257 results on '"Netto CA"'
Search Results
2. Arundic acid administration protects astrocytes, recovers histological damage and memory deficits induced by neonatal hypoxia ischemia in rats
- Author
-
Mari, C, primary, Odorcyk, FK, additional, Sanches, EF, additional, Wartchow, KM, additional, Martini, AP, additional, Nicola, F, additional, Zanotto, C, additional, Wyse, AT, additional, Gonçalves, CA, additional, and Netto, CA, additional
- Published
- 2019
- Full Text
- View/download PDF
3. Pre-conditioning to global cerebral ischemia changes hippocampal acetylcholinesterase in the rat
- Author
-
Schetinger, Mrc, Carla Denise Bonan, Frassetto, Ss, Wyse, Ats, Schierholt, Rc, Webber, A., Dias, Rd, Sarkis, Jjf, and Netto, Ca
4. Construction and reconstruction of memories
- Author
-
Izquierdo, I., Netto, Ca, Dalmaz, C., Marcia Chaves, Pereira, Me, and Siegfried, B.
5. Resveratrol prevents oxidative stress and inhibition of Na(+)K(+)-ATPase activity induced by transient global cerebral ischemia in rats.
- Author
-
Simao F, Matté A, Matté C, Soares FM, Wyse AT, Netto CA, and Salbego CG
- Published
- 2011
6. Protective effects of the PPAR agonist bezafibrate against disruption of redox and energy homeostasis, neuronal death, astroglial reactivity, and neuroinflammation induced in vivo by D-2-hydroxyglutaric acid in rat brain.
- Author
-
Ribeiro RT, Marcuzzo MB, Carvalho AVS, Palavro R, Castro ET, Pinheiro CV, Bobermin LD, Amaral AU, Leipnitz G, Netto CA, and Wajner M
- Abstract
The biochemical hallmark of D-2-hydroxyglutaric aciduria is brain accumulation of D-2-hydroxyglutaric acid (D2HG). Patients present predominantly neurological manifestations, whose pathogenesis is still unknown. Thus, we examined the impact of elevated brain levels of D2HG, induced by intracerebral injection of this metabolite in juvenile rats, on redox and mitochondrial homeostasis and histochemical landmarks in the cerebral cortex. D2HG administration disrupted redox homeostasis by increasing the levels of reactive oxygen species and lipid peroxidation and the activities of superoxide dismutase, glutathione peroxidase, and glutathione reductase and decreasing reduced glutathione levels. Furthermore, the complex IV and mitochondrial creatine kinase activities, as well as the protein contents of voltage-dependent anion channel 1, translocase of outer mitochondrial membrane 20, and peroxisome proliferator-activated receptor-γ coactivator 1-α, were diminished by D2HG, indicating bioenergetics dysfunction and disrupted mitochondrial biogenesis. D2HG also reduced neuronal nuclear protein content and augmented cleaved caspase-3, S100 calcium-binding protein B, glial fibrillary acidic protein, and ionized calcium-binding adaptor molecule 1, indicating neuronal loss, apoptosis, astrogliosis, and microglial activation, respectively. The tumor necrosis factor alpha expression was also significantly augmented, reflecting an increased inflammatory response. We also evaluated whether bezafibrate (BEZ) pretreatment could prevent the alterations induced by D2HG. BEZ normalized most of the D2HG-induced deleterious effects. Therefore, bioenergetics and redox status disruption caused by D2HG, associated with neuronal death, glial reactivity, and increased inflammatory response, may potentially represent pathomechanisms of brain damage in D-2-HGA. Finally, it is proposed that BEZ may be potentially used as therapy for D-2-HGA., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
7. Impact of peripheral immune cells in experimental neonatal hypoxia-ischemia: A systematic review and meta-analysis.
- Author
-
Nunes RR, Durán-Carabali LE, Ribeiro NH, Sirena DH, Tassinari ID, Netto CA, Paz AH, and de Fraga LS
- Abstract
Infiltration of peripheral immune cells into the brain following neonatal hypoxia-ischemia (HI) contributes to increased neuroinflammation and brain injury. However, the specific roles of different immune cell types in neonatal brain injury remain poorly understood. Although existing evidence suggests a potential role for sexual dimorphism in HI outcomes, this aspect has been insufficiently investigated. In this systematic review and meta-analysis, we examined the brain infiltration of peripheral immune cells in rodents of both sexes following neonatal HI. A total of 25 studies were included. Our analysis revealed significant increases in the infiltration of various subtypes of leukocytes after HI, along with increased brain injury, cell death, and neuroinflammation, and reduced neuronal survival. Notably, males exhibited a greater degree of immune cell infiltration and more pronounced neuroinflammation compared to females. These findings suggest that infiltrating leukocytes contribute significantly to the pathophysiology of neonatal HI, with sexually dimorphic responses further influencing the outcomes. It is crucial that future research focuses on elucidating the specific roles of immune cell subtypes to better understand the mechanisms underlying brain damage after HI and identify novel therapeutic targets. Moreover, the observed sex differences highlight the need to consider sex as a key factor when developing strategies for the treatment of neonatal HI., (Copyright © 2024 Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
8. Impairment of neuromotor development and cognition associated with histopathological and neurochemical abnormalities in the cerebral cortex and striatum of glutaryl-CoA dehydrogenase deficient mice.
- Author
-
Castro ET, Ribeiro RT, Carvalho AVS, Machado DN, Zemniaçak ÂB, Palavro R, de Azevedo Cunha S, Tavares TQ, de Souza DOG, Netto CA, Leipnitz G, Amaral AU, and Wajner M
- Subjects
- Animals, Mice, Cognition physiology, Cognition drug effects, Brain Diseases, Metabolic pathology, Brain Diseases, Metabolic metabolism, Male, Mice, Inbred C57BL, Glutaryl-CoA Dehydrogenase deficiency, Glutaryl-CoA Dehydrogenase genetics, Corpus Striatum drug effects, Corpus Striatum pathology, Corpus Striatum metabolism, Cerebral Cortex pathology, Cerebral Cortex metabolism, Cerebral Cortex drug effects, Mice, Knockout, Amino Acid Metabolism, Inborn Errors pathology
- Abstract
Patients with glutaric acidemia type I (GA I) manifest motor and intellectual disabilities whose pathogenesis has been so far poorly explored. Therefore, we evaluated neuromotor and cognitive abilities, as well as histopathological and immunohistochemical features in the cerebral cortex and striatum of glutaryl-CoA dehydrogenase (GCDH) deficient knockout mice (Gcdh
-/- ), a well-recognized model of GA I. The effects of a single intracerebroventricular glutaric acid (GA) injection in one-day-old pups on the same neurobehavioral and histopathological/immunohistochemical endpoints were also investigated. Seven-day-old Gcdh-/- mice presented altered gait, whereas those receiving a GA neonatal administration manifested other sensorimotor deficits, including an abnormal response to negative geotaxis, cliff aversion and righting reflex, and muscle tone impairment. Compared to the WT mice, adult Gcdh-/- mice exhibited motor impairment, evidenced by poor performance in the Rota-rod test. Furthermore, neonatal GA administration provoked long-standing short- and long-term memory impairment in adult Gcdh-/- mice. Regarding the histopathological features, a significant increase in vacuoles and neurodegenerative cells was observed in both the cerebral cortex and striatum of 15- and 60-day-old Gcdh-/- mice and was more pronounced in mice injected with GA. Neuronal loss (decrease of NeuN staining) was also significantly increased in the cerebral cortex and striatum of Gcdh-/- mice, particularly in those neonatally injected with GA. In contrast, immunohistochemistry of MBP, astrocytic proteins GFAP and S100B, and the microglial marker Iba1 was not changed in 60-day-old Gcdh-/- mice, suggesting no myelination disturbance, reactive astrogliosis, and microglia activation, respectively. These data highlight the neurotoxicity of GA and the importance of early treatment aiming to decrease GA accumulation at early stages of development to prevent brain damage and learning/memory disabilities in GA I patients., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier Ltd. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
9. Lactate administration causes long-term neuroprotective effects following neonatal hypoxia-ischemia.
- Author
-
Tassinari ID, Zang J, Ribeiro NH, Martins BB, Tauffer JVM, Nunes RR, Sanches EF, Sizonenko S, Netto CA, Paz AH, and de Fraga LS
- Subjects
- Animals, Rats, Female, Male, Disease Models, Animal, Recognition, Psychology drug effects, Exploratory Behavior drug effects, Hypoxia-Ischemia, Brain pathology, Hypoxia-Ischemia, Brain metabolism, Animals, Newborn, Rats, Wistar, Neuroprotective Agents pharmacology, Neuroprotective Agents therapeutic use, Lactic Acid metabolism
- Abstract
Neonatal hypoxia-ischemia (HI) is one of the main causes of mortality and long-term disabilities in newborns, and the only clinical approach to treat this condition is therapeutic hypothermia, which shows some limitations. Thus, putative neuroprotective agents have been tested in animal models of HI. Lactate is a preferential metabolic substrate of the neonatal brain and has already been shown to produce beneficial neuroprotective outcomes in neonatal animals exposed to HI. Here, we administered lactate as a treatment in neonatal rats previously exposed to HI and evaluated the impact of this treatment in adulthood. Seven-day-old (P7) male and female Wistar rats underwent permanent common right carotid occlusion combined with an exposition to a hypoxic atmosphere (8% oxygen) for 60 min. Animals were assigned to one of four experimental groups: HI, HI+LAC, SHAM, SHAM+LAC. Lactate was administered intraperitoneally 30 min and 2 h after hypoxia in HI+LAC and SHAM+LAC groups, whereas HI and SHAM groups received vehicle. Animals were tested in the behavioral tasks of negative geotaxis and righting reflex (P8), cylinder test (P24), and the modified neurological severity score was calculated (P25). Open field (OF), and novel object recognition (NOR) were evaluated in adulthood. Animals were killed at P60, and the brains were harvested and processed to evaluate the volume of brain injury. Our results showed that lactate administration reduced the volume of brain lesion and improved sensorimotor and cognitive behaviors in neonatal, juvenile, and adult life in HI animals from both sexes. Thus, lactate administration might be considered as a potential neuroprotective strategy for the treatment of neonatal HI, which is a prevalent disorder affecting newborns., Competing Interests: Declaration of competing interest The authors declare that they have no conflict of interest., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
10. Methylphenidate Exposing During Neurodevelopment Alters Amino Acid Profile, Astrocyte Marker and Glutamatergic Excitotoxicity in the Rat Striatum.
- Author
-
Schmitz F, Durán-Carabali LE, Rieder AS, Silveira JS, Ramires Junior OV, Bobermin LD, Quincozes-Santos A, Alves VS, Coutinho-Silva R, Savio LEB, Coelho DM, Vargas CR, Netto CA, and Wyse ATS
- Subjects
- Animals, Male, Rats, Lipid Peroxidation drug effects, Rats, Wistar, Corpus Striatum drug effects, Corpus Striatum metabolism, Astrocytes drug effects, Astrocytes metabolism, Methylphenidate toxicity, Methylphenidate pharmacology, Glutamic Acid metabolism, Central Nervous System Stimulants toxicity, Central Nervous System Stimulants pharmacology, Amino Acids metabolism
- Abstract
There is a public health concern about the use of methylphenidate (MPH) since the higher prescription for young individuals and non-clinical purposes is addressed to the limited understanding of its neurochemical and psychiatric consequences. This study aimed to evaluate the impact of early and chronic MPH treatment on the striatum focusing on amino acid profile, glutamatergic excitotoxicity, redox status, neuroinflammation and glial cell responses. Male Wistar rats were treated with MPH (2.0 mg/kg) or saline solution from the 15th to the 44th postnatal day. Biochemical and histological analyses were conducted after the last administration. MPH altered the amino acid profile in the striatum, increasing glutamate and ornithine levels, while decreasing the levels of serine, phenylalanine, and branched-chain amino acids (leucine, valine, and isoleucine). Glutamate uptake and Na
+ ,K+ -ATPase activity were decreased in the striatum of MPH-treated rats as well as increased ATP levels, as indicator of glutamatergic excitotoxicity. Moreover, MPH caused lipid peroxidation and nitrative stress, increased TNF alpha expression, and induced high levels of astrocytes, and led to a decrease in BDNF levels. In summary, our results suggest that chronic early-age treatment with MPH induces parallel activation of damage-associated pathways in the striatum and increases its vulnerability during the juvenile period. In addition, data presented here contribute to shedding light on the mechanisms underlying MPH-induced striatal damage and its potential implications for neurodevelopmental disorders., (© 2024. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)- Published
- 2024
- Full Text
- View/download PDF
11. 3-Hydroxy-3-Methylglutaric Acid Disrupts Brain Bioenergetics, Redox Homeostasis, and Mitochondrial Dynamics and Affects Neurodevelopment in Neonatal Wistar Rats.
- Author
-
Silveira JA, Marcuzzo MB, da Rosa JS, Kist NS, Hoffmann CIH, Carvalho AS, Ribeiro RT, Quincozes-Santos A, Netto CA, Wajner M, and Leipnitz G
- Abstract
3-Hydroxy-3-methylglutaric acidemia (HMGA) is a neurometabolic inherited disorder characterized by the predominant accumulation of 3-hydroxy-3-methylglutaric acid (HMG) in the brain and biological fluids of patients. Symptoms often appear in the first year of life and include mainly neurological manifestations. The neuropathophysiology is not fully elucidated, so we investigated the effects of intracerebroventricular administration of HMG on redox and bioenergetic homeostasis in the cerebral cortex and striatum of neonatal rats. Neurodevelopment parameters were also evaluated. HMG decreased the activity of glutathione reductase (GR) and increased catalase (CAT) in the cerebral cortex. In the striatum, HMG reduced the activities of superoxide dismutase, glutathione peroxidase, CAT, GR, glutathione S-transferase, and glucose-6-phosphate dehydrogenase. Regarding bioenergetics, HMG decreased the activities of succinate dehydrogenase and respiratory chain complexes II-III and IV in the cortex. HMG also decreased the activities of citrate synthase and succinate dehydrogenase, as well as complex IV in the striatum. HMG further increased DRP1 levels in the cortex, indicating mitochondrial fission. Finally, we found that the HMG-injected animals showed impaired performance in all sensorimotor tests examined. Our findings provide evidence that HMG causes oxidative stress, bioenergetic dysfunction, and neurodevelopmental changes in neonatal rats, which may explain the neuropathophysiology of HMGA.
- Published
- 2024
- Full Text
- View/download PDF
12. Acrobatic training prevents learning impairments and astrocyte remodeling in the hippocampus of rats undergoing chronic cerebral hypoperfusion: sex-specific benefits.
- Author
-
Martini APR, Schlemmer LM, Lucio Padilha JA, Fabres RB, Couto Pereira NS, Pereira LO, Dalmaz C, and Netto CA
- Abstract
Background: Chronic cerebral hypoperfusion (CCH) leads to memory and learning impairments associated with degeneration and gliosis in the hippocampus. Treatment with physical exercise carries different therapeutic benefits for each sex. We investigated the effects of acrobatic training on astrocyte remodeling in the CA1 and CA3 subfields of the hippocampus and spatial memory impairment in male and female rats at different stages of the two-vessel occlusion (2VO) model., Methods: Wistar rats were randomly allocated into four groups of males and females: 2VO acrobatic, 2VO sedentary, sham acrobatic, and sham sedentary. The acrobatic training was performed for 4 weeks prior to the 2VO procedure. Brain samples were collected for morphological and biochemical analysis at 3 and 7 days after 2VO. The dorsal hippocampi were removed and prepared for Western blot quantification of Akt, p-Akt, COX IV, cleaved caspase-3, PARP, and GFAP. GFAP immunofluorescence was performed on slices of the hippocampus to count astrocytes and apply the Sholl's circle technique. The Morris water maze was run after 45 days of 2VO., Results: Acutely, the trained female rats showed increased PARP expression, and the 2VO-trained rats of both sexes presented increased GFAP levels in Western blot. Training, mainly in males, induced an increase in the number of astrocytes in the CA1 subfield. The 2VO rats presented branched astrocytes, while acrobatic training prevented branching. However, the 2VO-induced spatial memory impairment was partially prevented by the acrobatic training., Conclusion: Acrobatic training restricted the astrocytic remodeling caused by 2VO in the CA1 and CA3 subfields of the hippocampus. The improvement in spatial memory was associated with more organized glial scarring in the trained rats and better cell viability observed in females., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (© 2024 Martini, Schlemmer, Lucio Padilha, Fabres, Couto Pereira, Pereira, Dalmaz and Netto.)
- Published
- 2024
- Full Text
- View/download PDF
13. Acrobatic training prevents motor deficits and neuronal loss in male and female rats following chronic cerebral hypoperfusion.
- Author
-
Martini APR, Hoeper E, Dos Santos DP, Norman T, Dos Santos AS, Pereira LO, and Netto CA
- Subjects
- Rats, Animals, Male, Female, Brain, Ischemia, Disease Models, Animal, Maze Learning, Brain Ischemia pathology, Dementia, Vascular, Motor Cortex
- Abstract
Background: Chronic cerebral hypoperfusion in vascular dementia leads to memory and motor deficits; Physical exercise improves these aspects and promotes neuroprotection. Sexual dimorphism may significantly influence both ischemic and exercise outcomes., Aims: The aim of this study was to investigate the effects of 2VO (Two-Vessel occlusion) and the acrobatic training on motor function, functional performance, and tissue loss in male and female rats., Methods: Male and female rats were randomly divided into 4 groups: sham acrobatic, sham sedentary, 2VO acrobatic and 2VO sedentary. After 45 days of 2VO surgery, the animals received 4 weeks of acrobatic training. At the end, open field, beam balance and horizontal ladder tests were performed. Brain samples were taken for histological and morphological evaluation., Results: Spontaneous motor activity in the open field was not affected by 2VO, on the other hand, an impairment in forelimb placement was observed after 2VO and acrobatic training prevented errors and improved hindlimb placement. Neuronal loss was found in the motor cortex and striatum after 2VO, especially in females, which was prevented by acrobatic training., Conclusion: Mild motor damage was found in animals after 2VO when refined movement was evaluated, probably associated to neuronal death in the motor cortex and striatum. The acrobatic exercise showed a neuroprotective effect, promoting neuronal survival and attenuating the motor deficit., (Copyright © 2024 Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
14. Neutrophil-Lymphocyte Ratio as a Predictor of the Risk of Death in Severe Cases of COVID-19.
- Author
-
Oliveira MJS, Anschau F, Kopittke L, Worm PV, Vargas T, Silva PSD, Cristaldo JCC, Goncalves CAS, Wyse A, and Netto CA
- Subjects
- Humans, Aged, SARS-CoV-2, Neutrophils, Prospective Studies, Lymphocytes, Biomarkers, Retrospective Studies, COVID-19 epidemiology
- Abstract
Background: Identifying clinical characteristics and risk factors, comorbid conditions, and complications arising from SARS-CoV-2 infection is important to predict the progression to more severe forms of the disease among hospitalized individuals to enable timely intervention and to prevent fatal outcomes. The aim of the study is to assess the possible role of the neutrophil/lymphocyte ratio (NLR) as a biomarker of the risk of death in patients with comorbidities hospitalized with COVID-19 in a tertiary hospital in southern Brazil., Methods: This is a prospective cohort study on patients with SARS-CoV-2 infection admitted to a hospital in the metropolitan region of Porto Alegre from September 2020 to March 2022., Results: The sample consisted of 185 patients with associated comorbidities, namely, hypertension, diabetes mellitus, obesity, cardiovascular, pulmonary, and renal diseases, hospitalized with COVID-19. Of these, 78 died and 107 were discharged alive. The mean age was 66.5 years for the group that died and 60.1 years for the group discharged. Statistical analysis revealed that a difference greater than or equal to 1.55 in the NLR, from hospitalization to the 5th day, was associated with a relative risk of death greater than 2., Conclusions: Measuring a simple inflammatory marker such as NLR may improve the risk stratification of comorbid patients with COVID-19 and can be considered a useful biomarker.
- Published
- 2024
- Full Text
- View/download PDF
15. Maternal environmental enrichment protects neonatal brains from hypoxic-ischemic challenge by mitigating brain energetic dysfunction and modulating glial cell responses.
- Author
-
Durán-Carabali LE, Odorcyk FK, Grun LK, Schmitz F, Ramires Junior OV, de Oliveria MR, Campos KF, Hoeper E, Carvalho AVS, Greggio S, Venturine GT, Zimmer ER, Barbé-Tuana F, Wyse ATS, and Netto CA
- Subjects
- Animals, Rats, Female, Male, Pregnancy, Animals, Newborn, Rats, Wistar, Brain metabolism, Astrocytes metabolism, Glucose metabolism, Adenosine Triphosphatases metabolism, Hypoxia-Ischemia, Brain metabolism, Neuromuscular Diseases
- Abstract
There is evidence that maternal milieu and changes in environmental factors during the prenatal period may exert a lasting impact on the brain health of the newborn, even in case of neonatal brain hypoxia-ischemia (HI). The present study aimed to investigate the effects of maternal environmental enrichment (EE) on HI-induced energetic and metabolic failure, along with subsequent neural cell responses in the early postnatal period. Male Wistar pups born to dams exposed to maternal EE or standard conditions (SC) were randomly divided into Sham-SC, HI-SC, Sham-EE, and HI-EE groups. Neonatal HI was induced on postnatal day (PND) 3. The Na
+, K+ -ATPase activity, mitochondrial function and neuroinflammatory related-proteins were assessed at 24 h and 48 h after HI. MicroPET-FDG scans were used to measure glucose uptake at three time points: 24 h post-HI, PND18, and PND24. Moreover, neuronal preservation and glial cell responses were evaluated at PND18. After HI, animals exposed to maternal EE showed an increase in Na+ ,K+ -ATPase activity, preservation of mitochondrial potential/mass ratio, and a reduction in mitochondrial swelling. Glucose uptake was preserved in HI-EE animals from PND18 onwards. Maternal EE attenuated HI-induced cell degeneration, white matter injury, and reduced astrocyte immunofluorescence. Moreover, the HI-EE group exhibited elevated levels of IL-10 and a reduction in Iba-1 positive cells. Data suggested that the regulation of AKT/ERK1/2 signaling pathways could be involved in the effects of maternal EE. This study evidenced that antenatal environmental stimuli could promote bioenergetic and neural resilience in the offspring against early HI damage, supporting the translational value of pregnancy-focused environmental treatments., Competing Interests: Declaration of competing interest A conflicting interest exists when professional judgment concerning a primary interest (such as patient's welfare or the validity of research) may be influenced by a secondary interest (such as financial gain or personal rivalry). It may arise for the authors when they have financial interest that may influence their interpretation of their results or those of others. Examples of potential conflicts of interest include employment, consultancies, stock ownership, honoraria, paid expert testimony, patent applications/registrations, and grants or other funding., (Copyright © 2024 Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
16. Simvastatin Differentially Modulates Glial Functions in Cultured Cortical and Hypothalamic Astrocytes Derived from Interferon α/β Receptor Knockout mice.
- Author
-
Bobermin LD, Sesterheim P, da Costa DS, Rezena E, Schmitz I, da Silva A, de Moraes ADM, Souza DO, Wyse AT, Leipnitz G, Netto CA, Quincozes-Santos A, and Gonçalves CA
- Subjects
- Mice, Animals, Mice, Knockout, Tumor Necrosis Factor-alpha metabolism, Interferon-alpha metabolism, Interferon-alpha pharmacology, Anti-Inflammatory Agents pharmacology, Cholesterol metabolism, Cells, Cultured, Astrocytes metabolism, Simvastatin pharmacology
- Abstract
Astrocytes have key regulatory roles in central nervous system (CNS), integrating metabolic, inflammatory and synaptic responses. In this regard, type I interferon (IFN) receptor signaling in astrocytes can regulate synaptic plasticity. Simvastatin is a cholesterol-lowering drug that has shown anti-inflammatory properties, but its effects on astrocytes, a main source of cholesterol for neurons, remain to be elucidated. Herein, we investigated the effects of simvastatin in inflammatory and functional parameters of primary cortical and hypothalamic astrocyte cultures obtained from IFNα/β receptor knockout (IFNα/βR
-/- ) mice. Overall, simvastatin decreased extracellular levels of tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β), which were related to a downregulation in gene expression in hypothalamic, but not in cortical astrocytes. Moreover, there was an increase in anti-inflammatory interleukin-10 (IL-10) in both structures. Effects of simvastatin in inflammatory signaling also involved a downregulation of cyclooxygenase 2 (COX-2) gene expression as well as an upregulation of nuclear factor κB subunit p65 (NFκB p65). The expression of cytoprotective genes sirtuin 1 (SIRT1) and nuclear factor erythroid derived 2 like 2 (Nrf2) was also increased by simvastatin. In addition, simvastatin increased glutamine synthetase (GS) activity and glutathione (GSH) levels only in cortical astrocytes. Our findings provide evidence that astrocytes from different regions are important cellular targets of simvastatin in the CNS, even in the absence of IFNα/βR, which was showed by the modulation of cytokine production and release, as well as the expression of cytoprotective genes and functional parameters., (© 2023. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)- Published
- 2024
- Full Text
- View/download PDF
17. Bumetanide Attenuates Cognitive Deficits and Brain Damage in Rats Subjected to Hypoxia-Ischemia at Two Time Points of the Early Postnatal Period.
- Author
-
Machado DN, Durán-Carabali LE, Odorcyk FK, Carvalho AVS, Martini APR, Schlemmer LM, de Mattos MM, Bernd GP, Dalmaz C, and Netto CA
- Subjects
- Rats, Animals, Male, Rats, Wistar, Solute Carrier Family 12, Member 2 metabolism, Ischemia drug therapy, Hypoxia drug therapy, Brain metabolism, Cognition, Animals, Newborn, Bumetanide pharmacology, Bumetanide therapeutic use, Hypoxia-Ischemia, Brain complications, Hypoxia-Ischemia, Brain drug therapy
- Abstract
Neonatal hypoxia-ischemia (HI) is one of the main causes of tissue damage, cell death, and imbalance between neuronal excitation and inhibition and synaptic loss in newborns. GABA, the major inhibitory neurotransmitter of the central nervous system (CNS) in adults, is excitatory at the onset of neurodevelopment and its action depends on the chloride (Cl
- ) cotransporters NKCC1 (imports Cl- ) and KCC2 (exports Cl- ) expression. Under basal conditions, the NKCC1/KCC2 ratio decreases over neurodevelopment. Thus, changes in this ratio caused by HI may be related to neurological disorders. The present study evaluated the effects of bumetanide (NKCC cotransporters inhibitor) on HI impairments in two neurodevelopmental periods. Male Wistar rat pups, 3 (PND3) and 11 (PND11) days old, were submitted to the Rice-Vannucci model. Animals were divided into 3 groups: SHAM, HI-SAL, and HI-BUM, considering each age. Bumetanide was administered intraperitoneally at 1, 24, 48, and 72 h after HI. NKCC1, KCC2, PSD-95, and synaptophysin proteins were analyzed after the last injection by western blot. Negative geotaxis, righting reflex, open field, object recognition test, and Morris water maze task were performed to assess neurological reflexes, locomotion, and memory function. Tissue atrophy and cell death were evaluated by histology. Bumetanide prevented neurodevelopmental delay, hyperactivity, and declarative and spatial memory deficits. Furthermore, bumetanide reversed HI-induced brain tissue damage, reduced neuronal death and controlled GABAergic tone, maintained the NKCC1/KCC2 ratio, and synaptogenesis close to normality. Thereby, bumetanide appears to play an important therapeutic role in the CNS, protecting the animals against HI damage and improving functional performance., (© 2023. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)- Published
- 2023
- Full Text
- View/download PDF
18. Serum of COVID-19 patients changes neuroinflammation and mitochondrial homeostasis markers in hippocampus of aged rats.
- Author
-
Montenegro YHA, Bobermin LD, Sesterheim P, Salvato RS, Anschau F, de Oliveira MJS, Wyse ATS, Netto CA, Gonçalves CS, Quincozes-Santos A, and Leipnitz G
- Subjects
- Humans, Animals, Rats, Neuroinflammatory Diseases, Post-Acute COVID-19 Syndrome, SARS-CoV-2, Homeostasis, Hippocampus, COVID-19 genetics
- Abstract
Patients affected by COVID-19 present mostly with respiratory symptoms but acute neurological symptoms are also commonly observed. Furthermore, a considerable number of individuals develop persistent and often remitting symptoms months after infection, characterizing the condition called long-COVID. Since the pathophysiology of acute and persistent neurological manifestations is not fully established, we evaluated the expression of different genes in hippocampal slices of aged rats exposed to the serum of a post-COVID (sPC) individual and to the serum of patients infected by SARS-CoV-2 [Zeta (sZeta) and Gamma (sGamma) variants]. The expression of proteins related to inflammatory process, redox homeostasis, mitochondrial quality control and glial reactivity was determined. Our data show that the exposure to sPC, sZeta and sGamma differentially altered the mRNA levels of most inflammatory proteins and reduced those of antioxidant response markers in rat hippocampus. Furthermore, a decrease in the expression of mitochondrial biogenesis genes was induced by all serum samples, whereas a reduction in mitochondrial dynamics was only caused by sPC. Regarding the glial reactivity, S100B expression was modified by sPC and sZeta. These findings demonstrate that changes in the inflammatory response and a reduction of mitochondrial biogenesis and dynamics may contribute to the neurological damage observed in COVID-19 patients., (© 2023. The Author(s) under exclusive licence to The Journal of NeuroVirology, Inc.)
- Published
- 2023
- Full Text
- View/download PDF
19. Consequences of oxygen deprivation on myelination and sex-dependent alterations.
- Author
-
Fabres RB, Cardoso DS, Aragón BA, Arruda BP, Martins PP, Ikebara JM, Drobyshevsky A, Kihara AH, de Fraga LS, Netto CA, and Takada SH
- Subjects
- Infant, Newborn, Humans, Pregnancy, Female, Oxygen metabolism, Neuroinflammatory Diseases, Infant, Premature, Myelin Sheath metabolism, Brain metabolism, Oligodendroglia metabolism, White Matter metabolism, Brain Injuries metabolism
- Abstract
Oxygen deprivation is one of the main causes of morbidity and mortality in newborns, occurring with a higher prevalence in preterm infants, reaching 20 % to 50 % mortality in newborns in the perinatal period. When they survive, 25 % exhibit neuropsychological pathologies, such as learning difficulties, epilepsy, and cerebral palsy. White matter injury is one of the main features found in oxygen deprivation injury, which can lead to long-term functional impairments, including cognitive delay and motor deficits. The myelin sheath accounts for much of the white matter in the brain by surrounding axons and enabling the efficient conduction of action potentials. Mature oligodendrocytes, which synthesize and maintain myelination, also comprise a significant proportion of the brain's white matter. In recent years, oligodendrocytes and the myelination process have become potential therapeutic targets to minimize the effects of oxygen deprivation on the central nervous system. Moreover, evidence indicate that neuroinflammation and apoptotic pathways activated during oxygen deprivation may be influenced by sexual dimorphism. To summarize the most recent research about the impact of sexual dimorphism on the neuroinflammatory state and white matter injury after oxygen deprivation, this review presents an overview of the oligodendrocyte lineage development and myelination, the impact of oxygen deprivation and neuroinflammation on oligodendrocytes in neurodevelopmental disorders, and recent reports about sexual dimorphism regarding the neuroinflammation and white matter injury after neonatal oxygen deprivation., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
20. Formation of the Brazilian Network for Research and Training in Neuroscience and Physical Activity: a meeting report.
- Author
-
Bento-Torres NVO, Bento-Torres J, Deslandes AC, Netto CA, Royes LFF, Schuch F, Siqueira IR, and Mello-Carpes PB
- Subjects
- Humans, Brazil, Exercise, Neurosciences
- Published
- 2023
- Full Text
- View/download PDF
21. Glioprotective effects of resveratrol in hypothalamic astrocyte cultures obtained from interferon receptor knockout (IFNα/βR -/- ) mice.
- Author
-
Sovrani V, Bobermin LD, Sesterheim P, Rezena E, Cioccari MS, Netto CA, Gonçalves CA, Leipnitz G, and Quincozes-Santos A
- Subjects
- Rats, Animals, Mice, Resveratrol pharmacology, Resveratrol metabolism, Phosphatidylinositol 3-Kinases metabolism, Rats, Wistar, Cells, Cultured, Astrocytes metabolism, Receptors, Interferon metabolism
- Abstract
Astrocytes play essential roles in the central nervous system (CNS), such as the regulation of glutamate metabolism, antioxidant defenses, and inflammatory/immune responses. Moreover, hypothalamic astrocytes seem to be crucial in the modulation of inflammatory processes, including those related to type I interferon signaling. In this regard, the polyphenol resveratrol has emerged as an important glioprotective molecule to regulate astrocyte functions. Therefore, this study aimed to investigate the immunomodulatory and protective effects of resveratrol in hypothalamic astrocyte cultures obtained from mouse depleted of type I interferon receptors (INF-α/β
-/- ), a condition that can impair immune and inflammatory functions. Resveratrol upregulated glutamate transporter and glutamine synthetase gene expression, as well as modulated the release of wide range of cytokines and genes involved in the control of inflammatory response, besides the expression of adenosine receptors, which display immunomodulatory functions. Resveratrol also increased genes associated with redox balance, mitochondrial processes, and trophic factors signaling. The putative genes associated with glioprotective effects of resveratrol, including nuclear factor erythroid derived 2 like 2 (Nrf2), heme oxygenase 1 (HO-1), sirtuin 1 (SIRT1), and phosphoinositide 3-kinase (PI3K)/Akt, were further upregulated by resveratrol. Thus, our data show that resveratrol was able to modulate key genes associated with glial functionality and inflammatory response in astrocyte cultures derived from IFNα/βR-/- mice. These data are in agreement with previous results, reinforcing its glioprotective effects even in hypothalamic astrocytes with altered inflammatory and immune signaling. Finally, this polyphenol can prepare astrocytes to better respond to injuries, including those associated with neuroimmunology defects., (© 2023. The Society for In Vitro Biology.)- Published
- 2023
- Full Text
- View/download PDF
22. L-2-Hydroxyglutaric Acid Administration to Neonatal Rats Elicits Marked Neurochemical Alterations and Long-Term Neurobehavioral Disabilities Mediated by Oxidative Stress.
