet, no workable treatments have been discovered to prevent or reverse AD. Curcumin (CUR), the major polyphenolic compound of turmeric (Curcuma longa) rhizomes, and Ginkgo biloba extract (GBE) are natural substances derived from conventional Chinese herbs that have long been shown to provide therapeutic advantages for AD. The uptake of curcumin into the brain is severely restricted by its low ability to cross the blood-brain barrier (BBB). Meanwhile, GBE has been shown to improve BBB permeability. The present study evaluated the neuroprotective effects and pharmacokinetic profile of curcumin and GBE combination to find out whether GBE can enhance curcumin's beneficial effects in AD by raising its brain concentration. Results revealed that CUR + GBE achieved significantly higher levels of curcumin in the brain and plasma after 30 min and 1 h of oral administration, compared to curcumin alone, and this was confirmed by reversed phase high-performance liquid chromatography (RP-HPLC). The effect of combined oral treatment, for 28 successive days, on cognitive function and other AD-like alterations was studied in scopolamine-heavy metal mixtures (SCO + HMM) AD model in rats. The combination reversed at least, partially on the learning and memory impairment induced by SCO + HMM. This was associated with a more pronounced inhibitory effect on acetylcholinesterase (AChE), caspase-3, hippocampal amyloid beta (Aß1-42), and phosphorylated tau protein (p-tau) count, and pro-inflammatory cytokines tumor necrosis factor-alpha (TNF-α) and interleukine-1beta (IL-1ß), as compared to the curcumin alone-treated group. Additionally, the combined treatment significantly decreased lipid peroxidation (MDA) and increased levels of reduced glutathione (GSH), when compared with the curcumin alone. These findings support the concept that the combination strategy might be an alternative therapy in the management/prevention of neurological disorders. This study sheds light on a new approach for exploring new phyto-therapies for AD and emphasizes that more research should focus on the synergic effects of herbal drugs in future.


Subject(s)
Alzheimer Disease , Curcumin , Neuroprotective Agents , Animals , Rats , Acetylcholinesterase/metabolism , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Curcumin/pharmacokinetics , Curcumin/pharmacology , Curcumin/therapeutic use , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Ginkgo Extract/pharmacokinetics , Ginkgo Extract/pharmacology , Ginkgo Extract/therapeutic use , Humans , Blood-Brain Barrier
2.
Molecules ; 27(3)2022 Feb 07.
Article in English | MEDLINE | ID: mdl-35164373

ABSTRACT

Acanthopanax senticosus (AS) is a medicinal and food homologous plant with many biological activities. In this research, we generated a brain injury model by 60Co -γ ray radiation at 4 Gy, and gavaged adult mice with the extract with AS, Acanthopanax senticocus polysaccharides (ASPS), flavones, syringin and eleutheroside E (EE) to explore the therapeutic effect and metabolic characteristics of AS on the brain injury. Behavioral tests and pathological experiments showed that the AS prevented the irradiated mice from learning and memory ability impairment and protected the neurons of irradiated mice. Meanwhile, the functional components of AS increased the antioxidant activity of irradiated mice. Furthermore, we found the changes of neurotransmitters, especially in the EE and syringin groups. Finally, distribution and pharmacokinetic analysis of AS showed that the functional components, especially EE, could exert their therapeutic effects in brain of irradiated mice. This lays a theoretical foundation for the further research on the treatment of radiation-induced brain injury by AS.


Subject(s)
Antioxidants/pharmacology , Brain Injuries/drug therapy , Eleutherococcus/chemistry , Neuroprotective Agents/pharmacology , Neurotransmitter Agents/metabolism , Plant Extracts/pharmacology , Radiation Injuries/drug therapy , Animals , Antioxidants/pharmacokinetics , Brain/drug effects , Brain Injuries/etiology , Brain Injuries/pathology , Cobalt Radioisotopes/toxicity , Male , Mice , Neuroprotective Agents/pharmacokinetics , Plant Extracts/pharmacokinetics , Radiation Injuries/etiology , Radiation Injuries/pathology , Tissue Distribution
3.
Nutrients ; 14(2)2022 Jan 07.
Article in English | MEDLINE | ID: mdl-35057429

ABSTRACT

For thousands of years, mankind has been using plant extracts or plants themselves as medicinal herbs. Currently, there is a great deal of public interest in naturally occurring medicinal substances that are virtually non-toxic, readily available, and have an impact on well-being and health. It has been noted that dietary curcumin is one of the regulators that may positively influence changes in the brain after ischemia. Curcumin is a natural polyphenolic compound with pleiotropic biological properties. The observed death of pyramidal neurons in the CA1 region of the hippocampus and its atrophy are considered to be typical changes for post-ischemic brain neurodegeneration and for Alzheimer's disease. Additionally, it has been shown that one of the potential mechanisms of severe neuronal death is the accumulation of neurotoxic amyloid and dysfunctional tau protein after cerebral ischemia. Post-ischemic studies of human and animal brains have shown the presence of amyloid plaques and neurofibrillary tangles. The significant therapeutic feature of curcumin is that it can affect the aging-related cellular proteins, i.e., amyloid and tau protein, preventing their aggregation and insolubility after ischemia. Curcumin also decreases the neurotoxicity of amyloid and tau protein by affecting their structure. Studies in animal models of cerebral ischemia have shown that curcumin reduces infarct volume, brain edema, blood-brain barrier permeability, apoptosis, neuroinflammation, glutamate neurotoxicity, inhibits autophagy and oxidative stress, and improves neurological and behavioral deficits. The available data suggest that curcumin may be a new therapeutic substance in both regenerative medicine and the treatment of neurodegenerative disorders such as post-ischemic neurodegeneration.


Subject(s)
Alzheimer Disease/drug therapy , Brain Ischemia/complications , Curcumin/pharmacology , Neuroprotective Agents/pharmacology , Alzheimer Disease/etiology , Amyloid/drug effects , Amyloid/metabolism , Animals , Apoptosis/drug effects , Atrophy/etiology , Biological Availability , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain Edema/drug therapy , Brain Ischemia/drug therapy , Curcumin/chemistry , Curcumin/pharmacokinetics , Disease Models, Animal , Gastrointestinal Microbiome/physiology , Gerbillinae , Hippocampus/pathology , Humans , Mice , Neuroinflammatory Diseases/drug therapy , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacokinetics , Oxidative Stress/drug effects , Rats , tau Proteins/drug effects , tau Proteins/metabolism
4.
Int J Mol Sci ; 22(22)2021 Nov 12.
Article in English | MEDLINE | ID: mdl-34830148

ABSTRACT

Phytosterols constitute a class of natural products that are an important component of diet and have vast applications in foods, cosmetics, and herbal medicines. With many and diverse isolated structures in nature, they exhibit a broad range of biological and pharmacological activities. Among over 200 types of phytosterols, stigmasterol and ß-sitosterol were ubiquitous in many plant species, exhibiting important aspects of activities related to neurodegenerative diseases. Hence, this mini-review presented an overview of the reported studies on selected phytosterols related to neurodegenerative diseases. It covered the major phytosterols based on biosynthetic considerations, including other phytosterols with significant in vitro and in vivo biological activities.


Subject(s)
Brain/metabolism , Neurodegenerative Diseases/prevention & control , Phytosterols/therapeutic use , Phytotherapy/methods , Plants, Medicinal/chemistry , Brain/pathology , Humans , Molecular Structure , Neurodegenerative Diseases/metabolism , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/therapeutic use , Phytosterols/chemistry , Phytosterols/pharmacokinetics , Sitosterols/chemistry , Sitosterols/pharmacokinetics , Sitosterols/therapeutic use , Stigmasterol/chemistry , Stigmasterol/pharmacokinetics , Stigmasterol/therapeutic use
5.
Cell Mol Biol (Noisy-le-grand) ; 67(1): 101-105, 2021 Jan 31.
Article in English | MEDLINE | ID: mdl-34817361

ABSTRACT

Parkinson's disease (PD) is one of the most prevalent neurodegenerative disorders worldwide. It is caused by the degeneration of dopaminergic neurons from the substantia nigra pars compacta. This neuronal loss causes the dopamine deficiency that leads to a series of functional changes within the basal ganglia, producing motor control abnormalities. L-DOPA is considered the gold standard for PD treatment, and it may alleviate its clinical manifestations for some time. However, its prolonged administration produces tolerance and several severe side effects, including dyskinesias and gastrointestinal disorders. Thus, there is an urgent need to find effective medications, and current trends have proposed some natural products as emerging options for this purpose. Concerning this, curcumin represents a promising bioactive compound with high therapeutic potential. Diverse studies in cellular and animal models have suggested that curcumin could be employed for the treatment of PD. Therefore, the objective of this narrative mini-review is to present an overview of the possible therapeutic effects of curcumin and the subjacent molecular mechanisms. Moreover, we describe several possible nanocarrier-based approaches to improve the bioavailability of curcumin and enhance its biological activity.


Subject(s)
Brain/drug effects , Curcumin/administration & dosage , Nanoparticles/administration & dosage , Parkinson Disease/drug therapy , Animals , Biological Availability , Brain/metabolism , Curcumin/chemistry , Curcumin/pharmacokinetics , Drug Liberation , Glutathione Peroxidase/metabolism , Humans , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , Nanoparticles/chemistry , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacokinetics , Parkinson Disease/metabolism , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , Treatment Outcome , Up-Regulation/drug effects
6.
Food Funct ; 12(19): 9211-9228, 2021 Oct 04.
Article in English | MEDLINE | ID: mdl-34606547

ABSTRACT

The present study aims to investigate the protective effects of N-(3-methoxybenzyl)-(9Z,12Z,15Z)-octadecatrienamide (M 18:3) on corticosterone-induced neurotoxicity. A neurotoxic model was established by subcutaneous injection of corticosterone (40 mg per kg bw) for 21 days. Depressive behaviors (the percentage of sucrose consumption, the immobility time in the forced swimming test, and the total distance in the open field test) were observed. The levels of the brain-derived neurotrophic factor, the contents of tumor necrosis factor-α and interleukin-6, and the numbers of positive cells of doublecortin and bromodeoxyuridine in the hippocampus were measured. The density of hippocampal neurons was calculated. The morphological changes of hippocampal neurons (the density of dendritic spines, the dendritic length, and the area and volume of dendritic cell bodies) were observed. The expression levels of synaptophysin, synapsin I, and postsynaptic density protein 95 were measured. Behavioral experiments showed that M 18:3 (5 and 25 mg per kg bw) could remarkably improve the depressive behaviors. The enzyme-linked immunosorbent assay showed that M 18:3 could considerably reduce hippocampal neuroinflammation and increase hippocampal neurotrophy. Nissl staining showed that M 18:3 could remarkably improve the corticosterone-induced decrease in the hippocampal neuron density. Immunofluorescence analysis showed that M 18:3 could considerably promote hippocampal neurogenesis. Golgi staining showed that M 18:3 could remarkably improve the corticosterone-induced changes in the hippocampal dendritic structure. Western blotting showed that M 18:3 could considerably increase the expression levels of synaptic-structure-related proteins in the hippocampus. In conclusion, the protective effects of M 18:3 may be attributed to the anti-inflammatory, neurotrophic, and synaptic protection properties.


Subject(s)
Alkenes/pharmacology , Benzyl Compounds/pharmacology , Hippocampus/drug effects , Lepidium , Neuroprotective Agents/pharmacology , Alkenes/pharmacokinetics , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antidepressive Agents/pharmacology , Behavior, Animal/drug effects , Benzyl Compounds/pharmacokinetics , Blood-Brain Barrier/metabolism , Cell Count , Cell Shape , Corticosterone , Depression/drug therapy , Hippocampus/cytology , Hippocampus/metabolism , Hippocampus/physiology , Male , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurogenesis , Neurons/cytology , Neuroprotective Agents/pharmacokinetics , Plant Extracts/chemistry , Plant Extracts/pharmacokinetics , Plant Extracts/pharmacology , Rats , Rats, Wistar , Synapses/drug effects , Synapses/physiology
7.
Mar Drugs ; 19(6)2021 May 26.
Article in English | MEDLINE | ID: mdl-34073184

ABSTRACT

Euphausia superba, commonly known as krill, is a small marine crustacean from the Antarctic Ocean that plays an important role in the marine ecosystem, serving as feed for most fish. It is a known source of highly bioavailable omega-3 polyunsaturated fatty acids (eicosapentaenoic acid and docosahexaenoic acid). In preclinical studies, krill oil showed metabolic, anti-inflammatory, neuroprotective and chemo preventive effects, while in clinical trials it showed significant metabolic, vascular and ergogenic actions. Solvent extraction is the most conventional method to obtain krill oil. However, different solvents must be used to extract all lipids from krill because of the diversity of the polarities of the lipid compounds in the biomass. This review aims to provide an overview of the chemical composition, bioavailability and bioaccessibility of krill oil, as well as the mechanisms of action, classic and non-conventional extraction techniques, health benefits and current applications of this marine crustacean.


Subject(s)
Anti-Inflammatory Agents , Antineoplastic Agents , Dietary Supplements , Euphausiacea , Fatty Acids, Omega-3 , Fish Oils/chemistry , Neuroprotective Agents , Animals , Anti-Inflammatory Agents/pharmacokinetics , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Exercise , Fatty Acids, Omega-3/pharmacokinetics , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-3/therapeutic use , Fish Oils/pharmacology , Gastrointestinal Microbiome/drug effects , Humans , Inflammatory Bowel Diseases/diet therapy , Inflammatory Bowel Diseases/prevention & control , Metabolic Diseases/diet therapy , Metabolic Diseases/prevention & control , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use
8.
ChemistryOpen ; 10(6): 630-638, 2021 06.
Article in English | MEDLINE | ID: mdl-34102706

ABSTRACT

Two novel alkaloids compounds together with fifteen know metabolites were identified from Aspergillus ochraceus. The stereochemistry features of the new molecules were determined via HRESIMS, NMR, ECD, and XRD analyses. Amongst these, compounds two compounds exhibited potential efficacy as anti-Parkinson's disease with the EC50 values of 2.30 and 2.45 µM, respectively. ADMET prediction showed that these compounds owned favorable drug-like characteristics and safe toxicity scores towards CNS drugs. Virtual screening analyses manifested that the compounds exhibited not only robust and reliable interactions to adenosine receptors A2A , but also higher binding selectivity to A2A receptors than to A1 and A3 receptors. Molecular dynamics simulation demonstrated the reliability of molecular docking results and the stability of the complexes obtained with the novel compounds and A2A receptors in natural environments. It is the first time that anti-PD lead compounds have been identified from Aspergillus ochraceus and targeting adenosine A2A receptors.


