Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 232
1.
J Enzyme Inhib Med Chem ; 35(1): 1206-1214, 2020 Dec.
Article En | MEDLINE | ID: mdl-32401069

Carbonic anhydrases (CAs, EC 4.2.1.1) are metalloenzymes which use CO2 as substrate, catalysing its interconversion to bicarbonate and a proton. In humans 15 CAs are expressed, 12 of which are catalytically active: the cytosolic CA I-III, VII, XIII, the membrane-bound CA IV, the mitochondrial CA VA and VB, the secreted CA VI, and the transmembrane CA IX, XII, XIV. Nine isoforms are present in the mammalian brain. Evidence supporting that CA inhibitors impair memory in humans has come from studies on topiramate and acetazolamide during acute high-altitude exposure. In contrast, administration of CA activators in animal models enhances memory and learning. Here we review the involvement of selective CA inhibition/activation in cognition-related disorders. CAs may represent a crucial family of new targets for improving cognition as well as in therapeutic areas, such as phobias, obsessive-compulsive disorder, generalised anxiety, and post-traumatic stress disorders, for which few efficient therapies are available.


Carbonic Anhydrases/metabolism , Cognition Disorders/drug therapy , Emotions , Brain/enzymology , Cognition Disorders/enzymology , Cognition Disorders/metabolism , Humans , Structure-Activity Relationship
2.
Future Med Chem ; 11(9): 993-1014, 2019 05.
Article En | MEDLINE | ID: mdl-31141392

The epigenetic control of gene expression could be affected by addition and/or removal of post-translational modifications such as phosphorylation, acetylation and methylation of histone proteins, as well as methylation of DNA (5-methylation on cytosines). Misregulation of these modifications is associated with altered gene expression, resulting in various disease conditions. G9a belongs to the protein lysine methyltransferases that specifically methylates the K9 residue of histone H3, leading to suppression of several tumor suppressor genes. In this review, G9a functions, role in various diseases, structural biology aspects for inhibitor design, structure-activity relationship among the reported inhibitors are discussed which could aid in the design and development of potent G9a inhibitors for cancer treatment in the future.


Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cognition Disorders/drug therapy , Cognition Disorders/enzymology , Histocompatibility Antigens/chemistry , Histocompatibility Antigens/metabolism , Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/metabolism , Humans , Models, Molecular , Neoplasms/drug therapy , Neoplasms/enzymology , Structure-Activity Relationship
3.
Cardiovasc Res ; 114(12): 1595-1604, 2018 10 01.
Article En | MEDLINE | ID: mdl-29931148

Anti-PCSK9 (proprotein convertase subtilisin kexin 9) monoclonal antibodies (Mab) are novel, potent lipid-lowering drugs. They demonstrated to improve the lipid profile in high cardiovascular risk patients. Anti-PCSK9 Mab inhibit the targeted low-density lipoprotein (LDL)-receptor degradation induced by PCSK9 protein and are able to reduce LDL cholesterol (LDL-C) levels on top of conventional lipid-lowering therapy. Though these drugs proved to be very safe in the short-term, little is known about the possible long-term effects, due to the short period of their marketing. The genetic low cholesterol syndromes (LCS) represent the natural models of the lipid-lowering anti-PCSK9 therapy, and a valuable opportunity to predict the long-term effects of these drugs. By looking at the clinical features of such models, we could be able to foresee possible drug-induced side effects. In the present review, the correspondences and discordances between the side effects of anti-PCSK9 therapy and the corresponding LCS models will be examined in the attempt to forecast possible long-term consequences of these novel lipid-lowering agents.


Antibodies, Monoclonal/therapeutic use , Anticholesteremic Agents/therapeutic use , Cholesterol, LDL/blood , Dyslipidemias/drug therapy , PCSK9 Inhibitors , Serine Proteinase Inhibitors/therapeutic use , Animals , Antibodies, Monoclonal/adverse effects , Anticholesteremic Agents/adverse effects , Biomarkers/blood , Cognition Disorders/blood , Cognition Disorders/enzymology , Cognition Disorders/epidemiology , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/epidemiology , Dyslipidemias/blood , Dyslipidemias/enzymology , Dyslipidemias/epidemiology , Fatty Liver/blood , Fatty Liver/enzymology , Fatty Liver/epidemiology , Humans , Proprotein Convertase 9/immunology , Proprotein Convertase 9/metabolism , Risk Assessment , Risk Factors , Serine Proteinase Inhibitors/adverse effects , Time Factors , Treatment Outcome
4.
Prim Care Diabetes ; 12(1): 51-58, 2018 02.
Article En | MEDLINE | ID: mdl-28732654

We performed a PRISMA systematic review of incretin-based therapies and effects on neurocognitive function in humans. There was observational evidence to support dipeptidyl peptidase-IV inhibitors in improving cognition, whilst glucagon-like peptide-1 had positive effects on cerebral glucose metabolism. Powered clinical trials are now needed in patients with- and without diabetes.


Brain/drug effects , Cognition Disorders/drug therapy , Cognition/drug effects , Dipeptidyl Peptidase 4/metabolism , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Glucagon-Like Peptide-1 Receptor/agonists , Glucose/metabolism , Incretins/therapeutic use , Adult , Aged , Brain/metabolism , Brain/physiopathology , Cognition Disorders/enzymology , Cognition Disorders/physiopathology , Cognition Disorders/psychology , Dipeptidyl-Peptidase IV Inhibitors/adverse effects , Female , Glucagon-Like Peptide-1 Receptor/metabolism , Humans , Incretins/adverse effects , Male , Middle Aged , Treatment Outcome
5.
Mol Neurobiol ; 55(5): 4136-4159, 2018 May.
Article En | MEDLINE | ID: mdl-28597397

Accumulating evidence has suggested that repeated treatment with methamphetamine (MA) resulted in cognitive impairments. Importantly, we show that selective upregulation of protein kinase Cδ (PKCδ) in the prefrontal cortex (PFC) of wild-type mice persisted for 28 days post withdrawal of MA. On day 28, the MA-induced increase in phospho-PKCδ expression and decrease in phospho-ERK1/2 expression were significantly attenuated by both the Src inhibitor PP2 and the dopamine D1 receptor antagonist SCH 23390. However, neither protein kinase A inhibitor H89 nor calmodulin-dependent protein kinase II inhibitor KN93 attenuated MA-induced alterations in phospho-PKCδ expression and phospho-ERK1/2 expression. Since PKCδ knockout (KO) significantly increased the expression of glutathione peroxidase (GPx)-1, we also utilized GPx-1 KO and GPx-1-overexpressing transgenic (GPx-1 TG) mice. Repeated MA treatment induced cognitive impairment, as assessed by the novel object recognition test. Moreover, the extent of cognitive impairment correlated with the extent of increased phospho-PKCδ expression and decreased GPx1 expression. In the absence of MA, exposure to novel objects increased phospho-ERK1/2 and GPx-1 expression in the PFC; however, these expression levels were decreased in the presence of MA. PKCδ KO and GPx-1 TG mice each exhibited significantly attenuated MA-induced decreases in phospho-ERK1/2 and GPx-1 expression. Consistently, PKCδ inhibition induces GPx/GSH-dependent antioxidant systems. More importantly, the antipsychotic drug clozapine significantly protected against cognitive impairment and was associated with alterations in phospho-ERK1/2 and phospho-PKCδ expression. However, GPx-1 KO potentiated MA-induced cognitive deficits and alterations in phospho-ERK1/2 and phospho-PKCδ expression. These results suggest that MA induces cognitive impairment by inhibiting ERK1/2 signaling, activating PKCδ, and inactivating GPx-1 by upregulating Src kinase or the D1 receptor. They also suggest that clozapine requires activation of ERK1/2 signaling via positive modulation between the phospho-PKCδ and GPx-1 genes to restore cognitive function.


Gene Deletion , Glutathione Peroxidase/genetics , MAP Kinase Signaling System , Memory Disorders/enzymology , Protein Kinase C-delta/genetics , Up-Regulation/genetics , Animals , Behavior, Animal , Butadienes/pharmacology , Clozapine/pharmacology , Cognition Disorders/enzymology , Cognition Disorders/physiopathology , DNA-Binding Proteins , Glutathione Disulfide/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Isoquinolines/pharmacology , Male , Memory Disorders/physiopathology , Methamphetamine , Mice, Inbred C57BL , Mice, Knockout , Motor Activity , Nerve Tissue Proteins/metabolism , Nitriles/pharmacology , Nuclear Proteins/metabolism , Phosphorylation , Prefrontal Cortex/enzymology , Prefrontal Cortex/pathology , Protein Kinase C-delta/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Up-Regulation/drug effects , Glutathione Peroxidase GPX1
6.
Mol Neurobiol ; 55(5): 3889-3900, 2018 May.
Article En | MEDLINE | ID: mdl-28547530

Among five members of the K+-dependent Na+/Ca2+ exchanger (NCKX) family (NCKX1-5), only NCKX2 is highly expressed in mouse brain. NCKX2 in plasma membranes mediates cytosolic calcium excretion through electrogenic exchange of 4 Na+ for 1 Ca2+ and 1 K+. Here, we observed significantly decreased levels of NCKX2 protein and mRNA in the CA1 region of APP23 mice, a model of Alzheimer's disease. We also found that, like APP23 mice, heterozygous NCKX2-mutant mice exhibit mildly impaired hippocampal LTP and memory acquisition, the latter based on novel object recognition and passive avoidance tasks. When we addressed underlying mechanisms, we found that both CaMKII autophosphorylation and CaMKIV phosphorylation significantly decreased in CA1 regions of NCKX2+/- relative to control mice. Likewise, phosphorylation of GluA1 (Ser-831) and CREB (Ser-133), respective downstream targets of CaMKII and CaMKIV, also significantly decreased in the CA1 region. BDNF protein and mRNA levels significantly decreased in CA1 of NCKX2+/- relative to control mice. Finally, CaN activity increased in CA1 of NCKX2+/- mice. Our findings suggest that like APP23 mice, NCKX2+/- mice may exhibit impaired learning and hippocampal LTP due to decreased CaM kinase II and CaM kinase IV activities.


Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 4/metabolism , Cognition Disorders/enzymology , Sodium-Calcium Exchanger/genetics , Animals , Astrocytes/metabolism , Behavior, Animal , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , CA1 Region, Hippocampal/metabolism , Calcineurin/metabolism , Cognition Disorders/pathology , Cognition Disorders/physiopathology , Heterozygote , Humans , Long-Term Potentiation , Male , Memory , Mice, Transgenic , Models, Biological , Neurons/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sodium-Calcium Exchanger/metabolism , Synapses/metabolism
7.
J Head Trauma Rehabil ; 33(1): 25-32, 2018.
Article En | MEDLINE | ID: mdl-28060207

OBJECTIVE: To investigate quantitative positron emission tomography (PET) findings and to study whether the cholinergic function differs between respondents to cholinergic medication versus nonrespondents. SETTING: Outpatient clinic and university PET imaging center. PARTICIPANTS: We studied 17 subjects for more than 1 year after at least moderate traumatic brain injury. Ten of the subjects were respondents and 7 nonrespondents to cholinergic medication. DESIGN: Cholinergic function was assessed with [methyl-C] N-methylpiperidyl-4-acetate-PET (C-MP4A-PET), which reflects the activity of the acetylcholinesterase (AChE) enzyme. The subjects were PET scanned twice: without medication and after a 4-week treatment with rivastigmine 1.5 mg twice a day. MEASURES: Regional cerebral AChE activity was measured with PET. RESULTS: At baseline Statistical Parametric Mapping analyses showed significantly lower AChE activity in respondents bilaterally in the frontal cortex as compared with nonrespondents. Region of interest (ROI) analysis revealed that the difference was most pronounced in the lateral frontal cortex (-9.4%, P = .034) and anterior cingulate (-6.0%, P = .049). After rivastigmine treatment, AChE activity was notably lower throughout the cortex in both respondents and nonrespondents, without significant differences between them. CONCLUSION: Our study suggests that frontal cholinergic dysfunction is associated with the clinical response to cholinergic stimulation in patients with traumatic brain injury.


Acetylcholinesterase/metabolism , Brain Injuries, Traumatic/diagnostic imaging , Brain Injuries, Traumatic/enzymology , Cholinesterase Inhibitors/therapeutic use , Cognition Disorders/drug therapy , Rivastigmine/therapeutic use , Adult , Brain Injuries, Traumatic/psychology , Chronic Disease , Cognition Disorders/diagnostic imaging , Cognition Disorders/enzymology , Cohort Studies , Female , Humans , Male , Middle Aged , Positron-Emission Tomography
8.
Exp Gerontol ; 102: 93-100, 2018 02.
Article En | MEDLINE | ID: mdl-29248560

Vascular endothelial growth factor (VEGF) has been identified as a potential treatment for effectively improving cognitive function in several neuropathological conditions. However, the underlying mechanism and the relevant downstream protective pathways that are activated in neurons by VEGF remain elusive, especially in chronic global cerebral ischemia. In this study, we intended to investigate the signaling mechanisms of VEGF in cognitive protection and anti-apoptosis in a rat model of chronic global cerebral ischemia induced by permanent bilateral common carotid artery occlusion (2-VO). The results showed that intranasal administration of VEGF (72h post-ischemia for 6 successive days) caused a significant improvement in the cognitive deficits induced by 2-VO, accompanied by a reversal of oxidative stress and VEGF depletion in the hippocampus. In addition, VEGF-treatment decreased the expression of Bax and Caspase-3, increased the expression of anti-apoptotic Bcl-xl and the main protein involved in energy homeostasis AMP-activated protein kinase (AMPK), which may account for the anti-apoptotic effects of VEGF. Importantly, VEGF administration upregulated the phosphorylation levels of Akt (pAkt) and PI3K, activated Notch1 pathway in 2-VO hippocampus. These findings suggested that intranasal administration of VEGF alleviated cognitive impairment induced by 2-VO injury, and attenuated oxidative damage and neuronal injury in hippocampus associated with the regulation of PI3K/Akt and Notch1 signaling pathway, which might be the underlying mechanisms of VEGF on global chronic cerebral ischemia.


Behavior, Animal/drug effects , Carotid Artery, Common , Cognition Disorders/prevention & control , Cognition/drug effects , Coronary Stenosis/drug therapy , Hippocampus/drug effects , Neuroprotective Agents/administration & dosage , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Notch1/metabolism , Vascular Endothelial Growth Factor A/administration & dosage , Administration, Intranasal , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Carotid Artery, Common/surgery , Cognition Disorders/enzymology , Cognition Disorders/pathology , Cognition Disorders/psychology , Coronary Stenosis/enzymology , Coronary Stenosis/pathology , Coronary Stenosis/psychology , Disease Models, Animal , Hippocampus/enzymology , Hippocampus/pathology , Hippocampus/physiopathology , Male , Maze Learning/drug effects , Memory/drug effects , Neurons/drug effects , Neurons/enzymology , Neurons/pathology , Oxidative Stress/drug effects , Phosphorylation , Rats, Wistar , Signal Transduction/drug effects
9.
Physiol Res ; 66(6): 1029-1039, 2017 12 20.
Article En | MEDLINE | ID: mdl-28937253

Hyperbaric oxygen preconditioning (HBO-PC) has been proposed as a safe and practical approach for neuroprotection in ischemic stroke. However, it is not known whether HPO-PC can improve cognitive deficits induced by cerebral ischemia, and the mechanistic basis for any beneficial effects remains unclear. We addressed this in the present study using rats subjected to middle cerebral artery occlusion (MCAO) as an ischemic stroke model following HBO-PC. Cognitive function and expression of phosphorylated neurofilament heavy polypeptide (pNF-H) and doublecortin (DCX) in the hippocampus were evaluated 14 days after reperfusion and after short interfering RNA-mediated knockdown of sirtuin1 (Sirt1). HBO-PC increased pNF-H and DCX expression and mitigated cognitive deficits in MCAO rats. However, these effects were abolished by Sirt1 knockdown. Our results suggest that HBO-PC can protect the brain from injury caused by ischemia-reperfusion and that Sirt1 is a potential molecular target for therapeutic approaches designed to minimize cognitive deficits caused by cerebral ischemia.


Behavior, Animal , Cognition Disorders/prevention & control , Cognition , Hippocampus/enzymology , Hyperbaric Oxygenation , Infarction, Middle Cerebral Artery/therapy , Sirtuin 1/metabolism , Animals , Cognition Disorders/enzymology , Cognition Disorders/physiopathology , Cognition Disorders/psychology , Disease Models, Animal , Doublecortin Domain Proteins , Doublecortin Protein , Hippocampus/physiopathology , Infarction, Middle Cerebral Artery/enzymology , Infarction, Middle Cerebral Artery/physiopathology , Infarction, Middle Cerebral Artery/psychology , Male , Microtubule-Associated Proteins/metabolism , Neurofilament Proteins/metabolism , Neuropeptides/metabolism , Phosphorylation , RNA Interference , Rats, Sprague-Dawley , Sirtuin 1/genetics , Time Factors
10.
J Med Chem ; 60(18): 7658-7676, 2017 09 28.
Article En | MEDLINE | ID: mdl-28759228

Herein, we describe the discovery of a potent, selective, brain-penetrating, in vivo active phosphodiesterase (PDE) 2A inhibitor lead series. To identify high-quality leads suitable for optimization and enable validation of the physiological function of PDE2A in vivo, structural modifications of the high-throughput screening hit 18 were performed. Our lead generation efforts revealed three key potency-enhancing functionalities with minimal increases in molecular weight (MW) and no change in topological polar surface area (TPSA). Combining these structural elements led to the identification of 6-methyl-N-((1R)-1-(4-(trifluoromethoxy)phenyl)propyl)pyrazolo[1,5-a]pyrimidine-3-carboxamide (38a), a molecule with the desired balance of preclinical properties. Further characterization by cocrystal structure analysis of 38a bound to PDE2A uncovered a unique binding mode and provided insights into its observed potency and PDE selectivity. Compound 38a significantly elevated 3',5'-cyclic guanosine monophosphate (cGMP) levels in mouse brain following oral administration, thus validating this compound as a useful pharmacological tool and an attractive lead for future optimization.


Brain/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 2/antagonists & inhibitors , Phosphodiesterase Inhibitors/pharmacology , Phosphodiesterase Inhibitors/pharmacokinetics , Pyrimidines/pharmacology , Pyrimidines/pharmacokinetics , Administration, Oral , Animals , Brain/metabolism , Cognition Disorders/drug therapy , Cognition Disorders/enzymology , Cognition Disorders/metabolism , Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 2/metabolism , Drug Discovery , Humans , Male , Mice , Mice, Inbred ICR , Molecular Docking Simulation , Phosphodiesterase Inhibitors/administration & dosage , Phosphodiesterase Inhibitors/chemistry , Pyrimidines/administration & dosage , Pyrimidines/chemistry , Rats
11.
Lipids Health Dis ; 16(1): 117, 2017 Jun 15.
Article En | MEDLINE | ID: mdl-28724385

BACKGROUND: Autism is neurodevelopmental disorder that is characterized by developmental, behavioral, social and sensory abnormalities. Researchers have focused in last years in immunological alteration and inflammation as a hot subject in autism field. This work aims to study the alteration in phospholipids (PE, PS, and PC) together with the change in cPLA2 concentration as the main phospholipid hydrolytic enzyme in autistic patients compared to control. It was also extended to find a correlation between these biomarkers and severity of autism measured as childhood autism rating scale (CARS), Social responsiveness scale (SRS), and Short sensory profile (SSP). METHODS: Phospholipids (PE, PS, PC) and cPLA2 as biochemical parameters were determined in the plasma of 48 Saudi autistic male patients, categorized as mild-moderate and severe as indicated by their Childhood Autism Rating Scale (CARS), social responsiveness scale (SRS) and short sensory profile (SSP) and compared to 40 age- and gender-matched control samples. RESULTS: The reported data demonstrate significantly lower levels of PE, PS, and PC together with a significant increase in cPLA2. While association between severity of autism and impaired phospholipid concentration was completely lacked, an association between cPLA2 and impaired sensory processing was observed. CONCLUSIONS: The impaired phospholipid level and remarkable increased in cPLA2 concentration asserted their roles in the etiology of autism. Receiver operating characteristic analysis together with predictiveness diagrams proved that the measured parameters could be used as predictive biomarkers of clinical symptoms and provide significant guidance for future therapeutic strategy to re-establish physiological homeostasis.


Autistic Disorder/blood , Autistic Disorder/enzymology , Cognition Disorders/blood , Cognition Disorders/enzymology , Phospholipases A2, Cytosolic/blood , Phospholipids/blood , Severity of Illness Index , Social Behavior , Case-Control Studies , Child , Humans , Hydrolysis , Male , ROC Curve
12.
Brain Res Bull ; 132: 190-198, 2017 06.
Article En | MEDLINE | ID: mdl-28599877

Published studies have shown that cognitive deficit is a characteristic manifestation of neurodegenerative disease in diabetes. However, there is no effective prevention and treatment for this diabetes-associated behavior disorder. In the present study, we attempted to elucidate the effect of zeaxanthin on cognitive deficit and the change in the hippocampus correlated with cognitive decline in diabetic rats. Diabetic rats in this study were induced by high-fat diet and low-dose streptozocin (STZ), cognitive ability of rats were evaluated use morris water maze (MWM) and morphology change in hippocampus was assessed by cresyl violet stain. Moreover, we detected the expression of phosphorylated serine/threonine kinase (p-AKT) and Cleaved caspase-3, and the activity of nuclear factor-κB (NF-κB) use western-blot (WB). Results displayed that supplementation with zeaxanthin reduce blood glucose, improve cognitive deficit, survive neural cell, increase p-AKT level, inhibit Cleaved caspase-3 level and NF-κB nuclear transcription in hippocampus. This study demonstrated that zeaxanthin ameliorate diabetes-related cognitive deficit may by means of protecting neural cell from hyperglycemia involved in AKT/NF-κB signaling pathway. This study may provide a potential therapeutic approach for the prevention of diabetes- associated cognitive deficit.


Cognition Disorders/drug therapy , Cognition Disorders/enzymology , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/enzymology , Nootropic Agents/pharmacology , Zeaxanthins/pharmacology , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/physiology , Animals , Caspase 3/metabolism , Cell Survival/drug effects , Cell Survival/physiology , Cognition Disorders/etiology , Cognition Disorders/pathology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/psychology , Hippocampus/drug effects , Hippocampus/enzymology , Hippocampus/pathology , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology , Male , Maze Learning/drug effects , Maze Learning/physiology , NF-kappa B/metabolism , Neurons/drug effects , Neurons/enzymology , Neurons/pathology , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Nootropic Agents/chemistry , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects , Zeaxanthins/chemistry
13.
J Alzheimers Dis ; 58(2): 521-535, 2017.
Article En | MEDLINE | ID: mdl-28482641

Bryostatin 1, a potent activator of protein kinase C epsilon (PKCɛ), has been shown to reverse synaptic loss and facilitate synaptic maturation in animal models of Alzheimer's disease (AD), Fragile X, stroke, and other neurological disorders. In a single-dose (25 µg/m2) randomized double-blind Phase IIa clinical trial, bryostatin levels reached a maximum at 1-2 h after the start of infusion. In close parallel with peak blood levels of bryostatin, an increase of PBMC PKCɛ was measured (p = 0.0185) within 1 h from the onset of infusion. Of 9 patients with a clinical diagnosis of AD, of which 6 received drug and 3 received vehicle within a double-blind protocol, bryostatin increased the Mini-Mental State Examination (MMSE) score by +1.83±0.70 unit at 3 h versus -1.00±1.53 unit for placebo. Bryostatin was well tolerated in these AD patients and no drug-related adverse events were reported. The 25 µg/m2 administered dose was based on prior clinical experience with three Expanded Access advanced AD patients treated with bryostatin, in which return of major functions such as swallowing, vocalization, and word recognition were noted. In one Expanded Access patient trial, elevated PKCɛ levels closely tracked cognitive benefits in the first 24 weeks as measured by MMSE and ADCS-ADL psychometrics. Pre-clinical mouse studies showed effective activation of PKCɛ and increased levels of BDNF and PSD-95. Together, these Phase IIa, Expanded Access, and pre-clinical results provide initial encouragement for bryostatin 1 as a potential treatment for AD.


Alzheimer Disease , Antipsychotic Agents/therapeutic use , Bryostatins/therapeutic use , Cognition Disorders , Protein Kinase C-epsilon/metabolism , Adult , Aged , Aged, 80 and over , Alzheimer Disease/complications , Alzheimer Disease/drug therapy , Alzheimer Disease/enzymology , Analysis of Variance , Animals , Brain/drug effects , Brain/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Cognition Disorders/drug therapy , Cognition Disorders/enzymology , Cognition Disorders/etiology , Disks Large Homolog 4 Protein/metabolism , Double-Blind Method , Female , Humans , Male , Mental Status Schedule , Mice , Mice, Inbred C57BL , Middle Aged , Neuropsychological Tests , Phosphopyruvate Hydratase/metabolism , Psychometrics , Synaptophysin/metabolism , Time Factors
14.
Clin Sci (Lond) ; 131(6): 425-437, 2017 Mar 01.
Article En | MEDLINE | ID: mdl-28265034

Vascular cognitive impairment and dementia (VCID) include a wide spectrum of chronic manifestations of vascular disease related to large vessel strokes and small vessel disease (SVD). Lacunar strokes and white matter (WM) injury are consequences of SVD. The main vascular risk factor for SVD is brain hypoperfusion from cerebral blood vessel narrowing due to chronic hypertension. The hypoperfusion leads to activation and degeneration of astrocytes with the resulting fibrosis of the extracellular matrix (ECM). Elasticity is lost in fibrotic cerebral vessels, reducing the response of stiffened blood vessels in times of increased metabolic need. Intermittent hypoxia/ischaemia activates a molecular injury cascade, producing an incomplete infarction that is most damaging to the deep WM, which is a watershed region for cerebral blood flow. Neuroinflammation caused by hypoxia activates microglia/macrophages to release proteases and free radicals that perpetuate the damage over time to molecules in the ECM and the neurovascular unit (NVU). Matrix metalloproteinases (MMPs) secreted in an attempt to remodel the blood vessel wall have the undesired consequences of opening the blood-brain barrier (BBB) and attacking myelinated fibres. This dual effect of the MMPs causes vasogenic oedema in WM and vascular demyelination, which are the hallmarks of the subcortical ischaemic vascular disease (SIVD), which is the SVD form of VCID also called Binswanger's disease (BD). Unravelling the complex pathophysiology of the WM injury-related inflammation in the small vessel form of VCID could lead to novel therapeutic strategies to reduce damage to the ECM, preventing the progressive damage to the WM.


Cognition Disorders/etiology , Dementia, Vascular/etiology , Extracellular Matrix/pathology , Inflammation/complications , Animals , Cognition Disorders/enzymology , Cognition Disorders/physiopathology , Dementia, Vascular/enzymology , Dementia, Vascular/physiopathology , Disease Models, Animal , Extracellular Matrix/enzymology , Extracellular Matrix/physiology , Humans , Hypertension/complications , Inflammation/drug therapy , Inflammation/enzymology , Inflammation/pathology , Inflammation/physiopathology , Matrix Metalloproteinase Inhibitors/therapeutic use , Matrix Metalloproteinases/metabolism
15.
Behav Brain Res ; 322(Pt A): 70-82, 2017 03 30.
Article En | MEDLINE | ID: mdl-28077315

Diabetes mellitus often results in a number of complications involving impaired brain function, including cognitive deficits and depression. However, the potential mechanisms for diabetes-related cognitive deficits and depression are not fully understood. Neurons in the hippocampal, cortical and amygdala functional regions are more susceptible to damage during hyperglycemia. Neuroprotection in the brain can rescue cognitive deficits and depression induced by hyperglycemia. This study investigated the potential mechanisms underlying diabetes-related congnitive deficits and depression, determined whether the inflammatory factor inducible nitric oxide synthase (iNOS) and the nitric oxide (NO)/soluble guanylyl cyclases (sGC)/cyclic guanosine monophosphate (cGMP)/protein kinase G (PKG) pathway, play key roles in cognitive deficits and depression associated. In the present study, diabetic animal models were induced by streptozotocin (STZ, 150mg/kg) in mice, and aminoguanidine (AG), a selective inhibitor of iNOS, was given by intraperitoneal injection for 10 weeks. Blood glucose, activities of NOS and the levels of NO in serum and brain regions were measured. The spatial memory was detected using the Morris water maze test, depressive behavior was evaluated by the tail suspension test (TST), forced swimming test (FST), closed field test (CFT) and open field test (OFT). We also detected neuronal survival and cleaved caspase-3 positive ratios in three brain regions and the levels of iNOS, sGC, cGMP and PKG in hippocampus and frontal cortex. Data indicated that diabetic mice exerted impairments in spatial memory, decreased locomotor activity and increased immobile time in diabetic mice. In addition, diabetic mice had significantly decreased surviving neuronal density and showed signs of obvious neuronal injury in the hippocampus, frontal cortex and amygdala. iNOS overexpression and its associated signaling pathway NO/sGC/cGMP/PKG in the hippocampus and frontal cortex were implicated during hyperglycemia. However, AG improved the behavior disorders, reduced the activity of iNOS, protected nerve cells and inhibited the level of iNOS, sGC, PKG and cleaved caspase-3 in the hippocampus and cortex. These results suggested that iNOS/NO/sGC/cGMP/PKG signal pathway is a key feature of cognitive deficits and depression associated with diabetes. AG ameliorated cognitive deficits and depression in diabetic mice by exerting anti-inflammatory and neuroprotective effects by suppressing iNOS-associated signaling pathways.


Cognition Disorders/drug therapy , Depressive Disorder/drug therapy , Diabetes Complications/drug therapy , Diabetes Mellitus, Experimental/drug therapy , Nitric Oxide Synthase Type II/antagonists & inhibitors , Psychotropic Drugs/pharmacology , Animals , Brain/drug effects , Brain/enzymology , Brain/pathology , Cognition Disorders/complications , Cognition Disorders/enzymology , Cognition Disorders/psychology , Depressive Disorder/complications , Depressive Disorder/enzymology , Depressive Disorder/psychology , Diabetes Complications/enzymology , Diabetes Complications/psychology , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/psychology , Enzyme Inhibitors/pharmacology , Guanidines/pharmacology , Hypoglycemic Agents/pharmacology , Male , Mice, Inbred ICR , Nitric Oxide Synthase Type II/metabolism , Signal Transduction/drug effects
16.
Naunyn Schmiedebergs Arch Pharmacol ; 390(2): 139-148, 2017 Feb.
Article En | MEDLINE | ID: mdl-27807596

The aim of this study was to investigate the effect of nerolidol-loaded nanospheres (N-NS) on the treatment of memory impairment caused by Trypanosoma evansi in mice, as well as oxidative stress, and Na+, K+-ATPase and acetylcholinesterase (AChE) activities in brain tissue. Animals were submitted to behavioral tasks (inhibitory avoidance task and open-field test) 4 days postinfection (PI). Reactive oxygen species (ROS) and thiobarbituric acid-reactive substance (TBARS) levels and catalase (CAT), superoxide dismutase (SOD), Na+, K+-ATPase and AChE activities were measured on the fifth-day PI. T. evansi-infected mice showed memory deficit, increased ROS and TBARS levels and SOD and AChE activities, and decreased CAT and Na+, K+-ATPase activities compared to uninfected mice. N-NS prevented memory impairment and oxidative stress parameters (except SOD activity), while free nerolidol (N-F) restored only CAT activity. Also, N-NS treatment was able to prevent alterations in Na+, K+-ATPase and AChE activities caused by T. evansi infection. A significantly negative correlation was observed between memory and ROS production (p < 0.001; r = -0.941), as well as between memory and AChE activity (p < 0.05; r = -0.774). On the contrary, a significantly positive correlation between memory and Na+, K+-ATPase activity was observed (p < 0.01; r = 0.844). In conclusion, N-NS was able to reverse memory impairment and to prevent increased ROS and TBARS levels due to amelioration of Na+, K+-ATPase and AChE activities and to activation of the antioxidant enzymes, respectively. These results suggest that N-NS treatment may be a useful strategy to treat memory dysfunction and oxidative stress caused by T. evansi infection.


Antioxidants/administration & dosage , Behavior, Animal/drug effects , Brain/drug effects , Central Nervous System Protozoal Infections/drug therapy , Cholinesterase Inhibitors/administration & dosage , Nanospheres , Oxidative Stress/drug effects , Sesquiterpenes/administration & dosage , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Trypanosoma/pathogenicity , Trypanosomiasis/drug therapy , Acetylcholinesterase/metabolism , Animals , Avoidance Learning/drug effects , Brain/enzymology , Brain/pathology , Catalase/metabolism , Central Nervous System Protozoal Infections/enzymology , Central Nervous System Protozoal Infections/parasitology , Central Nervous System Protozoal Infections/psychology , Cognition Disorders/drug therapy , Cognition Disorders/enzymology , Cognition Disorders/parasitology , Cognition Disorders/psychology , Disease Models, Animal , Female , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/metabolism , Memory/drug effects , Memory Disorders/drug therapy , Memory Disorders/enzymology , Memory Disorders/parasitology , Memory Disorders/psychology , Mice , Motor Activity/drug effects , Nootropic Agents/administration & dosage , Reaction Time/drug effects , Reactive Oxygen Species/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Superoxide Dismutase/metabolism , Thiobarbituric Acid Reactive Substances/metabolism , Trypanosomiasis/enzymology , Trypanosomiasis/parasitology , Trypanosomiasis/psychology
17.
Physiol Behav ; 170: 115-123, 2017 03 01.
Article En | MEDLINE | ID: mdl-28017679

Although Metformin, a first-line antidiabetic drug, can ameliorate ischemia/reperfusion (I/R) induced brain damage, but how metformin benefits injured hippocampus and the mechanisms are still largely unknown. Therefore, the aim of this study was to investigate the neuroprotective mechanisms of metformin against ischemic brain damage induced by cerebral I/R and to explore whether the Akt-mediated down-regulation of the phosphorylation of JNK3 signaling pathway contributed to the protection provided by metformin. Transient global brain ischemia was induced by 4-vessel occlusion in adult male Sprague-Dawley rats. The open field tasks and Morris water maze were used to assess the effect of metformin on anxiety-like behavioral and cognitive impairment after I/R. Cresyl Violet staining was used to examine the survival of hippocampal CA1 pyramidal neurons. Immunoblotting was performed to measure the phosphorylation of Akt1, JNK3, c-Jun and the expression of cleaved caspase-3. Through ischemia/reperfusion (I/R) rat model, we found that metformin could attenuate the deficits of hippocampal related behaviors and inhibit cell apoptosis. The western blot data showed that metformin could promote the activation of Akt1 and reduce the phosphorylation of JNK3 and c-Jun as well as elevation of cleaved caspase-3 in I/R brains. PI3K inhibitor reversed all the protective effects, further indicating that metformin protect hippocampus from ischemic damage through PI3K/Akt1/JNK3/c-Jun signaling pathway.


Brain Ischemia/drug therapy , Brain Ischemia/enzymology , Metformin/pharmacology , Neuroprotective Agents/pharmacology , Reperfusion Injury/drug therapy , Reperfusion Injury/enzymology , Animals , Anxiety/drug therapy , Anxiety/enzymology , Anxiety/etiology , Anxiety/pathology , Apoptosis/drug effects , Apoptosis/physiology , Brain Ischemia/complications , Brain Ischemia/pathology , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/enzymology , CA1 Region, Hippocampal/pathology , Caspase 3/metabolism , Cognition Disorders/drug therapy , Cognition Disorders/enzymology , Cognition Disorders/etiology , Cognition Disorders/pathology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Male , Maze Learning/drug effects , Maze Learning/physiology , Mitogen-Activated Protein Kinase 10/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Pyramidal Cells/drug effects , Pyramidal Cells/enzymology , Pyramidal Cells/pathology , Rats, Sprague-Dawley , Reperfusion Injury/complications , Reperfusion Injury/pathology , Signal Transduction/drug effects
18.
Eur Neuropsychopharmacol ; 27(2): 180-191, 2017 02.
Article En | MEDLINE | ID: mdl-27986355

Cognitive deficits are considered a key feature of schizophrenia, and they usually precede the onset of the illness and continue after psychotic symptoms appear. Current antipsychotic drugs have little or no effect on the cognitive deficits of this disorder. Prolyl oligopeptidase (POP) is an 81-kDa monomeric serine protease that is expressed in brain and other tissues. POP inhibitors have shown neuroprotective, anti-amnesic and cognition-enhancing properties. Here we studied the potential of IPR19, a new POP inhibitor, for the treatment of the cognitive symptoms related to schizophrenia. The efficacy of the inhibitor was evaluated in mouse models based on subchronic phencyclidine and acute dizocilpine administration, and in adult offspring from mothers with immune reaction induced by polyinosinic:polycytidylic acid administration during pregnancy. Acute IPR19 administration (5mg/kg, i.p.) reversed the cognitive performance deficits of the three mouse models in the novel object recognition test, T-maze, and eight-arm radial maze. The compound also ameliorates deficits of the prepulse inhibition response. The in vitro inhibitory efficacy and selectivity, brain penetration and exposure time after injection of IPR19 were also addressed. Our results indicate that the inhibition of POP using IPR19 may offer a promising strategy to develop drugs to ameliorate the cognitive deficits of schizophrenia.


Cognition Disorders/drug therapy , Proline/analogs & derivatives , Psychotropic Drugs/pharmacology , Schizophrenia/drug therapy , Schizophrenic Psychology , Animals , Cell Line, Tumor , Cognition/drug effects , Cognition/physiology , Cognition Disorders/enzymology , Cognition Disorders/etiology , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Male , Maze Learning/drug effects , Maze Learning/physiology , Mice, Inbred C57BL , Motor Activity/drug effects , Motor Activity/physiology , Poly I-C , Prepulse Inhibition/drug effects , Prepulse Inhibition/physiology , Proline/chemistry , Proline/pharmacokinetics , Proline/pharmacology , Proline/toxicity , Prolyl Oligopeptidases , Psychotropic Drugs/chemistry , Psychotropic Drugs/pharmacokinetics , Psychotropic Drugs/toxicity , Recognition, Psychology/drug effects , Recognition, Psychology/physiology , Schizophrenia/complications , Schizophrenia/enzymology , Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/pharmacokinetics , Serine Proteinase Inhibitors/pharmacology , Serine Proteinase Inhibitors/toxicity
19.
Brain Res ; 1657: 355-360, 2017 02 15.
Article En | MEDLINE | ID: mdl-27998794

Increasing evidence demonstrates an association between diabetes and hippocampal neuron damage. This study aimed to determine the effects of troxerutin on cognitive deficits and glutamate cysteine ligase subunits (GCLM and GCLC) in the hippocampus of streptozotocin-induced type 1 diabetes mellitus (T1DM) rats. At 12weeks after streptozotocin injection, T1DM rats were randomly divided into 4 groups (n=15 each group) to receive no treatment (T1DM), saline (T1DM+saline), alpha-lipoic acid (T1DM+alpha-lipoic acid), and troxerutin (T1DM+troxerutin), respectively, for 6weeks. Meanwhile, 10 control animals (NC group) were assessed in parallel. Learning performance was evaluated by the Morris water maze. After treatment, hippocampi were collected for pathological examination by hematoxylin and eosin (H&E) staining. Next, hippocampal superoxide dismutase (SOD) activity, and malondialdehyde (MDA) and glutathione (GSH) levels were assessed. Finally, glutamate cysteine ligase catalytic (GCLC) and glutamate cysteine ligase modifier (GCLM) subunit mRNA and protein levels were quantified by reverse transcription polymerase chain reaction (RT-PCR) and Western blot, respectively. Compared with T1DM and T1DM+saline groups, escape latency was overtly reduced in T1DM+alpha-lipoic acid and T1DM+troxerutin groups. Significantly increased GCLM and GCLC mRNA levels, GCLC protein amounts, SOD activity, and GSH levels, and reduced MDA amounts were observed in T1DM+alpha-lipoic acid and T1DM+troxerutin groups. In T1DM and T1DM+saline groups, H&E staining showed less pyramidal cells in the hippocampus, with disorganized layers, karyopyknosis, decreased endochylema, and cavitation, effects relieved in T1DM+alpha-lipoic acid and T1DM+troxerutin groups. Troxerutin alleviates oxidative stress and promotes learning in streptozotocin-induced T1DM rats, a process involving GCLC expression.


Cognition Disorders/drug therapy , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 1/drug therapy , Glutamate-Cysteine Ligase/metabolism , Hydroxyethylrutoside/analogs & derivatives , Hypoglycemic Agents/pharmacology , Animals , Cognition Disorders/enzymology , Cognition Disorders/etiology , Cognition Disorders/pathology , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/psychology , Diabetes Mellitus, Type 1/enzymology , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/psychology , Drug Evaluation, Preclinical , Hippocampus/drug effects , Hippocampus/enzymology , Hippocampus/pathology , Hydroxyethylrutoside/pharmacology , Male , Maze Learning/drug effects , Maze Learning/physiology , Nootropic Agents/pharmacology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Random Allocation , Rats, Sprague-Dawley , Thioctic Acid/pharmacology
20.
Brain Res ; 1657: 9-15, 2017 02 15.
Article En | MEDLINE | ID: mdl-27923634

Cerebral ischemia/reperfusion injury can result in neuronal death, which further results in brain damage and can even lead to death. Although recent studies showed that rosmarinic acid (RA) exerts neuroprotective effects and attenuates ischemia-induced brain injury and neuronal cell death, little is known about the precise mechanisms that occur during cerebral ischemia/reperfusion (I/R). Therefore, the aim of this study was to examine the underlying mechanism of the neuroprotective effects of RA against ischemic brain injury induced by cerebral I/R. Transient global brain ischemia was induced by 4-vessel occlusion in adult male Sprague-Dawley rats. We randomly divided rats into five groups: sham, I/R, I/R+RA, I/R+Vehicle and I/R+RA+LY. Open-field, closed-field and Morris water maze tests were carried our separately to examine the anxiety and cognitive behavior of each group. Cresyl violet staining was used to examine the survival of hippocampal CA1 pyramidal neurons. The levels of p-Akt, p-JNK3 and cleaved caspase-3 in the hippocampus were also examined by Western blotting. Our results showed that administration of RA protected locomotive ability, relieved anxiety behavior and protected cognitive ability in cerebral I/R-injured rats. Additionally, RA significantly protected neurons in the hippocampal CA1 region against cerebral I/R-induced damage. Furthermore, RA increased the phosphorylation of Akt1, downregulated the phosphorylation of JNK3 and reduced the expression of cleaved caspase-3. Finally, the Akt inhibitor LY294002 reversed all the protective effects of RA, indicating that RA protects neurons in the hippocampal CA1 region from ischemic damage through the Akt/JNK3/caspase-3 signaling pathway.


Brain Ischemia/drug therapy , CA1 Region, Hippocampal/drug effects , Cinnamates/pharmacology , Depsides/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Reperfusion Injury/drug therapy , Animals , Anxiety/drug therapy , Anxiety/enzymology , Anxiety/pathology , Apoptosis/drug effects , Apoptosis/physiology , Brain Ischemia/enzymology , Brain Ischemia/pathology , Brain Ischemia/psychology , CA1 Region, Hippocampal/enzymology , CA1 Region, Hippocampal/pathology , Caspase 3/metabolism , Cell Survival/drug effects , Cell Survival/physiology , Cognition Disorders/drug therapy , Cognition Disorders/enzymology , Cognition Disorders/pathology , Disease Models, Animal , Male , Maze Learning/drug effects , Maze Learning/physiology , Mitogen-Activated Protein Kinase 10/metabolism , Neurons/enzymology , Neurons/pathology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Random Allocation , Rats, Sprague-Dawley , Reperfusion Injury/enzymology , Reperfusion Injury/pathology , Reperfusion Injury/psychology , Rosmarinic Acid
...