Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters











Publication year range
1.
Pharmaceutics ; 15(12)2023 Nov 29.
Article in English | MEDLINE | ID: mdl-38140038

ABSTRACT

Progesterone (P4) is a neuroactive hormone having pleiotropic effects, supporting its pharmacological potential to treat global (cardiac-arrest-related) cerebral ischemia, a condition associated with an elevated risk of dementia. This review examines the current biochemical, morphological, and functional evidence showing the neuroprotective/neurorestorative effects of P4 against global cerebral ischemia (GCI). Experimental findings show that P4 may counteract pathophysiological mechanisms and/or regulate endogenous mechanisms of plasticity induced by GCI. According to this, P4 treatment consistently improves the performance of cognitive functions, such as learning and memory, impaired by GCI. This functional recovery is related to the significant morphological preservation of brain structures vulnerable to ischemia when the hormone is administered before and/or after a moderate ischemic episode; and with long-term adaptive plastic restoration processes of altered brain morphology when treatment is given after an episode of severe ischemia. The insights presented here may be a guide for future basic research, including the study of P4 administration schemes that focus on promoting its post-ischemia neurorestorative effect. Furthermore, considering that functional recovery is a desired endpoint of pharmacological strategies in the clinic, they could support the study of P4 treatment for decreasing dementia in patients who have suffered an episode of GCI.

2.
Neurotox Res ; 41(4): 311-323, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36922461

ABSTRACT

Phosphodiesterase 4 inhibitors (PDE4-I), which selectively increase cyclic adenosine monophosphate (cAMP) levels, have shown neuroprotective effects after several neurological injuries inducing blood-brain barrier (BBB) damage including local/focal cerebral ischemia. The present investigated whether roflumilast confers BBB neuroprotection in the hippocampus after transient global cerebral ischemia (TGCI) in rats. TGCI resulted in whole BBB disruption as measured by the increase of Evans blue (EB) and IgG extravasation, neurodegeneration, and downregulation of claudin-5 and endothelial nitric oxide synthase (eNOS) levels in the CA1 hippocampal subfield of ischemic rats. Roflumilast attenuated BBB disruption and restored the levels of eNOS in the CA1 hippocampal area. Moreover, roflumilast increased the levels of B2 cell lymphoma (BcL-2) and neuron-glial antigen-2 (NG2) in the CA1 subfield after global ischemia in rats. The protective effects of roflumilast against TGCI-induced BBB breakdown might involve preservation of BBB integrity, vascularization and angiogenesis, and myelin repair.


Subject(s)
Brain Ischemia , Ischemic Attack, Transient , Rats , Animals , Blood-Brain Barrier/metabolism , Brain Ischemia/metabolism , Hippocampus/metabolism
3.
Mol Neurobiol ; 58(10): 5338-5355, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34302281

ABSTRACT

Evidence for the clinical use of neuroprotective drugs for the treatment of cerebral ischemia (CI) is still greatly limited. Spatial/temporal disorientation and cognitive dysfunction are among the most prominent long-term sequelae of CI. Cannabidiol (CBD) is a non-psychotomimetic constituent of Cannabis sativa that exerts neuroprotective effects against experimental CI. The present study investigated possible neuroprotective mechanisms of action of CBD on spatial memory impairments that are caused by transient global cerebral ischemia (TGCI) in rats. Hippocampal synaptic plasticity is a fundamental mechanism of learning and memory. Thus, we also evaluated the impact of CBD on neuroplastic changes in the hippocampus after TGCI. Wistar rats were trained to learn an eight-arm aversive radial maze (AvRM) task and underwent either sham or TGCI surgery. The animals received vehicle or 10 mg/kg CBD (i.p.) 30 min before surgery, 3 h after surgery, and then once daily for 14 days. On days 7 and 14, we performed a retention memory test. Another group of rats that received the same pharmacological treatment was tested in the object location test (OLT). Brains were removed and processed to assess neuronal degeneration, synaptic protein levels, and dendritic remodeling in the hippocampus. Cannabidiol treatment attenuated ischemia-induced memory deficits. In rats that were subjected to TGCI, CBD attenuated hippocampal CA1 neurodegeneration and increased brain-derived neurotrophic factor levels. Additionally, CBD protected neurons against the deleterious effects of TGCI on dendritic spine number and the length of dendritic arborization. These results suggest that the neuroprotective effects of CBD against TGCI-induced memory impairments involve changes in synaptic plasticity in the hippocampus.


Subject(s)
Cannabidiol/therapeutic use , Hippocampus/drug effects , Ischemic Attack, Transient/prevention & control , Neuronal Plasticity/drug effects , Neuroprotection/drug effects , Synapses/drug effects , Animals , Cannabidiol/pharmacology , Disease Models, Animal , Hippocampus/metabolism , Hippocampus/pathology , Ischemic Attack, Transient/metabolism , Ischemic Attack, Transient/pathology , Male , Neuronal Plasticity/physiology , Neuroprotection/physiology , Organ Culture Techniques , Rats , Rats, Wistar , Spatial Memory/drug effects , Spatial Memory/physiology , Synapses/metabolism , Synapses/pathology
4.
Eur J Neurosci ; 53(4): 1171-1188, 2021 02.
Article in English | MEDLINE | ID: mdl-33340424

ABSTRACT

Phosphodiesterase 4 (PDE4) inhibitors have been shown to present beneficial effects in cerebral ischemic injury because of their ability to improve cognition and target different phases and mechanisms of cerebral ischemia, including apoptosis, neurogenesis, angiogenesis, and inflammation. The present study investigated whether repeated treatment with the PDE4 inhibitor roflumilast rescued memory loss and attenuated neuroinflammation in rats following transient global cerebral ischemia (TGCI). TGCI caused memory impairments, neuronal loss (reflected by Neuronal nuclei (NeuN) immunoreactivity), and compensatory neurogenesis (reflected by doublecortin (DCX) immunoreactivity) in the hippocampus. Also, increases in the protein expression of the phosphorylated response element-binding protein (pCREB) and inflammatory markers such as the glial fibrillary acidic protein (GFAP) and ionized calcium-binding adaptor molecule 1 (Iba-1), were detected in the hippocampus in TGCI rats. Repeated treatment with roflumilast (0.003 and 0.01 mg/kg) prevented spatial memory deficits without promoting hippocampal protection in ischemic animals. Roflumilast increased the levels of pCREB, arginase-1, interleukin (IL) 4, and IL-10 in the hippocampus 21 days after TGCI. These data suggest a protective effect of roflumilast against functional sequelae of cerebral ischemia, which might be related to its anti-inflammatory properties.


Subject(s)
Brain Ischemia , Ischemic Attack, Transient , Aminopyridines/pharmacology , Aminopyridines/therapeutic use , Animals , Benzamides , Brain Ischemia/drug therapy , Cyclopropanes , Doublecortin Protein , Hippocampus , Rats , Spatial Memory
5.
Biochim Biophys Acta Mol Basis Dis ; 1866(12): 165934, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32827650

ABSTRACT

Cerebral ischemia-induced hyperglycemia has been reported to accentuate neurological damage following focal or global cerebral ischemia. Hyperglycemia found in rats following focal brain ischemia occurs in the first 24 h and has been claimed to be caused by increased liver gluconeogenesis and insulin resistance. However, liver gluconeogenesis and the mechanisms leading to hyperglycemia after global cerebral ischemia remain uncertain. This study investigated the glycemic homeostasis and hepatic metabolism in rats after transient four-vessel occlusion (4-VO)-induced global cerebral ischemia, an event that mimics to a certain degree the situation during cardiac arrest. Several metabolic fluxes were measured in perfused livers. Activities and mRNA expressions of hepatic glycolysis and glyconeogenesis rate-limiting enzymes were assessed as well as respiratory activity of hepatic isolated mitochondria. Global cerebral ischemia was associated with hyperglycemia and hyperinsulinemia 24 h after ischemia. Insulin resistance developed later and was prominent after the 5th day. Hepatic anabolism and catabolism were both modified in a complex and time-dependent way. Gluconeogenesis, ß-oxidation, ketogenesis and glycolysis were diminished at 24 h after ischemia. At 5 days after ischemia glycolysis had normalized, but gluconeogenesis, ketogenesis and ß-oxidation were accelerated. The overall metabolic modifications suggest that a condition of depressed metabolism was established in response to the new conditions generated by the cerebral global ischemia. Whether the modifications in the liver metabolism found in rats after the ischemic insult can be translated to individuals following global brain ischemia remains uncertain, but the results of this study are hoped to encourage further investigations.


Subject(s)
Blood Glucose/metabolism , Brain Ischemia/metabolism , Homeostasis , Liver/metabolism , Animals , Male , Rats , Rats, Wistar
6.
J Chem Neuroanat ; 101: 101683, 2019 11.
Article in English | MEDLINE | ID: mdl-31499173

ABSTRACT

We previously found that fish oil (FO) facilitated memory recovery in the absence of pyramidal neuron rescue after transient, global cerebral ischemia (TGCI). Fish oil preserved the expression of microtubule-associated protein 2 (MAP-2), suggesting a relationship between dendritic plasticity and memory recovery that is mediated by FO after TGCI. The present study examined whether postischemic treatment with FO prevents ischemia-induced loss of dendritic processes in remaining pyramidal neurons. The effects of FO on neuroplasticity-related proteins were also examined after TGCI. Rats were subjected to TGCI (15 min, four-vessel occlusion model) and then received vehicle or FO (300 mg/kg docosahexaenoic acid) once daily for 7 days. The first dose was administered 4 h postischemia. Golgi-Cox staining was used to evaluate dentrict morphology in the pyramidal neurons of hippocampus (CA1 and CA3 subfields) and prefrontal cortex (PFC). Neuronal nuclei protein (NeuN), brain-derived neurotrophic factor (BDNF), growth-associated protein 43 (GAP-43), synaptophysin (SYP), and postsynaptic density protein 95 (PSD-95) levels were measured by Western blot in both structures. Fifteen minutes of TGCI reduced consistently the length of dendrites, number of dendritic branches and dendritic spine density (average of 25%, 43%, 32%, respectively) 7, 14, and 21 days postischemia, indicating that they did not recover spontaneously. This outcome of TGCI was reversed by FO treatment, an effect that was sustained even after treatment cessation. The NeuN and BDNF protein levels were reduced in both the hippocampus and PFC, which were recovered by FO treatment. GAP-43 protein levels decreased after ischemia in the PFC only, and this effect was also mitigated by FO. Neither SYP nor PSD-95 levels were altered by ischemia, but PDS-95 levels almost doubled after FO treatment in the ischemic group. These data support our hypothesis that synaptic plasticity at the level of dendrites may at least partially underlie the memory-protective effect of FO after TGCI and strengthen the possibility that FO has therapeutic potential for treating the sequelae of brain ischemia/reperfusion injury.


Subject(s)
Dendrites/drug effects , Fish Oils/pharmacology , Ischemic Attack, Transient/pathology , Neuronal Plasticity/drug effects , Synapses/drug effects , Animals , Dendrites/pathology , Docosahexaenoic Acids/pharmacology , Hippocampus/drug effects , Hippocampus/pathology , Male , Neuroprotective Agents/pharmacology , Rats , Rats, Wistar , Synapses/pathology
7.
Curr Pharm Biotechnol ; 19(6): 514-519, 2018.
Article in English | MEDLINE | ID: mdl-30019642

ABSTRACT

BACKGROUND: Stroke is a major cause of severe and long-term disability in adult individuals. Treatment of this disease is limited by the narrow therapeutic window in which intervention is crucial. An alternative therapy for stroke could be cellular growth factors, which participate in several pathways that mediate neuronal cell death. METHODS: We evaluated the neuroprotective ability of different doses of granulocyte colonystimulating factor (G-CSF; 5, 50 and 100 µg/kg/day) in the mouse model of global cerebral ischemia induced by bilateral occlusion of the common carotid arteries for 80 minutes. The control group received vehicle (5% glucose solution) and the treated group was administered with G-CSF at two postsurgery time-points: immediately after and 24 hours after. Subsequently, muscle strength, leukocyte count, infarcted cortical area, and apoptosis/TUNEL were evaluated. RESULTS: The global ischemia promoted an impairment of the strength (16%) and a cerebral infarction (0.437±0.08 cm2) which were accompanied by apoptosis evaluated by TUNEL in control mice. In mice treated with G-CSF the strength function was maintained, the infarcted area (~70%) and apoptosis were decreased in a similar magnitude in all treated groups. Accordingly, the cytokine activities were confirmed by blood leukocyte count that was increased approximately 2-fold than that observed in the control group. CONCLUSION: The results indicate a neuroprotective effect of G-CSF, even in small doses, in mice subjected to global cerebral ischemia, thereby reducing the neurofunctional impairment caused by stroke, when considering the maintenance of muscle strength in the treated animals.


Subject(s)
Brain Ischemia/drug therapy , Cerebral Infarction/drug therapy , Granulocyte Colony-Stimulating Factor/therapeutic use , Muscle Strength/drug effects , Neuroprotective Agents/therapeutic use , Stroke/drug therapy , Animals , Apoptosis/drug effects , Disease Models, Animal , Male , Mice
8.
J Ethnopharmacol ; 221: 109-118, 2018 Jul 15.
Article in English | MEDLINE | ID: mdl-29660468

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Trichilia catigua A. Juss (Meliaceae) preparations have been used in folk medicine to alleviate fatigue, stress, and improve memory. Antinociceptive, antiinflammatory, and in vitro neuroprotective effects have been observed in animals. Cerebral ischemia/reperfusion (I/R) leads to severe neuropsychological deficits that are largely associated with oxidative stress, inflammation and neurodegeneration. We reported previously that an ethyl-acetate fraction (EAF) of T. catigua reduced brain ischemia-induced learning and memory impairments in the absence of histological protection. AIM OF THE STUDY: Continuing those studies, here we aimed to investigate the antioxidant and antiinflammatory properties of T. catigua in an in vivo model of I/R. MATERIAL AND METHODS: Rats were subjected to 15 min of brain ischemia (4-VO model) followed by up to 15 days of reperfusion. Vehicle was given by gavage 30 min before ischemia and at 1 h of reperfusion. In a first experiment, brain ischemia-induced changes in oxidative stress markers, i.e., reduced glutathione (GSH), oxidized glutathione (GSSG), superoxide dismutase (SOD), catalase (CAT), malondialdehyde (MDA), and protein carbonyl groups (PCGs) were measured on days 1, 3, and 5 post-ischemia. Similar time course analysis was done for neuroinflammation markers, i.e., microglia (OX42 immunorreactivity) and astrocytes (GFAP immunorreactivity), in the hippocampus. In a second experiment, the time points at which these markers of oxidative stress and neuroinflammation peaked were used to test the effects of T. catigua (400 mg/kg, p.o.). RESULTS: Oxidative stress markers peaked on day 1 post-ischemia. GSH decreased (-23.2%) while GSSG increased (+ 71.1%), which yielded a significant reduction in the GSH/GSSG ratio (-39.1%). The activity of CAT was largely reduced by ischemia (-54.6% to -65.1%), while the concentration of PCG almost doubled in the brain of ischemic rats (+99.10%) in comparison to sham. Treatment with the EAF of T. catigua normalized these changes in oxidative markers to the control levels (GSH: +27.5%; GSSG: -23.8%; GSH/GSSG: +44.6%; PCG: -80.3%). In the hippocampus, neuroinflammation markers peaked on day 5 post-ischemia, with microglial and astrocytic responses increasing to 54.8% and 37.1%, respectively. The elevation in glial cells response was completely prevented by EAF. CONCLUSION: These results demonstrate that T. catigua has both antioxidant and antiinflammatory activities after transient global cerebral ischemia in rats, which may contribute to the previously reported memory protective effect of T. catigua.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Brain Ischemia/drug therapy , Meliaceae , Neuroprotective Agents/therapeutic use , Plant Extracts/therapeutic use , Reperfusion Injury/drug therapy , Acetates/chemistry , Animals , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Brain Ischemia/metabolism , CD11b Antigen/metabolism , Catalase/metabolism , Glial Fibrillary Acidic Protein/metabolism , Glutathione/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Male , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Phytotherapy , Plant Extracts/pharmacology , Plant Stems/chemistry , Rats, Wistar , Reperfusion Injury/metabolism , Solvents/chemistry , Superoxide Dismutase/metabolism
9.
Behav Brain Res ; 337: 173-182, 2018 Jan 30.
Article in English | MEDLINE | ID: mdl-28919157

ABSTRACT

We originally reported that an ethyl-acetate fraction (EAF) of Trichilia catigua prevented the impairment of water maze learning and hippocampal neurodegeneration after transient global cerebral (TGCI) in mice. We extended that previous study by evaluating whether T. catigua (i) prevents the loss of long-term retrograde memory assessed in the aversive radial maze (AvRM), (ii) confers hippocampal and cortical neuroprotection, and (iii) mitigates oxidative stress and neuroinflammation in rats that are subjected to the four vessel occlusion (4-VO) model of TGCI. In the first experiment, naive rats were trained in the AvRM and then subjected to TGCI. The EAF was administered orally 30min before and 1h after TGCI, and administration continued once per day for 7days post-ischemia. In the second experiment, the EAF was administered 30min before and 1h after TGCI, and protein carbonylation and myeloperoxidase (MPO) activity were assayed 24h and 5days later, respectively. Retrograde memory performance was assessed 8, 15, and 21days post-ischemia. Ischemia caused persistent retrograde amnesia, and this effect was prevented by T. catigua. This memory protection (or preservation) persisted even after the treatment was discontinued, despite the absence of histological neuroprotection. Protein carbonyl group content and MPO activity increased around 43% and 100%, respectively, after TGCI, which were abolished by the EAF of T. catigua. The administration of EAF did not coincide with the days of memory testing. The data indicate that antioxidant and/or antiinflammatory actions in the early phase of ischemia/reperfusion contribute to the long-term antiamnesic effect of T. catigua.