Subject(s)
Adenosine A2 Receptor Antagonists/pharmacology , Antiparkinson Agents/pharmacology , Aspergillus ochraceus/chemistry , Receptor, Adenosine A2A/metabolism , Adenosine A2 Receptor Antagonists/chemistry , Adenosine A2 Receptor Antagonists/metabolism , Adenosine A2 Receptor Antagonists/pharmacokinetics , Animals , Antiparkinson Agents/chemistry , Antiparkinson Agents/metabolism , Antiparkinson Agents/pharmacokinetics , Cell Line, Tumor , Drug Evaluation, Preclinical , Female , Humans , Male , Mice , Molecular Docking Simulation , Molecular Dynamics Simulation , Neuroprotective Agents/chemistry , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/pharmacology , Rats , Stereoisomerism
9.
Pharmacol Res ; 169: 105690, 2021 07.
Article in English | MEDLINE | ID: mdl-34029711

ABSTRACT

Traditional Chinese medicine plays a significant role in the treatment of various diseases and has attracted increasing attention for clinical applications. Forsythiae Fructus, the dried fruit of Forsythia suspensa (Thunb.) Vahl, is a widely used Chinese medicinal herb in clinic for its extensive pharmacological activities. Forsythiaside A is the main active index component isolated from Forsythiae Fructus and possesses prominent bioactivities. Modern pharmacological studies have confirmed that Forsythiaside A exhibits significant activities in treating various diseases, including inflammation, virus infection, neurodegeneration, oxidative stress, liver injury, and bacterial infection. In this review, the pharmacological activities of Forsythiaside A have been comprehensively reviewed and summarized. According to the data, Forsythiaside A shows remarkable anti-inflammation, antivirus, neuroprotection, antioxidant, hepatoprotection, and antibacterial activities through regulating multiple signaling transduction pathways such as NF-κB, MAPK, JAK/STAT, Nrf2, RLRs, TRAF, TLR7, and ER stress. In addition, the toxicity and pharmacokinetic properties of Forsythiaside A are also discussed in this review, thus providing a solid foundation and evidence for further studies to explore novel effective drugs from Chinese medicine monomers.


Subject(s)
Glycosides/pharmacology , Animals , Anti-Inflammatory Agents/pharmacokinetics , Anti-Inflammatory Agents/pharmacology , Forsythia/chemistry , Glycosides/adverse effects , Glycosides/pharmacokinetics , Humans , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/pharmacology , Signal Transduction/drug effects
10.
Molecules ; 26(6)2021 Mar 23.
Article in English | MEDLINE | ID: mdl-33807056

ABSTRACT

Kashmir saffron (Crocus sativus L.), also known as Indian saffron, is an important Asian medicinal plant with protective therapeutic applications in brain health. The main bioactive in Kashmir or Indian Saffron (KCS) and its extract (CSE) are apocarotenoids picrocrocin (PIC) and safranal (SAF) with carotenoids, crocetin esters (crocins), and crocetins. The ultra-fast liquid chromatography(UFLC)- photodiode array standardization confirmed the presence of biomarkers PIC, trans-4-GG-crocin (T4C), trans-3-Gg-crocin (T3C), cis-4-GG-crocin (C4C), trans-2-gg-crocin (T2C), trans-crocetin (TCT), and SAF in CSE. This study's objectives were to develop and validate a sensitive and rapid UFLC-tandem mass spectrometry method for PIC and SAF along T4C and TCT in rat plasma with internal standards (IS). The calibration curves were linear (R2 > 0.990), with the lower limit of quantification (LLOQ) as 10 ng/mL. The UFLC-MS/MS assay-based precision (RSD, <15%) and accuracy (RE, -11.03-9.96) on analytical quality control (QC) levels were well within the acceptance criteria with excellent recoveries (91.18-106.86%) in plasma samples. The method was applied to investigate the in vivo pharmacokinetic parameters after oral administration of 40 mg/kg CSE in the rats (n = 6). The active metabolite TCT and T4C, PIC, SAF were quantified for the first time with T3C, C4C, T2C by this validated bioanalytical method, which will be useful for preclinical/clinical trials of CSE as a potential neuroprotective dietary supplement.


Subject(s)
Carotenoids , Crocus/chemistry , Neuroprotective Agents , Plant Extracts , Animals , Carotenoids/chemistry , Carotenoids/pharmacokinetics , Carotenoids/pharmacology , Chromatography, High Pressure Liquid , Male , Mass Spectrometry , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/pharmacology , Plant Extracts/chemistry , Plant Extracts/pharmacokinetics , Plant Extracts/pharmacology , Rats , Rats, Sprague-Dawley
11.
Biomed Res Int ; 2021: 6645720, 2021.
Article in English | MEDLINE | ID: mdl-33575335

ABSTRACT

PURPOSE: The study evaluated the neuroprotective effect and pharmacokinetic profile of turmeric extract and their metabolites in the blood and brain in an aluminum-induced neurotoxic animal model. METHODS: Swiss albino mice received turmeric extract (TE), TE-essential oil combination (TE+EO) at doses of 25 and 50 mg/kg/day orally, vehicle (control), and a positive control group. Neurotoxicity was induced by injecting aluminum chloride (40 mg/kg/day, i.p.), and the effect of the intervention was studied for 45 days. The pharmacokinetic and behavioral biochemical markers of brain function and brain histopathological changes were evaluated. RESULTS: The AUC 0-t showed a 30.1 and 54.2 times higher free curcumin concentration in plasma with 25 mg/kg and 50 mg/kg of TE+EO vs. TE, respectively. The concentration of free curcumin in the brain was 11.01 and 13.71-fold higher for 25 mg/kg and 50 mg/kg of TE+EO vs. TE, respectively. Aluminum impairs spatial learning and memory, which was significantly reversed with TE+EO by 28.6% (25 mg/kg) and 39.4% (50 mg/kg). In the elevated plus maze test, 44.8% (25 mg/kg) and 67.1% (50 mg/kg) improvements were observed. A significant reduction in aluminum-induced lipid peroxidation was observed. Also, the levels of glutathione, acetylcholinesterase, and catalase were improved with TE+EO. Damage to the hippocampal pyramidal cells was averted with TE+EO. CONCLUSION: The neuroprotective and antioxidant response confirms the benefits of TE+EO against aluminum-induced neurotoxicity. The presence of free curcumin and its metabolites in the brain and plasma establishes its improved bioavailability and tissue distribution. Therefore, the benefits of TE+EO could be harnessed in neurodegenerative diseases.


Subject(s)
Brain , Curcuma , Neuroprotective Agents , Plant Extracts , Animals , Biological Availability , Brain/drug effects , Brain/metabolism , Brain Chemistry/drug effects , CA1 Region, Hippocampal/drug effects , Curcumin , Maze Learning/drug effects , Mice , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/pharmacology , Oils, Volatile/pharmacokinetics , Oils, Volatile/pharmacology , Plant Extracts/pharmacokinetics , Plant Extracts/pharmacology , Tissue Distribution
12.
Neurochem Res ; 46(3): 535-549, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33548035

ABSTRACT

Extensive data have reported the involvement of oxidative stress in the pathogenesis of neuropsychiatric disorders, prompting the pursuit of antioxidant molecules that could become adjuvant pharmacological agents for the management of oxidative stress-associated disorders. The 3-[(4-chlorophenyl)selanyl]-1-methyl-1H-indole (CMI) has been reported as an antioxidant and immunomodulatory compound that improves depression-like behavior and cognitive impairment in mice. However, the exact effect of CMI on specific brain cells is yet to be studied. In this context, the present study aimed to evaluate the antioxidant activity of CMI in H2O2-induced oxidative stress on human dopaminergic neuroblastoma cells (SH-SY5Y) and to shed some light into its possible mechanism of action. Our results demonstrated that the treatment of SH-SY5Y cells with 4 µM CMI protected them against H2O2 (343 µM)-induced oxidative stress. Specifically, CMI prevented the increased number of reactive oxygen species (ROS)-positive cells induced by H2O2 exposure. Furthermore, CMI treatment increased the levels of reduced glutathione in SH-SY5Y cells. Molecular docking studies demonstrated that CMI might interact with enzymes involved in glutathione metabolism (i.e., glutathione peroxidase and glutathione reductase) and H2O2 scavenging (i.e., catalase). In silico pharmacokinetics analysis predicted that CMI might be well absorbed, metabolized, and excreted, and able to cross the blood-brain barrier. Also, CMI was not considered toxic overall. Taken together, our results suggest that CMI protects dopaminergic neurons from H2O2-induced stress by lowering ROS levels and boosting the glutathione system. These results will facilitate the clinical application of CMI to treat nervous system diseases associated with oxidative stress.


Subject(s)
Hydrogen Peroxide/toxicity , Indoles/pharmacology , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Selenium Compounds/pharmacology , Catalytic Domain , Cell Line, Tumor , Glutathione/metabolism , Glutathione Transferase/chemistry , Glutathione Transferase/metabolism , Humans , Indoles/chemistry , Indoles/metabolism , Indoles/pharmacokinetics , Molecular Docking Simulation , Neuroprotective Agents/chemistry , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacokinetics , Oxidoreductases/chemistry , Oxidoreductases/metabolism , Protein Binding , Reactive Oxygen Species/metabolism , Selenium Compounds/chemistry , Selenium Compounds/metabolism , Selenium Compounds/pharmacokinetics
13.
J Med Chem ; 64(2): 1103-1115, 2021 01 28.
Article in English | MEDLINE | ID: mdl-33404239

ABSTRACT

O-GlcNAcase (OGA) has received increasing attention as an attractive therapeutic target for tau-mediated neurodegenerative disorders; however, its role in these pathologies remains unclear. Therefore, potent chemical tools with favorable pharmacokinetic profiles are desirable to characterize this enzyme. Herein, we report the discovery of a potent and novel OGA inhibitor, compound 5i, comprising an aminopyrimidine scaffold, identified by virtual screening based on multiple methodologies combining structure-based and ligand-based approaches, followed by sequential optimization with a focus on ligand lipophilicity efficiency. This compound was observed to increase the level of O-GlcNAcylated protein in cells and display suitable pharmacokinetic properties and brain permeability. Crystallographic analysis revealed that the chemical series bind to OGA via characteristic hydrophobic interactions, which resulted in a high affinity for OGA with moderate lipophilicity. Compound 5i could serve as a useful chemical probe to help establish a proof-of-concept of OGA inhibition as a therapeutic target for the treatment of tauopathies.


Subject(s)
Acetylglucosamine/antagonists & inhibitors , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/pharmacology , beta-N-Acetylhexosaminidases/antagonists & inhibitors , Animals , Brain/metabolism , Cell Line , Computer Simulation , Drug Design , Drug Evaluation, Preclinical , Enzyme Inhibitors/pharmacokinetics , Humans , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Docking Simulation , Molecular Dynamics Simulation , Neuroprotective Agents/pharmacokinetics , Structure-Activity Relationship , Tauopathies/drug therapy
14.
Biomed Pharmacother ; 135: 111167, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33383373

ABSTRACT

Alzheimer's disease (AD) is a common progressive neuro-degenerative disease, and the morbidity and mortality are still on the rise. In spite of recent advances in AD treatment, their clinical efficacy has been limited, non-curative and easy to drug resistance. Alpiniae oxyphyllae Fructus (AOF), derived from the dried and mature fruits of the Zingiberaceae plant Alpinia oxyphylla Miq, is a choice in traditional Chinese medicine to treat AD, which has a good effect and has been used for a long time. Recent studies have demonstrated its potent activities in modulating multiple signaling pathways associated with ß-amyloid deposition, tau protein phosphorylation, chronic inflammation, oxidative stress. The neuropharmacological mechanism of AOF in AD have been fully illustrated in numerous studies. In this review, we first briefly described the active components of AOF and related mechanism for treating AD. And we also provide a systematic overview of recent progress on the pharmacokinetic characteristics of the active ingredients of AOF and analyzed their bioavailability differences in the development of AD. Thus, AOF hold a great therapeutic potential in the treatment of AD and is worthy of further research and promotion.


Subject(s)
Alpinia , Alzheimer Disease/drug therapy , Brain/drug effects , Medicine, Chinese Traditional , Neurons/drug effects , Neuroprotective Agents/therapeutic use , Plant Extracts/therapeutic use , Alpinia/chemistry , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Animals , Brain/metabolism , Brain/physiopathology , Humans , Nerve Degeneration , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/isolation & purification , Neuroprotective Agents/pharmacokinetics , Plant Extracts/isolation & purification , Plant Extracts/pharmacokinetics , Rats , Signal Transduction
15.
Mol Pharm ; 18(2): 610-626, 2021 02 01.
Article in English | MEDLINE | ID: mdl-32584043

ABSTRACT

Polyglutamine (polyQ) diseases, such as Huntington's disease and several types of spinocerebellar ataxias, are dominantly inherited progressive neurodegenerative disorders and characterized by the presence of expanded CAG trinucleotide repeats in the respective disease locus of the patient genomes. Patients with polyQ diseases currently need to rely on symptom-relieving treatments because disease-modifying therapeutic interventions remain scarce. Many disease-modifying therapeutic agents are now under clinical testing for treating polyQ diseases, but their delivery to the brain is often too invasive (e.g., intracranial injection) or inefficient, owing to in vivo degradation and clearance by physiological barriers (e.g., oral and intravenous administration). Nanoparticles provide a feasible solution for improving drug delivery to the brain, as evidenced by an increasing number of preclinical studies that document the efficacy of nanomedicines for polyQ diseases over the past 5-6 years. In this review, we present the pathogenic mechanisms of polyQ diseases, the common animal models of polyQ diseases for evaluating the efficacy of nanomedicines, and the common administration routes for delivering nanoparticles to the brain. Next, we summarize the recent preclinical applications of nanomedicines for treating polyQ diseases and improving neurological conditions in vivo, placing emphasis on antisense oligonucleotides, small peptide inhibitors, and small molecules as the disease-modifying agents. We conclude with our perspectives of the burgeoning field of "nanomedicines for polyQ diseases", including the use of inorganic nanoparticles and potential drugs as next-generation nanomedicines, development of higher-order animal models of polyQ diseases, and importance of "brain-nano" interactions.


Subject(s)
Drug Carriers/chemistry , Huntington Disease/drug therapy , Nanoparticles/chemistry , Neuroprotective Agents/administration & dosage , Peptides/antagonists & inhibitors , Spinocerebellar Ataxias/drug therapy , Administration, Intranasal , Administration, Oral , Animals , Animals, Genetically Modified , Biological Availability , Blood-Brain Barrier/metabolism , Brain/drug effects , Brain/pathology , Disease Models, Animal , Drug Evaluation, Preclinical , Genetic Loci/genetics , Humans , Huntington Disease/genetics , Huntington Disease/pathology , Injections, Intraperitoneal , Injections, Intravenous , Injections, Intraventricular , Injections, Spinal , Neuroprotective Agents/pharmacokinetics , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/pharmacokinetics , Peptides/genetics , Peptides/metabolism , Permeability , Spinal Cord/drug effects , Spinal Cord/pathology , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/pathology , Tissue Distribution , Trinucleotide Repeat Expansion
16.
Neuromolecular Med ; 23(1): 184-198, 2021 03.
Article in English | MEDLINE | ID: mdl-33067719

