Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
1.
Exp Neurol ; 372: 114641, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-38065231

RESUMO

This study was to explore the mechanism of ferroptosis and hypoxic-ischemic brain damage in neonatal rats. The neonatal rat hypoxic-ischemic brain damage (HIBD) model was established using the Rice-Vannucci method and treated with the ferroptosis inhibitor liproxstatin-1. Cognitive assessment was performed through absentee field experiments to confirm the successful establishment of the model. Brain tissue damage was evaluated by comparing regional cerebral blood flow and quantifying tissue staining. Neuronal cell morphological changes in the rats' cortical and hippocampal regions were observed using HE and Nissl staining. ELISA was performed to determine GPX4, GSH and ROS expression levels in the rats' brain tissues, and Western blotting to assess the expression levels of 4-HNE, GPX4, GSS, ACSL4, SLC7A11, SLC3A2, TFRC, FHC, FLC, HIF-1α, and Nrf2 proteins in rat brain tissues. Compared to the Sham group, the HIBD group exhibited a significant decrease in cerebral blood perfusion, reduced brain nerve cells, and disordered cell arrangement. The use of the ferroptosis inhibitor effectively improved brain tissue damage and preserved the shape and structure of nerve cells. The oxidative stress products ROS and 4-HNE in the brain tissue of the HIBD group increased significantly, while the expression of antioxidant indicators GPX4, GSH, SLC7A11, and GSS decreased significantly. Furthermore, the expression of iron metabolism-related proteins TFRC, FHC, and FLC increased significantly, whereas the expression of the ferroptosis-related transcription factors HIF-1α and Nrf2 decreased significantly. Treatment with liproxstatin-1 exhibited therapeutic effects on HIBD and downregulated tissue ferroptosis levels. This study shows the involvement of ferroptosis in hypoxic-ischemic brain damage in neonatal rats through the System Xc--GSH-GPX4 functional axis and iron metabolism pathway, with the HIF-1α and Nrf2 transcription factors identified as the regulators of ferroptosis involved in the HIBD process in neonatal rats.


Assuntos
Ferroptose , Hipóxia-Isquemia Encefálica , Ratos , Animais , Animais Recém-Nascidos , Ratos Sprague-Dawley , Fator 2 Relacionado a NF-E2/metabolismo , Espécies Reativas de Oxigênio , Hipóxia-Isquemia Encefálica/metabolismo , Encéfalo/metabolismo , Ferro
2.
Brain Res ; 1822: 148640, 2024 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-37863169

RESUMO

Previous studies have showed that the permeability of blood brain barrier (BBB) increased after hypoxia ischemia (HI). The current research uncovered the mechanism of altered BBB permeability after hypoxic-ischemic brain damage (HIBD) through AKT/GSK-3ß/CREB signaling pathway in neonatal rats. Firstly, Magnetic resonance imaging (MRI) combined with hematoxylin-eosin (H&E) staining was used to assess brain injury. Initial findings showed abnormal signals in T2-weighted imaging (T2WI) and diffusion weighted imaging (DWI). Changes also happened in the morphology of nerve cells. Subsequently, we found that BBB damage is manifested as leakage of immunoglobulin G (IgG) and destruction of BBB-related proteins and ultrastructure. Meanwhile, the levels of matrix metalloproteinase-9 (MMP-9) significantly increased at 24 h after HIBD compared to a series of time points. Additionally, immunohistochemical (IHC) staining combined with Western blot (WB) was used to verify the function of the AKT/GSK-3ß/CREB signaling pathway in BBB damage after HI in neonatal rats. Results showed that less Claudin-5, ZO-1, p-AKT, p-GSK-3ß and p-CREB, along with more MMP-9 protein expression were visible on the damaged side of the cerebral cortex in the HIBD group in contrast to the sham and HIBD + SC79 groups. Together, our findings demonstrated that HI in neonatal rats might upregulate the levels of MMP-9 protein and downregulate the levels of Claudin-5 and ZO-1 by inhibiting the AKT/GSK-3ß/CREB pathway, thus disrupting the BBB, which in turn aggravates brain damage after HI in neonatal rats.


Assuntos
Barreira Hematoencefálica , Hipóxia-Isquemia Encefálica , Animais , Ratos , Animais Recém-Nascidos , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Claudina-5/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Hipóxia-Isquemia Encefálica/metabolismo , Isquemia/complicações , Isquemia/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos Sprague-Dawley , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo
3.
World J Stem Cells ; 15(6): 607-616, 2023 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-37424948

RESUMO

BACKGROUND: Timing of passaging, passage number, passaging approaches and methods for cell identification are critical factors influencing the quality of neural stem cells (NSCs) culture. How to effectively culture and identify NSCs is a continuous interest in NSCs study while these factors are comprehensively considered. AIM: To establish a simplified and efficient method for culture and identification of neonatal rat brain-derived NSCs. METHODS: First, curved tip operating scissors were used to dissect brain tissues from new born rats (2 to 3 d) and the brain tissues were cut into approximately 1 mm3 sections. Filter the single cell suspension through a nylon mesh (200-mesh) and culture the sections in suspensions. Passaging was conducted with TrypLTM Express combined with mechanical tapping and pipetting techniques. Second, identify the 5th generation of passaged NSCs as well as the revived NSCs from cryopreservation. BrdU incorporation method was used to detect self-renew and proliferation capabilities of cells. Different NSCs specific antibodies (anti-nestin, NF200, NSE and GFAP antibodies) were used to identify NSCs specific surface markers and muti-differentiation capabilities by immunofluorescence staining. RESULTS: Brain derived cells from newborn rats (2 to 3 d) proliferate and aggregate into spherical-shaped clusters with sustained continuous and stable passaging. When BrdU was incorporated into the 5th generation of passaged cells, positive BrdU cells and nestin cells were observed by immunofluorescence staining. After induction of dissociation using 5% fetal bovine serum, positive NF200, NSE and GFAP cells were observed by immunofluorescence staining. CONCLUSION: This is a simplified and efficient method for neonatal rat brain-derived neural stem cell culture and identification.

4.
Brain Res ; 1817: 148495, 2023 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-37481153

RESUMO

BACKGROUND: White matter injury (WMI) is an important type of preterm brain injury, which may result in severe neurological sequelae and lack of effective treatments. It is ascertained that selective vulnerability of oligodendrocytes is closely related to the WMI in preterm infants. But the alteration of the endogenous oligodendrogenesis over long time after hypoxic-ischemic WMI is still not clearly elucidated. METHODS: We adopted an animal model of hypoxic-ischemic WMI in 3-day-old neonatal Sprague-Dawley rats. Immunofluorescence staining and western blotting were used to detect dynamic changes of oligodendrogenesis in the white matter region on postoperative day (POD) 1, 3, 7, 14, 28, 56 and 84. RESULTS: In the sham group, the oligodendrocyte lineage in the white matter reached a developmental peak from POD 3 to 14. The proliferation and development of oligodendrocyte precursor cells (OPCs) occurred primarily within POD 14. The number of mature oligodendrocytes showed an upward trend and a dynamic change in proliferation over time. While in the WMI group, the oligodendrocyte lineage was upregulated on POD1 and 3 but downregulated on POD 7 and 14. The proliferation of OPCs increased on POD 1 and decreased on POD 3 and 7, with the total number of OPCs significantly reduced from POD 3 to 14. The number of mature oligodendrocytes decreased from POD 3 to 28, and return to the level of the sham group on POD 56 and 84, whereas the MBP expression was still significantly downregulated on POD 56 and 84. CONCLUSIONS: Hypoxia-ischemia can have a long-term dynamic effect on the endogenous oligodendrogenesis of neonatal rat brain white matter. The proliferation of OPCs was promoted on POD 1 but inhibited from POD 3 to 14, which may be an early intervention target to improve oligodendrogenesis. The number of mature oligodendrocytes recover to the normal on POD 56 and 84 but the myelination is still blocked, which suggests it is essential to promote the maturation of oligodendrocyte and its function recovery at the same time within POD 28. Such efforts will provide the opportunity to test new interventions in pre-clinical studies for their promising clinical application.


Assuntos
Lesões Encefálicas , Substância Branca , Recém-Nascido , Humanos , Animais , Ratos , Animais Recém-Nascidos , Ratos Sprague-Dawley , Substância Branca/metabolismo , Recém-Nascido Prematuro , Hipóxia/metabolismo , Oligodendroglia/metabolismo , Lesões Encefálicas/metabolismo , Isquemia/metabolismo
5.
Neuroscience ; 522: 121-131, 2023 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-37196978

RESUMO

Perinatal hypoxic-ischemic (HI) brain injury leads to mortality and morbidity in neonates and children. There are no effective and practical methods to attenuate this brain injury. This study determined whether desflurane, a volatile anesthetic with limited effect on the cardiovascular system, protected against HI-induced brain damage and the role of transient receptor potential ankyrin 1 (TRPA1), a mediator for simulated ischemia-induced myelin damage, in this protection. Seven-day-old male and female Sprague-Dawley rats had brain HI. They were exposed to 4.8%, 7.6% or 11.4% desflurane immediately or 4.8% desflurane at 0.5, 1 or 2 h after the HI. Brain tissue loss was evaluated 7 days later. Neurological functions and brain structures of rats with HI and 4.8% desflurane post-treatment were evaluated 4 weeks after the HI. TRPA1 expression was determined by Western blotting. HC-030031, a TRPA1 inhibitor, was used to determine the role of TRPA1 in the HI-induced brain injury. HI induced brain tissue and neuronal loss, which was attenuated by all tested concentrations of desflurane. Desflurane post-treatment also improved motor function, learning and memory in rats with brain HI. Brain HI increased the expression of TRPA1 and this increase was inhibited by desflurane. TRPA1 inhibition reduced HI-induced brain tissue loss and impairment of learning and memory. However, the combination of TRPA1 inhibition and desflurane post-treatment did not preserve brain tissues, learning and memory better than TRPA1 inhibition or desflurane post-treatment alone. Our results suggest that desflurane post-treatment induces neuroprotection against neonatal HI. This effect may be mediated by inhibiting TRPA1.


Assuntos
Lesões Encefálicas , Hipóxia-Isquemia Encefálica , Fármacos Neuroprotetores , Gravidez , Ratos , Animais , Masculino , Feminino , Animais Recém-Nascidos , Ratos Sprague-Dawley , Anquirinas/metabolismo , Desflurano , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/metabolismo , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Hipóxia-Isquemia Encefálica/metabolismo , Encéfalo/metabolismo , Lesões Encefálicas/metabolismo , Proteínas do Citoesqueleto/metabolismo
6.
BMC Neurosci ; 24(1): 27, 2023 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-37098490

RESUMO

BACKGROUND: Neonatal hyperoxic brain injury is caused by exposure to hyperphysiological oxygen content during the period of incomplete development of the oxidative stress defence system, resulting in a large number of reactive oxygen species (ROS) and causing damage to brain tissue. Mitochondrial biogenesis refers to the synthesis of new mitochondria from existing mitochondria, mostly through the PGC-1α/Nrfs/TFAM signalling pathway. Resveratrol (Res), a silencing information regulator 2-related enzyme 1 (Sirt1) agonist, has been shown to upregulate the level of Sirt1 and the expression of peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α). We speculate that Res has a protective effect on hyperoxia-induced brain injury through mitochondrial biogenesis. METHODS: Sprague-Dawley (SD) pups were randomly divided into the nonhyperoxia (NN) group, the nonhyperoxia with dimethyl sulfoxide (ND) group, the nonhyperoxia with Res (NR) group, the hyperoxia (HN) group, the hyperoxia with dimethyl sulfoxide (HD) group, and the hyperoxia with Res (HR) group within 12 h after birth. The HN, HD, and HR groups were placed in a high-oxygen environment (80‒85%), and the other three groups were placed in the standard atmosphere. The NR and HR groups were given 60 mg/kg Res every day, the ND and HD groups were given the same dose of dimethyl sulfoxide (DMSO) every day, and the NN and HN groups were given the same dose of normal saline every day. On postnatal day (PN) 1, PN7, and PN14, brain samples were acquired for HE staining to assess pathology, TUNEL to detect apoptosis, and real-time quantitative polymerase chain reaction and immunoblotting to detect the expression levels of Sirt1, PGC-1α, nuclear respiratory factor 1 (Nrf1), nuclear respiratory factor 2 (Nrf2) and mitochondrial transcription factor A (TFAM) in brain tissue. RESULTS: Hyperoxia induced brain tissue injury; increased brain tissue apoptosis; inhibited Sirt1, PGC-1α, Nrf1, Nrf2, TFAM mRNA expression in mitochondria; diminished the ND1 copy number and ND4/ND1 ratio; and decreased Sirt1, PGC-1α, Nrf1, Nrf2, and TFAM protein levels in the brain. In contrast, Res reduced brain injury and attenuated brain tissue apoptosis in neonatal pups and increased the levels of the corresponding indices. CONCLUSION: Res has a protective effect on hyperoxia-induced brain injury in neonatal SD pups by upregulating Sirt1 and stimulating the PGC-1α/Nrfs/TFAM signalling pathway for mitochondrial biogenesis.


Assuntos
Lesões Encefálicas , Hiperóxia , Humanos , Resveratrol/farmacologia , Sirtuína 1/metabolismo , Biogênese de Organelas , Hiperóxia/complicações , Fator 2 Relacionado a NF-E2/metabolismo , Dimetil Sulfóxido , Lesões Encefálicas/etiologia , Oxigênio/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo
7.
Toxicol Mech Methods ; 33(4): 279-292, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36127839

RESUMO

Damage to hippocampus, cerebellum, and cortex associated with cognitive functions due to anesthetic-induced toxicity early in life may cause cognitive decline later. Aquaporin 4 (AQP4), a key protein in waste clearance pathway of brain, is involved in synaptic plasticity and neurocognition. We investigated the effects of single and repeated isoflurane (Iso) anesthesia on AQP4 levels and brain damage. Postnatal-day (P)7 Wistar albino rats were randomly assigned to Iso or Control (C) groups. For single-exposure, pups were exposed to 1.5% Iso in 30% oxygenated-air for 3-h at P7 (Iso1). For repeated-exposure, pups were exposed to Iso for 3 days, 3-h each day, at 1-day intervals (P7 + 9 + 11) starting at P7 (Iso3). C1 and C3 groups received only 30% oxygenated-air. Based on HE-staining and immunoblotting (Bax/Bcl-2, cleaved-caspase3 and PARP1) analyses, Iso exposures caused a higher degree of apoptosis in hippocampus. Anesthesia increased 4-hydroxynonenal (4HNE), oxidative stress marker; the highest ROS accumulation was determined in cerebellum. Increased inflammation (TNF-α, NF-κB) was detected. Multiple Iso-exposures caused more significant damage than single exposure. Moreover, 4HNE and TNF-α contributed synergistically to Iso-induced neurotoxicity. After anesthesia, higher expression of AQP4 was detected in cortex than hippocampus and cerebellum. There was an inverse correlation between increased AQP4 levels and apoptosis/ROS/inflammation. Correlation analysis indicated that AQP4 had a more substantial protective profile against oxidative stress than apoptosis. Remarkably, acutely increased AQP4 against Iso exhibited a more potent neuroprotective effect in cortex, especially frontal cortex. These findings promote further research to understand better the mechanisms underlying anesthesia-induced toxicity in the developing brain.


Assuntos
Isoflurano , Animais , Ratos , Isoflurano/toxicidade , Espécies Reativas de Oxigênio/metabolismo , Aquaporina 4/metabolismo , Aquaporina 4/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Ratos Wistar , Hipocampo , Apoptose , Encéfalo/metabolismo , Inflamação/metabolismo , Animais Recém-Nascidos
8.
Neurotoxicology ; 93: 140-151, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36155068

RESUMO

Sevoflurane anesthesia induces neurocognitive impairment and pyroptosis in the developing brain. Pleckstrin homology-like domain, family A, member 1 (PHLDA1) was involved in neuronal apoptosis, oxidative stress and inflammation during ischemic stroke. The role of PHLDA1 in sevoflurane-induced pyroptosis in developing rats was investigated. Firstly, neonatal rats at day 7 was exposed to 2.0% sevoflurane for 6 h to induce neurotoxicity. Pathological analysis showed that sevoflurane anesthesia induced hippocampal injury and reduced the number of neurons. The expression of PHLDA1 was elevated in hippocampus of sevoflurane-treated rats. Secondly, sevoflurane anesthesia-treated neonatal rats were injected with adeno-associated virus serotype (AAV) to mediate knockdown of PHLDA1. Injection with AAV-shPHLDA1 ameliorated sevoflurane-induced hippocampal injury and neurocognitive impairment in rats. Moreover, knockdown of PHLDA1 increased the number of neurons in sevoflurane-treated rats. Silence of PHLDA1 suppressed neuronal apoptosis, and inhibited pyroptosis in sevoflurane-treated rats. Thirdly, PHLDA1 was also elevated in sevoflurane-treated primary neuronal cells. Loss of PHLDA1 also enhanced cell viability and suppressed pyroptosis of sevoflurane-treated primary neuronal cells. Lastly, silence of PHLDA1 reduced protein expression of TRAF6 and p-Rac1 in sevoflurane-treated rats and neuronal cells. Over-expression of TRAF6 attenuated PHLDA1 silence-induced increase of cell viability and decreased pyroptosis in neuronal cells. In conclusion, loss of PHLDA1 protected against sevoflurane-induced pyroptosis in developing rats through inhibition of TRAF6-mediated activation of Rac1.


Assuntos
Piroptose , Fator 6 Associado a Receptor de TNF , Animais , Ratos , Sevoflurano/toxicidade , Fator 6 Associado a Receptor de TNF/metabolismo , Ratos Sprague-Dawley , Neurônios/metabolismo , Hipocampo/metabolismo , Apoptose , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo
9.
Biomed Pharmacother ; 152: 113229, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35679721

RESUMO

Ketamine is a widely-used anesthetic in the field of pediatrics and obstetrics. Multiple studies have revealed that ketamine causes neurotoxicity in developing animals. However, further studies are needed to determine whether clinical doses of ketamine (20 mg/kg) are able to cause kidney damage in developing animals. Herein, we investigated the effects of continuous ketamine exposure on kidney injury and pyroptosis in seven-day-old rats. Serum renal function indicators, renal histopathological analysis, pyroptosis, as well as oxidative stress indicators, were tested. Additionally, the NLRP3 inhibitor MCC950 and the Caspase-1 inhibitor VX765 were used to evaluate the role of the NLRP3/Caspase-1 axis in ketamine-induced kidney injury among developing rats. Our findings indicate that ketamine exposure causes renal histopathological injury, increased the levels of blood urea nitrogen (BUN) and creatinine (Cre), and led to upregulation in the levels of pyroptosis. Furthermore, we found that ketamine induced an increase in levels of reactive oxygen species (ROS) and malonaldehyde (MDA), as well as a decrease in the content of glutathione (GSH) and catalase (CAT) in the kidneys of neonatal rats. Moreover, targeting NLRP3 and caspase-1 with MCC950 or VX765 improved pyroptosis and reduced renal damage after continuous ketamine exposure. In conclusion, this study suggested that continued exposure to ketamine caused kidney damage among neonatal rats and that the NLRP3/Caspase-1 axis-related pyroptosis may be involved in this process.


Assuntos
Ketamina , Piroptose , Animais , Animais Recém-Nascidos , Caspase 1 , Criança , Humanos , Inflamassomos , Ketamina/toxicidade , Rim/fisiologia , Proteína 3 que Contém Domínio de Pirina da Família NLR , Ratos
10.
J Pediatr Surg ; 57(7): 1264-1268, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35379491

RESUMO

BACKGROUND: Intestinal ischemia and reperfusion (IR) injury like that seen in midgut volvulus can be life-threatening in the pediatric population. Human breast milk-derived exosomes (HMDEs) can prevent intestinal inflammation in experimental necrotizing enterocolitis and other intestinal diseases. The aim of this study is to investigate the effects of HMDEs on intestinal damage related to IR injury. METHODS: Exosomes were isolated from human breast milk by ultracentrifugation then confirmed by Nanoparticle tracking analysis and detection of exosome membrane markers. 2-weeks old Sprague Dawley rats were randomly divided into 4 groups: a) Sham (n = 8) with laparotomy alone, b) Sham with HMDEs administration by gavage (n = 8), c) Intestinal IR injury (n = 8) by occlusion of the superior mesenteric artery (SMA) for 30 min followed by reperfusion, and d) Intestinal IR by SMA occlusion with HMDEs administration by gavage (n = 8). Six hours after laparotomy, animals were euthanized, and the ilea (10 cm to cecum) were harvested. Mucosal injury was scored histologically. The intestines were further examined for inflammatory cytokine TNFα, and epithelial proliferation marker Ki67. RESULTS: Compared to sham, the small intestine of IR rats had more intestinal damage, increased expression of inflammatory cytokine TNFα and decreased intestinal proliferation. HMDEs significantly counteracted all these changes. CONCLUSIONS: Human breast milk-derived exosomes protect the intestine against damage by IR injury. This beneficial effect is associated with decreased intestinal inflammation and enhanced epithelial proliferation. This study implicates the potential novel application of HMDEs in preventing intestinal damage in infants with intestinal IR injury.


Assuntos
Exossomos , Traumatismo por Reperfusão , Animais , Animais Recém-Nascidos , Biomarcadores/metabolismo , Criança , Citocinas/metabolismo , Exossomos/metabolismo , Humanos , Inflamação/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/patologia , Isquemia , Leite Humano/metabolismo , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/etiologia , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/prevenção & controle , Fator de Necrose Tumoral alfa/metabolismo
11.
Int J Mol Med ; 49(2)2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34935055

RESUMO

Persistent pulmonary hypertension of the newborn (PPHN) is a common pulmonary vascular disease during the neonatal period, and it is associated with a high clinical mortality rate and a poor prognosis. At present, the treatment of PPHN is based mainly on inhaled nitric oxide (iNO), high­frequency ventilation, and pulmonary vasodilators. Sildenafil has gradually begun to be used in recent years for the treatment of PPHN and has exhibited some success; however, its detailed mechanism of action requires further elucidation. An animal model of neonatal pulmonary hypertension (neonatal rats, 48 h after birth, 10% O2, 14 days) as well as a cell model [human pulmonary artery smooth muscle cells (PASMCs), 4% O2, 60 h] were established. The effects of sildenafil on pulmonary hypertension in neonatal rats were evaluated by hematoxylin and eosin staining, immunofluorescence analysis, western blotting and PCR, and the changes in peroxisome proliferator­activated receptor Î³ (PPARγ), transient receptor potential canonical (TRPC)1, TRPC6 and Ki67 expression levels were detected under hypoxic conditions. The results revealed that sildenafil reversed the increases in the right ventricular mean pressure and right ventricular hypertrophy index induced by hypoxia, and attenuated pulmonary arterial remodeling as well as PASMC proliferation. The inhibitory effects of sildenafil on TRPC expression and PASMC proliferation were attenuated by GW9662 and PPARγ small interfering RNA. In conclusion, sildenafil protects against hypoxia­induced pulmonary hypertension and right ventricular hypertrophy in neonatal rats by upregulating PPARγ expression and downregulating TRPC1 and TRPC6 expression.


Assuntos
Regulação para Baixo , Hipertensão Pulmonar/tratamento farmacológico , Hipertensão Pulmonar/etiologia , Hipóxia/complicações , PPAR gama/metabolismo , Citrato de Sildenafila/uso terapêutico , Canais de Cátion TRPC/metabolismo , Animais , Animais Recém-Nascidos , Pressão Sanguínea/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Feminino , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/fisiopatologia , Humanos , Hipertensão Pulmonar/fisiopatologia , Hipóxia/fisiopatologia , Antígeno Ki-67/metabolismo , Masculino , Miócitos de Músculo Liso/metabolismo , Artéria Pulmonar/patologia , Ratos Sprague-Dawley , Citrato de Sildenafila/farmacologia , Regulação para Cima/efeitos dos fármacos , Remodelação Vascular/efeitos dos fármacos
12.
J Affect Disord ; 297: 455-462, 2022 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-34715171

RESUMO

BACKGROUND: Whilst concerns have been raised about the detrimental effects of general anaesthetics on the brain's development and function in the young, reports have indicated that thyroid hormones are able to promote neurogenesis in the developing brain. This present study aimed to investigate the effects of triiodothyronine (T3) on the neonatal rat brain, following sevoflurane exposure. METHODS: Postnatal day 7 (P7) ratpups were treated with Triiodothyronine (T3) (1 µg/100 g body weight, i.p. injection, once/day for 3 days) after 2% sevoflurane exposure for 6 h. They were sacrificed at either P7 (immediately), P15 or P30 and their brains were harvested to assess cell death, proliferation in the hippocampus, N-methyl-D-aspartate (NMDA) receptor subunit A and B, and a post-synaptic protein (PSD-95 in the hippocampus,). Neuro-behavioral changes in other cohorts between P27 and P30 were evaluated with Morris water maze and open field tests. RESULTS: Sevoflurane exposure caused cell death and suppressed the proliferation of astrocytes and neurons, as well as the dendritic growth of neurons in the hippocampus which were all reversed by the administration of T3. Moreover, cognitive function, including learning, memory, and adaptability to a new environment, were impaired by sevoflurane exposure, which was also negated by T3 treatment. Furthermore, sevoflurane decreased the expression of NMDA receptor subunits NR2A and NR2B, as well as PSD-95 in the hippocampus at P15 and those effects of sevoflurane were abolished by T3 administration. CONCLUSIONS: A potential therapeutic role of T3 in protecting general anesthetic induced neuronal injury in the developing brain is likely to occur through enhancing expression of PSD-95 and the NMDA NR2A and NR2B expression.


Assuntos
Anestésicos Inalatórios , Éteres Metílicos , Animais , Animais Recém-Nascidos , Hipocampo , Aprendizagem em Labirinto , Ratos , Ratos Sprague-Dawley , Sevoflurano/farmacologia , Tri-Iodotironina
13.
Nutrients ; 13(12)2021 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-34959827

RESUMO

Given that combined vitamin A (VA) and retinoic acid (RA) supplementation stimulated the intestinal uptake of plasma retinyl esters in neonatal rats, we administrated an RA dose as a pretreatment before VA supplementation to investigate the distinct effect of RA on intestinal VA kinetics. On postnatal days (P) 2 and 3, half of the pups received an oral dose of RA (RA group), while the remaining received canola oil as the control (CN). On P4, after receiving an oral dose of 3H-labeled VA, pups were euthanized at selected times (n = 4-6/treatment/time) and intestine was collected. In both CN and RA groups, intestinal VA mass increased dramatically after VA supplementation; however, RA-pretreated pups had relatively higher VA levels from 10 h and accumulated 30% more VA over the 30-h study. Labeled VA rapidly peaked in the intestine of CN pups and then declined from 13 h, while a continuous increase was observed in the RA group, with a second peak at 10 h and nearly twice the accumulation of 3H-labeled VA compared to CN. Our findings indicate that RA pretreatment may stimulate the influx of supplemental VA into the intestine, and the increased VA accumulation suggests a potential VA storage capacity in neonatal intestine.


Assuntos
Transporte Biológico/efeitos dos fármacos , Suplementos Nutricionais , Tretinoína/administração & dosagem , Vitamina A/metabolismo , Animais , Animais Recém-Nascidos , Feminino , Intestino Delgado/metabolismo , Cinética , Masculino , Gravidez , Óleo de Brassica napus/administração & dosagem , Ratos , Ratos Sprague-Dawley
14.
Zhongguo Ying Yong Sheng Li Xue Za Zhi ; 37(6): 699-704, 2021 Nov.
Artigo em Chinês | MEDLINE | ID: mdl-34821109

RESUMO

Objective: To establish a stable, rapid and improved method for isolation and culture of primary cardiomyocytes from neonatal rats. Methods: Ventricular tissues from neonatal SD rats were digested with 0.12% collagenase Ⅱ, and then subjected to Percoll density gradient centrifugation. The original cardiomyocytes were cultured in modified DMEM/F12 containing 5% horse serum and 5-bromodeoxyuracil(5-BrdU) in vitro for further purification, and medium was changed to normal high glucose DMEM with 10% FBS the next day. The difference between the improved method and traditional differential attachment one used for isolation and culture of primary cardiomyocytes was compared. Results: Cardiacmyocytes obtained through the improved method grew well. 24 hours after plating, most cells adhered to the dishes, with shapes looked triangular, fusiform or irregular, and some of them showed spontaneously contract at a frequency varying from 10~30 times/min. After 48 h culture, the cardiomyocytes became longer and stretched out pseudopodia. Some cells showed synchronous beats with the frequency close to 50~80 times/min. 72 hours later, cardiomyocytes were interwoven into a network in chrysanthemum patterns, and spontaneous beats tended to be more synchronous, with a frequency of 80-100 times/min. After 96 h, cells gathered into clusters as islands, with synchronous beat at a frequency of around 100~120 times/min. All cardiomyocytes were in good condition within one week. Yields((1.17±0.15)×106 vs (1.21±0.22)×106,P>0.05)and survival rate of primary cardiomyocytes obtained by the improved method was comparable to that gained using traditional differential attachment way (93.3%±1.4% vs 92.2%±0.7%, P>0.05), but the purity of primary cardiomyocytes obtained through the improved method was much higher (94.7%±2.1% vs 89.5%±1.3%, P<0.05), while with less time consuming ((3.1±0.4)h vs (4.3±0.3)h, P<0.01). Conclusion: This improved method is an ideal and simple method for the isolation and culture of primary cardiomyocytes with shorter time-consuming, high purity, intact structure and function, and with great repeatability and stability.


Assuntos
Ventrículos do Coração , Miócitos Cardíacos , Animais , Animais Recém-Nascidos , Células Cultivadas , Ratos , Ratos Sprague-Dawley
15.
J Inflamm Res ; 14: 5393-5401, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34703276

RESUMO

PURPOSE: To evaluate whether ferroptosis is involved in hyperoxic acute lung injury (HALI) and its mechanisms through the HALI model. METHODS: HE staining was used to assess lung injury pathology after the establishment of neonatal rat HALI model. ELISA was used to detect ROS, GPX4, and GSH expression. Prussian blue staining and Western Blot were used to detect iron deposition and the expression of ferroptosis-related proteins, respectively. RESULTS: The HALI group showed pathological changes with larger and fewer alveoli and thicker alveolar septa after HE staining. Prussian blue staining detected significant iron deposition in the lung tissue of the HALI group. GPX4, GSH, GSS, and SLC7A11 expressions were significantly decreased in the HALI group than in the normal control group. In contrast, ROS, TFRC, FHC, and FLC expressions showed opposite results (p<0.05). CONCLUSION: Ferroptosis may be involved in the pathological process of hyperoxic lung injury in neonatal rats.

16.
Mol Neurobiol ; 58(12): 6272-6289, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34480336

RESUMO

The Notch signaling pathway plays an important role in the regulation of neurogenesis. The objective of this study was to investigate whether the Notch signaling pathway was involved in the neurogenesis impairment and long-term neurocognitive dysfunction caused by neonatal exposure to ketamine. On postnatal day 7 (PND-7), male Sprague-Dawley (SD) rats were intraperitoneally injected with 40 mg/kg ketamine four consecutive times (40 mg/kg × 4) at 1-h intervals. Notch ligand Jagged1 (0.5 mg/kg) and lentivirus overexpressing the Notch1 intracellular domain (LV-NICD1) were microinjected into the hippocampal dentate gyrus (DG) 1 h or 4 days before ketamine administration, respectively. The expression of Notch1 signaling pathway-related proteins was detected by Western blotting 24 h after ketamine administration. The proliferation and differentiation of the neural stem cells (NSCs) in the hippocampal DG were evaluated by double immunofluorescence staining 24 h after treatment. Moreover, changes in hippocampus-dependent spatial memory of 2-month-old rats were investigated with the Morris water maze test. Ketamine anesthesia in neonatal rats decreased the expression levels of Jagged1, Notch1, NICD1, and hairy enhancer of split 1 (Hes1); inhibited the proliferation and astrocytic differentiation of NSCs; and promoted the differentiation of neurons. Neonatal exposure to ketamine caused deficits in hippocampus-dependent spatial reference memory tasks in 2-month-old rats. Microinjection of Jagged1 or LV-NICD1 reversed the inhibitory effect of ketamine on the expression of Notch1-related proteins in the hippocampal DG, attenuated the ketamine-mediated decrease in NSC proliferation and differentiation, and improved the cognitive function of 2-month-old rats after neonatal exposure to ketamine. These results suggest that neonatal exposure to ketamine in rats inhibits the proliferation and differentiation of hippocampal NSCs and impairs neurocognitive function in adulthood. The Notch1 signaling pathway may be involved in the impairment of hippocampus-dependent learning and memory during adulthood caused by neonatal exposure to ketamine. These findings contribute to further understanding the neurotoxicity induced by neonatal exposure to ketamine and the underlying mechanisms.


Assuntos
Anestésicos Dissociativos/farmacologia , Proliferação de Células/efeitos dos fármacos , Cognição/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Ketamina/farmacologia , Células-Tronco Neurais/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Hipocampo/metabolismo , Masculino , Células-Tronco Neurais/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor Notch1/metabolismo , Transdução de Sinais/efeitos dos fármacos
17.
Environ Toxicol ; 36(12): 2454-2466, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34464015

RESUMO

The pesticide 2,4-dichlorophenoxyacetic acid (2,4-D) has neurotoxic effects, but its mechanism is not clear. In this study, a 2,4-D (75 mg/kg. b.w) exposure model was established in SD rats with colostrum. Lipopolysaccharide (1 mg/kg b.w) was used as the positive control, and Lycium barbarum polysaccharide (LBP, 50 mg/kg b.w) was used as an intervention factor to explore the neurotoxic effect of 2,4-D and the neuroprotective effect of LBP. Our research results show that 2,4-D causes a decrease in the number of hippocampal CA3 pyramidal cells and pyknosis in nuclei with a triangular or irregular shape and that rats show signs of anxiety or depression. In rat serum, superoxide dismutase, and glutathione peroxidase activity decreased, while malondialdehyde content increased. Protein and mRNA levels of TNFα, IL-6, IL-1ß, IL-18, NLRP3, ASC, caspase-1, IL-1ß, IL-18, and p62 increased, while those of LC3-II/LC3-I and Beclin-1 decreased in hippocampal tissues. In conclusion, 2,4-D increased the oxidative stress level, induced neuroinflammatory response, and decreased the autophagy level in experimental rats. LBP may have upregulated the autophagy level in the body by inhibiting the activation of the NLRP3 inflammasome, thus playing a neuroprotective role.


Assuntos
Inflamassomos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Ácido 2,4-Diclorofenoxiacético/toxicidade , Animais , Animais Recém-Nascidos , Proteínas Relacionadas à Autofagia , Medicamentos de Ervas Chinesas , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Ratos , Ratos Sprague-Dawley
18.
Brain Res ; 1768: 147588, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34310937

RESUMO

Preterm infant brain injury is a leading cause of morbidity and disability in survivors of preterm infants. Unfortunately, the effective treatment remains absent. Recent evidence suggests that GSK-3ß inhibitor TWS119 has a neuroprotectiverole in adult brain injury by activation of Wnt/ß-catenin signaling pathway. However, the role on neonatal brain injury is not yet explored. The study aims to evaluate the effect of TWS119 at 7 d after hypoxic-ischemic brain damage and investigate the mechanism that it regulates Wnt and Notch signaling pathways at 24 h after hypoxic-ischemic brain damage in neonatal rats. Three-day-old rats were randomly divided into 3 groups: sham group, HI group and TWS119 group. The neonatal rats were subjected to left carotid artery ligation followed by 2 h of hypoxia (8.0% O2). A single dose of TWS119 (30 mg/kg) was intraperitoneally injected 20 min prior to hypoxia-ischemia (HI). At 7 d after HI, TWS119 improved the tissue structure, reduced cell apoptosis, up-regulated bcl-2 expression, up-regulated the expression of PSD-95 and Synapsin-1. At 24 h after HI, it activated Wnt/ß-catenin signaling pathway by up-regulation of ß-catenin protein expression and wnt3a/wnt5a/wnt7a mRNA expression. Simultaneously, it suppressed Notch signaling pathway by down-regulation of Notch1 and HES-1 proteins expression. Our study suggested that TWS119 performed a neuroprotective function at 7 d after hypoxic-ischemic brain damage via a crosstalk with Wnt/ß-catenin and Notch signaling pathways at 24 h after hypoxic-ischemic brain damage in neonatal rats.


Assuntos
Glicogênio Sintase Quinase 3 beta/antagonistas & inibidores , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Pirimidinas/farmacologia , Pirróis/farmacologia , Animais , Animais Recém-Nascidos/metabolismo , Encéfalo/metabolismo , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/metabolismo , Feminino , Glicogênio Sintase Quinase 3 beta/efeitos dos fármacos , Glicogênio Sintase Quinase 3 beta/metabolismo , Hipóxia-Isquemia Encefálica/metabolismo , Masculino , Modelos Animais , Ratos , Ratos Sprague-Dawley , Receptores Notch/efeitos dos fármacos , Receptores Notch/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Via de Sinalização Wnt/efeitos dos fármacos , Via de Sinalização Wnt/fisiologia
19.
Am J Transl Res ; 13(4): 2784-2793, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34017441

RESUMO

OBJECTIVE: To explore the effect of miR-21-5p on the MAP2K3 expressions and cellular apoptosis in the lung tissues of neonatal rats with hyperoxia-induced lung injuries (HILI). METHODS: Twenty Sprague-Dawley neonatal rats were assigned to the normal group, and 120 rats were used to create a HILI model and were divided into the following six groups of 20 rats each: the model group, the miR-21-5p NC group, the miR-21-5p agomir group, the oe-NC group (MAP2K3 overexpression NC), the oe-MAP2K3 group, and the miR-21-5p agomir+oe-MAP2K3 group. RESULTS: miR-21-5p can target MAP2K3. Compared with the normal rats, the rats with HILI had lower miR-21-5p expression levels and higher MAP2K3 expression levels in the lung tissues (both P<0.05). Unlike the normal group, the other groups all presented different degrees of lung injuries, lower Bcl-2 expression levels, higher cellular apoptosis rates, and higher expression levels of cleaved caspase-3, Bax, IL-6, and TNF-α (all P<0.05). Compared with the model and the miR-21-5p NC groups, the miR-21-5p agomir group had better results in terms of the aforementioned markers; compared with the oe-NC group, the oe-MAP2K3 group had worse results in terms of these markers (all P<0.05). Moreover, we found that the protective effects of miR-21-5p overexpression on the lung tissues of HILI rats can be partially blocked by MAP2K3 overexpression. CONCLUSION: miR-21-5p can inhibit MAP2K3 expression and reduce cellular apoptosis in HILI, thereby exerting protective effects on neonatal rats with HILI.

20.
Respir Physiol Neurobiol ; 284: 103582, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33197605

RESUMO

Prenatal cigarette smoke (CS) exposure causes numerous respiratory health problems in infants. This study aimed to investigate the effect of prenatal CS exposure on sevoflurane-induced respiratory suppression in neonatal rats and the protective role of H2S. We found that at baseline, minute ventilation (V'E), respiratory frequency (fR), and tidal volume (VT) were similar among tested groups, whereas sigh frequency (fS) was lower in CS group than in the Control group. During 3 % sevoflurane anesthesia, V'E was decreased, fR was slowed, VT was increased, and fS was reduced in all groups; however, the decline in fR and increase in VT was greater in CS group than in the Control group. During the recovery, fS remained lower in CS group. The above changes of respiratory response caused by prenatal CS exposure were alleviated by NaHS pretreatment (a donor of H2S, 56 µmol/kg/d, intraperitoneal injection). These results indicated that prenatal CS exposure alters the breathing into a much slower and deeper manner in neonatal rats during sevoflurane anesthesia, and H2S mitigates this respiratory change.


Assuntos
Sulfeto de Hidrogênio/farmacologia , Efeitos Tardios da Exposição Pré-Natal , Transtornos Respiratórios , Sulfetos/farmacologia , Poluição por Fumaça de Tabaco/efeitos adversos , Anestésicos Inalatórios/farmacologia , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Feminino , Masculino , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Efeitos Tardios da Exposição Pré-Natal/prevenção & controle , Ratos , Ratos Sprague-Dawley , Transtornos Respiratórios/induzido quimicamente , Transtornos Respiratórios/fisiopatologia , Transtornos Respiratórios/prevenção & controle , Sevoflurano/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA