Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 205
1.
Biomed Pharmacother ; 143: 112175, 2021 Nov.
Article En | MEDLINE | ID: mdl-34649336

Withania somnifera (L.) Dunal (Solanaceae) has been used as a traditional Rasayana herb for a long time. Traditional uses of this plant indicate its ameliorative properties against a plethora of human medical conditions, viz. hypertension, stress, diabetes, asthma, cancer etc. This review presents a comprehensive summary of the geographical distribution, traditional use, phytochemistry, and pharmacological activities of W. somnifera and its active constituents. In addition, it presents a detailed account of its presence as an active constituent in many commercial preparations with curative properties and health benefits. Clinical studies and toxicological considerations of its extracts and constituents are also elucidated. Comparative analysis of relevant in-vitro, in-vivo, and clinical investigations indicated potent bioactivity of W. somnifera extracts and phytochemicals as anti-cancer, anti-inflammatory, apoptotic, immunomodulatory, antimicrobial, anti-diabetic, hepatoprotective, hypoglycaemic, hypolipidemic, cardio-protective and spermatogenic agents. W. somnifera was found to be especially active against many neurological and psychological conditions like Parkinson's disease, Alzheimer's disease, Huntington's disease, ischemic stroke, sleep deprivation, amyotrophic lateral sclerosis, attention deficit hyperactivity disorder, bipolar disorder, anxiety, depression, schizophrenia and obsessive-compulsive disorder. The probable mechanism of action that imparts the pharmacological potential has also been explored. However, in-depth studies are needed on the clinical use of W. somnifera against human diseases. Besides, detailed toxicological analysis is also to be performed for its safe and efficacious use in preclinical and clinical studies and as a health-promoting herb.


Ethnopharmacology , Phytochemicals/pharmacology , Plant Extracts/pharmacology , Withania , Animals , Antiviral Agents/isolation & purification , Antiviral Agents/pharmacology , COVID-19/virology , Humans , Neuroprotective Agents/isolation & purification , Neuroprotective Agents/pharmacology , Neuroprotective Agents/toxicity , Patient Safety , Phytochemicals/isolation & purification , Phytochemicals/toxicity , Plant Extracts/isolation & purification , Plant Extracts/toxicity , Plant Roots , Psychotropic Drugs/isolation & purification , Psychotropic Drugs/pharmacology , Psychotropic Drugs/toxicity , Risk Assessment , SARS-CoV-2/drug effects , SARS-CoV-2/pathogenicity , Withania/chemistry , COVID-19 Drug Treatment
2.
ACS Appl Mater Interfaces ; 13(39): 46213-46224, 2021 Oct 06.
Article En | MEDLINE | ID: mdl-34546708

Acute ischemic stroke has become the major cause of mortality and disability worldwide. Following ischemic stroke, the reperfusion injury is mainly mediated by the burst of reactive oxygen and nitrogen species (RONS). Therefore, blocking the excessive production or removing RONS holds great promise as a potential therapeutic strategy. Herein, we developed a Co-doped Fe3O4 nanozyme that is capable of scavenging H2O2, O2•-, •NO, and ONOO- in vitro and in vivo and provides neuroprotection against ischemic stroke. In vitro experiments showed that pre-incubation with the Co-Fe3O4 nanozyme could prevent neurotoxicity and neuroinflammation induced by H2O2 or lipopolysaccharide, respectively, in HT22 cells. After intravenous administration, the Co-Fe3O4 nanozyme showed no signs of toxicity in peripheral organs of C57BL/6J mice, even after prolonged delivery for 4 weeks. In permanent photothrombotic stroke model and transient middle cerebral artery occlusion stroke model, the Co-Fe3O4 nanozyme specifically accumulated in the infarct rim at 72 h post-stroke and was endocytosed by neurons, astrocytes, microglia, and endothelial cells. Importantly, the Co-Fe3O4 nanozyme delivery reduced the infarct volume in both stroke models. The observation that the Co-Fe3O4 nanozyme was efficacious in two well-characterized ischemic stroke models provides strong evidence that it represents a powerful tool for targeting oxidative and nitrosative stress in the ischemic brain.


Free Radical Scavengers/therapeutic use , Ischemic Stroke/drug therapy , Magnetite Nanoparticles/therapeutic use , Neuroprotective Agents/therapeutic use , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Animals , Catalysis , Cell Line , Cobalt/chemistry , Cobalt/toxicity , Free Radical Scavengers/chemistry , Free Radical Scavengers/toxicity , Lipopolysaccharides , Magnetite Nanoparticles/chemistry , Magnetite Nanoparticles/toxicity , Male , Mice, Inbred C57BL , Neuroinflammatory Diseases/chemically induced , Neuroinflammatory Diseases/drug therapy , Neuroprotection/drug effects , Neuroprotective Agents/chemistry , Neuroprotective Agents/toxicity , Oxidation-Reduction , Reactive Nitrogen Species/chemistry , Reactive Oxygen Species/chemistry
3.
ACS Appl Mater Interfaces ; 13(31): 37746-37760, 2021 Aug 11.
Article En | MEDLINE | ID: mdl-34318658

Mitochondrial dysfunction, which is directly involved in Parkinson's disease (PD), is characterized by the production of reactive oxygen species (ROS) and aberrant energy metabolism. Thus, regulating mitochondrial function might be an effective strategy to treat PD. However, the blood-brain barrier (BBB) presents a significant challenge for the intracerebral delivery of drugs. Here, we synthesized a zeolitic imidazolate framework 8-coated Prussian blue nanocomposite (ZIF-8@PB), which was encapsulated with quercetin (QCT), a natural antioxidant, to treat PD. ZIF-8@PB-QCT exhibited superior near-infrared radiation (NIR) response and penetrated through the BBB to the site of mitochondrial damage guided by the photothermal effect. In the mice model of PD, the QCT released from ZIF-8@PB-QCT significantly increased the adenosine triphosphate levels, reduced the oxidative stress levels, and reversed dopaminergic neuronal damage as well as PD-related behavioral deficits without any damage to the normal tissues. Furthermore, we explored the underlying neuroprotective mechanism of ZIF-8@PB-QCT that was mediated by activating the PI3K/Akt signaling pathway. Thus, combined with noninvasive NIR radiation, the biocompatible ZIF-8@PB-QCT nanocomposite could be used to treat neurodegenerative diseases.


Antioxidants/therapeutic use , Nanocomposites/therapeutic use , Neuroprotective Agents/therapeutic use , Parkinson Disease, Secondary/drug therapy , Quercetin/therapeutic use , Animals , Antioxidants/chemistry , Antioxidants/pharmacokinetics , Antioxidants/toxicity , Blood-Brain Barrier/physiology , Cell Line, Tumor , Drug Liberation , Ferrocyanides/chemistry , Ferrocyanides/radiation effects , Ferrocyanides/therapeutic use , Ferrocyanides/toxicity , Humans , Imidazoles/chemistry , Imidazoles/therapeutic use , Imidazoles/toxicity , Infrared Rays , Male , Mice, Inbred C57BL , Mitochondria/drug effects , Nanocomposites/chemistry , Nanocomposites/radiation effects , Nanocomposites/toxicity , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/toxicity , Oxidative Stress/drug effects , Quercetin/chemistry , Quercetin/pharmacokinetics , Quercetin/toxicity , Rats, Sprague-Dawley , Zeolites/chemistry , Zeolites/therapeutic use , Zeolites/toxicity
4.
Bioorg Med Chem Lett ; 49: 128212, 2021 10 01.
Article En | MEDLINE | ID: mdl-34153471

In this study, a series of multifunctional hybrids (6a-6l) against Alzheimer's disease were designed and obtained by conjugating the pharmacophores of deoxyvasicinone and indole. These analogs of deoxyvasicinone-indole were evaluated as inhibitors of acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE), and as inhibitors of amyloid aggregation (Aß1-42) for treatment of Alzheimer's disease (AD). Subsequently, AChE induced Aß aggregation inhibition test was also performed for selected compounds. Biological activity results demonstrated that compound 6b was the most potent and balanced dual ChEs inhibitor with IC50 values 0.12 µM and 0.15 µM for eeAChE and eqBuChE, respectively. Kinetic analysis and docking study indicated that compound 6b was a mixed-type inhibitor for both AChE and BuChE. Compound 6b also found to be the best inhibitors of self-induced Aß1-42 aggregation with IC50 values of 1.21 µM. Compound 6b also afforded excellent inhibition of AChE-induced Aß1-42 aggregation by 81.1%. Overall, these results indicate that 6b may be considered as lead compound for the development of highly effective anti-AD drugs.


Alzheimer Disease/drug therapy , Cholinesterase Inhibitors/pharmacology , Indoles/pharmacology , Neuroprotective Agents/pharmacology , Quinazolines/pharmacology , Acetylcholinesterase/chemistry , Acetylcholinesterase/metabolism , Amyloid beta-Peptides/metabolism , Animals , Butyrylcholinesterase/chemistry , Butyrylcholinesterase/metabolism , Catalytic Domain , Cell Line, Tumor , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/metabolism , Cholinesterase Inhibitors/toxicity , Drug Design , Electrophorus , Horses , Indoles/chemical synthesis , Indoles/metabolism , Indoles/toxicity , Kinetics , Mice , Molecular Docking Simulation , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/metabolism , Neuroprotective Agents/toxicity , Peptide Fragments/metabolism , Protein Binding , Protein Multimerization/drug effects , Quinazolines/chemical synthesis , Quinazolines/metabolism , Quinazolines/toxicity , Rats
5.
ACS Appl Mater Interfaces ; 13(26): 30373-30382, 2021 Jul 07.
Article En | MEDLINE | ID: mdl-34180234

Phytotherapeutic approaches are of immense value in the treatment of advanced Alzheimer's disease (AD) because of their diverse biological components and potential multitarget mechanisms. In this study, quercetin, a natural neuroprotective flavonoid, was encapsulated in human serum albumin to obtain HSA@QC nanoparticles (HQ NPs) as a natural phyto-antioxidant albumin nanoagent for the treatment of advanced AD. HQ NPs showed excellent antioxidant effects and protected PC12 cells from H2O2-induced oxidative damage. The intranasal administration of HQ NPs in 11-month-old APP/PS1 mice, which represented advanced AD, effectively prevented the loss of body weight, increased survival rates, and significantly reduced oxidative stress, Aß aggregation, neuronal apoptosis, and synaptic damage in the brain. It also ultimately reversed severely impaired cognitive function. In addition to their favorable anti-AD effects, HQ NPs exhibited excellent biosafety and biocompatibility owing to their natural composition and are expected to become an ideal choice for future drug development and clinical applications.


Alzheimer Disease/drug therapy , Drug Carriers/chemistry , Free Radical Scavengers/therapeutic use , Nanoparticles/chemistry , Quercetin/therapeutic use , Serum Albumin, Human/chemistry , Alzheimer Disease/complications , Alzheimer Disease/pathology , Animals , Apoptosis/drug effects , Body Weight/drug effects , Brain/pathology , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Cognitive Dysfunction/pathology , Drug Carriers/chemical synthesis , Drug Carriers/toxicity , Female , Free Radical Scavengers/toxicity , Humans , Mice, Inbred C57BL , Morris Water Maze Test/drug effects , Nanoparticles/toxicity , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/toxicity , Oxidative Stress/drug effects , PC12 Cells , Quercetin/toxicity , Rats , Serum Albumin, Human/toxicity
6.
Neurochem Res ; 46(8): 2066-2078, 2021 Aug.
Article En | MEDLINE | ID: mdl-34019198

Gamma-decanolactone (GD) has been shown to reduce epileptic behavior in different models, inflammatory decreasing, oxidative stress, and genotoxic parameters. This study assessed the GD effect on the pentylenetetrazole (PTZ) model after acute and subchronic treatment. We evaluated the expression of the inflammatory marker cyclooxygenase-2 (COX-2), GluN2B, a subunit of the NMDA glutamate receptor, adenosine A1 receptor, and GD genotoxicity and mutagenicity. Male and female mice were treated with GD (300 mg/kg) for 12 days. On the tenth day, they were tested in the Hot Plate test. On the thirteenth day, all animals received PTZ (90 mg/kg), and epileptic behavior PTZ-induced was observed for 30 min. Pregabalin (PGB) (30 mg/kg) was used as a positive control. Samples of the hippocampus and blood were collected for Western Blotting analyses and Comet Assay and bone marrow to the Micronucleus test. Only the acute treatment of GD reduced the seizure occurrence and increased the latency to the first stage 3 seizures. Males treated with GD for 12 days demonstrated a significant increase in the expression of the GluN2B receptor and a decrease in the COX-2 expression. Acute and subchronic treatment with GD and PGB reduced the DNA damage produced by PTZ in males and females. There is no increase in the micronucleus frequency in bone marrow after subchronic treatment. This study suggests that GD, after 12 days, could not reduce PTZ-induced seizures, but it has been shown to protect against DNA damage, reduce COX-2 and increase GluN2B expression.


Cyclooxygenase 2/metabolism , Lactones/therapeutic use , Neuroprotective Agents/therapeutic use , Receptor, Adenosine A1/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Seizures/drug therapy , Animals , Body Weight/drug effects , DNA Damage/drug effects , Female , Lactones/toxicity , Male , Mice , Neuroprotective Agents/toxicity , Pentylenetetrazole , Seizures/chemically induced , Seizures/metabolism
7.
Bioorg Med Chem Lett ; 40: 127929, 2021 05 15.
Article En | MEDLINE | ID: mdl-33705903

A small set of trehalose-centered putative autophagy inducers was rationally designed and synthesized, with the aim to identify more potent and bioavailable autophagy inducers than free trehalose, and to acquire information about their molecular mechanism of action. Several robust, high yield routes to key trehalose intermediates and small molecule prodrugs (2-5), putative probes (6-10) and inorganic nanovectors (12a - thiol-PEG-triazole-trehalose constructs 11) were successfully executed, and compounds were tested for their autophagy-inducing properties. While small molecules 2-11 showed no pro-autophagic behavior at sub-millimolar concentrations, trehalose-bearing PEG-AuNPs 12a caused measurable autophagy induction at an estimated 40 µM trehalose concentration without any significant toxicity at the same concentration.


Autophagy/drug effects , Neuroprotective Agents/pharmacology , Trehalose/analogs & derivatives , Trehalose/pharmacology , Drug Design , Gold/chemistry , Gold/toxicity , HeLa Cells , Humans , Metal Nanoparticles/chemistry , Metal Nanoparticles/toxicity , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/toxicity , Polyethylene Glycols/chemistry , Polyethylene Glycols/toxicity , Trehalose/toxicity
8.
Eur J Med Chem ; 215: 113278, 2021 Apr 05.
Article En | MEDLINE | ID: mdl-33662757

Alzheimer's disease (AD) is an age-related multifactorial neurodegenerative disorder characterized by severe central cholinergic neuronal loss, gradually contributing to cognitive dysfunction and impaired motor activity, resulting in the brain's cell death at the later stages of AD. Although the etiology of AD is not well understood, however, several factors such as oxidative stress, deposition of amyloid-ß (Aß) peptides to form Aß plaques, intraneuronal accumulation of hyperphosphorylated tau protein, and low level of acetylcholine are thought to play a major role in the pathogenesis of AD. There is practically no drug for AD treatment that can address the basic factors responsible for the neurodegeneration and slow down the disease progression. The currently available therapies for AD in the market focus on providing only symptomatic relief without addressing the aforesaid basic factors responsible for the neurodegeneration. Ferulic acid (FA) is a phenol derivative from natural sources and serves as a potential pharmacophore that exerts multiple pharmacological properties such as antioxidant, neuroprotection, Aß aggregation modulation, and anti-inflammatory. Several FA based hybrid analogs are under investigation as a multi-target directed ligand (MTDLs) to develop novel hybrid compounds for the treatment of AD. In the present review article, we are focused on the critical pathogenic factors responsible for the onset of AD followed by the developments of FA pharmacophore-based hybrids compounds as a novel multifunctional therapeutic agent to address the limitations associated with available treatment for AD. The rationale behind the development of these compounds and their pharmacological activities in particular to their ChE inhibition (ChEI), neuroprotection, antioxidant property, Aß aggregation modulation, and metal chelation ability, are discussed in detail. We have also discussed the discovery of caffeic and cinnamic acids based MTDLs for AD. This review paper provides an in-depth insight into the research progress and current status of these novel therapeutics in AD and prospects for developing a druggable molecule with desired pharmacological affinity and reduced toxicity for the management of AD.


Alzheimer Disease/drug therapy , Coumaric Acids/therapeutic use , Neuroprotective Agents/therapeutic use , Amyloid beta-Peptides/metabolism , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Antioxidants/toxicity , Cell Line, Tumor , Coumaric Acids/pharmacology , Coumaric Acids/toxicity , Humans , Neuroprotective Agents/pharmacology , Neuroprotective Agents/toxicity , Oxidative Stress/drug effects , Protein Multimerization/drug effects
9.
Mol Neurobiol ; 58(8): 3641-3652, 2021 Aug.
Article En | MEDLINE | ID: mdl-33788167

Repurposing PARP-1 inhibitors (PARPi) for non-oncological applications offers an attractive therapeutic strategy for pathological conditions characterized by PARP-1 hyperactivity. In the context of Parkinson's disease (PD), PARP-1 hyperactivity has been linked to neuronal death and disease progression. From a therapy perspective, the evaluation of PARPi as neuroprotective agents may offer a new therapeutic alternative for neurodegenerative disorders. An ideal PARPi needs to inhibit PARP-1 hyperactivity while also limiting downstream DNA damage and cellular toxicity-an effect that is attractive in cancer but far from ideal in neurological disease applications. Consequently, in this study, we set out to evaluate the neuroprotective properties of a previously reported low-toxicity PARPi (10e) using in vitro neuronal models of PD. 10e is a structural analogue of FDA-approved PARPi olaparib, with high PARP-1 affinity and selectivity. Our studies revealed that 10e protects neuronal cells from oxidative stress and DNA damage. In addition, 10e exhibits neuroprotective properties against α-synuclein pre-formed fibrils (αSyn PFF) mediated effects, including reduction in the levels of phosphorylated αSyn and protection against abnormal changes in NAD+ levels. Our in vitro studies with 10e provide support for repurposing high-affinity and low-toxicity PARPi for neurological applications and lay the groundwork for long-term therapeutic studies in animal models of PD.


Neuroprotective Agents/pharmacology , Parkinson Disease/prevention & control , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Cell Line, Tumor , Dose-Response Relationship, Drug , Gene Knockout Techniques/methods , Humans , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/toxicity , Parkinson Disease/metabolism , Phthalazines/pharmacology , Phthalazines/therapeutic use , Phthalazines/toxicity , Piperazines/pharmacology , Piperazines/therapeutic use , Piperazines/toxicity , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors/toxicity
10.
J Ethnopharmacol ; 272: 113923, 2021 May 23.
Article En | MEDLINE | ID: mdl-33617968

ETHNOPHARMACOLOGICAL RELEVANCE: Tanshinone-Ⅰ (TSNⅠ), a member of the mainly active components of Salvia miltiorrhiza Bunge (Dan Shen), which is widely used for the treatment for modern clinical diseases including cardiovascular and cerebrovascular diseases, has been reported to show the properties of anti-oxidation, anti-inflammation, neuroprotection and other pharmacological actions. However, whether TSNⅠ can improve neuron survival and neurological function against transient focal cerebral ischemia (tMCAO) in mice is still a blank field. AIM OF THE STUDY: This study aims to investigate the neuroprotective effects of TSNⅠ on ischemic stroke (IS) induced by tMCAO in mice and explore the potential mechanism of TSNⅠ against IS by combining network pharmacology approach and experimental verification. MATERIALS AND METHODS: In this study, the pivotal candidate targets of TSNⅠ against IS were screened by network pharmacology firstly. Enrichment analysis and molecular docking of those targets were performed to identify the possible mechanism of TSNⅠ against IS. Afterwards, experiments were carried out to further verify the mechanism of TSNⅠ against IS. The infarct volume and neurological deficit were evaluated by 2, 3, 5-triphenyl tetrazolium chloride (TTC) staining and Longa respectively. Immunohistochemistry was used to observe neuronal death in the hippocampus and cortical regions by detecting the change of NeuN. The predicting pathways of signaling-related proteins were assessed by Western blot in vitro and in vivo experiments. RESULTS: In vivo, TSNⅠ was found to dose-dependently decrease mice's cerebral infarct volume induced by tMCAO. In vitro, pretreatment with TSNⅠ could increase cell viability of HT-22 cell following oxygen-glucose deprivation (OGD/R). Moreover, the results showed that 125 candidate targets were identified, Protein kinase B (AKT) signaling pathway was significantly enriched by Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis and mitogen-activated protein kinases 1 (MAPK1) and AKT1 could be bound to TSNⅠ more firmly by molecular docking analysis, which implies that TSNⅠ may play a role in neuroprotection through activating AKT and MAPK signaling pathways. Meanwhile, TSNⅠ was confirmed to significantly protect neurons from injury induced by IS through activating AKT and MAPK signaling pathways. CONCLUSION: In conclusion, our study clarifies that the mechanism of TSNⅠ against IS might be related to AKT and MAPK signaling pathways, which may provide the basic evidence for further development and utilization of TSNⅠ.


Abietanes/pharmacology , Ischemic Stroke/prevention & control , Neuroprotective Agents/pharmacology , Abietanes/therapeutic use , Abietanes/toxicity , Animals , Brain Ischemia/complications , Cell Line , Cell Survival/drug effects , Disease Models, Animal , Glycogen Synthase Kinase 3 beta/metabolism , Hippocampus/metabolism , Ischemic Stroke/etiology , Ischemic Stroke/genetics , MAP Kinase Signaling System/drug effects , Male , Mice, Inbred ICR , Mitogen-Activated Protein Kinase Kinases/metabolism , Molecular Docking Simulation , Neurons/drug effects , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/toxicity , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Phosphoinositide-3 Kinase Inhibitors/therapeutic use , Protein Interaction Maps , Proto-Oncogene Proteins c-akt/metabolism , beta Catenin/metabolism , raf Kinases/metabolism
11.
J Inorg Biochem ; 217: 111393, 2021 04.
Article En | MEDLINE | ID: mdl-33610031

Alzheimer's disease (AD) is a neurodegenerative disorder of the central nervous system. The main pathophysiological mechanisms involve cholinergic neurotransmission, beta-amyloid (Αß) and Tau proteins, several metal ions and oxidative stress, among others. Current drugs offer only relief of symptoms and not a cure of AD. Accumulating evidence suggests that multifunctional compounds, targeting multiple pathophysiological mechanisms, may have a great potential for the treatment of AD. In this study, we report on the synthesis and physicochemical characterization of four quinoline-based metal chelators and their respective copper(II) complexes. Most compounds were non-toxic at concentrations ≤5 µM. In neuroprotection studies employing undifferentiated and differentiated SH-SY5Y cells, the metal chelator N2,N6-di(quinolin-8-yl)pyridine-2,6-dicarboxamide (H2dqpyca) appeared to exert significant neuroprotection against both, Aß peptide- and H2O2-induced toxicities. The copper(II) complex [CuII(H2bqch)Cl2].3H2O (H2bqch = N,N'-Bis(8-quinolyl)cyclohexane-1,2-diamine) also protected against H2O2-induced toxicity, with a half-maximal effective concentration of 80 nM. Molecular docking simulations, using the crystal structure of the acetylcholinesterase (AChE)-rivastigmine complex as a template, indicated a strong interaction of the metal chelator H2dqpyca, followed by H2bqch, with both the peripheral anionic site and the catalytic active site of AChE. In conclusion, the sufficient neuroprotection provided by the metal chelator H2dqpyca and the copper(II) complex [CuII(H2bqch)Cl2].3H2O along with the evidence for interaction between H2dqpyca and AChE, indicate that these compounds have the potential and should be further investigated in the framework of preclinical studies employing animal models of AD as candidate multifunctional lead compounds for the treatment of the disease.


Alzheimer Disease/drug therapy , Coordination Complexes/pharmacology , Neuroprotective Agents/pharmacology , Quinolines/pharmacology , Acetylcholinesterase/chemistry , Acetylcholinesterase/metabolism , Animals , CHO Cells , Catalytic Domain , Cell Line, Tumor , Coordination Complexes/chemical synthesis , Coordination Complexes/metabolism , Coordination Complexes/toxicity , Copper/chemistry , Cricetulus , Humans , Hydrogen Peroxide/toxicity , Ligands , Molecular Docking Simulation , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/metabolism , Neuroprotective Agents/toxicity , Protein Binding , Quinolines/chemical synthesis , Quinolines/metabolism , Quinolines/toxicity
12.
Pak J Pharm Sci ; 34(6): 2197-2203, 2021 Nov.
Article En | MEDLINE | ID: mdl-35034881

Naproxen, known as the most potent non-steroid anti-inflammatory drugs, is vitally crucial in the treatment of neurodegenerative diseases. However, due to the poor brain penetration ability, it causes serious adverse effects with the therapeutic doses. Predictably, these unfavorable factors have hindered its further application. In this study, a novel brain-targeting conjugate, Nap-Pro, was designed and synthesized. The chemical stability and metabolic stability of this conjugate were determined in phosphate buffer, blood serum and brain homogenate, respectively. The cytotoxicity of Nap-Pro was evaluated in b End. 3 cells. In addition, the brain targeting capacity of Nap-Pro was also investigated in vivo. Importantly, Nap-Pro showed excellent capacity to cross the brain-blood barrier (BBB), suggesting probenecid was a super carrier that enhanced the delivery of drugs into brain. Collectively, the probenecid modification was a promising strategy to develop the novel drug delivery systems for brain targeting.


Anti-Inflammatory Agents, Non-Steroidal/metabolism , Brain/metabolism , Neuroprotective Agents/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/toxicity , Biological Transport , Blood-Brain Barrier/metabolism , Cell Line , Drug Compounding , Drug Stability , Mice , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/toxicity , Permeability , Tissue Distribution
13.
Curr Comput Aided Drug Des ; 17(3): 351-359, 2021.
Article En | MEDLINE | ID: mdl-32303174

AIM: To screen the zinc database for structurally similar molecules to compound 23 that targets DJ1 for use as a neuroprotective agent for Parkinson's disease. BACKGROUND: Parkinson's disease (PD) is the second most common chronic neurodegenerative disorder characterized by progressive loss of dopaminergic neurons of the substantia nigra. To date, several proteins account for the recessive familial PD-forms, namely, Parkin, PINK-1, DJ-1, SNCA, PARK2, and LRRK2 Genes. DJ1 is one of the important central points that may be targeted for PD therapy. Recently, Compound 23 has been observed to exert the neuroprotective effect against neurodegeneration in a PD model, but due to its toxic substructure, the hunt for better nontoxic compounds continues. OBJECTIVE: The overall objective of our work is to apply in silico approaches to screen structure similar compounds that interact potentially with DJ1 and may serve as a good therapeutic molecule for PD. METHODS: Initial data mining was done from the zinc database and then screened compounds were additionally screened with toxicity checker, carcinopred, ADMET analysis and docking analysis. RESULTS: The basic screening of database for structurally similar chemicals to compound 23 resulted in 50 compounds, which were further screened to twenty-three and finally seven compounds have been screened based on the toxicity and carcinopred test. Later, the seven compounds were docked and analysed for docking efficiency with DJ1. Our result of molecular docking and molecular simulation analysis highlights Molecule 42(SS2) to exhibit best binding affinity against DJ-1 protein target and can be proposed to be used as a therapeutic agent to modulate neurodegenerative proteins. CONCLUSION: Therefore, we conclude the discovery of novel, non-toxic, non-carcinogenic, ADMET investigated, capable of crossing BB barrier but structurally similar compounds to Compound-23, specifically molecule 42(SS2) and potentially molecule 34(SS1) to be used as neuroprotective agents for Parkinson's disease.


Antiparkinson Agents/pharmacology , Neuroprotective Agents/pharmacology , Parkinson Disease/drug therapy , Protein Deglycase DJ-1/metabolism , Antiparkinson Agents/pharmacokinetics , Antiparkinson Agents/toxicity , Computer Simulation , Dopaminergic Neurons/pathology , Drug Discovery/methods , Humans , Molecular Docking Simulation , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/toxicity , Parkinson Disease/physiopathology
14.
Int J Toxicol ; 40(1): 4-14, 2021.
Article En | MEDLINE | ID: mdl-33131343

Glial cell line-derived neurotrophic factor (GDNF) is a potent neuroprotective biologic in Parkinson's disease models. Adeno-associated viral vector serotype 2 (AAV2)-human GDNF safety was assessed in rats treated with a single intracerebral dose of vehicle, 6.8 × 108, 6.8 × 109, or 5.2 × 1010 vector genomes (vg)/dose followed by interim sacrifices on day 7, 31, 90, and 376. There were no treatment-related effects observed on food consumption, body weight, hematology, clinical chemistry, coagulation parameters, neurobehavioral parameters, organ weights, or serum GDNF and anti-GDNF antibody levels. Increased serum anti-AAV2 neutralizing antibody titers were observed in the 5.2 × 1010 vg/dose group. Histopathological lesions were observed at the injection site in the 6.8 × 109 vg/dose (day 7) and 5.2 × 1010 vg/dose groups (days 7 and 31) and consisted of gliosis, mononuclear perivascular cuffing, intranuclear inclusion bodies, and/or apoptosis on day 7 and mononuclear perivascular cuffing on day 31. GDNF immunostaining was observed in the injection site in all dose groups through day 376 indicating no detectable impacts of anti-AAV2 neutralizing antibody. There was no evidence of increased expression of calcitonin gene-related peptide or Swann cell hyperplasia in the cervical and lumbar spinal cord or medulla oblongata at the 5.2 × 1010 vg/dose level indicating lack of hyperplastic effects. In conclusion, no systemic toxicity was observed, and the local toxicity observed at the injection site appeared to be reversible demonstrating a promising safety profile of intracerebral AAV2-GDNF delivery. Furthermore, an intracerebral dose of 6.8 × 108 AAV2-GDNF vg/dose was considered to be a no observed adverse effect level in rats.


Glial Cell Line-Derived Neurotrophic Factor/administration & dosage , Glial Cell Line-Derived Neurotrophic Factor/toxicity , Glial Cell Line-Derived Neurotrophic Factor/therapeutic use , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/toxicity , Neuroprotective Agents/therapeutic use , Parkinson Disease/drug therapy , Animals , Disease Models, Animal , Female , Humans , Male , Rats , Rats, Sprague-Dawley
15.
Int J Biol Macromol ; 166: 1352-1364, 2021 Jan 01.
Article En | MEDLINE | ID: mdl-33161083

In recent years, butyrylcholinesterase (BChE) has gradually gained worldwide interests as a novel target for treating Alzheimer's disease (AD). Here, two pharmacophore models were generated using Schrödinger suite and used to virtually screen ChemDiv database, from which three hits were obtained. Among them, 2513-4169 displayed the highest inhibitory activity and selectivity against BChE (eeAChE IC50 > 10 µM, eqBChE IC50 = 3.73 ± 1.90 µM). Molecular dynamic (MD) simulation validated the binding pattern of 2513-4169 in BChE, and it could form a various of receptor-ligand interactions with adjacent residues. In vitro cytotoxicity assay proved the safety of 2513-4169 on diverse neural cell lines. Moreover, the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium (MTT) assay performed on SH-SY5Y cells proved the neuroprotective effect of 2513-4169 against toxic Aß1-42. In vivo behavioral study further confirmed the great efficacy of 2513-4169 on reversing Aß1-42-induced cognitive impairment of mice and clearing the toxic Aß1-42 in brains. Moreover, 2513-4169 was proved to be able to cross blood-brain barrier (BBB) through a parallel artificial membrane permeation assay of BBB (PAMPA-BBB). Taken together, 2513-4169 is a promising lead compound for future optimization to discover anti-AD treating agents.