ABSTRACT

Ergothioneine (ET) is a naturally occurring antioxidant that is synthesized by non-yeast fungi and certain bacteria. ET is not synthesized by animals, including humans, but is avidly taken up from the diet, especially from mushrooms. In the current study, we elucidated the effect of ET on the hCMEC/D3 human brain endothelial cell line. Endothelial cells are exposed to high levels of the cholesterol oxidation product, 7-ketocholesterol (7KC), in patients with cardiovascular disease and diabetes, and this process is thought to mediate pathological inflammation. 7KC induces a dose-dependent loss of cell viability and an increase in apoptosis and necrosis in the endothelial cells. A relocalization of the tight junction proteins, zonula occludens-1 (ZO-1) and claudin-5, towards the nucleus of the cells was also observed. These effects were significantly attenuated by ET. In addition, 7KC induces marked increases in the mRNA expression of pro-inflammatory cytokines, IL-1ß IL-6, IL-8, TNF-α and cyclooxygenase-2 (COX2), as well as COX2 enzymatic activity, and these were significantly reduced by ET. Moreover, the cytoprotective and anti-inflammatory effects of ET were significantly reduced by co-incubation with an inhibitor of the ET transporter, OCTN1 (VHCL). This shows that ET needs to enter the endothelial cells to have a protective effect and is unlikely to act via extracellular neutralizing of 7KC. The protective effect on inflammation in brain endothelial cells suggests that ET might be useful as a nutraceutical for the prevention or management of neurovascular diseases, such as stroke and vascular dementia. Moreover, the ability of ET to cross the blood-brain barrier could point to its usefulness in combatting 7KC that is produced in the CNS during neuroinflammation, e.g. after excitotoxicity, in chronic neurodegenerative diseases, and possibly COVID-19-related neurologic complications.


Subject(s)
Antioxidants/pharmacology , COVID-19/complications , Endothelial Cells/drug effects , Ergothioneine/pharmacology , Ketocholesterols/toxicity , Nervous System Diseases/prevention & control , Neuroprotective Agents/pharmacology , Antioxidants/pharmacokinetics , Apoptosis/drug effects , Biological Transport , Blood-Brain Barrier , Brain/blood supply , Brain/cytology , Cell Line , Cholesterol/metabolism , Claudin-5 , Cyclooxygenase 2/biosynthesis , Cyclooxygenase 2/genetics , Cytokines/biosynthesis , Cytokines/genetics , Drug Evaluation, Preclinical , Ergothioneine/pharmacokinetics , Humans , Microvessels/cytology , Nervous System Diseases/etiology , Neuroprotective Agents/pharmacokinetics , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type III/metabolism , Organic Cation Transport Proteins , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Symporters , Zonula Occludens-1 Protein
17.
Arch Pharm Res ; 43(12): 1259-1275, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33206346

ABSTRACT

Harmine is isolated from the seeds of the medicinal plant, Peganum harmala L., and has been used for thousands of years in the Middle East and China. Harmine has many pharmacological activities including anti-inflammatory, neuroprotective, antidiabetic, and antitumor activities. Moreover, harmine exhibits insecticidal, antiviral, and antibacterial effects. Harmine derivatives exhibit pharmacological effects similar to those of harmine, but with better antitumor activity and low neurotoxicity. Many studies have been conducted on the pharmacological activities of harmine and harmine derivatives. This article reviews the pharmacological effects and associated mechanisms of harmine. In addition, the structure-activity relationship of harmine derivatives has been summarized.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Harmine/pharmacology , Hypoglycemic Agents/pharmacology , Neuroprotective Agents/pharmacology , Peganum , Animals , Anti-Inflammatory Agents/isolation & purification , Anti-Inflammatory Agents/pharmacokinetics , Antineoplastic Agents, Phytogenic/isolation & purification , Antineoplastic Agents, Phytogenic/pharmacokinetics , Harmine/analogs & derivatives , Harmine/isolation & purification , Harmine/pharmacokinetics , Humans , Hypoglycemic Agents/isolation & purification , Hypoglycemic Agents/pharmacokinetics , Molecular Structure , Neuroprotective Agents/isolation & purification , Neuroprotective Agents/pharmacokinetics , Peganum/chemistry , Seeds , Structure-Activity Relationship
18.
Phytomedicine ; 79: 153316, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32942205

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is currently incurable and there is an urgent need to develop new AD drugs. Many studies have revealed the potential neuroprotective effect of Epigallocatechin-3-O-gallate (EGCG), the main antioxidant in green tea, on animal models of AD. However, a systematic review of these reports is lacking. PURPOSE: To assess the effectiveness of EGCG for AD treatment using systematic review and meta-analysis of pre-clinical trials. METHODS: We conducted a systematic search of all available randomized controlled trials (RCTs) performed up to November 2019 in the following electronic databases: ScienceDirect, Web of Science, and PubMed. 17 preclinical studies assessing the effect of EGCG on animal AD models have been identified. Meta-analysis and subgroup analysis was performed to evaluate cognition improvement of various types of AD models. The study quality was assessed using the CAMARADES checklist and the criteria of published studies. RESULTS: Our analysis shows that the methodological quality ranges from 3 to 5, with a median score of 4. According to meta-analysis of random-effects method, EGCG showed a positive effect in AD with shorter escape latency (SMD= -9.24, 95%CI= -12.05 to -6.42) and decreased Aß42 level (SD= -25.74,95%CI= -42.36 to -9.11). Regulation of α-, ß-, γ-secretase activity, inhibition of tau phosphorylation, anti-oxidation, anti-inflammation, anti-apoptosis, and inhibition of AchE activity are reported as the main neuroprotective mechanisms. Though more than 100 clinical trials have been registered on the ClinicalTrials.gov, only one clinical trial has been conducted to test the therapeutic effects of EGCG on the AD progression and cognitive performance. CONCLUSION: Here, we conducted this review to systematically describe the therapeutic potential of EGCG in animal models of AD and hope to provide a more comprehensive assessment of the effects in order to design future clinical trials. Besides, the safety, blood-brain barrier (BBB) penetration and bioavailability issues in conducting clinical trials were also discussed.


Subject(s)
Alzheimer Disease/drug therapy , Catechin/analogs & derivatives , Neuroprotective Agents/pharmacology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/metabolism , Animals , Antioxidants/pharmacology , Blood-Brain Barrier/drug effects , Catechin/pharmacokinetics , Catechin/pharmacology , Cognition/drug effects , Disease Models, Animal , Humans , Neuroprotective Agents/pharmacokinetics , Phosphorylation/drug effects
19.
Biomed Pharmacother ; 129: 110458, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32603893

ABSTRACT

Salidroside (Sal) is a bioactive extract principally from traditional herbal medicine such as Rhodiola rosea L., which has been commonly used for hundreds of years in Asia countries. The excellent neuroprotective capacity of Sal has been illuminated in recent studies. This work focused on the source, pharmacokinetics, safety and anti-ischemic stroke (IS) effect of Sal, especially emphasizing its mechanism of action and BBB permeability. Extensive databases, including Pubmed, Web of science (WOS), Google Scholar and China National Knowledge Infrastructure (CNKI), were applied to obtain relevant online literatures. Sal exerts powerful therapeutic effects on IS in experimental models either in vitro or in vivo due to its neuroprotection, with significantly diminishing infarct size, preventing cerebral edema and improving neurological function. Also, the findings suggest the underlying mechanisms involve anti-oxidation, anti-inflammation and anti-apoptosis by regulating multiple signaling pathways and key molecules, such as NF-κB, TNF-α and PI3K/Akt pathway. In pharmacokinetics, although showing a rapid absorption and elimination, bioavailability of Sal is elevated under some non-physiological conditions. The component and its metabolite (tyrosol) are capable of distributing to brain tissue and the later keeps a higher level of concentration. Moreover, Sal scarcely has obvious toxicity or side effects in a variety of animal experiments and clinical trials, but combination of drugs and perinatal use of medicine should be taken more attentions. Finally, as an active ingredient, not only is Sal isolated from diverse plants with limited yield, but also large batches of the products can be harvested by biological and chemical synthesis. With higher efficacy and better safety profiles, Sal could sever as a promising neuroprotectant for preventing and treating IS. Nevertheless, further investigations are still required to explore the pharmacodynamic and pharmacokinetic properties of Sal in the treatment of IS.


Subject(s)
Brain/drug effects , Glucosides/pharmacokinetics , Ischemic Stroke/drug therapy , Neuroprotective Agents/pharmacokinetics , Phenols/pharmacokinetics , Animals , Blood-Brain Barrier/metabolism , Brain/metabolism , Brain/pathology , Brain/physiopathology , Capillary Permeability , Glucosides/administration & dosage , Glucosides/adverse effects , Humans , Ischemic Stroke/metabolism , Ischemic Stroke/pathology , Ischemic Stroke/physiopathology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/adverse effects , Phenols/administration & dosage , Phenols/adverse effects , Treatment Outcome
20.
Bioorg Med Chem ; 28(13): 115531, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32386953

ABSTRACT

The M3 muscarinic acetylcholine receptor (mAChR) is a member of the family of mAChRs, which are associated with a variety of physiological functions including the contraction of various smooth muscle tissues, stimulation of glandular secretion, and regulation of a range of cholinergic processes in the central nerve system. We report here the discovery and a comprehensive structure--activity relationships (SARs) study of novel positive allosteric modulators (PAMs) of the M3 mAChR through a high throughput screening (HTS) campaign. Compound 9 exhibited potent in vitro PAM activity towards the M3 mAChR and significant enhancement of muscle contraction in a concentration-dependent manner when applied to isolated smooth muscle strips of rat bladder. Compound 9 also showed excellent subtype selectivity over other subtypes of mAChRs including M1, M2, and M4 mAChRs, and moderate selectivity over the M5 mAChR, indicating that compound 9 is an M3-preferring M3/M5 dual PAM. Moreover, compound 9 displayed acceptable pharmacokinetics profiles after oral dosing to rats. These results suggest that compound 9 may be a promising chemical probe for the M3 mAChR for further investigation of its pharmacological function both in vitro and in vivo.


Subject(s)
Muscarinic Agonists/chemical synthesis , Neuroprotective Agents/chemical synthesis , Receptors, Muscarinic/metabolism , Thiazoles/chemical synthesis , Allosteric Regulation , Amines/chemistry , Animals , CHO Cells , Central Nervous System/drug effects , Cricetulus , Drug Evaluation, Preclinical , High-Throughput Screening Assays , Humans , Muscarinic Agonists/pharmacology , Neuroprotective Agents/pharmacokinetics , Piperidines/chemistry , Pyrrolidines/chemistry , Rats , Stereoisomerism , Structure-Activity Relationship , Thiazoles/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL