Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 24(19)2023 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-37833970

RESUMO

Kynurenic acid (KYNA), an endogenous neuroprotectant with antiexcitotoxic, antioxidant, and anti-inflammatory effects, is synthesized through the tryptophan-kynurenine (KYN) pathway. We investigated whether brain KYN or KYNA levels were affected by asphyxia in a translational piglet model of hypoxic-ischemic encephalopathy (HIE). We also studied brain levels of the putative blood-brain barrier (BBB) permeable neuroprotective KYNA analogue SZR72, and whether SZR72 or therapeutic hypothermia (TH) modified KYN or KYNA levels. KYN, KYNA, and SZR72 levels were determined using ultra-high-performance liquid chromatography coupled with tandem mass spectrometry in five brain regions 24 h after 20 min of asphyxia in vehicle-, SZR72- and TH-treated newborn piglets (n = 6-6-6) and naive controls (n = 4). Endogenous brain KYN levels (median range 311.2-965.6 pmol/g) exceeded KYNA concentrations (4.5-6.0 pmol/g) ~100-fold. Asphyxia significantly increased cerebral KYN and KYNA levels in all regions (1512.0-3273.9 and 16.9-21.2 pmol/g, respectively), increasing the KYN/Tryptophan-, but retaining the KYNA/KYN ratio. SZR72 treatment resulted in very high cerebral SZR72 levels (13.2-33.2 nmol/g); however, KYN and KYNA levels remained similar to those of the vehicle-treated animals. However, TH virtually ameliorated asphyxia-induced elevations in brain KYN and KYNA levels. The present study reports for the first time that the KYN pathway is altered during HIE development in the piglet. SZR72 readily crosses the BBB in piglets but fails to affect cerebral KYNA levels. Beneficial effects of TH may include restoration of the tryptophan metabolism to pre-asphyxia levels.


Assuntos
Hipotermia , Hipóxia-Isquemia Encefálica , Suínos , Animais , Cinurenina/metabolismo , Triptofano/metabolismo , Ácido Cinurênico/metabolismo , Asfixia , Hipóxia-Isquemia Encefálica/terapia
2.
Int J Mol Sci ; 22(9)2021 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-34062911

RESUMO

Hypoxic-ischemic encephalopathy (HIE) remains to be a major cause of long-term neurodevelopmental deficits in term neonates. Hypothermia offers partial neuroprotection warranting research for additional therapies. Kynurenic acid (KYNA), an endogenous product of tryptophan metabolism, was previously shown to be beneficial in rat HIE models. We sought to determine if the KYNA analog SZR72 would afford neuroprotection in piglets. After severe asphyxia (pHa = 6.83 ± 0.02, ΔBE = -17.6 ± 1.2 mmol/L, mean ± SEM), anesthetized piglets were assigned to vehicle-treated (VEH), SZR72-treated (SZR72), or hypothermia-treated (HT) groups (n = 6, 6, 6; Tcore = 38.5, 38.5, 33.5 °C, respectively). Compared to VEH, serum KYNA levels were elevated, recovery of EEG was faster, and EEG power spectral density values were higher at 24 h in the SZR72 group. However, instantaneous entropy indicating EEG signal complexity, depression of the visual evoked potential (VEP), and the significant neuronal damage observed in the neocortex, the putamen, and the CA1 hippocampal field were similar in these groups. In the caudate nucleus and the CA3 hippocampal field, neuronal damage was even more severe in the SZR72 group. The HT group showed the best preservation of EEG complexity, VEP, and neuronal integrity in all examined brain regions. In summary, SZR72 appears to enhance neuronal activity after asphyxia but does not ameliorate early neuronal damage in this HIE model.


Assuntos
Asfixia Neonatal/tratamento farmacológico , Isquemia Encefálica/tratamento farmacológico , Ácido Cinurênico/análogos & derivados , Neurônios/metabolismo , Animais , Asfixia Neonatal/metabolismo , Asfixia Neonatal/patologia , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Região CA1 Hipocampal/diagnóstico por imagem , Região CA1 Hipocampal/efeitos dos fármacos , Região CA3 Hipocampal/diagnóstico por imagem , Região CA3 Hipocampal/efeitos dos fármacos , Modelos Animais de Doenças , Eletroencefalografia , Potenciais Evocados Visuais/efeitos dos fármacos , Humanos , Ácido Cinurênico/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Ratos , Pesquisa Translacional Biomédica
3.
Int J Mol Sci ; 21(18)2020 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-32948011

RESUMO

Hypoxic-ischemic encephalopathy (HIE) is still a major cause of neonatal death and disability as therapeutic hypothermia (TH) alone cannot afford sufficient neuroprotection. The present study investigated whether ventilation with molecular hydrogen (2.1% H2) or graded restoration of normocapnia with CO2 for 4 h after asphyxia would augment the neuroprotective effect of TH in a subacute (48 h) HIE piglet model. Piglets were randomized to untreated naïve, control-normothermia, asphyxia-normothermia (20-min 4%O2-20%CO2 ventilation; Tcore = 38.5 °C), asphyxia-hypothermia (A-HT, Tcore = 33.5 °C, 2-36 h post-asphyxia), A-HT + H2, or A-HT + CO2 treatment groups. Asphyxia elicited severe hypoxia (pO2 = 19 ± 5 mmHg) and mixed acidosis (pH = 6.79 ± 0.10). HIE development was confirmed by altered cerebral electrical activity and neuropathology. TH was significantly neuroprotective in the caudate nucleus but demonstrated virtually no such effect in the hippocampus. The mRNA levels of apoptosis-inducing factor and caspase-3 showed a ~10-fold increase in the A-HT group compared to naïve animals in the hippocampus but not in the caudate nucleus coinciding with the region-specific neuroprotective effect of TH. H2 or CO2 did not augment TH-induced neuroprotection in any brain areas; rather, CO2 even abolished the neuroprotective effect of TH in the caudate nucleus. In conclusion, the present findings do not support the use of these medical gases to supplement TH in HIE management.


Assuntos
Asfixia Neonatal/terapia , Dano Encefálico Crônico/prevenção & controle , Dióxido de Carbono/uso terapêutico , Hidrogênio/uso terapêutico , Hipotermia Induzida , Hipóxia-Isquemia Encefálica/terapia , Neuroproteção/efeitos dos fármacos , Fármacos Neuroprotetores/uso terapêutico , Acidose/sangue , Acidose/etiologia , Acidose/prevenção & controle , Administração por Inalação , Animais , Animais Recém-Nascidos , Fator de Indução de Apoptose/biossíntese , Fator de Indução de Apoptose/genética , Asfixia Neonatal/complicações , Asfixia Neonatal/tratamento farmacológico , Dano Encefálico Crônico/etiologia , Fator Neurotrófico Derivado do Encéfalo/biossíntese , Fator Neurotrófico Derivado do Encéfalo/genética , Dióxido de Carbono/administração & dosagem , Dióxido de Carbono/toxicidade , Caspase 3/biossíntese , Caspase 3/genética , Núcleo Caudado/patologia , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Eletroencefalografia , Potenciais Evocados Visuais/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Hipocampo/patologia , Hidrogênio/administração & dosagem , Hidrogênio/análise , Hipóxia-Isquemia Encefálica/complicações , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Hipóxia-Isquemia Encefálica/patologia , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Fármacos Neuroprotetores/administração & dosagem , Especificidade de Órgãos , Distribuição Aleatória , Suínos
4.
Acta Pharmacol Sin ; 39(8): 1273-1283, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29565041

RESUMO

Cyclooxygenase-2 (COX-2) has an established role in the pathogenesis of hypoxic-ischemic encephalopathy (HIE). In this study we sought to determine whether COX-2 was induced by asphyxia in newborn pigs, and whether neuronal COX-2 levels were affected by H2 treatment. Piglets were subjected to either 8 min of asphyxia or a more severe 20 min of asphyxia followed by H2 treatment (inhaling room air containing 2.1% H2 for 4 h). COX-2 immunohistochemistry was performed on brain samples from surviving piglets 24 h after asphyxia. The percentages of COX-2-immunopositive neurons were determined in cortical and subcortical areas. Only in piglets with more severe HIE, we observed significant, region-specific increases in neuronal COX-2 expression within the parietal and occipital cortices and in the CA3 hippocampal subfield. H2 treatment essentially prevented the increases in COX-2-immunopositive neurons. In the parietal cortex, the attenuation of COX-2 induction was associated with reduced 8'-hydroxy-2'-deoxyguanozine immunoreactivity and retained microglial ramifcation index, which are markers of oxidative stress and neuroinfiammation, respectively. This study demonstrates for the first time that asphyxia elevates neuronal COX-2 expression in a piglet HIE model. Neuronal COX-2 induction may play region-specific roles in brain lesion progression during HIE development, and inhibition of this response may contribute to the antioxidant/anti-infiammatory neuroprotective effects of H2 treatment.


Assuntos
Asfixia/prevenção & controle , Ciclo-Oxigenase 2/metabolismo , Hidrogênio/uso terapêutico , Hipóxia-Isquemia Encefálica/prevenção & controle , Fármacos Neuroprotetores/uso terapêutico , Animais , Animais Recém-Nascidos , Hipocampo/fisiopatologia , Masculino , Microglia/metabolismo , Neurônios/metabolismo , Lobo Parietal/fisiopatologia , Suínos
5.
Arterioscler Thromb Vasc Biol ; 33(4): 752-9, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23329133

RESUMO

OBJECTIVE: Mitochondrial depolarization after ATP-sensitive potassium channel activation has been shown to induce cerebral vasodilation by the generation of calcium sparks in smooth muscle. It is unclear, however, whether mitochondrial depolarization in endothelial cells is capable of promoting vasodilation by releasing vasoactive factors. Therefore, we studied the effect of endothelial mitochondrial depolarization by mitochondrial ATP-sensitive potassium channel activators, BMS-191095 (BMS) and diazoxide, on endothelium-dependent vasodilation. APPROACH AND RESULTS: Diameter studies in isolated rat cerebral arteries showed BMS- and diazoxide-induced vasodilations that were diminished by endothelial denudation. Mitochondrial depolarization-induced vasodilation was reduced by inhibition of mitochondrial ATP-sensitive potassium channels, phosphoinositide-3 kinase, or nitric oxide synthase. Scavenging of reactive oxygen species, however, diminished vasodilation induced by diazoxide, but not by BMS. Fluorescence studies in cultured rat brain microvascular endothelial cells showed that BMS elicited mitochondrial depolarization and enhanced nitric oxide production; diazoxide exhibited largely similar effects, but unlike BMS, increased mitochondrial reactive oxygen species production. Measurements of intracellular calcium ([Ca(2+)]i) in cultured rat brain microvascular endothelial cells and arteries showed that both diazoxide and BMS increased endothelial [Ca(2+)]i. Western blot analyses revealed increased phosphorylation of protein kinase B and endothelial nitric oxide synthase (eNOS) by BMS and diazoxide. Increased phosphorylation of eNOS by diazoxide was abolished by phosphoinositide-3 kinase inhibition. Electron spin resonance spectroscopy confirmed vascular nitric oxide generation in response to diazoxide and BMS. CONCLUSIONS: Pharmacological depolarization of endothelial mitochondria promotes activation of eNOS by dual pathways involving increased [Ca(2+)]i as well as by phosphoinositide-3 kinase-protein kinase B-induced eNOS phosphorylation. Both mitochondrial reactive oxygen species-dependent and -independent mechanisms mediate activation of eNOS by endothelial mitochondrial depolarization.


Assuntos
Artérias Cerebrais/metabolismo , Circulação Cerebrovascular , Células Endoteliais/metabolismo , Potencial da Membrana Mitocondrial , Mitocôndrias/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Canais de Potássio/metabolismo , Vasodilatação , Animais , Benzopiranos/farmacologia , Western Blotting , Cálcio/metabolismo , Células Cultivadas , Artérias Cerebrais/efeitos dos fármacos , Circulação Cerebrovascular/efeitos dos fármacos , Diazóxido/farmacologia , Relação Dose-Resposta a Droga , Espectroscopia de Ressonância de Spin Eletrônica , Células Endoteliais/efeitos dos fármacos , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Sequestradores de Radicais Livres/farmacologia , Imidazóis/farmacologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/antagonistas & inibidores , Fosfatidilinositol 3-Quinase/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/agonistas , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia
6.
Heliyon ; 10(7): e28607, 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38571587

RESUMO

Brain interstitial pH (pHbrain) alterations play a crucial role in the development of hypoxic-ischemic (HI) encephalopathy (HIE) caused by asphyxia in neonates. The newborn pig is one of the most suitable large animal models for studying HIE, however, compared to rats, experimental data on pHbrain alterations during HIE induction are limited. The major objective of the present study was thus to compare pHbrain changes during HIE development induced by experimental normocapnic hypoxia (H) or asphyxia (A), elicited with ventilation of a gas mixture containing 6%O2 or 6%O2/20%CO2, respectively for 20 min, under either normothermia (NT) or hypothermia (HT) (38.5 ± 0.5 °C or 33.5 ± 0.5 °C core temperature, respectively) in anesthetized piglets yielding four groups: H-NT, A-NT, H-HT, and A-HT. pHbrain changes during HI stress and the 60 min reoxygenation period were measured using a pH-selective microelectrode inserted into the parietal cortex through an open cranial window. In all groups, the pHbrain response to HI stress was acidosis, at the nadir pHbrain values dropped from the baseline of 7.27 ± 0.02 to H-NT:5.93 ± 0.30, A-NT:5.90 ± 0.52, H-HT:6.81 ± 0.27, and A-HT:6.27 ± 0.24 indicating that (1) H and A elicited similar, severe brain acidosis under NT greatly exceeding pH changes in arterial blood (pHa dropped to 7.24 ± 0.07 and 6.78 ± 0.03 from 7.52 ± 0.06 and 7.50 ± 0.05, respectively), and (2) HT ameliorated more the brain acidosis induced by H than by A. In all four groups, pHbrain was restored to baseline values without an alkalotic overshoot during the observed reoxygenation, Our findings suggest that under NT either H or A - both commonly employed HI stresses to elicit HIE in piglet models - would result in a similar acidotic pHbrain response without an alkalotic component either during the HI stress or the early reoxygenation period.

7.
Cells ; 13(8)2024 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-38667268

RESUMO

Subarachnoid hemorrhage (SAH) remains a major cause of cerebrovascular morbidity, eliciting severe headaches and vasospasms that have been shown to inversely correlate with vasodilator calcitonin gene-related peptide (CGRP) levels. Although dura mater trigeminal afferents are an important source of intracranial CGRP, little is known about the effects of SAH on these neurons in preclinical models. The present study evaluated changes in CGRP levels and expression in trigeminal primary afferents innervating the dura mater 72 h after experimentally induced SAH in adult rats. SAH, eliciting marked damage revealed by neurological examination, significantly reduced the density of CGRP-immunoreactive nerve fibers both in the dura mater and the trigeminal caudal nucleus in the medulla but did not affect the total dural nerve fiber density. SAH attenuated ex vivo dural CGRP release by ~40% and in the trigeminal ganglion, reduced both CGRP mRNA levels and the number of highly CGRP-immunoreactive cell bodies. In summary, we provide novel complementary evidence that SAH negatively affects the integrity of the CGRP-expressing rat trigeminal neurons. Reduced CGRP levels suggest likely impaired meningeal neurovascular functions contributing to SAH complications. Further studies are to be performed to reveal the importance of impaired CGRP synthesis and its consequences in central sensory processing.


Assuntos
Peptídeo Relacionado com Gene de Calcitonina , Dura-Máter , Neurônios , Ratos Sprague-Dawley , Hemorragia Subaracnóidea , Gânglio Trigeminal , Animais , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Dura-Máter/metabolismo , Masculino , Ratos , Hemorragia Subaracnóidea/metabolismo , Hemorragia Subaracnóidea/patologia , Neurônios/metabolismo , Gânglio Trigeminal/metabolismo , RNA Mensageiro/metabolismo , RNA Mensageiro/genética , Nervo Trigêmeo/metabolismo
8.
Sci Rep ; 13(1): 10089, 2023 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-37344545

RESUMO

The present study demonstrates the importance of converting signal intensity maps of organic tissues collected by laser-induced breakdown spectroscopy (LIBS) to elemental concentration maps and also proposes a methodology based on machine learning for its execution. The proposed methodology employs matrix-matched external calibration supported by a pixel-by-pixel automatic matrix (tissue type) recognition performed by linear discriminant analysis of the spatially resolved LIBS hyperspectral data set. On a swine (porcine) brain sample, we successfully performed this matrix recognition with an accuracy of 98% for the grey and white matter and we converted a LIBS intensity map of a tissue sample to a correct concentration map for the elements Na, K and Mg. Found concentrations in the grey and white matter agreed the element concentrations published in the literature and our reference measurements. Our results revealed that the actual concentration distribution in tissues can be quite different from what is suggested by the LIBS signal intensity map, therefore this conversion is always suggested to be performed if an accurate concentration distribution is to be assessed.


Assuntos
Encéfalo , Lasers , Animais , Suínos , Análise Espectral/métodos
9.
Microcirculation ; 19(8): 749-56, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22845548

RESUMO

OBJECTIVE: Previously, we have shown that IR impairs the vascular reactivity of the major cerebral arteries of ZO rats prior to the occurrence of Type-II diabetes mellitus. However, the functional state of the microcirculation in the cerebral cortex is still being explored. METHODS: We tested the local CoBF responses of 11-13-week-old ZO (n = 31) and control ZL (n = 32) rats to several stimuli measured by LDF using a closed cranial window setup. RESULTS: The topical application of 1-100 µm bradykinin elicited the same degree of CoBF elevation in both ZL and ZO groups. There was no significant difference in the incidence, latency, and amplitude of the NMDA-induced CSD-related hyperemia between the ZO and ZL groups. Hypercapnic CoBF response to 5% carbon-dioxide ventilation did not significantly change in the ZO compared with the ZL. Topical bicuculline-induced cortical seizure was accompanied by the same increase of CoBF in both the ZO and ZL at all bicuculline doses. CONCLUSIONS: CoBF responses of the microcirculation are preserved in the early period of the metabolic syndrome, which creates an opportunity for intervention to prevent and restore the function of the major cerebral vascular beds.


Assuntos
Bicuculina/farmacologia , Circulação Cerebrovascular/efeitos dos fármacos , Convulsivantes/farmacologia , Diabetes Mellitus Tipo 2/fisiopatologia , Resistência à Insulina , Microcirculação/efeitos dos fármacos , Animais , Bicuculina/efeitos adversos , Convulsivantes/efeitos adversos , Diabetes Mellitus Tipo 2/metabolismo , Hipercapnia/metabolismo , Hipercapnia/fisiopatologia , Masculino , Ratos , Convulsões/induzido quimicamente , Convulsões/metabolismo , Convulsões/fisiopatologia
10.
Microvasc Res ; 83(3): 311-7, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22306444

RESUMO

A new laser speckle-contrast analysis (LASCA) technique based on multi-exposure imaging was employed to simultaneously study pial arteriolar responses with cerebrocortical perfusion changes to various vasodilator (5-10% CO(2) ventilation, bradykinin (1-10 µM), N-methyl-D-aspartate (100 µM)) vasoconstrictor (10-100 µM noradrenaline, 1M KCl), or neutral (2.1% H(2) ventilation) stimuli as well as to asphyxia in the newborn piglet. Anesthetized, ventilated animals (n=20) were fitted with closed cranial windows. Multiple exposure laser-speckle image series (1-100 ms) were obtained using a near infrared diode laser (λ=808 nm). The autocorrelation decay time (τ) of speckle fluctuations was determined over pial arterioles and parenchymal areas to express 1/τ being proportional to blood flow velocity by two different LASCA techniques: our novel multi-exposure or a single exposure (2 and 20 ms) approach. 1/τ values yielded by different LASCA techniques were not significantly different at most points. LASCA easily detected both increases and decreases in cortical blood flow (CoBF). Cortical 1/τ changes to hypercapnia closely matched quantitative CoBF data determined previously, and were also in accordance with increases of pial arteriolar blood flow, calculated from arteriolar flow velocity and cross sectional area changes. In summary, LASCA emerges as an appealing method to simultaneously study microvascular reactivity and cortical perfusion changes in the piglet.


Assuntos
Processamento de Imagem Assistida por Computador/métodos , Microcirculação , Algoritmos , Animais , Animais Recém-Nascidos , Arteríolas/fisiologia , Velocidade do Fluxo Sanguíneo/fisiologia , Bradicinina/farmacologia , Circulação Cerebrovascular , Meios de Contraste/farmacologia , Relação Dose-Resposta a Droga , Lasers , Modelos Estatísticos , N-Metilaspartato/farmacologia , Pia-Máter/irrigação sanguínea , Suínos , Fatores de Tempo , Vasoconstritores/farmacologia , Vasodilatadores/farmacologia
11.
Acta Histochem Cytochem ; 45(3): 187-92, 2012 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-22829712

RESUMO

Cyclooxygenase (COX)-2 is the major constitutively expressed COX isoform in the newborn brain. COX-2 derived prostanoids and reactive oxygen species appear to play a major role in the mechanism of perinatal hypoxic-ischemic injury in the newborn piglet, an accepted animal model of the human term neonate. The study aimed to quantitatively determine COX-2 immunopositive neurons in different brain regions in piglets under normoxic conditions (n=15), and 4 hours after 10 min asphyxia (n=11). Asphyxia did not induce significant changes in neuronal COX-2 expression of any studied brain areas. In contrast, there was a marked regional difference in all experimental groups. Thus, significant difference was observed between fronto-parietal and temporo-occipital regions: 59±4% and 67±3% versus 41±2%* and 31±3%* respectively (mean±SEM, data are pooled from all subjects, n=26, *p<0.05, vs. fronto-parietal region). In the hippocampus, COX-2 immunopositivity was rare (highest expression in CA1 region: 14±2%). The studied subcortical areas showed negligible COX-2 staining. Our findings suggest that asphyxia does not significantly alter the pattern of neuronal COX-2 expression in the early reventilation period. Furthermore, based on the striking differences observed in cortical neuronal COX-2 distribution, the contribution of COX-2 mediated neuronal injury after asphyxia may also show region-specific differences.

12.
Am J Physiol Heart Circ Physiol ; 300(6): H2080-7, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21421821

RESUMO

Insulin resistance (IR) impairs cerebrovascular responses to several stimuli in Zucker obese (ZO) rats. However, cerebral artery responses after subarachnoid hemorrhage (SAH) have not been described in IR. We hypothesized that IR worsens vascular reactions after a mild SAH. Hemolyzed blood (300 µl) or saline was infused (10 µl/min) into the cisterna magna of 11-13-wk-old ZO (n = 25) and Zucker lean (ZL) rats (n = 25). One day later, dilator responses of the basilar artery (BA) and its side branch (BA-Br) to acetylcholine (ACh, 10(-6) M), cromakalim (10(-7) M, 10(-6) M), and sodium nitroprusside (10(-7) M) were recorded with intravital videomicroscopy. The baseline diameter of the BA was increased both in the ZO and ZL rats 24 h after the hemolysate injection. Saline-injected ZO animals showed reduced dilation to ACh (BA = 9 ± 3 vs. 22 ± 4%; and BA-Br = 23 ± 5 vs. 37 ± 7%) compared with ZL rats. Hemolysate injection blunted the response to ACh in both the ZO (BA = 4 ± 2%; and BA-Br = 12 ± 3%) and ZL (BA = 7 ± 2%; and BA-Br = 11 ± 3%) rats. Cromakalim (10(-6) M)-induced dilation was significantly reduced in the hemolysate-injected ZO animals compared with the saline control (BA = 13 ± 3 vs. 26 ± 5%; and BA-Br = 28 ± 8 vs. 44 ± 9%) and in the hemolysate-injected ZL rats compared with their saline control (BA = 24 ± 4 vs. 32 ± 4%; but not BA-Br = 39 ± 6 vs. 59 ± 9%). No significant difference in sodium nitroprusside reactivity was observed. Western blot analysis of the BA showed a lower baseline level of neuronal nitric oxide synthase expression and an enhanced cyclooxygenase-2 level in the hemolysate-injected ZO animals. In summary, cerebrovascular reactivity to both endothelium-dependent and -independent stimuli is severely compromised by SAH in IR animals.


Assuntos
Circulação Cerebrovascular/fisiologia , Resistência à Insulina/fisiologia , Obesidade/fisiopatologia , Hemorragia Subaracnóidea/fisiopatologia , Vasoespasmo Intracraniano/fisiopatologia , Acetilcolina/farmacologia , Animais , Circulação Cerebrovascular/efeitos dos fármacos , Cromakalim/farmacologia , Ciclo-Oxigenase 2/metabolismo , Modelos Animais de Doenças , Nitroprussiato/farmacologia , Obesidade/metabolismo , Ratos , Ratos Zucker , Vasodilatação/efeitos dos fármacos , Vasodilatação/fisiologia , Vasodilatadores/farmacologia
13.
Curr Pharm Des ; 27(5): 687-694, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33185158

RESUMO

Hypoxic-ischemic encephalopathy (HIE) remains to be a major cause of morbidity, mortality and severe neurodevelopmental disability in term neonates. Moderate whole body hypothermia is an established, effective neuroprotective therapy to reduce mortality and long-term disability associated with HIE, however, research for adjunct therapies is still warranted to complement the effect of hypothermia. In the last decade, molecular hydrogen emerged as a simple, available, inexpensive substance with advantageous pharmacokinetics to ameliorate hypoxic-ischemic cellular damage. The present review examines the preclinical studies employing hydrogen to combat the deleterious consequences of hypoxic-ischemic insults in rodent and piglet HIE models. Hydrogen exerted unequivocal neuroprotective actions shown by preserved neurovascular function, neuronal viability, and neurocognitive functions in virtually all model species and hypoxic-ischemic insult types tested. Administration of hydrogen started in most studies after the hypoxic-ischemic insult enhancing the translational value of the findings. Among the explored mechanisms of hydrogen-induced neuroprotection, antioxidant, anti- apoptotic and anti-inflammatory effects appeared to be dominant. Unfortunately, the additive neuroprotective effect of hydrogen and therapeutic hypothermia has not yet been demonstrated, thus such studies are warranted to promote the clinical testing of molecular hydrogen as an adjunct neuroprotective treatment of HIE.


Assuntos
Hipotermia Induzida , Hipóxia-Isquemia Encefálica , Fármacos Neuroprotetores , Animais , Humanos , Hidrogênio , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Recém-Nascido , Neuroproteção , Fármacos Neuroprotetores/farmacologia , Suínos
14.
Pediatr Res ; 68(5): 387-92, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20657346

RESUMO

Hydrogen (H2) has been reported to neutralize toxic reactive oxygen species. Oxidative stress is an important mechanism of neuronal damage after perinatal asphyxia. We examined whether 2.1% H2-supplemented room air (H2-RA) ventilation would preserve cerebrovascular reactivity (CR) and brain morphology after asphyxia/reventilation (A/R) in newborn pigs. Anesthetized, ventilated piglets were assigned to one of the following groups: A/R with RA or H2-RA ventilation (A/R-RA and A/R-H2-RA; n = 8 and 7, respectively) and respective time control groups (n = 9 and 7). Asphyxia was induced by suspending ventilation for 10 min, followed by reventilation with the respective gases for 4 h. After euthanasia, the brains were processed for neuropathological examination. Pial arteriolar diameter changes to graded hypercapnia (5-10% CO2 inhalation), and NMDA (10(-4) M) were determined using the closed cranial window/intravital microscopy before and 1 h after asphyxia. Neuropathology revealed that H2-RA ventilation significantly reduced neuronal injury induced by A/R in virtually all examined brain regions including the cerebral cortex, the hippocampus, basal ganglia, cerebellum, and the brainstem. Furthermore, H2-RA ventilation significantly increased CR to hypercapnia after A/R (% vasodilation was 23 ± 4% versus 41 ± 9%, p < 0.05). H2-RA ventilation did not affect reactive oxygen species-dependent CR to NMDA. In summary, H2-RA could be a promising approach to reduce the neurologic deficits after perinatal asphyxia.


Assuntos
Asfixia Neonatal/fisiopatologia , Encéfalo , Circulação Cerebrovascular/efeitos dos fármacos , Modelos Animais de Doenças , Hidrogênio/farmacologia , Fármacos Neuroprotetores/farmacologia , Animais , Animais Recém-Nascidos , Análise Química do Sangue , Encéfalo/irrigação sanguínea , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Hemodinâmica , Humanos , Hipercapnia/metabolismo , Recém-Nascido , Suínos
15.
Prog Neurobiol ; 86(4): 379-95, 2008 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-18835324

RESUMO

Cortical spreading depression (CSD) leads to dramatic changes in cerebral hemodynamics. However, mechanisms involved in promoting and counteracting cerebral vasodilator responses are unclear. Here we review the development and current status of this important field of research especially with respect to the role of perivascular nerves and nitric oxide (NO). It appears that neurotransmitters released from the sensory and the parasympathetic nerves associated with cerebral arteries, and NO released from perivascular nerves and/or parenchyma, promote cerebral hyperemia during CSD. However, the relative contributions of each of these factors vary according to species studied. Related to CSD, axonal and reflex responses involving trigeminal afferents on the pial surface lead to increased blood flow and inflammation of the overlying dura mater. Counteracting the cerebral vascular dilation is the production and release of constrictor prostaglandins, at least in some species, and other possibly yet unknown agents from the vascular wall. The cerebral blood flow response in healthy human cortex has not been determined, and thus it is unclear whether the cerebral oligemia associated with migraines represents the normal physiological response to a CSD-like event or represents a pathological response. In addition to promoting cerebral hyperemia, NO produced during CSD appears to initiate signaling events which lead to protection of the brain against subsequent ischemic insults. In summary, the cerebrovascular response to CSD involves multiple dilator and constrictor factors produced and released by diverse cells within the neurovascular unit, with the contribution of each of these factors varying according to the species examined.


Assuntos
Circulação Cerebrovascular/fisiologia , Depressão Alastrante da Atividade Elétrica Cortical/fisiologia , Vasodilatação/fisiologia , Animais
17.
PLoS One ; 15(5): e0233851, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32470084

RESUMO

Brain interstitial pH (pHbrain) alterations play an important role in the mechanisms of neuronal injury in neonatal hypoxic-ischemic encephalopathy (HIE) induced by perinatal asphyxia. The newborn pig is an established large animal model to study HIE, however, only limited information on pHbrain alterations is available in this species and it is restricted to experimental perinatal asphyxia (PA) and the immediate reventilation. Therefore, we sought to determine pHbrain over the first 24h of HIE development in piglets. Anaesthetized, ventilated newborn pigs (n = 16) were instrumented to control major physiological parameters. pHbrain was determined in the parietal cortex using a pH-selective microelectrode. PA was induced by ventilation with a gas mixture containing 6%O2-20%CO2 for 20 min, followed by reventilation with air for 24h, then the brains were processed for histopathology assessment. The core temperature was maintained unchanged during PA (38.4±0.1 vs 38.3±0.1°C, at baseline versus the end of PA, respectively; mean±SEM). In the arterial blood, PA resulted in severe hypoxia (PaO2: 65±4 vs 23±1*mmHg, *p<0.05) as well as acidosis (pHa: 7.53±0.03 vs 6.79±0.02*) that is consistent with the observed hypercapnia (PaCO2: 37±3 vs 160±6*mmHg) and lactacidemia (1.6±0.3 vs 10.3±0.7*mmol/L). Meanwhile, pHbrain decreased progressively from 7.21±0.03 to 5.94±0.11*. Reventilation restored pHa, blood gases and metabolites within 4 hours except for PaCO2 that remained slightly elevated. pHbrain returned to 7.0 in 29.4±5.5 min and then recovered to its baseline level without showing secondary alterations during the 24 h observation period. Neuropathological assessment also confirmed neuronal injury. In conclusion, in spite of the severe acidosis and alterations in blood gases during experimental PA, pHbrain recovered rapidly and notably, there was no post-asphyxia hypocapnia that is commonly observed in many HIE babies. Thus, the neuronal injury in our piglet model is not associated with abnormal pHbrain or low PaCO2 over the first 24 h after PA.


Assuntos
Encéfalo/metabolismo , Hipóxia-Isquemia Encefálica/metabolismo , Acidose/sangue , Acidose/complicações , Acidose/metabolismo , Acidose/fisiopatologia , Animais , Animais Recém-Nascidos , Asfixia Neonatal/sangue , Asfixia Neonatal/metabolismo , Asfixia Neonatal/fisiopatologia , Encéfalo/patologia , Encéfalo/fisiopatologia , Hemodinâmica , Concentração de Íons de Hidrogênio , Hipercapnia/sangue , Hipercapnia/complicações , Hipercapnia/metabolismo , Hipercapnia/fisiopatologia , Hipóxia-Isquemia Encefálica/sangue , Hipóxia-Isquemia Encefálica/complicações , Hipóxia-Isquemia Encefálica/fisiopatologia , Masculino , Neurônios/patologia , Oxigênio/metabolismo , Suínos
18.
Microcirculation ; 16(7): 629-39, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19657965

RESUMO

OBJECTIVE: N-methyl-d-aspartate (NMDA) is a powerful cerebrovascular dilator in vivo. Cortical spreading depression (CSD) has recently been shown to contribute to the pial arteriolar dilation in mice. Our main aim was to examine the participation of CSD in the overall cerebrovascular response to NMDA in the rat. METHODS: Anesthetized Wistar rats (eight weeks old) were equipped with a closed cranial window to allow topical application of NMDA (10(-5)-10(-3) M) to the parietal cortex. Cortical blood flow (CoBF) under and outside the cranial window was simultaneously monitored by using a two-channel laser-Doppler flowmeter. CSDs were detected by recording the changes in the cortical DC potential. RESULTS: Concentrations of 10(-4) and 10(-3) M of NMDA evoked single CSDs associated with rapid, transient hyperemia, followed by a sustained, but reduced, increase in CoBF. The latency and magnitude of the CoBF responses were dose dependent. The higher dose resulted in shorter latency (100+/-5* vs. 146+/-11 seconds, *P<0.05; mean+/-standard error of the mean) and larger overall flow response (77+/-12* vs. 28+/-3% from baseline) under, but not outside, the cranial window. CONCLUSIONS: NMDA elicits dose-dependent increases in CoBF that are composed of CSD-dependent and -independent components in rats.


Assuntos
Córtex Cerebral/irrigação sanguínea , Depressão Alastrante da Atividade Elétrica Cortical/fisiologia , Hiperemia/induzido quimicamente , N-Metilaspartato/farmacologia , Animais , Circulação Cerebrovascular , Relação Dose-Resposta a Droga , Ratos , Ratos Wistar , Fluxo Sanguíneo Regional
19.
Microvasc Res ; 78(2): 212-7, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19555699

RESUMO

Cerebral ischemia/reperfusion (I/R) results in cellular energy failure and dysfunction of the neurovascular unit that contribute to subsequent neuronal cell death in the neonate. PX-18 is a putative neuroprotective inhibitor of secretory phospholipase A(2) (sPLA(2)) but its in vivo testing has been limited by its poor solubility. Our purpose was to assess whether PX-18 preserved neuronal-vascular reactivity to I/R-sensitive endothelium-dependent (hypercapnia, bradykinin) and/or neuron-dependent (N-methyl-D-aspartate; NMDA) stimuli. To make the drug available for in vivo studies, PX-18 was formulated as a 3% nanosuspension applying high pressure homogenization. Newborn piglets (1-day old, n=40) were anesthetized and ventilated, and cerebrovascular reactivity to the above stimuli was determined by measuring changes in pial arteriolar diameters using the closed cranial window/intravital videomicroscopy technique. Intravenous infusion of PX-18 nanosuspension (6 mg/kg, 20 min) did not affect baseline arteriolar diameters, or hypercapnia-, bradykinin-, or NMDA-induced pial arteriolar vasodilation under normoxic conditions. Global cerebral ischemia (10 min) followed by 1 h of reperfusion significantly attenuated hypercapnia-, bradykinin-, and NMDA-induced vasodilation in untreated or vehicle-treated controls. However, PX-18 resulted in nearly full preservation of cerebrovascular reactivity to all these stimuli. In conclusion, inhibition of sPLA(2) by PX-18 improves neurovascular function both at the neuronal and the microvascular level following I/R. This effect of PX-18 likely contributes to its neuroprotective effect.


Assuntos
Ácidos Alcanossulfônicos/farmacologia , Isquemia Encefálica/fisiopatologia , Circulação Cerebrovascular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Ácidos Oleicos/farmacologia , Fosfolipases A2 Secretórias/antagonistas & inibidores , Animais , Animais Recém-Nascidos , Arteríolas/efeitos dos fármacos , Arteríolas/fisiologia , Bradicinina/farmacologia , Artérias Cerebrais/efeitos dos fármacos , Artérias Cerebrais/inervação , Química Farmacêutica/métodos , Avaliação Pré-Clínica de Medicamentos , Hipercapnia/fisiopatologia , Microscopia de Vídeo , N-Metilaspartato/farmacologia , Neurônios/efeitos dos fármacos , Tamanho da Partícula , Pia-Máter/irrigação sanguínea , Pia-Máter/efeitos dos fármacos , Traumatismo por Reperfusão/tratamento farmacológico , Suínos , Vasodilatação/efeitos dos fármacos , Vasodilatação/fisiologia , Vasodilatadores/farmacologia
20.
Sci Rep ; 9(1): 18900, 2019 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-31827200

RESUMO

Cortical spreading depolarization (SD) involves activation of NMDA receptors and elicit neurovascular unit dysfunction. NMDA cannot trigger SD in newborns, thus its effect on neurovascular function is not confounded by other aspects of SD. The present study investigated if NMDA affected hypercapnia-induced microvascular and electrophysiological responses in the cerebral cortex of newborn pigs. Anesthetized piglets were fitted with cranial windows over the parietal cortex to study hemodynamic and electrophysiological responses to graded hypercapnia before/after topically applied NMDA assessed with laser-speckle contrast imaging and recording of local field potentials (LFP)/neuronal firing, respectively. NMDA increased cortical blood flow (CoBF), suppressed LFP power in most frequency bands but evoked a 2.5 Hz δ oscillation. The CoBF response to hypercapnia was abolished after NMDA and the hypercapnia-induced biphasic changes in δ and θ LFP power were also altered. MK-801 prevented NMDA-induced increases in CoBF and the attenuation of microvascular reactivity to hypercapnia. The neuronal nitric oxide synthase (nNOS) inhibitor (N-(4 S)-4-amino-5-[aminoethyl]aminopentyl-N'-nitroguanidin) also significantly preserved the CoBF response to hypercapnia after NMDA, although it didn't reduce NMDA-induced increases in CoBF. In conclusion, excess activation of NMDA receptors alone can elicit SD-like neurovascular unit dysfunction involving nNOS activity.


Assuntos
Córtex Cerebral/efeitos dos fármacos , Circulação Cerebrovascular/efeitos dos fármacos , Hipercapnia/fisiopatologia , N-Metilaspartato/farmacologia , Neurônios/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Arteríolas/efeitos dos fármacos , Córtex Cerebral/fisiopatologia , Masculino , Suínos , Vasodilatação/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA