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










Publication year range
1.
Environ Pollut ; 295: 118677, 2022 Feb 15.
Article in English | MEDLINE | ID: mdl-34906594

ABSTRACT

Air pollution exposure positively correlates with increased cardiovascular morbidity and mortality rates, mainly due to myocardial infarction (MI). Herein, we aimed to study the metabolic mechanisms underlying this association, focusing on the evaluation of cardiac mitochondrial function and dynamics, together with its impact over MI progression. An initial time course study was performed in BALB/c mice breathing filtered air (FA) or urban air (UA) in whole-body exposure chambers located in Buenos Aires City downtown for up to 16 weeks (n = 8 per group and time point). After 12 weeks, lung inflammatory cell recruitment was evident in UA-exposed mice. Interestingly, impaired redox metabolism, characterized by decreased lung SOD activity and increased GSSG levels and NOX activity, precede local inflammation in this group. At this selected time point, additional mice were exposed to FA or UA (n = 12 per group) and alveolar macrophage PM uptake and nitric oxide (NO) production was observed in UA-exposed mice, together with increased pro-inflammatory cytokine levels (TNF-α and IL-6) in BAL and plasma. Consequently, impaired heart tissue oxygen metabolism and altered mitochondrial ultrastructure and function were observed in UA-exposed mice after 12 weeks, characterized by decreased active state respiration and ATP production rates, and enhanced mitochondrial H2O2 production. Moreover, disturbed cardiac mitochondrial dynamics was detected in this group. This scenario led to a significant increase in the area of infarcted tissue following myocardial ischemia reperfusion injury in vivo, from 43 ± 3% of the area at risk in mice breathing FA to 66 ± 4% in UA-exposed mice (n = 6 per group, p < 0.01), together with a sustained increase in LVEDP during myocardial reperfusion. Taken together, our data unravel cardiac mitochondrial mechanisms that contribute to the understanding of the adverse health effects of urban air pollution exposure, and ultimately highlight the importance of considering environmental factors in the development of cardiovascular diseases.


Subject(s)
Air Pollution , Myocardial Infarction , Air Pollution/analysis , Animals , Hydrogen Peroxide , Mice , Mitochondria , Myocardial Infarction/chemically induced , Particulate Matter/toxicity
2.
Adv Drug Alcohol Res ; 2: 10734, 2022.
Article in English | MEDLINE | ID: mdl-38390616

ABSTRACT

In the last few years, an increasing interest in the neuroprotective effect of cannabinoids has taken place. The aim of the present work was to study the effects of modulating cannabinoid receptor 1 (CB1) in the context of light induced retinal degeneration (LIRD), using an animal model that resembles many characteristics of human age-related macular degeneration (AMD) and other degenerative diseases of the outer retina. Sprague Dawley rats (n = 28) were intravitreally injected in the right eye with either a CB1 agonist (ACEA), or an antagonist (AM251). Contralateral eyes were injected with respective vehicles as controls. Then, rats were subjected to continuous illumination (12,000 lux) for 24 h. Retinas from 28 animals were processed by GFAP-immunohistochemistry (IHC), TUNEL technique, Western blotting (WB), or qRT-PCR. ACEA-treated retinas showed a significantly lower number of apoptotic nuclei in the outer nuclear layer (ONL), lower levels of activated Caspase-3 by WB, and lower levels of glial reactivity by both GFAP-IHC and WB. qRT-PCR revealed that ACEA significantly decreased the expression of Bcl-2 and CYP1A1. Conversely, AM251-treated retinas showed a higher number of apoptotic nuclei in the ONL, higher levels of activated Caspase-3 by WB, and higher levels of glial reactivity as determined by GFAP-IHC and WB. AM251 increased the expression of Bcl-2, Bad, Bax, Aryl hydrocarbon Receptor (AhR), GFAP, and TNFα. In summary, the stimulation of the CB1 receptor, previous to the start of the pathogenic process, improved the survival of photoreceptors exposed to LIRD. The modulation of CB1 activity may be used as a neuroprotective strategy in retinal degeneration and deserves further studies.

3.
Behav Brain Res ; 400: 113007, 2021 02 26.
Article in English | MEDLINE | ID: mdl-33171148

ABSTRACT

Cannabinoid receptor type 1 (CB1R) is the most abundant cannabinoid receptor in central nervous system. Clinical studies and animal models have shown that the attenuation of endocannabinoid system signaling correlates with the development of psychiatric disorders such as anxiety, depression and schizophrenia. In the present work, multiple behavioral tests were performed to evaluate behaviors related to anxiety and depression in CB1R+/- and CB1R-/-. CB1R+/- mice had anxiety-related behavior similar to wild type (CB1R+/+) mice, whereas CB1R-/- mice displayed an anxious-like phenotype, which indicates that lower expression of CB1R is sufficient to maintain the neural circuits modulating anxiety. In addition, CB1R-/- mice exhibited alterations in risk assessment and less exploration, locomotion, grooming, body weight and appetite. These phenotypic characteristics observed in CB1R-/- mice could be associated with symptoms observed in human psychiatric disorders such as depression. A better knowledge of the neuromodulatory role of CB1R may contribute to understand scope and limitations of the development of medical treatments.


Subject(s)
Anxiety/metabolism , Anxiety/physiopathology , Behavior, Animal/physiology , Depression/metabolism , Depression/physiopathology , Motivation/physiology , Receptor, Cannabinoid, CB1/physiology , Animals , Disease Models, Animal , Male , Mice , Mice, Transgenic , Receptor, Cannabinoid, CB1/genetics
4.
Ecotoxicol Environ Saf ; 205: 111186, 2020 Dec 01.
Article in English | MEDLINE | ID: mdl-32853868

ABSTRACT

Exposure to ambient air particulate matter (PM) is associated with increased cardiorespiratory morbidity and mortality. In this context, alveolar macrophages exhibit proinflammatory and oxidative responses as a result of the clearance of particles, thus contributing to lung injury. However, the mechanisms linking these pathways are not completely clarified. Therefore, the oxinflammation phenomenon was studied in RAW 264.7 macrophages exposed to Residual Oil Fly Ash (ROFA), a PM surrogate rich in transition metals. While cell viability was not compromised under the experimental conditions, a proinflammatory phenotype was observed in cells incubated with ROFA 100 µg/mL, characterized by increased levels of TNF-α and NO production, together with PM uptake. This inflammatory response seems to precede alterations in redox metabolism, characterized by augmented levels of H2O2, diminished GSH/GSSG ratio, and increased SOD activity. This scenario resulted in increased oxidative damage to phospholipids. Moreover, alterations in mitochondrial respiration were observed following ROFA incubation, such as diminished coupling efficiency and spare respiratory capacity, together with augmented proton leak. These findings were accompanied by a decrease in mitochondrial membrane potential. Finally, NADPH oxidase (NOX) and mitochondria were identified as the main sources of superoxide anion () in our model. These results indicate that PM exposure induces direct activation of macrophages, leading to inflammation and increased reactive oxygen species production through NOX and mitochondria, which impairs antioxidant defense and may cause mitochondrial dysfunction.


Subject(s)
Macrophages, Alveolar/drug effects , Mitochondria/drug effects , NADPH Oxidases/metabolism , Oxidative Stress/drug effects , Particulate Matter/toxicity , Superoxides/metabolism , Air Pollutants/toxicity , Animals , Antioxidants/metabolism , Coal Ash/toxicity , Hydrogen Peroxide/metabolism , Inflammation , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Mice , Mitochondria/immunology , Mitochondria/metabolism , Oxidation-Reduction , Oxidative Stress/immunology , RAW 264.7 Cells , Tumor Necrosis Factor-alpha/metabolism
5.
Cannabis Cannabinoid Res ; 5(2): 150-163, 2020 Jun 01.
Article in English | MEDLINE | ID: mdl-32656347

ABSTRACT

Introduction: Lafora disease (LD) is a rare form of progressive infantile epilepsy in which rapid neurological deterioration occurs as the disease advances, leading the patients to a vegetative state and then death, usually within the first decade of disease onset. Based on the capacity of the endogenous cannabinoid system (ECS) to modulate several cellular processes commonly altered in many neurodegenerative processes, as well as the antiepileptic properties of certain natural cannabinoids, the aim of this study was to evaluate the role of the ECS in LD progression. Materials and Methods: We tested whether a natural cannabis extract highly enriched in cannabidiol (CBD) might be effective in curbing the pathological phenotype of malin knockout (KO) mice as an animal model of LD. Results: Our results reveal for the first time that alterations in the ECS occur during the evolution of LD, mainly at the level of CB1, CB2, and G protein-coupled receptor 55 (GPR55) receptor expression, and that a CBD-enriched extract (CBDext) is able to reduce the cognitive impairment exhibited by malin KO mice. However, in contrast to what has previously been reported for other kinds of refractory epilepsy in childhood, the CBD-enriched extract does not reduce the severity of the epileptic seizures induced in this animal model of LD. Conclusions: In summary, this study reveals that the ECS might play a role in LD and that a CBD-enriched extract partially reduces the dementia-like phenotype, but not the increased vulnerability to epileptic seizures, exhibited by an animal model of such a life-threatening disease.

6.
J Neurosci Res ; 98(11): 2245-2262, 2020 11.
Article in English | MEDLINE | ID: mdl-32729959

ABSTRACT

Cannabinoid receptor type 1 (CB1R) modulates synaptic activity and is widely distributed in brain areas such as the hippocampus, cerebellum, cerebral cortex, and striatum, among others. CB1R is involved in processes such as memory, learning, motor coordination, and mood. Genetic deletion of CB1R causes behavioral alterations. In this work, we evaluated neuronal morphology and synaptic structure in the hippocampus of adult male CB1R knockout mice (CB1R-/- ). Morphological changes in the CB1R-/- hippocampus evidenced a decrease in the expression of cytoskeletal proteins neurofilaments 160 KDa, neurofilaments 200 KDa, and microtubule-associated protein 2. CA1 neurons showed decreased arborization and changes in synaptic structure such as lower thickness of postsynaptic density and a reduction in synaptophysin levels. Results obtained in the present work provide evidence of the participation of CB1R in the establishment of neuronal structure and networks that could have an important role in neuronal plasticity. In addition, these changes observed in CB1R-/- could be correlated with behavioral alterations reported.


Subject(s)
Hippocampus/anatomy & histology , Neurons/ultrastructure , Receptor, Cannabinoid, CB1/genetics , Synapses/ultrastructure , Animals , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Female , Hippocampus/ultrastructure , Immunohistochemistry , Male , Mice , Mice, Knockout , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Nerve Net/anatomy & histology , Nerve Net/ultrastructure , Neurofilament Proteins/genetics , Neurofilament Proteins/metabolism , Neuronal Plasticity/genetics , Neuronal Plasticity/physiology
7.
J Neurochem ; 135(3): 616-29, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26296704

ABSTRACT

Brain ischemia produces neuronal cell death and the recruitment of pro-inflammatory cells. In turn, the search for neuroprotection against this type of insult has rendered results involving a beneficial role of endocannabinoid receptor agonists in the Central Nervous System. In this work, to further elucidate the mechanisms associated to this neuroprotective effect, focal brain ischemia was generated by middle cerebral artery occlusion (MCAo) in C57Bl/6 mice. Three, 24 and 48 h after MCAo, animals received CB1R agonist ACEA (1 mg/kg), CB1R antagonist AM251 (1 mg/kg) or vehicle. To assess motor activity, neural deficit scores and motor tests were performed 1 day before and 3, 7, 14, 21, and 28 days after MCAo. At 7 and 28 days post lesion, cytoskeleton structure, astroglial and microglial reaction, and alterations in synapsis were studied in the cerebral cortex. ACEA treatment reduced astrocytic reaction, neuronal death, and dendritic loss. In contrast, AM251 treatment increased these parameters. Motor tests showed a progressive deterioration in motor activity in ischemic animals, which only ACEA treatment was able to counteract. Our results suggest that CB1R may be involved in neuronal survival and in the regulation of neuroprotection during focal cerebral ischemia in mice.


Subject(s)
Arachidonic Acids/therapeutic use , Brain Ischemia/prevention & control , Motor Skills/drug effects , Neuroprotective Agents/therapeutic use , Receptor, Cannabinoid, CB1/agonists , Recovery of Function/drug effects , Stroke/prevention & control , Animals , Arachidonic Acids/pharmacology , Brain Ischemia/pathology , Male , Mice , Mice, Inbred C57BL , Motor Skills/physiology , Neurons/drug effects , Neurons/pathology , Neuroprotective Agents/pharmacology , Receptor, Cannabinoid, CB1/physiology , Recovery of Function/physiology , Stroke/pathology
9.
J Mol Neurosci ; 56(4): 773-781, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25645684

ABSTRACT

Endocannabinoid receptors CB1R and CB2R are present in the CNS and modulate synaptic activity. By using an in vitro model, two concentrations of CB1R agonist ACEA at 0.5 and 5 µM doses and CB1R antagonist AM251 at 1 and 10 µM doses were administered in organotypic slice cultures of mouse hippocampus, and their effects on neurons and glial cells were analyzed at different time points. Exposure to low concentrations of ACEA (0.5 µM) did not seem to affect tissue organization, neuronal morphology, or glial response. In contrast, at a higher concentration of ACEA, many neurons in the dentate gyrus exhibited strong caspase-3 immunoreactivity. After treatment with AM251, we observed an increase in caspase-3 immunoreactivity and a downregulation of CB1R expression. Results show that long-term hippocampal slice cultures respond to both CB1R activation and inactivation by changing neuronal protein expression patterns. In the present study, we demonstrate that CB1R agonist ACEA promotes alterations in the neuronal cytoskeleton as well as changes in CB1R expression in organotypic hippocampal slice cultures, and that CB1R antagonist AM251 promotes neuronal death and astroglial reaction.


Subject(s)
Cannabinoids/metabolism , Hippocampus/drug effects , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB2/antagonists & inhibitors , Animals , Arachidonic Acids/pharmacology , Caspase 3/metabolism , Cells, Cultured , Cytoskeleton/metabolism , Hippocampus/metabolism , Mice , Mice, Inbred C57BL , Piperidines/pharmacology , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/genetics , Receptor, Cannabinoid, CB2/metabolism
10.
Neurochem Int ; 79: 79-87, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25280833

ABSTRACT

Ischemic brain injury is a dynamic process involving oxidative stress, inflammation, cell death and the activation of endogenous adaptive and regenerative mechanisms depending on the activation of transcription factors such as hypoxia-inducible factor 1-alpha. Accordingly, we have previously described a new focal hypoxia model by direct intracerebral cobalt chloride injection. In turn, oleanolic acid, a plant-derived triterpenoid, has been extensively used in Asian countries for its anti-inflammatory and anti-tumor properties. A variety of novel pharmacological effects have been attributed to this triterpenoid, including beneficial effects on neurodegenerative disorders--including experimental autoimmune encephalomyelitis--due to its immunomodulatory activities at systemic level, as well as within the central nervous system. In this context, we hypothesize that this triterpenoid may be capable of exerting neuroprotective effects in ischemic brain, suppressing glial activities that contribute to neurotoxicity while promoting those that support neuronal survival. In order to test this hypothesis, we used the intraperitoneal administration of oleanoic acid in adult rats for seven days previous to focal cortical hypoxia induced by cobalt chloride brain injection. We analyzed the neuroprotective effect of oleanoic acid from a morphological point of view, focusing on neuronal survival and glial reaction.


Subject(s)
Hypoxia, Brain/drug therapy , Neuroprotective Agents/pharmacology , Oleanolic Acid/pharmacology , Animals , Astrocytes/drug effects , Astrocytes/pathology , Brain/pathology , Cell Survival/drug effects , Cytoskeleton/drug effects , Hypoxia, Brain/pathology , Male , NADPH Dehydrogenase/metabolism , Neuroglia/drug effects , Neurons/drug effects , Neurons/pathology , Rats , Rats, Wistar
11.
J Neurochem ; 128(3): 431-44, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24382264

ABSTRACT

Rats with pre-hepatic portal hypertension because of partial portal vein ligation develop minimal hepatic encephalopathy (MHE) with hyperammonemia, impaired blood-brain barrier, mild brain edema, and severe mitochondrial changes in the hippocampus. The aim of this study was to evaluate changes of different neural cells in the cerebral cortex and the hippocampus. Animals were divided into two groups, MHE and sham. Astrocytes were studied by immunostaining with glial fibrillary acidic protein and S100ß protein; neurons were immunostained with neuronal nuclear marker, microtubule associated protein-2, and NF-200 and capillaries with Nestin. The hypoxia-inducible factor 1α (HIF-1α) and its downstream proteins, P-glycoprotein (P-gp) and erythropoietin receptor (Epo-R), were also evaluated. Astrocytes were increased in area and number only in the hippocampus, while S100ß increased in both brain areas in MHE animals. Microtubule associated protein-2 and NF-200 immunoreactivities (-ir) were significantly reduced in both areas. Hippocampal Nestin-ir was increased in MHE animals. These cellular changes were similar to those described in ischemic conditions, thus HIF-1α, P-gp, and Epo-R were also evaluated. A high expression of HIF-1α in cortical neurons was observed in the MHE group. It is likely that this hypoxia-like state is triggered via ammonia occupying the binding domain of HIF-1α and thereby preventing its degradation and inducing its stabilization, leading to the over-expression of P-gp and the Epo-R.


Subject(s)
Central Nervous System/pathology , Hyperammonemia/pathology , Hypertension, Portal/pathology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Alzheimer Disease/pathology , Ammonia/blood , Animals , Antigens, Nuclear/metabolism , Arterial Pressure/drug effects , Astrocytes/pathology , Blood Gas Analysis , Blood Pressure/drug effects , CA1 Region, Hippocampal/pathology , Cerebral Cortex/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Image Processing, Computer-Assisted , Immunohistochemistry , Male , Mitogen-Activated Protein Kinase 1/metabolism , Nerve Tissue Proteins/metabolism , Nestin/metabolism , Neurofilament Proteins/metabolism , Portal Vein/drug effects , Portal Vein/physiology , Rats , Rats, Inbred WKY , Tissue Fixation
12.
J Neurochem ; 129(4): 637-48, 2014 May.
Article in English | MEDLINE | ID: mdl-24329778

ABSTRACT

The endocannabinoid system, composed of cannabinoid receptors, endocannabinoids, and synthesis and degradation enzymes, is present since early stages of brain development. During this period, the endocannabinoid system is involved in the regulation of neural progenitor proliferation and specification as well as the migration and differentiation of pyramidal neurons and interneurons. Marijuana consumption during pregnancy represents a serious risk in relation to the fetal brain development since Δ(9) -tetrahidrocannabinol, the main active compound of cannabis, can reach the fetus through placenta and hemato-encephalic barrier. Cohort studies performed on children and adolescents of mothers who consumed marijuana during pregnancy reported cognitive and comportamental abnormalities. In the present study, we examined the expression of the cannabinoid receptor CB1 R during corticogenesis in radially and tangentially migrating post-mitotic neurons. We found that prenatal exposure to WIN impaired tangential and radial migration of post-mitotic neurons in the dorsal pallium. In addition, we described alterations of two transcription factors associated with proliferating and newly post-mitotic glutamatergic cells in the dorsal pallium, Tbr1 and Tbr2, and disruption in the number of Cajal-Retzius cells. The present results contribute to the knowledge of neurobiological substrates that determine neuro-comportamental changes that will persist through post-natal life.


Subject(s)
Benzoxazines/pharmacology , Cannabinoid Receptor Agonists/pharmacology , Cerebral Cortex/cytology , Endocannabinoids/physiology , Morpholines/pharmacology , Naphthalenes/pharmacology , Neurons/drug effects , Receptor, Cannabinoid, CB1/physiology , Animals , Apoptosis/drug effects , Cell Adhesion Molecules, Neuronal/analysis , Cell Division/drug effects , Cell Movement/physiology , Cerebral Cortex/drug effects , Cerebral Cortex/embryology , Doublecortin Domain Proteins , Extracellular Matrix Proteins/analysis , Female , GABAergic Neurons/cytology , GABAergic Neurons/drug effects , GABAergic Neurons/physiology , Glutamic Acid/physiology , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/embryology , Interneurons/cytology , Interneurons/drug effects , Interneurons/physiology , Microtubule-Associated Proteins/analysis , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/metabolism , Neurogenesis/drug effects , Neurons/cytology , Neurons/physiology , Neuropeptides/analysis , Pregnancy , Rats , Rats, Wistar , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/biosynthesis , Reelin Protein , Serine Endopeptidases/analysis , T-Box Domain Proteins/metabolism , Transcription, Genetic
13.
Neuropharmacology ; 73: 388-96, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23796670

ABSTRACT

In this study, the role of CB2r on aversive memory consolidation was further evaluated. Mice lacking CB2r (CB2KO) and their corresponding littermates (WT) were exposed to the step-down inhibitory avoidance test (SDIA). MAP2, NF200 and synaptophysin (SYN)-immunoreactive fibers were studied in the hippocampus (HIP) of both genotypes. The number of synapses, postsynaptic density thickness and the relation between the synaptic length across the synaptic cleft and the distance between the synaptic ends were evaluated in the HIP (dentate gyrus (DG) and CA1 fields) by electron microscopy. Brain-derived neurotrophic factor (BDNF), glucocorticoid receptor (NR3C1) gene expressions and mTOR/p70S6K signaling cascade were evaluated in the HIP and prefrontal cortex (PFC). Finally, the effects of acute administration of CB2r-agonist JWH133 or CB2r-antagonist AM630 on memory consolidation were evaluated in WT mice by using the SDIA. The lack of CB2r impaired aversive memory consolidation, reduced MAP2, NF200 and SYN-immunoreactive fibers and also reduced the number of synapses in DG of CB2KO mice. BDNF and NR3C1 gene expression were reduced in the HIP of CB2KO mice. An increase of p-p70S6K (T389 and S424) and p-AKT protein expression was observed in the HIP and PFC of CB2KO mice. Interestingly, administration of AM630 impaired aversive memory consolidation, whereas JWH133 enhanced it. Further functional and molecular assessments would have been helpful to further support our conclusions. These results revealed that CB2r are involved in memory consolidation, suggesting that this receptor could be a promising target for developing novel treatments for different cognitive impairment-related disorders.


Subject(s)
Memory Disorders/physiopathology , Neuronal Plasticity/physiology , Receptor, Cannabinoid, CB2/deficiency , Receptor, Cannabinoid, CB2/physiology , Synapses/ultrastructure , Animals , Avoidance Learning/physiology , Brain-Derived Neurotrophic Factor/biosynthesis , Cannabinoid Receptor Agonists/pharmacology , Cannabinoid Receptor Antagonists/pharmacology , Cannabinoids/pharmacology , Gene Expression/physiology , Hippocampus/metabolism , Hippocampus/physiopathology , Indoles/pharmacology , Memory/drug effects , Memory/physiology , Memory Disorders/genetics , Mice , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Neurofilament Proteins/metabolism , Neuronal Plasticity/genetics , Prefrontal Cortex/metabolism , Receptor, Cannabinoid, CB2/genetics , Receptors, Glucocorticoid/biosynthesis , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Synaptophysin/metabolism , TOR Serine-Threonine Kinases/metabolism
14.
PLoS One ; 7(12): e51068, 2012.
Article in English | MEDLINE | ID: mdl-23251423

ABSTRACT

Influenza virus (InfV) infection during pregnancy is a known risk factor for neurodevelopment abnormalities in the offspring, including the risk of schizophrenia, and has been shown to result in an abnormal behavioral phenotype in mice. However, previous reports have concentrated on neuroadapted influenza strains, whereas increased schizophrenia risk is associated with common respiratory InfV. In addition, no specific mechanism has been proposed for the actions of maternal infection on the developing brain that could account for schizophrenia risk. We identified two common isolates from the community with antigenic configurations H3N2 and H1N1 and compared their effects on developing brain with a mouse modified-strain A/WSN/33 specifically on the developing of dopaminergic neurons. We found that H1N1 InfV have high affinity for dopaminergic neurons in vitro, leading to nuclear factor kappa B activation and apoptosis. Furthermore, prenatal infection of mothers with the same strains results in loss of dopaminergic neurons in the offspring, and in an abnormal behavioral phenotype. We propose that the well-known contribution of InfV to risk of schizophrenia during development may involve a similar specific mechanism and discuss evidence from the literature in relation to this hypothesis.