Butyrylcholinesterase/chemistry , Cholinesterase Inhibitors/chemical synthesis , Neuroprotective Agents/chemical synthesis , Animals , Butyrylcholinesterase/metabolism , Cell Line, Tumor , Cholinesterase Inhibitors/pharmacology , Cholinesterase Inhibitors/toxicity , Cognition/drug effects , Drug Discovery/methods , Humans , Male , Mice , Mice, Inbred ICR , Molecular Docking Simulation/methods , Neurons/drug effects , Neuroprotective Agents/pharmacology , Neuroprotective Agents/toxicity , Quantitative Structure-Activity Relationship
16.
J Med Chem ; 63(17): 10030-10044, 2020 09 10.
Article En | MEDLINE | ID: mdl-32787113

To discover novel BChE inhibitors, a hierarchical virtual screening protocol followed by biochemical evaluation was applied. The most potent compound 8012-9656 (eqBChE IC50 = 0.18 ± 0.03 µM, hBChE IC50 = 0.32 ± 0.07 µM) was purchased and synthesized. It inhibited BChE in a noncompetitive manner and could occupy the binding pocket forming diverse interactions with the target. 8012-9656 was proven to be safe in vivo and in vitro and showed comparable performance in ameliorating the scopolamine-induced cognition impairment to tacrine. Additionally, treatment with 8012-9656 could almost entirely recover the Aß1-42 (icv)-impaired cognitive function to the normal level and showed better behavioral performance than donepezil. The evaluation of the Aß1-42 total amount confirmed its anti-amyloidogenic profile. Moreover, 8012-9656 possessed blood-brain barrier (BBB) penetrating ability, a long T1/2, and low intrinsic clearance. Hence, the novel potential BChE inhibitor 8012-9656 can be considered as a promising lead compound for further investigation of anti-AD agents.


Aminoquinolines/pharmacology , Benzimidazoles/pharmacology , Butyrylcholinesterase/metabolism , Cholinesterase Inhibitors/pharmacology , Neuroprotective Agents/pharmacology , Aminoquinolines/chemical synthesis , Aminoquinolines/metabolism , Aminoquinolines/toxicity , Animals , Benzimidazoles/chemical synthesis , Benzimidazoles/metabolism , Benzimidazoles/toxicity , Cell Line, Tumor , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/metabolism , Cholinesterase Inhibitors/toxicity , Drug Discovery , Drug Evaluation, Preclinical , Female , Humans , Male , Mice, Inbred ICR , Microsomes, Liver/metabolism , Molecular Docking Simulation , Molecular Dynamics Simulation , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/metabolism , Neuroprotective Agents/toxicity , Protein Binding , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacology , Small Molecule Libraries/toxicity
17.
Biomed Pharmacother ; 128: 110285, 2020 Aug.
Article En | MEDLINE | ID: mdl-32485569

Minocycline has been proposed as a neuroprotective agent with pleiotropic effects on several experimental models of neurodegenerative diseases, including microglial inhibition. However, although most studies have focused on the central actions of minocycline in affecting microglial functions, other central nervous system (CNS) cell types may also be affected by this drug toxicity. Hence, considering that glial cells play a pivotal role on CNS physiology and are the main responsible for neuronal integrity, a comprehensive investigation on the effects of minocycline treatment on human glial cells is mandatory before translational studies to afford neuroprotection in humans. Therefore, we explored the cytotoxic and genotoxic effects of minocycline at different concentrations in glial cells using an in vitro model. To achieve this, U87 glial cell were exposed to 10-50 µg/mL for 24 h. After exposure, cell viability, general metabolic status and genotoxic assays were performed. No changes were observed in cell viability, however, the general metabolic status decreased over 20 µg/mL. In addition, although no chromossome aberrations were observed, evidences of genotoxicity, such as increase on micronucleus, buds and bridges, were observed from 10 µg/mL. These results suggest that minocycline may induce genotoxic effects even at concentrations considered previously safe and should be used with caution in translational studies.


Micronuclei, Chromosome-Defective/chemically induced , Minocycline/toxicity , Neuroglia/drug effects , Neuroprotective Agents/toxicity , Cell Line , Cell Survival/drug effects , Chromosome Aberrations/chemically induced , Comet Assay , DNA Damage , Dose-Response Relationship, Drug , Humans , Micronucleus Tests , Neuroglia/metabolism , Neuroglia/pathology , Risk Assessment
18.
Eur J Med Chem ; 197: 112282, 2020 Jul 01.
Article En | MEDLINE | ID: mdl-32380361

Compounds capable of interacting with single or multiple targets involved in Alzheimer's disease (AD) pathogenesis are potential anti-Alzheimer's agents. In our aim to develop new anti-Alzheimer's agents, a series of 36 new N-alkylpiperidine carbamates was designed, synthesized and evaluated for the inhibition of cholinesterases [acetylcholinesterase (AChE) and butyrylcholinesterase (BChE)] and monoamine oxidases [monoamine oxidase A (MAO-A and monoamine oxidase B (MAO-B)]. Four compounds are very promising: multiple AChE (IC50 = 7.31 µM), BChE (IC50 = 0.56 µM) and MAO-B (IC50 = 26.1 µM) inhibitor 10, dual AChE (IC50 = 2.25 µM) and BChE (IC50 = 0.81 µM) inhibitor 22, selective BChE (IC50 = 0.06 µM) inhibitor 13, and selective MAO-B (IC50 = 0.18 µM) inhibitor 16. Results of enzyme kinetics experiments showed that despite the carbamate group in the structure, compounds 10, 13, and 22 are reversible and non-time-dependent inhibitors of AChE and/or BChE. The resolved crystal structure of the complex of BChE with compound 13 confirmed the non-covalent mechanism of inhibition. Additionally, N-propargylpiperidine 16 is an irreversible and time-dependent inhibitor of MAO-B, while N-benzylpiperidine 10 is reversible. Additionally, compounds 10, 13, 16, and 22 should be able to cross the blood-brain barrier and are not cytotoxic to human neuronal-like SH-SY5Y and liver HepG2 cells. Finally, compounds 10 and 16 also prevent amyloid ß1-42 (Aß1-42)-induced neuronal cell death. The neuroprotective effects of compound 16 could be the result of its Aß1-42 anti-aggregation effects.


Carbamates/pharmacology , Cholinesterase Inhibitors/pharmacology , Monoamine Oxidase Inhibitors/pharmacology , Neuroprotective Agents/pharmacology , Piperidines/pharmacology , Acetylcholinesterase/metabolism , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Butyrylcholinesterase/metabolism , Carbamates/chemical synthesis , Carbamates/toxicity , Cell Line, Tumor , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/toxicity , Drug Design , Humans , Molecular Structure , Monoamine Oxidase Inhibitors/chemical synthesis , Monoamine Oxidase Inhibitors/toxicity , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/toxicity , Peptide Fragments/metabolism , Piperidines/chemical synthesis , Piperidines/toxicity , Protein Multimerization/drug effects , Structure-Activity Relationship
19.
Neurotoxicology ; 79: 200-208, 2020 07.
Article En | MEDLINE | ID: mdl-32360092

In recent years, piperine has attracted much attention due to its various biological effects as a neuroprotective agent. Therefore, clarification of the possible side effects of piperine is important to identify its potential pharmacological action. Thus, the effects of piperine on the long-term plasticity of perforant pathway to dentate gyrus synapses were studied in hippocampus of an animal model of Alzheimer's disease (AD). Adult male rats were injected with intracerebroventricular (ICV) streptozotocin (STZ) bilaterally, on days 1 and 3 (3 mg/kg). The STZ-injected rats were treated with different doses of piperine for 4 weeks before being used in behavioral, electrophysiological and histopathological experiments. The passive-avoidance test was conducted on all animals in order to determine the cognitive performance. Rats were placed in a stereotaxic frame to implant a recording electrode in the hippocampal dentate gyrus and a stimulating electrode in the perforant path. Additionally, we assessed the density of survived neurons stained by cresyl violet. In this study, chronic administration of piperine low dose improved the ICV-STZ induced learning and long-term potentiation (LTP) impairments with no significant effect on baseline synaptic activity. In contrast, remarkable learning and long-term plasticity impairments were observed in rats treated by high dose of piperine in comparison to the other groups. Interestingly, this impaired hippocampal LTP was accompanied by an obvious alteration in baseline activity and significantly decreased neuronal numbers within the hippocampus. Therefore, our data provides a new understanding of the piperine supplementation effects on hippocampal electrophysiological profile although the consequences may be either beneficial or detrimental.


Alkaloids/toxicity , Behavior, Animal/drug effects , Benzodioxoles/toxicity , Hippocampus/drug effects , Memory Disorders/chemically induced , Memory/drug effects , Neuronal Plasticity/drug effects , Neuroprotective Agents/toxicity , Neurotoxicity Syndromes/etiology , Piperidines/toxicity , Polyunsaturated Alkamides/toxicity , Animals , Disease Models, Animal , Hippocampus/physiopathology , Long-Term Potentiation/drug effects , Male , Memory Disorders/physiopathology , Memory Disorders/psychology , Neurotoxicity Syndromes/physiopathology , Neurotoxicity Syndromes/psychology , Rats, Wistar , Streptozocin , Time Factors
20.
Eur J Pharmacol ; 879: 173124, 2020 Jul 15.
Article En | MEDLINE | ID: mdl-32339515

As a naturally occurring furanocoumarin, the medicinal value of imperatorin has been studied more and more. We hope to provide useful information for the further development of imperatorin by analyzing the literature of imperatorin in recent years. By collating the literature on the pharmacology of imperatorin, we found that the pharmacological activity of imperatorin is wide and imperatorin can be used for anti-cancer, neuroprotection, anti-inflammatory, anti-hypertension and antibacterial. In addition, we found that some researchers confirmed the toxicity of imperatorin. Pharmacokinetic studies demonstrated that oxidation metabolism is the main metabolic pathways of imperatorin. At present, the shortcomings of research on imperatorin mainly include: most pharmacological studies are concentrated in vitro, lacking enough in vivo experimental data; more and more studies showed that imperatorin has synergistic effect with other drugs in anticancer and other aspects, but lacking the detailed explanation of the mechanism of the synergistic effect; imperatorin has side effect, but it lacks enough experimental conclusions. Based on the above defects, we believe that more in vivo experiments of imperatorin should be carried out in the future; future research need to explore synergistic mechanisms of imperatorin with other drugs, especially in anticancer; the dose affects both the pharmacological activity and the side effect of imperatorin. The relationship between the dose and the two aspects need to be further studied in order to reduce the side effect. In addition, through structural modification of imperatorin, it is possible to improve the treatment effect and reduce side effect.


Anti-Inflammatory Agents , Antineoplastic Agents, Phytogenic , Furocoumarins , Neuroprotective Agents , Animals , Anti-Inflammatory Agents/pharmacokinetics , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/toxicity , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/toxicity , Furocoumarins/pharmacokinetics , Furocoumarins/pharmacology , Furocoumarins/toxicity , Humans , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/pharmacology , Neuroprotective Agents/toxicity
...