Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 15 de 15
1.
Eur J Pharmacol ; 899: 174025, 2021 May 15.
Article En | MEDLINE | ID: mdl-33722590

Cerebral ischemia constitutes the most frequent type of cerebrovascular disease. The reduction of blood supply to the brain initiates the ischemic cascade starting from ionic imbalance to subsequent glutamate excitotoxicity, neuroinflammation and oxidative stress, eventually causing neuronal death. Previously, the authors have demonstrated the in vitro cytoprotective and antioxidant effects of a new arylidene malonate derivative, KM-34, against oxidizing agents like hydrogen peroxide, glutamate or Fe3+/ascorbate. Here, we examined for the first time the neuroprotective effect of KM-34 on ischemia/reperfusion models. In vitro, treatment with 10 and 50 µM KM-34 reduced the cellular death (propidium iodide incorporation) induced by oxygen glucose deprivation (OGD) in rat organotypic hippocampal slices cultures. In vivo, stroke was induced in male Wistar rats through middle cerebral artery occlusion (MCAO), followed by 23 h of reperfusion. KM-34 was orally administered 105 min after MCAO onset. We noticed that 1 mg/kg KM-34 reduced infarct volume and neurological score, and increased the latency to fall in the Hanging Wire test compared to vehicle-treated ischemic animals. While ischemic and sham-operated groups showed similar horizontal locomotor activity, vertical counts decreased after MCAO, suggesting that vertical movements are more sensitive to the ischemic injury. Treatment with KM-34 also alleviated the mitochondrial impairment (ROS generation, swelling and membrane potential dissipation) induced by transient MCAO but not significant alterations were found in oxidative stress parameters. Overall, the study provides preclinical evidences confirming the neuroprotective effects of a novel synthetic molecule and paved the way for future investigations regarding its therapeutic potential against brain ischemia/reperfusion injury.


Brain/drug effects , Catechols/pharmacology , Infarction, Middle Cerebral Artery/drug therapy , Neuroprotective Agents/pharmacology , Reperfusion Injury/prevention & control , Animals , Behavior, Animal/drug effects , Brain/metabolism , Brain/pathology , Brain/physiopathology , Disease Models, Animal , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Locomotion/drug effects , Male , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Swelling/drug effects , Motor Activity/drug effects , Oxidative Stress/drug effects , Rats, Wistar , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Tissue Culture Techniques
2.
Neurotoxicology ; 82: 89-98, 2021 01.
Article En | MEDLINE | ID: mdl-33232743

We have previously shown that JM-20, a new chemical entity consisting of 1,5-benzodiazepine fused to a dihydropyridine moiety, protects against rotenone-induced neurotoxicity in an experimental model of Parkinson's disease (PD). The aim of this study was to investigate the effect of a novel hybrid molecule, named JM-20, in in vitro and in vivo models of PD induced by 6-hydroxydopamine (6-OHDA). PC-12 cells were exposed to 6-OHDA and treated with JM-20. Protection against mitochondrial damage induced by 6-OHDA was also investigated using isolated rat brain mitochondria. We found that JM-20 protected PC-12 cells against cytotoxicity induced by 6-OHDA and inhibited hydrogen peroxide generation, mitochondrial swelling and membrane potential dissipation. For in vivo experiments, adult male Wistar rats were lesioned in the substantia nigra pars compacta (SNpc) by 6-OHDA administration. JM-20 was orally administered (10, 20 or 40 mg/kg), intragastric via gavage, 24 h after surgery and daily for seven days. Treatment with JM-20 significantly reduced the percentage of motor asymmetry and increased vertical exploration. It improved the redox state of the SNpc and the striatal tissue of these animals. Also, JM-20 reduced glial fibrillary acidic protein overexpression and increased tyrosine hydroxylase-positive cell number, both in SNpc. Altogether, these results demonstrate that JM-20 is a potential neuroprotective agent against 6-OHDA-induced damage in both in vitro and in vivo models. The mechanism underlying JM-20 neuroprotection against 6-OHDA appears to be associated with the control of oxidative injury and mitochondrial impairment.