- Author
-
Ribeiro RT, Carvalho AVS, Palavro R, Durán-Carabali LE, Zemniaçak ÂB, Amaral AU, Netto CA, and Wajner M
- Subjects
- Rats, Animals, Animals, Newborn, Antioxidants pharmacology, Oxidative Stress
- Abstract
L-2-Hydroxyglutaric aciduria (L-2-HGA) is an inherited neurometabolic disorder caused by deficient activity of L-2-hydroxyglutarate dehydrogenase. L-2-Hydroxyglutaric acid (L-2-HG) accumulation in the brain and biological fluids is the biochemical hallmark of this disease. Patients present exclusively neurological symptoms and brain abnormalities, particularly in the cerebral cortex, basal ganglia, and cerebellum. Since the pathogenesis of this disorder is still poorly established, we investigated the short-lived effects of an intracerebroventricular injection of L-2-HG to neonatal rats on redox homeostasis in the cerebellum, which is mostly affected in this disorder. We also determined immunohistochemical landmarks of neuronal viability (NeuN), astrogliosis (S100B and GFAP), microglia activation (Iba1), and myelination (MBP and CNPase) in the cerebral cortex and striatum following L-2-HG administration. Finally, the neuromotor development and cognitive abilities were examined. L-2-HG elicited oxidative stress in the cerebellum 6 h after its injection, which was verified by increased reactive oxygen species production, lipid oxidative damage, and altered antioxidant defenses (decreased concentrations of reduced glutathione and increased glutathione peroxidase and superoxide dismutase activities). L-2-HG also decreased the content of NeuN, MBP, and CNPase, and increased S100B, GFAP, and Iba1 in the cerebral cortex and striatum at postnatal days 15 and 75, implying long-standing neuronal loss, demyelination, astrocyte reactivity, and increased inflammatory response, respectively. Finally, L-2-HG administration caused a delay in neuromotor development and a deficit of cognition in adult animals. Importantly, the antioxidant melatonin prevented L-2-HG-induced deleterious neurochemical, immunohistochemical, and behavioral effects, indicating that oxidative stress may be central to the pathogenesis of brain damage in L-2-HGA., (© 2022. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2023
- Full Text
- View/download PDF
23. Short-term effects of therapeutic hypothermia following hypoxia-ischemia in neonatal male and female rats.
- Author
-
Zang J, Colucci ACM, Tassinari ID, Nunes RR, Andrade MKG, Spies FF, de Oliveira MR, Durán-Carabali LE, Rigon P, Netto CA, Paz AH, and de Fraga LS
- Subjects
- Humans, Rats, Animals, Male, Female, Rats, Wistar, Animals, Newborn, Ischemia therapy, Hypoxia, Brain, Hypoxia-Ischemia, Brain therapy, Hypothermia, Induced, Brain Injuries
- Abstract
Nowadays, the only treatment for human babies suffering from hypoxia-ischemia (HI) is therapeutic hypothermia (TH). However, a better understanding of the specific effects of TH in males and females is important to improve its clinical application. The present study evaluated the short-term effects of TH on the brain injury and behavioral outcomes in male and female neonatal rats submitted to neonatal HI. Seven-day-old Wistar rats underwent a surgery for unilateral occlusion of the right common carotid artery and were exposed to a hypoxic atmosphere (8% oxygen) for 75 min. Then, the animals in the TH group were submitted to TH (scalp temperature of 32°C) for 5 h. In the behavioral tests, no remarkable differences triggered by HI or TH were observed relative to SHAM animals. Only females of the HI group presented lower latency to complete the righting reflex test. TH reduced the volume of brain injury in males, but not in females. The animals of the HI group showed a reduction in the number of neurons in the CA1 and dentate gyrus (DG) regions of the hippocampus and TH partially prevented neuronal death. In the CA1 region of the hippocampus, animals from the HI group showed more degenerating cells relative to the SHAM, which was reversed by TH. In the DG, animals from the HI group showed an increase in the number of degenerating neurons, which was partially reversed by TH only in males. Our data show that HI leads to a brain injury, which was attenuated by TH in a sex-dependent way and clarify the importance of the assessment of males and females in order to outline specific strategies for the treatment of each sex in newborns suffering from HI., (© 2022 International Society for Developmental Neuroscience.)
- Published
- 2023
- Full Text
- View/download PDF
24. Protective effect of sex steroid hormones on morphological and cellular outcomes after neonatal hypoxia-ischemia: A meta-analysis of preclinical studies.
- Author
-
Durán-Carabali LE, Da Silva JL, Colucci ACM, Netto CA, and De Fraga LS
- Subjects
- Animals, Animals, Newborn, Brain, Estradiol, Ischemia, Progesterone pharmacology, Progesterone therapeutic use, Gonadal Steroid Hormones, Hypoxia-Ischemia, Brain drug therapy, Hypoxia-Ischemia, Brain metabolism
- Abstract
Sex steroid hormones play an important role in fetal development, brain functioning and neuronal protection. Growing evidence highlights the positive effects of these hormones against brain damage induced by neonatal hypoxia-ischemia (HI). This systematic review with meta-analysis aims to verify the efficacy of sex steroid hormones in preventing HI-induced brain damage in rodent models. The protocol was registered at PROSPERO and a total of 22 articles were included. Moderate to large effects were observed in HI animals treated with sex steroid hormones in reducing cerebral infarction size and cell death, increasing neuronal survival, and mitigating neuroinflammatory responses and astrocyte reactivity. A small effect was evidenced for cognitive function, but no significant effect for motor function; moreover, a high degree of heterogeneity was observed. In summary, data suggest that sex steroid hormones, such as progesterone and 17β estradiol, improve morphological and cellular outcomes following neonatal HI. Further research is paramount to examine neurological function during HI recovery and standardization of methodological aspects is imperative to reduce the risk of spurious findings., (Copyright © 2023 Elsevier Ltd. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
25. SARS-CoV-2-Induced Amyloidgenesis: Not One, but Three Hypotheses for Cerebral COVID-19 Outcomes.
- Author
-
Gonçalves CA, Bobermin LD, Sesterheim P, and Netto CA
- Abstract
The main neuropathological feature of Alzheimer's disease (AD) is extracellular amyloid deposition in senile plaques, resulting from an imbalance between the production and clearance of amyloid beta peptides. Amyloid deposition is also found around cerebral blood vessels, termed cerebral amyloid angiopathy (CAA), in 90% of AD cases. Although the relationship between these two amyloid disorders is obvious, this does not make CAA a characteristic of AD, as 40% of the non-demented population presents this derangement. AD is predominantly sporadic; therefore, many factors contribute to its genesis. Herein, the starting point for discussion is the COVID-19 pandemic that we are experiencing and how SARS-CoV-2 may be able to, both directly and indirectly, contribute to CAA, with consequences for the outcome and extent of the disease. We highlight the role of astrocytes and endothelial cells in the process of amyloidgenesis, as well as the role of other amyloidgenic proteins, such as fibrinogen and serum amyloid A protein, in addition to the neuronal amyloid precursor protein. We discuss three independent hypotheses that complement each other to explain the cerebrovascular amyloidgenesis that may underlie long-term COVID-19 and new cases of dementia.
- Published
- 2022
- Full Text
- View/download PDF
26. A Review of In Silico Research, SARS-CoV-2, and Neurodegeneration: Focus on Papain-Like Protease.
- Author
-
Rieder AS, Deniz BF, Netto CA, and Wyse ATS
- Subjects
- Glycyrrhizic Acid, Humans, Ligands, Molecular Docking Simulation, Pandemics, Papain chemistry, Papain metabolism, Peptide Hydrolases metabolism, SARS-CoV-2, Viral Proteins chemistry, Viral Proteins metabolism, Curcumin, COVID-19 Drug Treatment
- Abstract
Since the appearance of SARS-CoV-2 and the COVID-19 pandemic, the search for new approaches to treat this disease took place in the scientific community. The in silico approach has gained importance at this moment, once the methodologies used in this kind of study allow for the identification of specific protein-ligand interactions, which may serve as a filter step for molecules that can act as specific inhibitors. In addition, it is a low-cost and high-speed technology. Molecular docking has been widely used to find potential viral protein inhibitors for structural and non-structural proteins of the SARS-CoV-2, aiming to block the infection and the virus multiplication. The papain-like protease (PLpro) participates in the proteolytic processing of SARS-CoV-2 and composes one of the main targets studied for pharmacological intervention by in silico methodologies. Based on that, we performed a systematic review about PLpro inhibitors from the perspective of in silico research, including possible therapeutic molecules in relation to this viral protein. The neurological problems triggered by COVID-19 were also briefly discussed, especially relative to the similarities of neuroinflammation present in Alzheimer's disease. In this context, we focused on two molecules, curcumin and glycyrrhizinic acid, given their PLpro inhibitory actions and neuroprotective properties and potential therapeutic effects on COVID-19., (© 2022. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2022
- Full Text
- View/download PDF
27. Therapeutic hypothermia for the treatment of neonatal hypoxia-ischemia: sex-dependent modulation of reactive astrogliosis.
- Author
-
Fabres RB, Nunes RR, de Medeiros de Mattos M, Andrade MKG, Martini APR, Tassinari ID, Sanches EF, de Fraga LS, and Netto CA
- Subjects
- Animals, Male, Female, Rats, Gliosis therapy, Gliosis pathology, Rats, Wistar, Animals, Newborn, Brain, Ischemia pathology, Ischemia therapy, Disease Models, Animal, Hypoxia-Ischemia, Brain therapy, Hypoxia-Ischemia, Brain pathology, Hypothermia, Induced
- Abstract
Therapeutic hypothermia (TH) is the standard treatment for neonatal hypoxia-ischemia (HI) with a time window limited up to 6 h post injury. However, influence of sexual dimorphism in the therapeutic window for TH has not yet been elucidated in animal models of HI. Therefore, the aim of this study was to investigate the most effective time window to start TH in male and female rats submitted to neonatal HI. Wistar rats (P7) were divided into the following groups: NAÏVE and SHAM (control groups), HI (submitted to HI) and TH (submitted to HI and TH; 32ºC for 5 h). TH was started at 2 h (TH-2 h group), 4 h (TH-4 h group), or 6 h (TH-6 h group) after HI. At P14, animals were subjected to behavioural tests, volume of lesion and reactive astrogliosis assessments. Male and female rats from the TH-2 h group showed reduction in the latency of behavioral tests, and decrease in volume of lesion and intensity of GFAP immunofluorescence. TH-2 h females also showed reduction of degenerative cells and morphological changes in astrocytes. Interestingly, females from the TH-6 h group showed an increase in volume of lesion and in number of degenerative hippocampal cells, associated with worse behavioral performance. Together, these results indicate that TH neuroprotection is time- and sex-dependent. Moreover, TH started later (6 h) can worsen volume of brain lesion in females. These data indicate the need to develop specific therapeutic protocols for each sex and reinforce the importance of early onset of the hypothermic treatment., (© 2022. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2022
- Full Text
- View/download PDF
28. Role of Brain Β-endorphin in Memory Modulation Revisited.
- Author
-
Netto CA
- Subjects
- Animals, Avoidance Learning, Brain, Humans, Morphine pharmacology, Naloxone pharmacology, Memory, beta-Endorphin physiology
- Abstract
A possible role for the brain β-endorphin system in memory modulation was proposed by Ivan Izquierdo more than 30 years ago. Along with pharmacologic evidence of the effects of morphine and naloxone administered immediately after training in avoidance tasks and with the demonstration of medial-basal hypothalamus β-endorphin release after novelty detection, it was hypothesized that an endogenous opioid state present in the labile period of consolidation will be part of the memory of the newly acquired information. The fact that pre-test novelty exposure, through release of β-endorphin, or the injection of opioids facilitate retrieval supports that. The mechanisms through which novelty exerts its retrieval-enhancing effect were studied; evidence that several forms of amnesia induced by post-training treatments are due to unavailability of retrieval and not to a storage deficit, challenging the memory consolidation framework is discussed. In this review some of the original papers in the subject are revisited. Recent studies on the memory beneficial effects of novelty, both in animal models and in humans, indicate this is line of investigation is worth of pursuing and demonstrate the importance of the seminal work of Ivan Izquierdo in the field of memory modulation., (Copyright © 2022 IBRO. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
29. Effects of acrobatic training on spatial memory and astrocytic scar in CA1 subfield of hippocampus after chronic cerebral hypoperfusion in male and female rats.
- Author
-
Martini APR, Hoeper E, Pedroso TA, Carvalho AVS, Odorcyk FK, Fabres RB, Pereira NSC, and Netto CA
- Subjects
- Animals, Female, Male, Rats, Astrocytes pathology, Cicatrix pathology, Disease Models, Animal, Hippocampus, Maze Learning, Brain Ischemia pathology, Spatial Memory physiology
- Abstract
Chronic cerebral hypoperfusion leads to neuronal loss in the hippocampus and spatial memory impairments. Physical exercise is known to prevent cognitive deficits in animal models; and there is evidence of sex differences in behavioral neuroprotective approaches. The aim of present study was to investigate the effects of acrobatic training in male and female rats submitted to chronic cerebral hypoperfusion. Males and females rats underwent 2VO (two-vessel occlusion) surgery and were randomly allocated into 4 groups of males and 4 groups of females, as follows: 2VO acrobatic, 2VO sedentary, Sham acrobatic and Sham sedentary. The acrobatic training started 45 days after surgery and lasted 4 weeks; animals were then submitted to object recognition and water maze testing. Brain samples were collected for histological and morphological assessment and flow cytometry. 2VO causes cognitive impairments and acrobatic training prevented spatial memory deficits assessed in the water maze, mainly for females. Morphological analysis showed that 2VO animals had less NeuN labeling and acrobatic training prevented it. Increased number of GFAP positive cells was observerd in females; moreover, males had more branched astrocytes and acrobatic training prevented the branching after 2VO. Flow cytometry showed higher mitochondrial potential in trained animals and more reactive oxygen species production in males. Acrobatic training promoted neuronal survival and improved mitochondrial function in both sexes, and influenced the glial scar in a sex-dependent manner, associated to greater cognitive benefit to females after chronic cerebral hypoperfusion., (Copyright © 2022 Elsevier B.V. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
30. Coumestrol pre-treatment improves spatial learning and memory deficits following transient cerebral ischemia recruiting hippocampal GluR2 AMPA receptors.
- Author
-
Castro CC, de Souza Pagnussat A, Munhoz CD, and Netto CA
- Subjects
- Animals, Coumestrol, Hippocampus metabolism, Ischemia, Memory Disorders drug therapy, Memory Disorders etiology, Mice, Receptors, AMPA metabolism, Spatial Learning, Brain Ischemia complications, Brain Ischemia drug therapy, Ischemic Attack, Transient complications, Ischemic Attack, Transient drug therapy, Ischemic Attack, Transient genetics, Neuroprotective Agents pharmacology
- Abstract
Transient global ischemia is a leading cause of learning and memory dysfunction and induces a pattern of delayed neuronal death in the CA1 subfield of the hippocampus by down-regulating GluR2 mRNA AMPA receptors in this cerebral area. This study sought to investigate the neuroprotective effect of coumestrol against spatial memory impairment induced by global ischemia that leads to neural death by reducing the GluR2 receptors content in the hippocampal CA1 area. Our studies demonstrated that coumestrol administration prevented spatial memory deficits in mice. These findings suggest a cognitive enhancement role of coumestrol against cognitive impairment in ischemic events., (© 2022 Wiley Periodicals LLC.)
- Published
- 2022
- Full Text
- View/download PDF
31. Arundic Acid (ONO-2506) Attenuates Neuroinflammation and Prevents Motor Impairment in Rats with Intracerebral Hemorrhage.
- Author
-
Cordeiro JL, Neves JD, Nicola F, Vizuete AF, Sanches EF, Gonçalves CA, and Netto CA
- Subjects
- Animals, Caprylates pharmacology, Cerebral Hemorrhage complications, Cerebral Hemorrhage drug therapy, Cerebral Hemorrhage metabolism, Microglia metabolism, Rats, Motor Disorders complications, Neuroinflammatory Diseases
- Abstract
Intracerebral hemorrhage (ICH) is a severe stroke subtype caused by the rupture of blood vessels within the brain. Increased levels of S100B protein may contribute to neuroinflammation after ICH through activation of astrocytes and resident microglia, with the consequent production of proinflammatory cytokines and reactive oxygen species (ROS). Inhibition of astrocytic synthesis of S100B by arundic acid (AA) has shown beneficial effects in experimental central nervous system disorders. In present study, we administered AA in a collagenase-induced ICH rodent model in order to evaluate its effects on neurological deficits, S100B levels, astrocytic activation, inflammatory, and oxidative parameters. Rats underwent stereotactic surgery for injection of collagenase in the left striatum and AA (2 μg/μl; weight × 0.005) or vehicle in the left lateral ventricle. Neurological deficits were evaluated by the Ladder rung walking and Grip strength tests. Striatal S100B, astrogliosis, and microglial activation were assessed by immunofluorescence analysis. Striatal levels of interleukin 1β (IL-1β) and tumor necrosis factor α (TNF-α) were measured by ELISA, and the ROS production was analyzed by dichlorofluorescein (DCF) oxidation. AA treatment prevented motor dysfunction, reduced S100B levels, astrogliosis, and microglial activation in the damaged striatum, thus decreasing the release of proinflammatory cytokines IL-1β and TNF-α, as well as ROS production. Taken together, present results suggest that AA could be a pharmacological tool to prevent the harmful effects of increased S100B, attenuating neuroinflammation and secondary brain damage after ICH., (© 2020. Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2022
- Full Text
- View/download PDF
32. Effect of environmental enrichment on behavioral and morphological outcomes following neonatal hypoxia-ischemia in rodent models: A systematic review and meta-analysis.
- Author
-
Durán-Carabali LE, Odorcyk FK, Sanches EF, de Mattos MM, Anschau F, and Netto CA
- Subjects
- Animals, Animals, Newborn, Disease Models, Animal, Environment, Ischemia, Rats, Rats, Wistar, Rodentia, Hypoxia-Ischemia, Brain pathology
- Abstract
Neonatal hypoxia-ischemia (HI) is a major cause of mortality and morbidity in newborns and, despite recent advances in neonatal intensive care, there is no definitive treatment for this pathology. Once preclinical studies have shown that environmental enrichment (EE) seems to be a promising therapy for children with HI, the present study conducts a systematic review and meta-analysis of articles with EE in HI rodent models focusing on neurodevelopmental reflexes, motor and cognitive function as well as brain damage. The protocol was registered a priori at PROSPERO. The search was conducted in PubMed, Embase and PsycINFO databases, resulting in the inclusion of 22 articles. Interestingly, EE showed a beneficial impact on neurodevelopmental reflexes (SMD= -0.73, CI= [-0.98; -0.47], p< 0.001, I
2 = 0.0%), motor function (SMD= -0.55, CI= [-0.81; -0.28], p< 0.001, I2 = 62.6%), cognitive function (SMD= -0.93, CI= [-1.14; -0.72], p< 0.001, I2 = 27.8%) and brain damage (SMD= -0.80, CI= [-1.03; -0.58], p< 0.001, I2 = 10.7%). The main factors that potentiate EE positive effects were enhanced study quality, earlier age at injury as well as earlier start and longer duration of EE exposure. Overall, EE was able to counteract the behavioral and histological damage induced by the lesion, being a promising therapeutic strategy for HI., (© 2022. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)- Published
- 2022
- Full Text
- View/download PDF
33. Plinia trunciflora Extract Administration Prevents HI-Induced Oxidative Stress, Inflammatory Response, Behavioral Impairments, and Tissue Damage in Rats.
- Author
-
Carvalho AVS, Ribeiro RT, Durán-Carabali LE, Martini APR, Hoeper E, Sanches EF, Konrath EL, Dalmaz C, Wajner M, and Netto CA
- Subjects
- Animals, Animals, Newborn, Behavior, Animal drug effects, Brain drug effects, Brain pathology, Brain physiopathology, Fruit chemistry, Glutathione Peroxidase metabolism, Hypoxia-Ischemia, Brain complications, Hypoxia-Ischemia, Brain physiopathology, Lipid Peroxidation drug effects, Male, Neurons pathology, Oxidation-Reduction drug effects, Oxidative Stress drug effects, Rats, Rats, Wistar, Hypoxia-Ischemia, Brain drug therapy, Myrtaceae chemistry, Neuroinflammatory Diseases prevention & control, Neuroprotective Agents, Plant Extracts administration & dosage
- Abstract
The disruption of redox homeostasis and neuroinflammation are key mechanisms in the pathogenesis of brain hypoxia-ischemia (HI); medicinal plants have been studied as a therapeutic strategy, generally associated with the prevention of oxidative stress and inflammatory response. This study evaluates the neuroprotective role of the Plinia trunciflora fruit extract (PTE) in neonatal rats submitted to experimental HI. The HI insult provoked a marked increase in the lipoperoxidation levels and glutathione peroxidase (GPx) activity, accompanied by a decrease in the brain concentration of glutathione (GSH). Interestingly, PTE was able to prevent most of the HI-induced pro-oxidant effects. It was also observed that HI increased the levels of interleukin-1β in the hippocampus, and that PTE-treatment prevented this effect. Furthermore, PTE was able to prevent neuronal loss and astrocyte reactivity induced by HI, as demonstrated by NeuN and GFAP staining, respectively. PTE also attenuated the anxiety-like behavior and prevented the spatial memory impairment caused by HI. Finally, PTE prevented neural tissue loss in the brain hemisphere, the hippocampus, cerebral cortex, and the striatum ipsilateral to the HI. Taken together our results provide good evidence that the PTE extract has the potential to be investigated as an adjunctive therapy in the treatment of brain insult caused by neonatal hypoxia-ischemia.
- Published
- 2022
- Full Text
- View/download PDF
34. Pre- and early postnatal enriched environmental experiences prevent neonatal hypoxia-ischemia late neurodegeneration via metabolic and neuroplastic mechanisms.
- Author
-
Durán-Carabali LE, Odorcyk FK, Greggio S, Venturin GT, Sanches EF, Schu GG, Carvalho AS, Pedroso TA, de Sá Couto-Pereira N, Da Costa JC, Dalmaz C, Zimmer ER, and Netto CA
- Subjects
- Animals, Animals, Newborn, Female, Hypoxia-Ischemia, Brain psychology, Lactation metabolism, Lactation psychology, Male, Maze Learning physiology, Neurodegenerative Diseases metabolism, Neurodegenerative Diseases prevention & control, Neurodegenerative Diseases psychology, Positron-Emission Tomography methods, Pregnancy, Prenatal Exposure Delayed Effects psychology, Rats, Rats, Wistar, Brain metabolism, Environment, Hypoxia-Ischemia, Brain metabolism, Hypoxia-Ischemia, Brain prevention & control, Neuronal Plasticity physiology, Prenatal Exposure Delayed Effects metabolism
- Abstract
Prenatal and early postnatal periods are important for brain development and neural function. Neonatal insults such as hypoxia-ischemia (HI) causes prolonged neural and metabolic dysregulation, affecting central nervous system maturation. There is evidence that brain hypometabolism could increase the risk of adult-onset neurodegenerative diseases. However, the impact of non-pharmacologic strategies to attenuate HI-induced brain glucose dysfunction is still underexplored. This study investigated the long-term effects of early environmental enrichment in metabolic, cell, and functional responses after neonatal HI. Thereby, male Wistar rats were divided according to surgical procedure, sham, and HI (performed at postnatal day 3), and the allocation to standard (SC) or enriched condition (EC) during gestation and lactation periods. In-vivo cerebral metabolism was assessed by means of [
18 F]-FDG micro-positron emission tomography, and cognitive, biochemical, and histological analyses were performed in adulthood. Our findings reveal that HI causes a reduction in glucose metabolism and glucose transporter levels as well as hyposynchronicity in metabolic brain networks. However, EC during prenatal or early postnatal period attenuated these metabolic disturbances. A positive correlation was observed between [18 F]-FDG values and volume ratios in adulthood, indicating that preserved tissue by EC is metabolically active. EC promotes better cognitive scores, as well as down-regulation of amyloid precursor protein in the parietal cortex and hippocampus of HI animals. Furthermore, growth-associated protein 43 was up-regulated in the cortex of EC animals. Altogether, results presented support that EC during gestation and lactation period can reduce HI-induced impairments that may contribute to functional decline and progressive late neurodegeneration., (© 2020 International Society for Neurochemistry.)- Published
- 2021
- Full Text
- View/download PDF
35. Pregnancy swimming prevents early brain mitochondrial dysfunction and causes sex-related long-term neuroprotection following neonatal hypoxia-ischemia in rats.
- Author
-
Sanches EF, Dos Santos TM, Odorcyk F, Untertriefallner H, Rezena E, Hoeper E, Avila T, Martini AP, Venturin GT, da Costa JC, Greggio S, Netto CA, and Wyse AT
- Subjects
- Animals, Animals, Newborn, Brain pathology, Female, Hypoxia-Ischemia, Brain pathology, Male, Maze Learning physiology, Mitochondria pathology, Pregnancy, Rats, Rats, Wistar, Time Factors, Brain metabolism, Hypoxia-Ischemia, Brain metabolism, Hypoxia-Ischemia, Brain prevention & control, Mitochondria metabolism, Neuroprotection physiology, Sex Characteristics, Swimming physiology
- Abstract
Neonatal hypoxia-ischemia (HI) is a major cause of cognitive impairments in infants. Antenatal strategies improving the intrauterine environment can have high impact decreasing pregnancy-derived intercurrences. Physical exercise alters the mother-fetus unity and has been shown to prevent the energetic challenge imposed by HI. This study aimed to reveal neuroprotective mechanisms afforded by pregnancy swimming on early metabolic failure and late cognitive damage, considering animals' sex as a variable. Pregnant Wistar rats were submitted to daily swimming exercise (20' in a tank filled with 32 °C water) during pregnancy. Neonatal HI was performed in male and female pups at postnatal day 7. Electron chain transport, mitochondrial mass and function and ROS formation were assessed in the right brain hemisphere 24 h after HI. From PND45, reference and working spatial memory were tested in the Morris water maze. MicroPET-FDG images were acquired 24 h after injury (PND8) and at PND60, following behavioral analysis. HI induced early energetic failure, decreased enzymatic activity in electron transport chain, increased production of ROS in cortex and hippocampus as well as caused brain glucose metabolism dysfunction and late cognitive impairments. Maternal swimming was able to prevent mitochondrial dysfunction and to improve spatial memory. The intergenerational effects of swimming were sex-specific, since male rats were benefited most. In conclusion, maternal swimming was able to affect the mitochondrial response to HI in the offspring's brains, preserving its function and preventing cognitive damage in a sex-dependent manner, adding relevant information on maternal exercise neuroprotection and highlighting the importance of mitochondria as a therapeutic target for HI neuropathology., (Copyright © 2021. Published by Elsevier Inc.)
- Published
- 2021
- Full Text
- View/download PDF
36. Differential Age-Dependent Mitochondrial Dysfunction, Oxidative Stress, and Apoptosis Induced by Neonatal Hypoxia-Ischemia in the Immature Rat Brain.
- Author
-
Odorcyk FK, Ribeiro RT, Roginski AC, Duran-Carabali LE, Couto-Pereira NS, Dalmaz C, Wajner M, and Netto CA
- Subjects
- Age Factors, Animals, Disease Models, Animal, Female, Homeostasis physiology, Oxidation-Reduction, Oxygen Consumption physiology, Rats, Rats, Wistar, Apoptosis physiology, Hippocampus metabolism, Hypoxia-Ischemia, Brain metabolism, Mitochondria metabolism, Oxidative Stress physiology
- Abstract
Neonatal hypoxia-ischemia (HI) is among the main causes of mortality and morbidity in newborns. Experimental studies show that the immature rat brain is less susceptible to HI injury, suggesting that changes that occur during the first days of life drastically alter its susceptibility. Among the main developmental changes observed is the mitochondrial function, namely, the tricarboxylic acid (TCA) cycle and respiratory complex (RC) activities. Therefore, in the present study, we investigated the influence of neonatal HI on mitochondrial functions, redox homeostasis, and cell damage at different postnatal ages in the hippocampus of neonate rats. For this purpose, animals were divided into four groups: sham postnatal day 3 (ShP3), HIP3, ShP11, and HIP11. We initially observed increased apoptosis in the HIP11 group only, indicating a higher susceptibility of these animals to brain injury. Mitochondrial damage, as determined by flow cytometry showing mitochondrial swelling and loss of mitochondrial membrane potential, was also demonstrated only in the HIP11 group. This was consistent with the decreased mitochondrial oxygen consumption, reduced TCA cycle enzymes, and RC activities and induction of oxidative stress in this group of animals. Considering that HIP3 and the sham animals showed no alteration of mitochondrial functions, redox homeostasis, and showed no apoptosis, our data suggest an age-dependent vulnerability of the hippocampus to hypoxia-ischemia. The present results highlight age-dependent metabolic differences in the brain of neonate rats submitted to HI indicating that different treatments might be needed for HI newborns with different gestational ages.
- Published
- 2021
- Full Text
- View/download PDF
37. Arundic acid (ONO-2526) inhibits stimulated-S100B secretion in inflammatory conditions.
- Author
-
Vizuete AFK, de Lima Cordeiro J, Neves JD, Seady M, Grun LK, Barbé-Tuana FM, Leite MC, Netto CA, and Gonçalves CA
- Subjects
- Animals, Astrocytes drug effects, Astrocytes metabolism, Cells, Cultured, Hippocampus cytology, Hippocampus metabolism, Lipopolysaccharides toxicity, Male, Rats, Rats, Wistar, S100 Calcium Binding Protein beta Subunit genetics, Tumor Necrosis Factor-alpha metabolism, Anti-Inflammatory Agents pharmacology, Caprylates pharmacology, Hippocampus drug effects, Neuroprotective Agents pharmacology, S100 Calcium Binding Protein beta Subunit metabolism
- Abstract
Astrocytes respond to injury by modifying the expression profile of several proteins, including the S100 calcium-binding protein B (S100B), assumed to be a marker as well as a mediator of brain injury. AA is an inhibitor of S100B synthesis and plays a protective role in different models of brain injury, as decreases in S100B expression cause decreases in extracellular S100B. However, S100B mRNA expression, S100B protein content and S100B secretion do not always occur in association; as such, we herein investigated the effect of AA on S100B secretion, using different approaches with three stimulating conditions for S100B secretion, namely, low potassium medium, TNF-α (in hippocampal slices) and LPS exposure (in astrocyte cultures). Our data indicate that AA directly affects S100B secretion, indicating that the extracellular levels of this astroglial protein may be mediating the action of this compound. More importantly, AA had no effect on basal S100B secretion, but inhibited stimulated S100B secretion (stimulated either by the proinflammatory molecules, LPS or TNF-α, or by low potassium medium). Data from hippocampal slices that were directly exposed to AA, or from animals that received the acid by intracerebroventricular infusion, contribute to understanding its neuroprotective effect., (Copyright © 2021 Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
38. Experimental cerebral palsy causes microstructural brain damage in areas associated to motor deficits but no spatial memory impairments in the developing rat.
- Author
-
Sanches EF, Carvalho AS, van de Looij Y, Toulotte A, Wyse AT, Netto CA, and Sizonenko SV
- Abstract
Introduction: Cerebral palsy (CP) is the major cause of motor and cognitive impairments during childhood. CP can result from direct or indirect structural injury to the developing brain. In this study, we aimed to describe brain damage and behavioural alterations during early adult life in a CP model using the combination of maternal inflammation, perinatal anoxia and postnatal sensorimotor restriction., Methods: Pregnant Wistar rats were injected intraperitoneally with 200 µg/kg LPS at embryonic days E18 and E19. Between 3 and 6 h after birth (postnatal day 0 - PND0), pups of both sexes were exposed to anoxia for 20 min. From postnatal day 2 to 21, hindlimbs of animals were immobilized for 16 h daily during their active phase. From PND40, locomotor and cognitive tests were performed using Rota-Rod, Ladder Walking and Morris water Maze. Ex-vivo MRI Diffusion Tensor Imaging (DTI) and Neurite Orientation Dispersion and Density Imaging (NODDI) were used to assess macro and microstructural damage and brain volume alterations induced by the model. Myelination and expression of neuronal, astroglial and microglial markers, as well as apoptotic cell death were evaluated by immunofluorescence., Results: CP animals showed decreased body weight, deficits in gross (rota-rod) and fine (ladder walking) motor tasks compared to Controls. No cognitive impairments were observed. Ex-vivo MRI showed decreased brain volumes and impaired microstructure in the cingulate gyrus and sensory cortex in CP brains. Histological analysis showed increased cell death, astrocytic reactivity and decreased thickness of the corpus callosum and altered myelination in CP animals. Hindlimb primary motor cortex analysis showed increased apoptosis in CP animals. Despite the increase in NeuN and GFAP, no differences between groups were observed as well as no co-localization with the apoptotic marker. However, an increase in Iba-1
+ microglia with co-localization to cleaved caspase 3 was observed., Conclusion: Our results suggest that experimental CP induces long-term brain microstructural alterations in myelinated structures, cell death in the hindlimb primary motor cortex and locomotor impairments. Such new evidence of brain damage could help to better understand CP pathophysiological mechanisms and guide further research for neuroprotective and neurorehabilitative strategies for CP patients., (Copyright © 2021. Published by Elsevier B.V.)- Published
- 2021
- Full Text
- View/download PDF
39. Lactate Administration Reduces Brain Injury and Ameliorates Behavioral Outcomes Following Neonatal Hypoxia-Ischemia.
- Author
-
Tassinari ID, Andrade MKG, da Rosa LA, Hoff MLM, Nunes RR, Vogt EL, Fabres RB, Sanches EF, Netto CA, Paz AH, and de Fraga LS
- Subjects
- Animals, Animals, Newborn, Brain, Disease Models, Animal, Female, Hypoxia, Ischemia, Lactic Acid, Male, Rats, Rats, Wistar, Brain Injuries, Hypoxia-Ischemia, Brain drug therapy, Neuroprotective Agents
- Abstract
Neonatal hypoxic-ischemic encephalopathy is a major cause of mortality and disability in newborns and the only standard approach for treating this condition is therapeutic hypothermia, which shows some limitations. Thus, putative neuroprotective agents have been tested in animal models. The present study evaluated the administration of lactate, a potential energy substrate of the central nervous system (CNS) in an animal model of hypoxia-ischemia (HI), that mimics in neonatal rats the brain damage observed in human newborns. Seven-day-old (P7) male and female Wistar rats underwent permanent common right carotid occlusion combined with an exposition to a hypoxic atmosphere (8% oxygen) for 60 min. Animals were assigned to four experimental groups: HI, HI + LAC, SHAM, SHAM + LAC. Lactate was administered intraperitoneally 30 min and 2 h after hypoxia in HI + LAC and SHAM + LAC groups. HI and SHAM groups received vehicle at the same time points. The volume of brain lesion was evaluated in P9. Animals underwent behavioral assessments: negative geotaxis, righting reflex (P8 and P14), and cylinder test (P20). Lactate administration reduced the volume of brain lesion and improved behavioral parameters after HI in both sexes. Thus, lactate administration could be a neuroprotective strategy for the treatment of neonatal HI, a disorder still affecting a significant percentage of human newborns., (Copyright © 2020 IBRO. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
40. Long-Lasting Actions of Progesterone Protect the Neonatal Brain Following Hypoxia-Ischemia.
- Author
-
Fabres RB, Montes NL, Camboim YM, de Souza SK, Nicola F, Tassinari ID, Ribeiro MFM, Netto CA, and de Fraga LS
- Subjects
- Animals, Animals, Newborn, Brain metabolism, Hypoxia drug therapy, Hypoxia metabolism, Hypoxia-Ischemia, Brain metabolism, Hypoxia-Ischemia, Brain pathology, Ischemia drug therapy, Male, Rats, Wistar, Brain drug effects, Hypoxia-Ischemia, Brain drug therapy, Ischemia metabolism, Neuroprotective Agents pharmacology, Progesterone pharmacology
- Abstract
Neonatal hypoxia-ischemia (HI) is the leading cause of mortality and morbidity in newborns, occurring in approximately 2% of live births. Neuroprotective actions of progesterone (PROG) have already been described in animal models of brain lesions. However, PROG actions on neonates are still controversial. Here, we treated male Wistar rats exposed to HI with PROG. Five experimental groups were defined (n = 6/group) according to the scheme of PROG administration (10 mg/kg): SHAM (animals submitted to a fictitious surgery, without ischemia induction, and maintained under normoxia), HI (animals undergoing HI), BEFORE (animals undergoing HI and receiving PROG immediately before HI), AFTER (animals undergoing HI and receiving PROG at 6 and 24 h after HI) and BEFORE/AFTER (animals undergoing HI and receiving PROG immediately before and 6 and 24 h after HI). At P14 (7 days following HI), the volumes of lesion of the cerebral hemisphere and the hippocampus ipsilateral to the cerebral ischemia were evaluated, along with p-Akt, cleaved caspase-3 and GFAP expression in the hippocampus. PROG reduces the loss of brain tissue caused by HI. Moreover, when administered after HI, PROG was able to increase p-Akt expression and reduce both cleaved caspase-3 and GFAP expression in the hippocampus. In summary, it was possible to observe a neuroprotective action of PROG on the brain of neonatal animals exposed to experimental HI. This is the first study suggesting PROG-dependent Akt activation is able to regulate negatively cleaved caspase-3 and GFAP expression protecting neonatal hypoxic-ischemic brain tissue from apoptosis and reactive gliosis.
- Published
- 2020
- Full Text
- View/download PDF
41. Differential glucose and beta-hydroxybutyrate metabolism confers an intrinsic neuroprotection to the immature brain in a rat model of neonatal hypoxia ischemia.
- Author
-
Odorcyk FK, Duran-Carabali LE, Rocha DS, Sanches EF, Martini AP, Venturin GT, Greggio S, da Costa JC, Kucharski LC, Zimmer ER, and Netto CA
- Subjects
- Animals, Animals, Newborn, Disease Models, Animal, Male, Rats, Rats, Wistar, 3-Hydroxybutyric Acid metabolism, Glucose metabolism, Hypoxia-Ischemia, Brain metabolism, Neuroprotection physiology
- Abstract
Neonatal hypoxia ischemia (HI) is the main cause of newborn mortality and morbidity. Preclinical studies have shown that the immature rat brain is more resilient to HI injury, suggesting innate mechanisms of neuroprotection. During neonatal period brain metabolism experience changes that might greatly affect the outcome of HI injury. Therefore, the aim of the present study was to investigate how changes in brain metabolism interfere with HI outcome in different stages of CNS development. For this purpose, animals were divided into 6 groups: HIP3, HIP7 and HIP11 (HI performed at postnatal days 3, 7 and 11, respectively), and their respective shams. In vivo [
18 F]FDG micro positron emission tomography (microPET) imaging was performed 24 and 72 h after HI, as well as ex-vivo assessments of glucose and beta-hydroxybutyrate (BHB) oxidation. At adulthood behavioral tests and histology were performed. Behavioral and histological analysis showed greater impairments in HIP11 animals, while HIP3 rats were not affected. Changes in [18 F]FDG metabolism were found only in the lesion area of HIP11, where a substantial hypometabolism was detected. Furthermore, [18 F]FDG hypometabolism predicted impaired cognition and worst histological outcomes at adulthood. Finally, substrate oxidation assessments showed that glucose oxidation remained unaltered and higher level of BHB oxidation found in P3 animals, suggesting a more resilient metabolism. Overall, present results show [18 F]FDG microPET predicts long-term injury outcome and suggests that higher BHB utilization is one of the mechanisms that confer the intrinsic neuroprotection to the immature brain and should be explored as a therapeutic target for treatment of HI., (Copyright © 2020 Elsevier Inc. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
42. Arundic Acid (ONO-2506), an Inhibitor of S100B Protein Synthesis, Prevents Neurological Deficits and Brain Tissue Damage Following Intracerebral Hemorrhage in Male Wistar Rats.
- Author
-
Cordeiro JL, Neves JD, Vizuete AF, Aristimunha D, Pedroso TA, Sanches EF, Gonçalves CA, and Netto CA
- Subjects
- Animals, Caprylates, Cerebral Hemorrhage complications, Cerebral Hemorrhage drug therapy, Disease Models, Animal, Male, Rats, Rats, Wistar, S100 Calcium Binding Protein beta Subunit, Brain Injuries, Neuroprotective Agents pharmacology
- Abstract
Stroke is one of the leading causes of mortality and neurological morbidity. Intracerebral hemorrhage (ICH) has the poorest prognosis among all stroke subtypes and no treatment has been effective in improving outcomes. Following ICH, the observed high levels of S100B protein have been associated with worsening of injury and neurological deficits. Arundic acid (AA) exerts neuroprotective effects through inhibition of astrocytic synthesis of S100B in some models of experimental brain injury; however, it has not been studied in ICH. The aim of this study was to evaluate the effects of intracerebroventricular (ICV) administration of AA in male Wistar rats submitted to ICH model assessing the following variables: reactive astrogliosis, S100B levels, antioxidant defenses, cell death, lesion extension and neurological function. Firstly, AA was injected at different doses (0.02, 0.2, 2 and 20 μg/μl) in the left lateral ventricle in order to observe which dose would decrease GFAP and S100B striatal levels in non-injured rats. Following determination of the effective dose, ICH damage was induced by IV-S collagenase intrastrial injection and 2 μg/μl AA was injected through ICV route immediately before injury. AA treatment prevented ICH-induced neurological deficits and tissue damage, inhibited excessive astrocytic activation and cellular apoptosis, reduced peripheral and central S100B levels (in striatum, serum and cerebrospinal fluid), improved neuronal survival and enhanced the antioxidant defences after injury. Altogether, these results suggest that S100B is a viable target for treating ICH and highlight AA as an interesting strategy for improving neurological outcome after experimental brain hemorrhage., (Copyright © 2020 IBRO. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
43. Neurometabolic effects of sweetened solution intake during adolescence related to depressive-like phenotype in rats.
- Author
-
Arcego DM, Olivo LB, Moraes RO, Garcia EDS, Silveira AC, Krolow R, Couto-Pereira NS, Lampert C, Toniazzo AP, Nicola FDC, Sanches EF, Aristimunha D, Hoppe JB, Klein CP, Fontella FU, Almeida RF, Gamaro GD, Fróes FCTDS, Leite MC, Netto CA, Zancan DM, and Dalmaz C
- Subjects
- Animals, Female, Male, Phenotype, Rats, Sucrose, Aspartame toxicity, Phosphatidylinositol 3-Kinases, Sweetening Agents toxicity
- Abstract
Objective: Exposure to artificial sweeteners, such as aspartame, during childhood and adolescence has been increasing in recent years. However, the safe use of aspartame has been questioned owing to its potentially harmful effects on the developing brain. The aim of this study was to test whether the chronic consumption of aspartame during adolescence leads to a depressive-like phenotype and to investigate the possible mechanisms underlying these behavioral changes., Methods: Adolescent male and female rats were given unlimited access to either water, solutions of aspartame, or sucrose in their home cages from postnatal day 21 to 55., Results: Forced swim test revealed that both chronic aspartame and sucrose intake induced depressive-like behaviord, which was more pronounced in males. Additionally, repeated aspartame intake was associated with increased cerebrospinal fluid (CSF) aspartate levels, decreased hippocampal neurogenesis, and reduced activation of the hippocampal leptin signaling pathways in males. In females, we observed a main effect of aspartame: reducing PI3K/AKT one of the brain-derived neurotrophic factor pathways; aspartame also increased CSF aspartate levels and decreased the immunocontent of the GluN2A subunit of the N-methyl-d-aspartic acid receptor., Conclusion: The findings revealed that repeated aspartame intake during adolescence is associated with a depressive-like phenotype and changes in brain plasticity. Interestingly, males appear to be more vulnerable to the adverse neurometabolic effects of aspartame than females, demonstrating a sexually dimorphic response. The present results highlighted the importance of understanding the effects caused by the constant use of this artificial sweetener in sensitive periods of development and contribute to regulation of its safe use., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
44. Long-term changes in metabolic brain network drive memory impairments in rats following neonatal hypoxia-ischemia.
- Author
-
Azevedo PN, Zanirati G, Venturin GT, Schu GG, Durán-Carabali LE, Odorcyk FK, Soares AV, Laguna GO, Netto CA, Zimmer ER, da Costa JC, and Greggio S
- Subjects
- Animals, Brain diagnostic imaging, Disease Models, Animal, Glucose metabolism, Hypoxia-Ischemia, Brain complications, Hypoxia-Ischemia, Brain diagnostic imaging, Male, Memory Disorders diagnostic imaging, Memory Disorders etiology, Nerve Net diagnostic imaging, Positron-Emission Tomography, Rats, Rats, Wistar, Brain metabolism, Hypoxia-Ischemia, Brain metabolism, Memory Disorders metabolism, Nerve Net metabolism
- Abstract
Background and Purpose: Hypoxia and cerebral ischemia (HI) events are capable of triggering important changes in brain metabolism, including glucose metabolism abnormalities, which may be related to the severity of the insult. Using positron emission microtomography (microPET) with [
18 F]fluorodeoxyglucose (18 F-FDG), this study proposes to assess abnormalities of brain glucose metabolism in adult rats previously submitted to the neonatal HI model. We hypothesize that cerebral metabolic outcomes will be associated with cognitive deficits and magnitude of brain injury., Methods: Seven-day-old rats were subjected to an HI model, induced by permanent occlusion of the right common carotid artery and systemic hypoxia.18 F-FDG-microPET was used to assess regional and whole brain glucose metabolism in rats at 60 postnatal days (PND 60). An interregional cross-correlation matrix was utilized to construct metabolic brain networks (MBN). Rats were also subjected to the Morris Water Maze (MWM) to evaluate spatial memory and their brains were processed for volumetric evaluation., Results: Brain glucose metabolism changes were observed in adult rats after neonatal HI insult, limited to the right brain hemisphere. However, not all HI animals exhibited significant cerebral hypometabolism. Hippocampal glucose metabolism was used to stratify HI animals into HI hypometabolic (HI-h) and HI non-hypometabolic (HI non-h) groups. The HI-h group had drastic MBN disturbance, cognitive deficit, and brain tissue loss, concomitantly. Conversely, HI non-h rats had normal brain glucose metabolism and brain tissue preserved, but also presented MBN changes and spatial memory impairment. Furthermore, data showed that brain glucose metabolism correlated with cognitive deficits and brain volume outcomes., Conclusions: Our findings demonstrated that long-term changes in MBN drive memory impairments in adult rats subjected to neonatal hypoxic ischemia, using in vivo imaging microPET-FDG. The MBN analyses identified glucose metabolism abnormalities in HI non-h animals, which were not detected by conventional18 F-FDG standardized uptake value (SUVr) measurements. These animals exhibited a metabolic brain signature that may explain the cognitive deficit even with no identifiable brain damage., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2020 Elsevier Inc. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
45. Previous adaptation triggers distinct molecular pathways and modulates early and long-term neuroprotective effects of pregnancy swimming preventing neonatal hypoxia-ischemia damage in rats.
- Author
-
Sanches EF, Fabres RB, Mazeron LS, Rezena E, Tassinari ID, Carvalho AS, Andrade MK, de Fraga LS, and Netto CA
- Subjects
- Animals, Animals, Newborn, Behavior, Animal, Female, Hypoxia-Ischemia, Brain prevention & control, Pregnancy, Rats, Wistar, Adaptation, Physiological, Hippocampus metabolism, Hypoxia-Ischemia, Brain metabolism, Neuroprotection, Swimming
- Abstract
Neonatal hypoxia-ischemia (HI) is one of the main causes of neurological damage in newborns. Pregnancy swimming (PS) alters brain maturation and has neuroprotective effects following HI; however, variables such as timing play a decisive role in its effects. Prior to mating, we tested if adaptation of female rats to a tank filled with water at 32 °C for 7 days before mating, modulates PS benefits. After mating, rats swam 20 min/day or remained in standard cages. Seven-day-old pups were subjected to HI (right common carotid artery occlusion followed by FiO
2 8% for 60 min). Animals were divided into 8 experimental groups, adaptation, swimming and injury. Astrocytic reactivity, apoptosis-related proteins, neurotrophins and cell survival markers expression were assessed in the hippocampus 24 h after HI. From PND45, animals performed behavioral tests followed by histological assessment. Three-way ANOVA showed a significant increase in astrogliosis only in non-adapted HI animals. Swimming decreased apoptotic cell death despite adaptation period in both exercised groups. Cylinder evidenced HI impairments; no effect of swimming or adaptation period were observed. In the open field, only HI animals whose mothers had been adapted had increased locomotion; moreover, swimming reversed HI damage. Hemisphere and hippocampus were preserved only in the HI group whose mothers swam before mating, suggesting a preconditioning effect mediated by the adaptation. In summary, adaptation period plays a major role in the mechanisms involving neuroprotection afforded by PS and needs to be further explored in future studies involving damage to the neonatal brain., (Copyright © 2020 Elsevier B.V. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
46. Chronic mild hyperhomocysteinemia induces anxiety-like symptoms, aversive memory deficits and hippocampus atrophy in adult rats: New insights into physiopathological mechanisms.
- Author
-
Wyse ATS, Sanches EF, Dos Santos TM, Siebert C, Kolling J, and Netto CA
- Subjects
- Adenosine Triphosphate metabolism, Animals, Anxiety pathology, Atrophy etiology, Atrophy pathology, Avoidance Learning, Chronic Disease, DNA Damage physiology, Electron Transport Complex IV metabolism, Hippocampus physiopathology, Homocysteine blood, Hyperhomocysteinemia chemically induced, Male, Memory Disorders physiopathology, Open Field Test, Oxidative Stress physiology, Rats, Rats, Wistar, Anxiety etiology, Hippocampus pathology, Hyperhomocysteinemia complications, Memory Disorders etiology
- Abstract
In the last decade, increased homocysteine levels have been implicated as a risk factor for neurodegenerative and psychiatric disorders. We have developed an experimental model of chronic mild hyperhomocysteinemia (HHcy) in order to observe metabolic impairments in the brain of adult rodents. Besides its known effects on brain metabolism, the present study sought to investigate whether chronic mild HHcy could induce learning/memory impairments associated with biochemical and histological damage to the hippocampus. Adult male Wistar rats received daily subcutaneous injections of homocysteine (0.03 μmol/g of body weight) twice a day, from the 30th to the 60th day of life or saline solution (Controls). After injections, anxiety-like and memory tests were performed. Following behavioral analyses, brains were sliced and hippocampal volumes assessed and homogenized for redox state assessment, antioxidant activity, mitochondrial functioning (chain respiratory enzymes and ATP levels) and DNA damage analyses. Behavioral analyses showed that chronic mild HHcy may induce anxiety-like behavior and impair long-term aversive memory (24 h) that was evaluated by inhibitory avoidance task. Mild HHcy decreased locomotor and/or exploratory activities in elevated plus maze test and caused hippocampal atrophy. Decrease in cytochrome c oxidase, DNA damage and redox state changes were also observed in hippocampus of adult rats subjected to mild HHcy. Our findings show that chronic mild HHcy alters biochemical and histological parameters in the hippocampus, leading to behavioral impairments. These findings might be considered in future studies aiming to search for alternative strategies for treating the behavioral impairments in patients with mild elevations in homocysteine levels., (Copyright © 2019 Elsevier B.V. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
47. Phytoestrogen coumestrol attenuates brain mitochondrial dysfunction and long-term cognitive deficits following neonatal hypoxia-ischemia.
- Author
-
Anastacio JBR, Sanches EF, Nicola F, Odorcyk F, Fabres RB, and Netto CA
- Subjects
- Animals, Brain metabolism, Cognitive Dysfunction metabolism, Coumestrol therapeutic use, Hypoxia-Ischemia, Brain metabolism, Male, Maze Learning drug effects, Mitochondria metabolism, Neuroprotective Agents therapeutic use, Phytoestrogens pharmacology, Phytoestrogens therapeutic use, Rats, Rats, Wistar, Reactive Oxygen Species metabolism, Brain drug effects, Cognition drug effects, Cognitive Dysfunction drug therapy, Coumestrol pharmacology, Hypoxia-Ischemia, Brain drug therapy, Mitochondria drug effects, Neuroprotective Agents pharmacology
- Abstract
Introduction: Neonatal Hypoxia-Ischemia (HI) is a major cause of morbidity and mortality, and is frequently associated with short and long-term neurologic and cognitive impairments. The HI injury causes mitochondrial damage leading to increased production of reactive oxygen species (ROS). Phytoestrogens are non-steroidal plant substances structurally and functionally similar to estrogen. Coumestrol is a potent isoflavonoid with a protective effect against ischemic brain damage in adult rats. Our aim was to determine if coumestrol treatment following neonatal HI attenuates the long-term cognitive deficits induced by neonatal HI, as well as to investigate one possible mechanism underlying its potential effect., Methods: On the 7th postnatal day, male Wistar rats were submitted to the Levine-Rice HI model. Intraperitoneal injections of 20 mg/kg of coumestrol, or vehicle, were administered immediately pre-hypoxia or 3 h post-hypoxia. At 12 h after HI the mitochondrial status and ROS levels were determined. At 60th postnatal day the cognitive deficits were revealed in the Morris water maze reference and working spatial memories. Following behavioral analysis, histological assessment was performed and reactive astrogliosis was measured by GFAP expression., Results: Results demonstrate that both pre- and post-HI administration of coumestrol were able to counteract the long-term cognitive and morphological impairments caused by HI, as well as to block the late reactive astrogliosis. The pre-HI administration of coumestrol was able to prevent the early mitochondrial dysfunction in the hippocampus of injured rat pups., Conclusion: Present data suggest that coumestrol exerts protection against experimental neonatal brain hypoxia-ischemia through, at least in part, early modulation of mitochondrial function., (Copyright © 2019 ISDN. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
48. Galantamine improves functional recovery and reduces lesion size in a rat model of spinal cord injury.
- Author
-
Sperling LE, Pires Reis K, Nicola F, Euzebio Teixeira C, Gulielmin Didó G, Garrido Dos Santos M, Konrath E, Netto CA, and Pranke P
- Subjects
- Animals, Contusions pathology, Galantamine metabolism, Hindlimb physiopathology, Locomotion drug effects, Male, Motor Activity drug effects, Neuroprotective Agents pharmacology, Rats, Rats, Wistar, Recovery of Function physiology, Spinal Cord metabolism, Spinal Cord Injuries physiopathology, Galantamine pharmacology, Recovery of Function drug effects, Spinal Cord Injuries drug therapy
- Abstract
Spinal cord injury (SCI) is a medical condition that currently lacks effective treatment. Galantamine is a reversible, competitive acetylcholinesterase inhibitor, used to treat patients with Alzheimeŕs disease. It has been demonstrated that galantamine increases cerebral neurogenesis and has a neuroprotective effect by binding to nicotinic receptors and has an anti-inflammatory effect due to its allosteric binding to the α7nAChR. In the present study, the effects of galantamine on functional recovery and histological outcome in a rat contusion model of SCI were analyzed. Male Wistar rats were submitted to SCI using a NYU/MASCIS impactor. The animals from the galantamine group were treated with 5 mg/kg galantamine intraperitoneally for 5 days. The Basso, Beattie and Bresnahan scale (BBB) was used to evaluate locomotor activity. The expression of beta3-tubulin, NFM, GFAP, O4, CD68 and CD3 was analyzed by flow cytometry. Rats that received galantamine had significantly higher BBB scores in comparison with the control lesion group. Galantamine treatment increased the percentage of NFM positive cells at 6 weeks post-injury and reduced the size of the lesion. The results indicate that galantamine increased tissue survival and accelerated hind limb motor function recovery. This is the first study that has shown the possibility of therapeutic use of galantamine in a model of acute spinal cord injury., (Copyright © 2019 Elsevier B.V. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
49. Tissue Injury and Astrocytic Reaction, But Not Cognitive Deficits, Are Dependent on Hypoxia Duration in Very Immature Rats Undergoing Neonatal Hypoxia-Ischemia.
- Author
-
Durán-Carabali LE, Sanches EF, Odorcyk FK, Nicola F, Mestriner RG, Reichert L, Aristimunha D, Pagnussat AS, and Netto CA
- Subjects
- Animals, Animals, Newborn, Brain pathology, Cognitive Dysfunction physiopathology, Female, Gliosis physiopathology, Hypoxia-Ischemia, Brain pathology, Male, Maze Learning physiology, Memory Disorders physiopathology, Rats, Wistar, Regression Analysis, Time Factors, Astrocytes physiology, Hypoxia-Ischemia, Brain physiopathology
- Abstract
Preterm birth and hypoxia-ischemia (HI) are major causes of neonatal death and neurological disabilities in newborns. The widely used preclinical HI model combines carotid occlusion with hypoxia exposure; however, the relationship between different hypoxia exposure periods with brain tissue loss, astrocyte reactivity and behavioral impairments following HI is lacking. Present study evaluated HI-induced behavioral and morphological consequences in rats exposed to different periods of hypoxia at postnatal day 3. Wistar rats of both sexes were assigned into four groups: control group, HI-120 min, HI-180 min and HI-210 min. Neurodevelopmental reflexes, exploratory abilities and cognitive function were assessed. At adulthood, tissue damage and reactive astrogliosis were measured. Animals exposed to HI-180 and HI-210 min had delayed neurodevelopmental reflexes compared to control group. Histological assessment showed tissue loss that was restricted to the ipsilateral hemisphere in lower periods of hypoxia exposure (120 and 180 min) but affected both hemispheres when 210 min was used. Reactive astrogliosis was increased only after 210 min of hypoxia. Interestingly, cognitive deficits were induced regardless the duration of hypoxia and there were correlations between behavioral parameters and cortex, hippocampus and corpus callosum volumes. These results show the duration of hypoxia has a close relationship with astrocytic response and tissue damage progression. Furthermore, the long-lasting cognitive memory deficit and its association with brain structures beyond the hippocampus suggests that complex anatomical changes should be involved in functional alterations taking place as hypoxia duration is increased, even when the cognitive impairment limit is achieved.
- Published
- 2019
- Full Text
- View/download PDF
50. The neuroprotective role of melatonin in a gestational hypermethioninemia model.
- Author
-
Figueiró PW, Moreira DS, Dos Santos TM, Prezzi CA, Rohden F, Faccioni-Heuser MC, Manfredini V, Netto CA, and Wyse ATS
- Subjects
- Animals, Brain drug effects, Brain metabolism, Disease Models, Animal, Female, Male, Maze Learning drug effects, Melatonin pharmacology, Neurons drug effects, Neurons metabolism, Neuroprotective Agents pharmacology, Oxidative Stress drug effects, Pregnancy, Rats, Rats, Wistar, Recognition, Psychology drug effects, Amino Acid Metabolism, Inborn Errors drug therapy, Glycine N-Methyltransferase deficiency, Melatonin therapeutic use, Neuroprotective Agents therapeutic use, Pregnancy Complications drug therapy
- Abstract
Elevated levels of methionine in blood characterize the hypermethioninemia, which may have genetic or non-genetic origin, as for example from high protein diet. Born rats from hypermethioninemic mothers presented cerebral oxidative stress, inhibition of Na
+ ,K+ -ATPase, memory deficit and ultrastructure cerebral changes. Melatonin is a hormone involved in circadian rhythm and has antioxidant effects. The aim of this study was to verify the possible neuroprotective effects of melatonin administration in hypermethioninemic pregnant rats on damage to biomolecules (Na+ ,K+ -ATPase, sulfhydryl content and DNA damage index) and behavior (open field, novel object recognition and water maze tasks), as well as its effect on cells morphology by electron microscopy in offspring. Wistar female rats received methionine (2.68 μmol/g body weight) and/or melatonin (10 mg/kg body weight) by subcutaneous injections during entire pregnancy. Control rats received saline. Biochemical analyzes were performed at 21 and 30 days of life of offspring and behavioral analyzes were performed only at 30 days of age in male pups. Results showed that gestational hypermethioninemia diminished Na+ ,K+ -ATPase activity and sulfhydryl content and increased DNA damage at 21 and 30 days of life. Melatonin was able to totally prevent Na+ ,K+ -ATPase activity alteration at 21 days and partially prevent its alteration at 30 days of rats life. Melatonin was unable in to prevent sulfhydryl and DNA damage at two ages. It also improved DNA damage, but not at level of saline animals (controls). Regarding to behavioral tests, data showed that pups exposed to gestational hypermethioninemia decreased reference memory in water maze, spent more time to the center of the open field and did not differentiate the objects in the recognition test. Melatonin was able to prevent the deficit in novel object recognition task. Electron microscopy revealed ultrastructure alterations in neurons of hypermethioninemic at both ages of offspring, whose were prevented by melatonin. These findings suggest that melatonin may be a good neuroprotective to minimize the harmful effects of gestational hypermethioninemia on offspring., (Copyright © 2019. Published by Elsevier Ltd.)- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.