Subject(s)
Amnesia, Retrograde/drug therapy , Amnesia, Retrograde/etiology , Brain Ischemia/complications , Inflammation/drug therapy , Inflammation/etiology , Meliaceae/chemistry , Oxidative Stress/drug effects , Plant Extracts/therapeutic use , Animals , Biomarkers/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Disease Models, Animal , Hippocampus/metabolism , Maze Learning/drug effects , Plant Extracts/chemistry , Rats
10.
Physiol Behav ; 177: 196-207, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28483394

ABSTRACT

A series of our previous studies demonstrated that fish oil (FO), equivalent to 300mg/kg docosahexahenoic acid (DHA), facilitates memory recovery after transient, global cerebral ischemia (TGCI) in the aversive radial maze (AvRM). The present study sought to address two main issues: (i) whether the memory-protective effect of FO that has been observed in the AvRM can be replicated in the passive avoidance test (PAT) and object location test (OLT) and (ii) whether FO at doses that are lower than those used previously can also prevent TGCI-induced memory loss. In Experiment 1, naive rats were trained in the PAT, subjected to TGCI (4-vessel occlusion model), and tested for retrograde memory performance 8 and 15days after ischemia. Fish oil (300mg/kg/day DHA) was given orally for 8days. The first dose was delivered 4h postischemia. In Experiment 2, the rats were subjected to TGCI, treated with the same FO regimen, and then trained and tested in the OLT. In Experiment 3, the rats were trained in the AvRM, subjected to TGCI, administered FO (100, 200, and 300mg/kg DHA), and tested for memory performance up to 3weeks after TGCI. At the end of the behavioral tests, the brains were examined for neurodegeneration and neuroblast proliferation. All of the behavioral tests (PAT, OLT, and AvRM) were sensitive to ischemia, but only the AvRM was able to detect the memory-protective effect of FO. Ischemia-induced neurodegeneration and neuroblast proliferation were unaffected by FO treatment. These results suggest that (i) the beneficial effect of FO on memory recovery after TGCI is task-dependent, (ii) doses of FO<300mg/kg DHA can protect memory function in the radial maze, and (iii) cognitive recovery occurs in the absence of neuronal rescue and/or hippocampal neurogenesis.


Subject(s)
Fish Oils/pharmacology , Hippocampus/drug effects , Ischemic Attack, Transient/drug therapy , Memory Disorders/drug therapy , Neuroprotective Agents/pharmacology , Recovery of Function/drug effects , Animals , Antioxidants/pharmacology , Avoidance Learning/drug effects , Avoidance Learning/physiology , Basolateral Nuclear Complex/drug effects , Basolateral Nuclear Complex/pathology , Disease Models, Animal , Hippocampus/pathology , Ischemic Attack, Transient/complications , Ischemic Attack, Transient/pathology , Ischemic Attack, Transient/psychology , Male , Memory Disorders/etiology , Memory Disorders/pathology , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/psychology , Neurogenesis/drug effects , Neurons/drug effects , Neurons/pathology , Rats, Wistar , Recovery of Function/physiology , Spatial Memory/drug effects , Spatial Memory/physiology
11.
Behav Brain Res ; 311: 425-439, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27235715

ABSTRACT

We reported that fish oil (FO) prevented the loss of spatial memory caused by transient, global cerebral ischemia (TGCI), provided the treatment covered the first days prior to and after ischemia. Continuing these studies, trained rats were subjected to TGCI, and FO was administered for 10days, with a time window of efficacy (TWE) of 4, 8 or 12h post-ischemia. Retrograde memory was assessed up to 43days after TGCI. In another experiment, ischemic rats received FO with a 4- or 12-h TWE, and dendritic density was assessed in the hippocampus and cerebral cortex. The brain lipid profile was evaluated in sham-operated and ischemic rats that were treated with FO or vehicle with a 4-h TWE. Ischemia-induced retrograde amnesia was prevented by FO administration that was initiated with either a 4- or 8-h TWE. Fish oil was ineffective after a 12-h TWE. Independent of the TWE, FO did not prevent ischemic neuronal death. In the hippocampus, but not cerebral cortex, TGCI-induced dendritic loss was prevented by FO with a 4-h TWE but not 12-h TWE. The level of docosahexaenoic acid almost doubled in the hippocampus in ischemic, FO-treated rats (4-h TWE). The data indicate that (i) the anti-amnesic effect of FO can be observed with a TWE of up to 8h, (ii) the stimulation of dendritic neuroplasticity may have contributed to this effect, and (iii) DHA in FO may be the main active constituent in FO that mediates the cognitive and neuroplasticity effects on TGCI.


Subject(s)
Dendrites/drug effects , Fish Oils/administration & dosage , Hippocampus/drug effects , Ischemic Attack, Transient/drug therapy , Memory, Long-Term/drug effects , Neuroprotective Agents/administration & dosage , Amnesia, Retrograde/drug therapy , Amnesia, Retrograde/etiology , Amnesia, Retrograde/metabolism , Amnesia, Retrograde/pathology , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Dendrites/metabolism , Dendrites/pathology , Disease Models, Animal , Docosahexaenoic Acids/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Ischemic Attack, Transient/metabolism , Ischemic Attack, Transient/pathology , Ischemic Attack, Transient/psychology , Male , Memory, Long-Term/physiology , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/psychology , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Nootropic Agents/administration & dosage , Rats, Wistar , Time Factors
12.
Anat Sci Int ; 91(3): 226-37, 2016 Jun.
Article in English | MEDLINE | ID: mdl-25966656

ABSTRACT

Animal models of cerebral ischemia have typically been established and performed using young animals, even though cerebral ischemia (CI) affects primarily elderly patients. This situation represents a discrepancy that complicates the translation of novel therapeutic strategies for CI. Models of transient global CI using aged animals have demonstrated an apparent neuroprotective effect on CA1 hippocampal neurons; however, this effect is not completely understood. Our study used a model in which young (3-6 months) and aged (18-21 months) male Wistar rats were subjected to 15 min of transient global CI using the four-vessel occlusion (4 VO) model. We determined that the 4 VO model can be performed on aged rats with a slight increase in mortality rate. In aged rats, the morphological damage was completely established by the 4th day after reperfusion, displaying no difference from their younger counterparts. These results demonstrated the lack of a neuroprotective effect of aging on CA1 hippocampal neurons in aged male Wistar rats. This study determined and characterized the morphological damage to the CA1 area after 15 min of 4 VO in aged male Wistar rats, validating the use of this model in CI and aging research.


Subject(s)
Aging/pathology , Brain Ischemia/pathology , Brain/blood supply , Cerebrovascular Disorders/pathology , Disease Models, Animal , Animals , CA1 Region, Hippocampal/pathology , Male , Neurons/pathology , Rats, Wistar
13.
J Ethnopharmacol ; 172: 232-7, 2015 Aug 22.
Article in English | MEDLINE | ID: mdl-26099636

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Trichilia catigua preparations have antinociceptive, antiinflammatory, and neuroprotective activity. Recently, a neuroprotective role for T. catigua was proposed using an in vitro model of ischemia-reperfusion in rat hippocampal slices. The aim of the present study was to evaluate the effects of an ethyl-acetate fraction (EAF) of T. catigua, which has potent antioxidant activity, in mice subjected to an in vivo model of cerebral ischemia. MATERIAL AND METHODS: Male Swiss mice were subject to the bilateral common carotid occlusion (BCCAO) model of cerebral ischemia. The animals were orally administered the T. catigua EAF (200, 400, or 800 mg/kg) 30 min before and once per day for 7 days after BCCAO. Histological and behavioral outcomes were assessed using Nissl staining and the Morris water maze test of cognition, respectively. RESULTS: Mice that were subjected to BCCAO exhibited cognitive impairments in the Morris water maze. The spatial cognitive deficits were counteracted by T. catigua EAF administration (200-800 mg/kg). The T. catigua EAF significantly increased the number of intact-appearing Nissl-stained cells in the hippocampus in BCCAO mice. CONCLUSIONS: These results show that the T. catigua EAF promoted functional recovery, decreased the delayed hippocampal cell loss, and mitigated the ongoing neurodegenerative processes induced by BCCAO in mice.


Subject(s)
Cognition Disorders/drug therapy , Meliaceae/chemistry , Plant Extracts/pharmacology , Acetates/chemistry , Animals , Brain Ischemia/complications , Brain Ischemia/drug therapy , Brain Ischemia/pathology , Carotid Artery, Common/pathology , Carotid Stenosis/complications , Cell Death/drug effects , Cognition Disorders/etiology , Disease Models, Animal , Dose-Response Relationship, Drug , Hippocampus/pathology , Male , Maze Learning/drug effects , Mice , Plant Extracts/administration & dosage
14.
J Neurosci Res ; 93(8): 1240-9, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25702923

ABSTRACT

4-Hydroxy-3-methoxy-acetophenone (apocynin) is a naturally occurring methoxy-substitute catechol that is isolated from the roots of Apocynin cannabinum (Canadian hemp) and Picrorhiza kurroa (Scrophulariaceae). It has been previously shown to have antioxidant and neuroprotective properties in several models of neurodegenerative disease, including cerebral ischemia. The present study investigates the effects of apocynin on transient global cerebral ischemia (TGCI)-induced retrograde memory deficits in rats. The protective effects of apocynin on neurodegeneration and the glial response to TGCI are also evaluated. Rats received a single intraperitoneal injection of apocynin (5 mg/kg) 30 min before TGCI and were tested 7, 14, and 21 days later in the eight-arm aversive radial maze (AvRM). After behavioral testing, the hippocampi were removed for histological evaluation. The present results confirm that TGCI causes memory impairment in the AvRM and that apocynin prevents these memory deficits and attenuates hippocampal neuronal death in a sustained way. Apocynin also decreases OX-42 and glial fibrillary acidic protein immunoreactivity induced by TGCI. These findings support the potential role of apocynin in preventing neurodegeneration and cognitive impairments following TGCI in rats. The long-term protective effects of apocynin may involve inhibition of the glial response.


Subject(s)
Acetophenones/therapeutic use , Hippocampus/metabolism , Ischemic Attack, Transient/metabolism , Memory Disorders/metabolism , Neuroglia/metabolism , Neuroprotective Agents/therapeutic use , Acetophenones/pharmacology , Animals , Cell Death/drug effects , Cell Death/physiology , Hippocampus/drug effects , Ischemic Attack, Transient/drug therapy , Ischemic Attack, Transient/psychology , Male , Memory Disorders/drug therapy , Memory Disorders/psychology , Neuroglia/drug effects , Neuroprotective Agents/pharmacology , Rats , Rats, Wistar
15.
Behav Brain Res ; 265: 101-10, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24561066

ABSTRACT

We reported that fish oil (FO) abolishes retrograde amnesia consistently following transient global cerebral ischemia (TGCI) in young rats, provided it covered the first days prior to and after ischemia. Here, we further evaluated whether FO given post-ischemia in older rats (15-18 months old) is equally effective in facilitating memory recovery. We also tested whether the antiamnesic effect of FO observed after TGCI can be reproduced after chronic cerebral hypoperfusion (CCH). FO (300 mg/kg docosahexaenoic acid [DHA]) was delivered orally 4h after TGCI and continued once per day for 9 days. In the CCH group, FO treatment began soon after the first stage of 4-VO/ICA and continued daily for 43 days. Two weeks after surgery, the animals were tested for retrograde memory performance across 5 weeks. Both TGCI and CCH caused persistent memory impairment and hippocampal and cortical neurodegeneration. TGCI-induced retrograde amnesia was reversed by FO, an effect that was sustained for at least 5 weeks after discontinuing treatment. In contrast, the memory deficit caused by CCH remained unchanged after FO treatment. Both hippocampal and cortical damage was not alleviated by FO. We conclude that the FO-mediated antiamnesic effect following TGCI can be extended to older rats, even when the treatment begins 4h postischemia. Such efficacy was not reproduced after CCH. Therefore, the present results support the notion that FO may have therapeutic utility in treating learning/memory dysfunction after acute/transient cerebral ischemia and suggest that such benefits may not apply when a state of chronic cerebrovascular insufficiency is present.


Subject(s)
Amnesia, Retrograde/drug therapy , Fish Oils/therapeutic use , Ischemic Attack, Transient/complications , Neuroprotective Agents/therapeutic use , Amnesia, Retrograde/etiology , Analysis of Variance , Animals , Dose-Response Relationship, Drug , Ischemic Attack, Transient/mortality , Nerve Degeneration/drug therapy , Nerve Degeneration/etiology , Rats , Rats, Wistar , Reaction Time/drug effects
16.
Brain Res ; 1545: 23-34, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-24316245

ABSTRACT

In this study, the effects of progesterone (P4) on the immunoreactivity to the neurite growth inhibitor Nogo-A, its receptor (Ng-R), and its effector Rho-A in the rat hippocampus, in association with parameters of spatial learning and memory following global cerebral ischemia, were assessed. Adult male rats were subjected to global cerebral ischemia (15 min), and treated with P4 or its vehicle at 15 min, 2, 6, 24, 48 and 72 h of reperfusion. Immunoreactivity to Nogo-A, Ng-R, and Rho-A was evaluated at 24 h, 72 h or 7 d, or at 14 d of reperfusion after rats were tested in the Morris Water Maze (MWM). Global cerebral ischemia induced an increase in Nogo-A, Ng-R, and Rho-A immunoreactivities in the cell bodies of CA1 pyramidal neurons at 24h after global cerebral ischemia, peaking at 72 h, and persisting 14 d later. In addition, at 72 h, a strong immunoreactivity was observed in the hippocampal layers where dendritic arborizations of CA1 pyramidal neurons are located. Treatment with P4 reduced Nogo-A, Ng-R, and Rho-A immunoreactivities in CA1, particularly at 72 h of reperfusion. These effects of P4 were consistent with the parameters of a more efficient spatial learning and memory in the MWM, as compared to vehicle-treated rats. Overall results suggest the reduction of neurite growth inhibitory molecules Nogo-A, Ng-R, and Rho-A, as a part of the restorative effects of progesterone possibly allowing the plastic phenomena to occur, able to support the functional preservation of the hippocampus following global cerebral ischemia.


Subject(s)
Brain Ischemia/metabolism , Growth Inhibitors/metabolism , Hippocampus/metabolism , Myelin Proteins/metabolism , Progesterone/therapeutic use , Receptors, Cell Surface/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Brain Ischemia/drug therapy , GPI-Linked Proteins/metabolism , Hippocampus/drug effects , Male , Maze Learning/drug effects , Nogo Proteins , Nogo Receptor 1 , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Rats , Rats, Sprague-Dawley
17.
Physiol Behav ; 119: 61-71, 2013 Jul 02.
Article in English | MEDLINE | ID: mdl-23770426

ABSTRACT

We previously reported that long-term treatment with fish oil (FO) facilitates memory recovery after transient, global cerebral ischemia (TGCI), despite the presence of severe hippocampal damage. The present study tested whether this antiamnesic effect resulted from an action of FO on behavioral performance itself, or whether it resulted from an anti-ischemic action. Different treatment regimens were used that were distinguished from each other by their initiation or duration with regard to the onset of TGCI and memory assessment. Naive rats were trained in an eight-arm radial maze, subjected to TGCI (4-VO model, 15 min), and tested for memory performance up to 6 weeks after TGCI. Fish oil (docosahexaenoic acid, 300 mg/kg/day) was given orally according to one of the following regimens: regimen 1 (from 3 days prior to ischemia until 4 weeks post-ischemia), regimen 2 (from 3 days prior to ischemia until 1 week post-ischemia), and regimen 3 (from week 2 to week 5 post-ischemia). When administered according to regimens 1 and 2, FO abolished amnesia completely. This effect persisted for at least 5 weeks after discontinuing the treatment. Such an effect did not occur, however, in the group treated according to regimen 3. Hippocampal and cortical damage was not alleviated by FO. The present results demonstrate that FO-mediated memory recovery (or preservation) following TGCI is a reproducible, robust, and long-lasting effect. Moreover, such an effect was found with a relatively short period of treatment, provided it covered the first days prior to and after ischemia. This suggests that FO prevented amnesia by changing some acute, ischemia/reperfusion-triggered process and not by stimulating memory performance on its own.


Subject(s)
Docosahexaenoic Acids/therapeutic use , Ischemic Attack, Transient/diet therapy , Memory Disorders/diet therapy , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Docosahexaenoic Acids/administration & dosage , Drug Administration Schedule , Hippocampus/drug effects , Hippocampus/pathology , Ischemic Attack, Transient/complications , Ischemic Attack, Transient/pathology , Male , Maze Learning/drug effects , Memory Disorders/complications , Memory Disorders/pathology , Nerve Degeneration/diet therapy , Nerve Degeneration/pathology , Rats
SELECTION OF CITATIONS
SEARCH DETAIL