Antioxidants/pharmacology , Benzodiazepines/pharmacology , Brain/drug effects , Mitochondria/drug effects , Neuroprotective Agents/pharmacology , Niacin/analogs & derivatives , Oxidopamine/toxicity , Parkinsonian Disorders/drug therapy , Animals , Male , Mitochondria/metabolism , Niacin/pharmacology , Open Field Test/drug effects , Oxidative Stress/drug effects , PC12 Cells/drug effects , Parkinsonian Disorders/chemically induced , Rats , Rats, Wistar
3.
Brain Res Bull ; 165: 185-197, 2020 12.
Article En | MEDLINE | ID: mdl-33096198

The present study examines the possible effect of the novel hybrid molecule JM-20 (3-ethoxycarbonyl-2-methyl-4-(2-nitrophenyl)-411-dihydro-1H-pyrido[2,3-b] [1,5] benzodiazepine) on pain-related behaviours in a persistent pain model (5% formalin test) and in the neutrophil migration events during the inflammatory process. It further introduces JM-20 in a chronic constriction injury (CCI) model to clarify the possible subjacent mechanisms with its consequent clinical relevance. A single administration of JM-20 (20 or 40 mg/kg, per os [p.o.]) decreased licking/biting exclusively in the tonic phase of the formalin test in a GABA/benzodiazepine (BZD) receptor antagonist flumazenil-sensitive manner. JM-20 reduced in vivo neutrophil migration, rolling and adhesion to the endothelium induced by intraperitoneal administration of carrageenan in mice. In addition, plasma extravasation and tumour necrosis factor alpha production in the peritoneal fluid were decreased. Treatment with JM-20 (20 mg/kg, p.o.) for 7 days after CCI reduced mechanical hypersensitivity in a NG-monomethyl-l-arginine (L-NMMA)/methylene blue/glibenclamide-sensitive manner. Histopathological signs of Wallerian degeneration (WD) of the sciatic nerve were also attenuated, as well as interleukin-1 beta release in the spinal cord. The nitrate/nitrite concentration was increased centrally and did not show differences at the peripheral nerve level. The findings of this study suggest JM-20 can decrease persistent pain. A transient activity of its BDZ portion on nociceptive pathways mediated by GABA/BDZ receptors in association with its anti-inflammatory properties could be at least partially involved in this effect. JM-20 decreased CCI-induced mechanical hypersensitivity via the l-arginine/nitric oxide (NO)/cyclic GMP-sensitive ATP-sensitive potassium channel pathway. Its neuroprotective ability by preventing WD could be implicated in its anti-neuropathic mechanisms.


Benzodiazepines/therapeutic use , Inflammation/drug therapy , Neuroprotective Agents/therapeutic use , Niacin/analogs & derivatives , Pain/drug therapy , Animals , Behavior, Animal/drug effects , Benzodiazepines/pharmacology , Cell Movement/drug effects , Inflammation/pathology , Male , Neuroprotective Agents/pharmacology , Neutrophils/drug effects , Niacin/pharmacology , Niacin/therapeutic use , Pain/pathology , Pain Measurement , Pain Threshold/drug effects , Rats , Rats, Sprague-Dawley , Sciatic Nerve/drug effects , Sciatic Nerve/pathology
4.
Mol Neurobiol ; 56(1): 502-512, 2019 Jan.
Article En | MEDLINE | ID: mdl-29725905

Stroke is frequently associated with severe neurological decline and mortality, and its incidence is expected to increase due to aging population. The only available pharmacological treatment for cerebral ischemia is thrombolysis, with narrow therapeutic windows. Efforts aimed to identify new therapeutics are crucial. In this study, we look into plausible molecular and cellular targets for JM-20, a new hybrid molecule, against ischemic stroke in vivo. Male Wistar rats were subjected to 90 min middle cerebral artery occlusion (MCAO) following 23 h of reperfusion. Animals treated with 8 mg/kg JM-20 (p.o., 1 h after reperfusion) showed minimal neurological impairment and lower GABA and IL-1ß levels in CSF when compared to damaged rats that received vehicle. Immunocontent of pro-survival, phosphorylated Akt protein decreased in the cortex after 24 h as result of the ischemic insult, accompanied by decreased number of NeuN+ cells in the peri-infarct cortex, cornu ammonis 1 (CA1) and dentate gyrus (DG) areas. Widespread reactive astrogliosis in both cortex and hippocampus (CA1, CA3, and DG areas) was observed 24 h post-ischemia. JM-20 prevented the activated Akt reduction, neuronal death, and astrocytes reactivity throughout the brain. Overall, the results reinforce the pharmacological potential of JM-20 as neuroprotective agent and provide important evidences about its molecular and cellular targets in this model of cerebral ischemia.


Astrocytes/pathology , Benzodiazepines/therapeutic use , Brain Infarction/drug therapy , Brain/pathology , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/pathology , Neurons/pathology , Niacin/analogs & derivatives , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Benzodiazepines/pharmacology , Brain Infarction/cerebrospinal fluid , Brain Infarction/pathology , CA3 Region, Hippocampal/drug effects , CA3 Region, Hippocampal/metabolism , CA3 Region, Hippocampal/pathology , Cell Death/drug effects , Dentate Gyrus/drug effects , Dentate Gyrus/metabolism , Dentate Gyrus/pathology , Glial Fibrillary Acidic Protein/metabolism , Gliosis/metabolism , Gliosis/pathology , Infarction, Middle Cerebral Artery/cerebrospinal fluid , Interleukin-10/cerebrospinal fluid , Interleukin-1beta/cerebrospinal fluid , Male , Neurons/drug effects , Niacin/pharmacology , Niacin/therapeutic use , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats, Wistar , Treatment Outcome , gamma-Aminobutyric Acid/cerebrospinal fluid
5.
Neurotox Res ; 36(2): 279-291, 2019 Aug.
Article En | MEDLINE | ID: mdl-29294239

The etiology of Parkinson's disease is not completely understood and is believed to be multifactorial. Neuronal disorders associated to oxidative stress and mitochondrial dysfunction are widely considered major consequences. The aim of this study was to investigate the effect of the synthetic arylidenmalonate derivative 5-(3,4-dihydroxybenzylidene)-2,2-dimethyl-1,3-dioxane-4,6-dione (KM-34), in oxidative stress and mitochondrial dysfunction induced by 6-hydroxydopamine (6-OHDA). Pretreatment (2 h) with KM-34 (1 and 10 µM) markedly attenuated 6-OHDA-induced PC12 cell death in a concentration-dependent manner. KM-34 also inhibited H2O2 generation, mitochondrial swelling, and membrane potential dissipation after 6-OHDA-induced mitochondrial damage. In vivo, KM-34 treatment (1 and 2 mg/Kg) reduced percentage of asymmetry (cylinder test) and increased the vertical exploration (open field) with respect to untreated injured animals; KM-34 also reduced glial fibrillary acidic protein overexpression and increased tyrosine hydroxylase-positive cell number, both in substantia nigra pars compacta. These results demonstrate that KM-34 present biological effects associated to mitoprotection and neuroprotection in vitro, moreover, glial response and neuroprotection in SNpc in vivo. We suggest that KM-34 could be a putative neuroprotective agent for inhibiting the progressive neurodegenerative disease associated to oxidative stress and mitochondrial dysfunction.


Antioxidants/therapeutic use , Catechols/therapeutic use , Mitochondria/drug effects , Neuroprotective Agents/therapeutic use , Oxidopamine/toxicity , Parkinsonian Disorders/prevention & control , Animals , Antioxidants/pharmacology , Catechols/pharmacology , Dose-Response Relationship, Drug , Male , Mitochondria/metabolism , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Oxidative Stress/physiology , PC12 Cells , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/metabolism , Rats , Rats, Wistar
6.
Pharmacol Rep ; 70(4): 699-704, 2018 Aug.
Article En | MEDLINE | ID: mdl-29933207

Ischemic stroke is a major cause of death and disability worldwide. Thrombolysis by tissue plasminogen activator is the only pharmacological treatment approved for clinical practice, but has a narrow therapeutic window and poor efficacy when the cell death cascade is activated. Numerous drugs that are thought to protect neurons against injury have previously failed in human trials despite showing efficacy in experimental models of stroke. Herein, we reviewed the main pre-clinical results of the neuroprotective effects of JM-20, a new hybrid molecule, against brain ischemia. JM-20 appears to protect the brain from ischemic damage by interfering with several elements of the ischemic cascade: antiexcitotoxic, anticalcic, antioxidant, antiapoptotic, and anti-inflammatory. Its ability to protect not only neurons but also glial cells together with its ability to target and preserve mitochondrial function makes JM-20 a promising molecule that may be able to shield the whole neurovascular unit. The multimodal and multi-cell action of JM-20 may explain the high degree of protection observed in a rat model of brain ischemia, as assayed through histological (hematoxylin-eosin, and luxol fast blue staining), neurochemical (glutamate and aspartate levels in cerebrospinal fluid), mitochondrial functionality and behavioural (neurological scale) analysis at doses of 4 and 8mg/kg. Furthermore, the wide therapeutic window of JM-20 of 8h also suggests that this molecule could be of potential interest in situations where brain perfusion is compromised.


Benzodiazepines/pharmacology , Brain Ischemia/prevention & control , Niacin/analogs & derivatives , Animals , Drug Evaluation, Preclinical , Neuroprotective Agents/pharmacology , Niacin/pharmacology
7.
Drug Res (Stuttg) ; 68(5): 263-269, 2018 May.
Article En | MEDLINE | ID: mdl-29100263

Free radicals are important mediators in a number of neurodegenerative diseases and molecules capable of scavenging reactive oxygen species (ROS) may be a feasible strategy for protecting neuronal cells. In this sense, polyphenols have been studied for their antioxidant effects, KM-34 (5-(3, 4-dydroxyl-benzylidene)-2, 2-dimethyl-1, 3-dioxane-4, 6-Dione) is a novel synthetic catechol with potential neuroprotective and antioxidant properties. We have assessed the antioxidant (as scavenging and iron-chelating compound) and neuroprotectant in vitro (in PC12 cell injury induced by H2O2, glutamate or FeSO4/AA) of KM-34. KM-34 was found to be a potent antioxidant, as shown by (i) inhibition of iron induced-brain lipid peroxidation, (ii) inhibition of 2-deoxyribose degradation, (iii) inhibition of superoxide radicals generation (IC50=11.04 µM) and (iv) inhibition of 1,1-diphenyl-2-picrylhydrazyl (DPPH) radical reduction (IC50=16.26 µM). The overall anti-oxidant action of KM-34 appears to be a combination of a direct reaction with free radicals and chelating the metal ions responsible for the production of ROS. Our work suggests that the antioxidant properties of KM-34 may provide future therapeutic approaches for neurodegenerative disorders.


Antioxidants/pharmacology , Catechols/chemical synthesis , Catechols/pharmacology , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Animals , Cell Survival/drug effects , Cells, Cultured , Free Radical Scavengers/pharmacology , Lipid Peroxidation/drug effects , Male , Rats
8.
Eur J Pharmacol ; 819: 198-206, 2018 Jan 15.
Article En | MEDLINE | ID: mdl-29221949

This study aims to examine the effects of a new 1,4-dihydropyridine derivative, VdiE-2N, on cell signaling pathways and mitochondrial events in head and neck squamous cell carcinoma (HNSCC) cells, and on a mice model of xenograft tumor growth/cell proliferation. Four HNSCC cell lines (HN13, HN12, HN6, and CAL27), HEK293 cells (human embryonic kidney 293 cells), and human oral healthy mucosa fibroblasts (OHMF) were used for in vitro assessment of cell viability (resazurin assay) and invasion capacity (modified Boyden chamber assay), and mitochondrial membrane potential (JC-1 fluorescence assay), morphology (transmission electron microscopy), and number of mitochondria (MitoTracker® imaging). SET and pDRP1 proteins were analyzed by immunofluorescence, and proteins involved in cell death/survival pathways were analyzed by Western blotting. HN12 xenograft tumors were established in the flank of Balb/c nude mice, and their characteristics and sensitivity to VdiE-2N were determined by immunohistochemistry and histology. VdiE-2N decreased cell viability in HNSCC cells (IC50 = 9.56 and 22.45µM for HN13 and HN12 cells, respectively) more strongly than it decreased cell viability in OHMF and HEK293 cells (IC50 = 32.90 and > 50µM, respectively). In HN13 cells, VdiE-2N dissipated mitochondrial membrane potential and altered the mitochondria size, shape, and number in a concentration-dependent manner, as well as it induced apoptosis and reduced their invasion capacity. Treatment of mice bearing xenograft tumors with VdiE-2N significantly diminished proliferation of cancer cells. Therefore, VdiE-2N induces HNSCC cell death in vitro through mitochondria-mediated apoptotic pathways and dampens tumor growth in vivo, thus supporting a potential anti-cancer effect.


Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/pathology , Dihydropyridines/chemistry , Dihydropyridines/pharmacology , Head and Neck Neoplasms/pathology , Animals , Apoptosis/drug effects , Carcinoma, Squamous Cell/blood supply , Carcinoma, Squamous Cell/drug therapy , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Genes, myc/genetics , Head and Neck Neoplasms/blood supply , Head and Neck Neoplasms/drug therapy , Humans , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondrial Size/drug effects , Neoplasm Invasiveness , Neovascularization, Pathologic/drug therapy , Squamous Cell Carcinoma of Head and Neck , Xenograft Model Antitumor Assays
9.
Neurochem Int ; 90: 215-23, 2015 Nov.
Article En | MEDLINE | ID: mdl-26361722

Cerebral ischemia is the third most common cause of death and a major cause of disability worldwide. Beyond a shortage of essential metabolites, ischemia triggers many interconnected pathophysiological events, including excitotoxicity, oxidative stress, inflammation and apoptosis. Here, we investigated the neuroprotective mechanisms of JM-20, a novel synthetic molecule, focusing on the phosphoinositide-3-kinase (PI3K)/Akt survival pathway and glial cell response as potential targets of JM-20. For this purpose, we used organotypic hippocampal slice cultures exposed to oxygen-glucose deprivation (OGD) to achieve ischemic/reperfusion damage in vitro. Treatment with JM-20 at 0.1 and 10 µM reduced PI incorporation (indicative of cell death) after OGD. OGD decreased the phosphorylation of Akt (pro-survival) and GSK 3ß (pro-apoptotic), resulting in respective inhibition and activation of these proteins. Treatment with JM20 prevented the reduced phosphorylation of these proteins after OGD, representing a shift from pro-apoptotic to pro-survival signaling. The OGD-induced activation of caspase-3 was also attenuated by JM-20 treatment at 10 µM. Moreover, in cultures treated with JM-20 and exposed to OGD conditioning, we observed a decrease in activated microglia, as well as a decrease in interleukin (IL)-1ß, IL-6 and tumor necrosis factor (TNF)-α release into the culture medium, while the level of the anti-inflammatory IL-10 increased. GFAP immunostaining and IB4 labeling showed that JM-20 treatment significantly augmented GFAP immunoreactivity after OGD, when compared with cultures exposed to OGD only, suggesting the activation of astroglial cells. Our results confirm that JM-20 has a strong neuroprotective effect against ischemic injury and suggest that the mechanisms involved in this effect may include the modulation of reactive astrogliosis, as well as neuroinflammation and the anti-apoptotic cell signaling pathway.


Benzodiazepines/pharmacology , Cell Death/drug effects , Glycogen Synthase Kinase 3/metabolism , Hippocampus/drug effects , Niacin/analogs & derivatives , Oxygen/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Animals, Newborn , Glucose/metabolism , Glycogen Synthase Kinase 3 beta , Hippocampus/metabolism , Male , Neurons/drug effects , Neuroprotection/drug effects , Neuroprotective Agents/pharmacology , Niacin/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Rats, Wistar
10.
Neurochem Int ; 81: 41-7, 2015 Feb.
Article En | MEDLINE | ID: mdl-25617730

JM-20 (3-ethoxycarbonyl-2-methyl-4-(2-nitrophenyl)-4,11-dihydro-1H-pyrido[2,3-b][1,5]benzodiazepine) is a novel benzodiazepine dihydropyridine hybrid molecule, which has been shown to be a neuroprotective agent in brain disorders involving glutamate receptors. However, the effect of JM-20 on the functionality of the glutamatergic system has not been investigated. In this study, by using different in vitro preparations, we investigated the effects of JM-20 on (i) rat brain synaptic vesicles (L-[(3)H]-glutamate uptake, proton gradient built-up and bafilomycin-sensitive H(+)-ATPase activity), (ii) rat brain synaptosomes (glutamate release) and (iii) primary cultures of rat cortical neurons, astrocytes and astrocyte-neuron co-cultures (L-[(3)H]-glutamate uptake and glutamate release). We observed here that JM-20 impairs H(+)-ATPase activity and consequently reduces vesicular glutamate uptake. This molecule also inhibits glutamate release from brain synaptosomes and markedly increases glutamate uptake in astrocytes alone, and co-cultured neurons and astrocytes. The impairment of vesicular glutamate uptake by inhibition of the H(+)-ATPase caused by JM-20 could decrease the amount of the transmitter stored in synaptic vesicles, increase the cytosolic levels of glutamate, and will thus down-regulate neurotransmitter release. Together, these results contribute to explain the anti-excitotoxic effect of JM-20 and its strong neuroprotective effect observed in different in vitro and in vivo models of brain ischemia.


Benzodiazepines/pharmacology , Brain/drug effects , Glutamic Acid/metabolism , Neurons/drug effects , Niacin/analogs & derivatives , Synaptic Vesicles/drug effects , Synaptosomes/drug effects , Animals , Brain/cytology , Brain/metabolism , Cells, Cultured , Male , Neurons/metabolism , Niacin/pharmacology , Rats , Rats, Wistar , Synaptic Vesicles/metabolism , Synaptosomes/metabolism
11.
Brain Res Bull ; 109: 68-76, 2014 Oct.
Article En | MEDLINE | ID: mdl-25305343

Because mitochondrial oxidative stress and impairment are important mediators of neuronal damage in neurodegenerative diseases and in brain ischemia/reperfusion, in the present study, we evaluated the antioxidant and mitoprotective effect of a new promising neuroprotective molecule, JM-20, in mitochondria and synaptosomes isolated from rat brains. JM-20 inhibited succinate-mediated H2O2 generation in both mitochondria and synaptosomes incubated in depolarized (high K(+)) medium at extremely low micromolar concentration and with identical IC50 values of 0.91 µM. JM-20 also repressed glucose-induced H2O2 generation stimulated by rotenone or by antimycin A in synaptosomes incubated in high sodium-polarized medium at extremely low IC50 values of 0.395 µM and 2.452 µM, respectively. JM-20 was unable to react directly with H2O2 or with superoxide anion radicals but displayed a cathodic reduction peak at -0.71V, which is close to that of oxygen (-0.8V), indicating high electron affinity. JM-20 also inhibited uncoupled respiration in mitochondria or synaptosomes and was a more effective inhibitor in the presence of the respiratory substrates glutamate/malate than in the presence of succinate. JM-20 also prevented Ca(2+)-induced mitochondrial permeability transition pore opening, membrane potential dissipation and cytochrome c release, which are key pathogenic events during stroke. This molecule also prevented Ca(2+) influx into synaptosomes and mitochondria; the former effect was a consequence of the latter because JM-20 inhibition followed the patterns of carbonyl cyanide p-trifluoromethoxyphenyl hydrazone (FCCP), which is a classic mitochondrial uncoupler. Because the mitochondrion is considered an important source and target of neuronal cell death signaling after an ischemic insult, the antioxidant and protective effects of JM-20 against the deleterious effects of Ca(2+) observed at the mitochondrial level in this study may endow this molecule with the ability to succeed in mitochondrion-targeted strategies to combat ischemic brain damage.


Antioxidants/pharmacology , Benzodiazepines/pharmacology , Calcium/toxicity , Mitochondria/drug effects , Niacin/analogs & derivatives , Prosencephalon/ultrastructure , Synaptosomes/drug effects , Adenosine Triphosphate/metabolism , Animals , Catalase/pharmacology , Cytochromes c/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Male , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Niacin/pharmacology , Oligomycins/pharmacology , Oxygen/metabolism , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Superoxides/metabolism
12.
Neuropharmacology ; 85: 517-27, 2014 Oct.
Article En | MEDLINE | ID: mdl-24953828

We previously showed that JM-20, a novel 1,5-benzodiazepine fused to a dihydropyridine moiety, possessed an anxiolytic profile similar to diazepam and strong neuroprotective activity in different cell models relevant to cerebral ischemia. Here, we investigated whether JM-20 protects against ischemic neuronal damage in vitro and in vivo. The effects of JM-20 were evaluated on hippocampal slices subjected to oxygen and glucose deprivation (OGD). For in vivo studies, Wistar rats were subjected 90 min of middle cerebral artery occlusion (MCAo) and oral administration of JM-20 at 2, 4 and 8 mg/kg 1 h following reperfusion. Twenty-four hours after cerebral blood flow restoration, neurological deficits were scored, and the infarct volume, histopathological changes in cortex, number of hippocampal and striatal neurons, and glutamate/aspartate concentrations in the cerebrospinal fluid were measured. Susceptibility to brain mitochondrial swelling, membrane potential dissipation, H2O2 generation, cytochrome c release, Ca2+ accumulation, and morphological changes in the organelles were assessed 24 h post-ischemia. In vitro, JM-20 (1 and 10 µM) administered during reperfusion significantly reduced cell death in hippocampal slices subjected to OGD. In vivo, JM-20 treatment (4 and 8 mg/kg) significantly decreased neurological deficit scores, edema formation, total infarct volumes and histological alterations in different brain regions. JM-20 treatment also protected brain mitochondria from ischemic damage, most likely by preventing Ca2+ accumulation in organelles. Moreover, an 8-mg/kg JM-20 dose reduced glutamate and aspartate concentrations in cerebrospinal fluid and the deleterious effects of MCAo even when delivered 8 h after blood flow restoration. These results suggest that in rats, JM-20 is a robust neuroprotective agent against ischemia/reperfusion injury with a wide therapeutic window. Our findings support the further examination of potential clinical JM-20 use to treat acute ischemic stroke.


Benzodiazepines/pharmacology , Brain Ischemia/drug therapy , Excitatory Amino Acids/metabolism , Mitochondria/drug effects , Neuroprotective Agents/pharmacology , Niacin/analogs & derivatives , Animals , Brain/drug effects , Brain/pathology , Brain/physiopathology , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Cell Death/drug effects , Cell Death/physiology , Disease Models, Animal , Glucose/deficiency , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/physiopathology , Male , Mitochondria/pathology , Mitochondria/physiology , Niacin/pharmacology , Random Allocation , Rats, Wistar , Tissue Culture Techniques
13.
Eur J Pharmacol ; 726: 57-65, 2014 Mar 05.
Article En | MEDLINE | ID: mdl-24462350

The ischemic stroke cascade is composed of several pathophysiological events, providing multiple targets for pharmacological intervention. JM-20 (3-ethoxycarbonyl-2-methyl-4-(2-nitrophenyl)-4,11-dihydro-1H-pyrido[2,3-b][1,5]benzodiazepine) is a novel hybrid molecule, in which a benzodiazepine portion is covalently linked to a dihydropyridine ring, forming a new chemical entity with potential multisite neuroprotective activity. In the present study, JM-20 prevented PC-12 cell death induced either by glutamate, hydrogen peroxide or KCN-mediated chemical hypoxia. This molecule also protected cerebellar granule neurons from glutamate or glutamate plus pentylenetetrazole-induced damage at very low micromolar concentrations. In rat liver mitochondria, JM-20, at low micromolar concentrations, prevented the Ca2+-induced mitochondrial permeability transition, as assessed by mitochondrial swelling, membrane potential dissipation and organelle release of the pro-apoptotic protein cytochrome c. JM-20 also inhibited the mitochondrial hydrolytic activity of F1F0-ATP synthase and Ca2+ influx. Therefore, JM-20 may be a multi-target neuroprotective agent, promoting reductions in neuronal excitotoxic injury and the protection of the mitochondria from Ca2+-induced impairment as well as the preservation of cellular energy balance.


Benzodiazepines/chemistry , Benzodiazepines/pharmacology , Brain Ischemia/pathology , Dihydropyridines/chemistry , Mitochondria/drug effects , Neurons/drug effects , Neurons/pathology , Niacin/analogs & derivatives , Animals , Brain Ischemia/complications , Calcium/metabolism , Cell Death/drug effects , Cerebellum/cytology , Cytochromes c/metabolism , Glutamic Acid/pharmacology , Hydrogen Peroxide/pharmacology , Hydrolysis/drug effects , Liver/drug effects , Liver/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Mitochondrial Proton-Translocating ATPases/antagonists & inhibitors , Mitochondrial Swelling/drug effects , Neurons/metabolism , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Niacin/chemistry , Niacin/pharmacology , Oxidative Stress/drug effects , PC12 Cells , Pentylenetetrazole/pharmacology , Phosphates/metabolism , Potassium Cyanide/pharmacology , Rats , Stroke/complications
14.
Rev. cuba. farm ; 40(3)sept.-dic. 2006. tab
Article Es | LILACS | ID: lil-465280

Se prepararon cinco hexahidroquinoleínas por una síntesis en un solo paso, a partir de un aldehído aromático, el ß -aminocrotonato de alquilo (enamina) y dimedona en etanol absoluto como disolvente. Se caracterizaron los efectos de las cinco hexahidroquinoleínas sobre la contractilidad en anillos de aorta de conejo y sobre la contractilidad de músculos papilares de ventrículo derecho de rata, de manera comparativa con la clásica nifedipina. Todas las hexahidroquinoleínas inhibieron la actividad contráctil tanto en músculo liso vascular como en músculo cardíaco, y mostraron efectos característicos de compuestos antagonistas del calcio, aunque con menor potencia de acción que la nifedipina. Sin embargo, dos de las hexahidroquinoleínas estudiadas presentaron cierta vasoselectividad, aunque no marcada


Calcium , Chemical Compounds , Dihydropyridines
15.
Rev. cuba. farm ; 40(3)sep.-dic. 2006. tab
Article Es | CUMED | ID: cum-33403

Se prepararon cinco hexahidroquinoleínas por una síntesis en un solo paso, a partir de un aldehído aromático, el ß -aminocrotonato de alquilo (enamina) y dimedona en etanol absoluto como disolvente. Se caracterizaron los efectos de las cinco hexahidroquinoleínas sobre la contractilidad en anillos de aorta de conejo y sobre la contractilidad de músculos papilares de ventrículo derecho de rata, de manera comparativa con la clásica nifedipina. Todas las hexahidroquinoleínas inhibieron la actividad contráctil tanto en músculo liso vascular como en músculo cardíaco, y mostraron efectos característicos de compuestos antagonistas del calcio, aunque con menor potencia de acción que la nifedipina. Sin embargo, dos de las hexahidroquinoleínas estudiadas presentaron cierta vasoselectividad, aunque no marcada(AU)


Calcium/antagonists & inhibitors , Dihydropyridines , Chemical Compounds
...