Subject(s)
Brain/virology , Dopaminergic Neurons/virology , Influenza A Virus, H1N1 Subtype , Influenza A Virus, H3N2 Subtype , Orthomyxoviridae Infections/virology , Prenatal Exposure Delayed Effects/virology , Schizophrenia/virology , Animals , Behavior, Animal/physiology , Brain/immunology , Brain/physiopathology , Cells, Cultured , Disease Models, Animal , Female , Maze Learning/physiology , Mice , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/physiopathology , Pregnancy , Prenatal Exposure Delayed Effects/immunology , Prenatal Exposure Delayed Effects/physiopathology , Recognition, Psychology/physiology
15.
Drug Metabol Drug Interact ; 26(2): 65-9, 2011.
Article in English | MEDLINE | ID: mdl-21756166

ABSTRACT

Stroke is a major human health problem without efficient available therapeutics. Ischemic brain injury can induce cell death as well as upregulation of endogenous adaptive mechanisms depending on the severity and duration of hypoxia, and the activity of transcription factors, such as hypoxia inducible factor 1-α (HIF-1α). HIF-1α induces gene expression as multidrug resistance (MDR-1) gene associated with drug-refractory phenotype, as well as erythropoietin (Epo) and erythropoietin receptor (Epo-R) associated with O(2) supply. The spontaneous stimulation of the Epo/Epo-R system is not enough for brain protection. Therefore, administration of exogenous recombinant human Epo (rHu-Epo) was suggested as an alternative therapy in stroke. In several experimental models of brain hypoxia, Epo and Epo variants, including rHu-Epo, showed neuroprotective effects. Intranasal administration of these Epo-compounds can reach the central nervous system and protect the brain against ischemia, avoiding hematopoietic effects. However, it has been reported that high expression of Epo-R in neurons must be available to be activated by Epo. According to these considerations, intranasal delivery of rHu-Epo could be an interesting approach in the treatment of cerebral hypoxias avoiding both (i) adverse peripheral effects of treatment with Epo in stroke, and (ii) the pharmacoresistant phenotype depending on MDR-1 expression.


Subject(s)
Erythropoietin/administration & dosage , Hypoxia, Brain/drug therapy , Neuroprotective Agents/administration & dosage , Administration, Intranasal , Animals , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Receptors, Erythropoietin/analysis , Recombinant Proteins/administration & dosage
16.
Neurotox Res ; 20(2): 182-92, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21116766

ABSTRACT

Stroke is a major human health problem inducing long-term disability without any efficient therapeutic option being currently available. Under hypoxia, hypoxia-inducible factor-1α (HIF-1α) activates several genes as erythropoietin receptor (Epo-R) related with O(2) supply, and the multidrug-resistance gene (MDR-1) related with drug-refractory phenotype. Brain cortical injection of CoCl(2) produces focal hypoxia-like lesion with neuronal and glial alterations, as well as HIF-1α stabilization and MDR-1 overexpression. Intranasal (IN) drug delivery can by-pass blood-brain barrier (BBB) where MDR-1 is normally expressed. We evaluated the effects of IN-rHu-Epo administration on spontaneous motor activity (SMA) and the brain pattern expression of HIF-1α, MDR-1, and Epo-R in our cobalt-induced hypoxia model. Adult male Wistar rats were injected by stereotaxic surgery in frontoparietal cortex, with CoCl(2) (2 µl-50 mM; n = 20) or saline (controls; n = 20). Ten rats of each group were treated with IN-rHu-Epo 24 U or IN-saline. In addition, erythropoietic stimulation was evaluated by reticulocytes (Ret) account during three consecutive days, after intraperitoneal (i.p.)-recombinant-human Epo (rHu-Epo) (950 U; n = 6) or IN-rHu-Epo (24 U; n = 6) administration. SMA was evaluated by open field and rotarod tests, before and after surgical procedures during five consecutive days. Histological and immunostaining studies of HIF-1α, MDR-1, and Epo-R were performed on brain slides. A significant difference in SMA was observed in the hypoxic rats of IN-rHu-Epo-administered group as compared with Co-Saline-treated subjects and controls (p < 0.001). HIF-1α, EPO-R, and MDR-1 were overexpressed in the hypoxic cortex areas, while in contralateral hemisphere or controls, they were negatives. Reticulocytes were only increased in intraperitoneal (i.p.)-rHu-Epo-administered group. In spite of MDR-1 overexpression being detected in neurons, the coexpression of Epo-R could explain the positive effects observed on SMA of IN-rHu-Epo-administered group.


Subject(s)
Cobalt/toxicity , Erythropoietin/administration & dosage , Exploratory Behavior/drug effects , Hypoxia, Brain/chemically induced , Hypoxia, Brain/drug therapy , Recovery of Function/drug effects , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Administration, Intranasal , Analysis of Variance , Animals , Disease Models, Animal , Gene Expression Regulation/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Motor Activity/drug effects , Rats , Rats, Wistar , Receptors, Erythropoietin/metabolism , Recombinant Proteins , Reticulocytes/drug effects , Rotarod Performance Test
17.
Neurotox Res ; 15(4): 348-58, 2009 May.
Article in English | MEDLINE | ID: mdl-19384568

ABSTRACT

Ischemic brain injury is a dynamic process that involves oxidative stress, inflammation, and cell death, as well as activation of endogenous adaptive and regenerative mechanisms depending on activation of transcription factors such as hypoxia inducible factor 1-alpha (HIF-1alpha). Because CoCl2 activates HIF-1alpha, we described a new focal-hypoxia model by direct intracerebral CoCl2 injection. Adult male Wistar rats were intracerebrally injected with CoCl2 (2 microl-50 mM), in frontoparietal cortex of right hemisphere, and saline (2 microl) in the contralateral hemisphere. In slides of fixed brains at 1, 6, 9, 24 h or 5 day after treatment, TTC, histochemistry (toluidine blue, Hoescht-33342, TUNEL), immunostaining (HIF-1alpha, GFAP), Lycopersicon esculentum lectin staining, and electron microscopy (EM) were performed. Immediately after 1 h post CoCl2 injection, HIF-1alpha stabilization and neuronal nuclear shrinkage and cromathin condensation were observed by immunostaining and EM, respectively. Neuronal apoptotic nuclear morphology and GFAP immunoreactivity and lectin maximal reactivity were detected during 6-9 h. Ultrastructural alterations of morphology included edematous perinuclear cytoplasm, organelles and endoplasmic reticulum (RE) enlargement, mitochondrial swelling with increased matrix density, and deposits of electron-dense material. Neurons showed particular nuclear indentations. Astrocytes and oligodendrocytes presented alterations in both nuclei and RE with dilated lumen and altered mitochondrias, and all these ultrastructural changes became detectable at day 5. CoCl2 cortical injection mimics focal brain ischemia, inducing neuronal death and glial activation. This model brings the opportunity to develop focal ischemia in selected brain areas to study their functional consequences and potential pharmacological therapies for in vivo models of stroke.


Subject(s)
Antimutagenic Agents/toxicity , Cobalt/toxicity , Hypoxia/chemically induced , Hypoxia/pathology , Neuroglia/drug effects , Neurons/drug effects , Analysis of Variance , Animals , Brain/drug effects , Brain/pathology , Cell Death/drug effects , Disease Models, Animal , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , In Situ Nick-End Labeling/methods , Lectins/drug effects , Male , Microscopy, Electron, Transmission/methods , Nerve Tissue Proteins/metabolism , Neuroglia/ultrastructure , Neurons/ultrastructure , Rats , Rats, Wistar , Staining and Labeling/methods , Time Factors
18.
J Neurol Sci ; 258(1-2): 84-92, 2007 Jul 15.
Article in English | MEDLINE | ID: mdl-17459414

ABSTRACT

Neuronal damage after stroke-associated brain hypoxia is a leading cause of long-term disability and death. The refractoriness to therapeutic strategies for neuroprotection after 3 h post brain ischemia is poorly understood. P-glycoprotein (P-gp), the multidrug resistance gene (MDR-1) product is normally expressed at blood-brain-barrier. P-gp neuronal expression has been demonstrated in refractory epilepsy and after brain ischemia. In this report we investigated the hypoxia-induced neuronal P-gp expression after local injection of CoCl(2) (1-200 mM) in the fronto-parietal cortex of male adult rats (Bregma -1.30 mm) by stereotaxic surgery. P-gp immunostaining of brain slides was analyzed using specific monoclonal antibodies and double immunolabeling was done with specific astrocytic and neuronal markers. Five days after injection of 1 mM CoCl(2), P-gp expression surrounding the lesion site was observed in neurons, astrocytic end-foot on capillary blood vessels and endothelial cells on blood vessels. Higher CoCl(2) doses (200 mM) resulted in additional P-gp immunostaining of the entire astrocytic and neuronal cytoplasm. Electron microscopy (EM) studies showed alterations in neurons as early as 6 h after the CoCl(2) injection. P-gp expression in hypoxic neurons and astrocytic end-foot could potentially impair of drugs access to the brain parenchyma thus suggesting the presence of two P-gp-based pumping systems (one in astrocytes and other in the hypoxic neurons) that are able to behave as a previously unnoticed obstacle for pharmacological strategies of neuroprotection.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Gene Expression/physiology , Hypoxia/metabolism , Hypoxia/physiopathology , Neurons/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Animals , Antimutagenic Agents/administration & dosage , Cobalt/administration & dosage , Disease Models, Animal , Gene Expression/drug effects , Glial Fibrillary Acidic Protein/metabolism , Hypoxia/chemically induced , Hypoxia/pathology , Male , Microscopy, Electron, Transmission/methods , Neurons/drug effects , Neurons/ultrastructure , Parietal Lobe/drug effects , Parietal Lobe/metabolism , Parietal Lobe/pathology , Phosphopyruvate Hydratase/metabolism , Rats , Rats, Wistar
19.
Exp Neurol ; 200(2): 438-59, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16631170

ABSTRACT

Little is known about the morphological effects of alcoholism on the developing adolescent brain and its consequences into adulthood. We studied here the relationship between two neurotransmitter systems (the serotoninergic and nitrergic) and the astrocytic and neuronal cytoskeleton immediately and long after drinking cessation of a chronic, but low, ethanol administration. Adolescent male Wistar rats were exposed to ethanol 6.6% (v/v) in drinking water for 6 weeks and studied after ending exposure or after a 10-week recovery period drinking water. Control animals received water. Brain sections were processed by immunohistochemistry using antibodies to serotonin (5-HT); glial fibrillary acidic protein (GFAP); astroglial S-100b protein; microtubule associated protein-2 (MAP-2); 200 kDa neurofilaments (Nf-200); and neuronal nitric oxide synthase (nNOS). The mesencephalic dorsal and median raphe nucleus (DRN; MRN) and three prosencephalic areas closely related to cognitive abilities (CA1 hippocampal area, striatum and frontal cortex) were studied by digital image analysis. 5-HT immunoreactivity (-ir) decreased in the DRN and recovered after abstinence and was not changed in the MRN. In the three prosencephalic areas, astrocytes' cell area (GFAP-ir cells) increased after EtOH exposure and tended to return to normality after abstinence, while cytoplasmic astroglial S100b protein-ir, relative area of MAP-2-ir and Nf-200-ir fibers decreased, and later partially recovered. In the striatum and frontal cortex, nNOS-ir decreased only after abstinence. In conclusion, in the adolescent brain, drinking cessation can partially ameliorate the ethanol-induced morphological changes on neurons and astrocytes but cannot fully return it to the basal state.


Subject(s)
Brain/drug effects , Central Nervous System Depressants/administration & dosage , Ethanol/administration & dosage , Nerve Tissue Proteins/metabolism , Alcohol Drinking/metabolism , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Brain/cytology , Brain/metabolism , Brain Chemistry/drug effects , Cell Count/methods , Central Nervous System Depressants/blood , Diagnostic Imaging/methods , Ethanol/blood , Gene Expression/drug effects , Immunohistochemistry , Male , Neurons/drug effects , Neurons/metabolism , Rats , Rats, Wistar , Time Factors
20.
J Neurosci Res ; 83(5): 787-800, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16493669

ABSTRACT

Chronic activation of the stress response during pregnancy has been shown to be injurious to the development of the offspring. We have previously demonstrated that restraint prenatal stress inflicted during the last week of pregnancy in rats increased dopamine and glutamate receptors in forebrain areas of the adult offsprings. In this study, the same prenatal insult was employed to assess morphological changes in astrocytes and in the dendritic arborization in frontal cortex, striatum, and hippocampus of the adult rat brain. On postnatal day 90, brains were processed for immunocytochemistry using primary antibodies to glial fibrillary acidic protein (GFAP; the main cytoskeletal astroglial protein), S100B protein (an astroglial-derived neurotrophic factor), MAP-2 (a microtubule-associated protein present almost exclusively in dendrites), and synaptophysin (Syn; one major integral protein of the synaptic vesicles membrane). The results show a significant increase in the cell area of GFAP-immunoreactive (-IR) astrocytes, with high levels of S100B protein and a significant decrease in the relative area of MAP-2-IR neuronal processes in prenatally stressed adult rats. The expression of synaptophysin decreased in all areas studied. These results demonstrate that prenatal stress induces a long-lasting astroglial reaction and a reduced dendritic arborization, with synaptic loss in the brain of adult offspring. In addition to the neurochemical alterations previously reported, these morphological changes might be underlying the behavioral and learning impairment previously observed in prenatally stressed rats.


Subject(s)
Astrocytes/pathology , Brain/pathology , Neurons/pathology , Prenatal Exposure Delayed Effects/physiopathology , Stress, Psychological/physiopathology , Animals , Astrocytes/metabolism , Brain/metabolism , Cell Communication/physiology , Female , Glial Fibrillary Acidic Protein/metabolism , Image Processing, Computer-Assisted , Immunohistochemistry , Microscopy, Electron, Transmission , Microtubule-Associated Proteins/metabolism , Nerve Growth Factors/metabolism , Neurons/metabolism , Pregnancy , Rats , Rats, Wistar , Restraint, Physical , S100 Calcium Binding Protein beta Subunit , S100 Proteins/metabolism , Synaptophysin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...