Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Int Immunopharmacol ; 125(Pt A): 111108, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37890380

ABSTRACT

Currently, there are no effective therapeutic targets for the treatment of chronic cerebral hypoperfusion(CCH)-induced cerebral ischemic injury. Vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) are discovered as the inducers of neurogenesis and angiogenesis. We previously made a nanofiber membrane (NFM), maintaining a long-term release of VEGF and bFGF up to 35 days, which might make VEGF and bFGF NFM as the potential protective agents against cerebral ischemic insult. In this study, the effects of VEGF and bFGF delivered by NFM into brain were investigated as well as their underlying mechanismsin a rat model of CCH. VEGF + bFGF NFM application increased the expressions of tight junction proteins, maintained BBB integrity, and alleviated vasogenic cerebral edema. Furthermore, VEGF + bFGF NFM sticking enhanced angiogenesis and elevated CBF. Besides, VEGF + bFGF NFM treatment inhibited neuronal apoptosis and decreased neuronal loss. Moreover, roofing of VEGF + bFGF NFM attenuated microglial activation and blocked the launch of NLRP3/caspase-1/IL-1ß pathway. In addition, VEGF + bFGF NFM administration prevented disruption to the pre/postsynaptic membranes and loss of myelin sheath, relieving synaptic injury and demyelination. Oligodendrogenesis, neurogenesis and PI3K/AKT/mTOR pathway were involved in the treatment of VEGF + bFGF NFM against CCH-induced neuronal injury and hypomyelination. These findings supported that VEGF + bFGF NFM application constitutes a neuroprotective strategy for the treatment of CCH, which may be worth further clinical translational research as a novel neuroprotective approach, benifiting indirect surgical revascularization.


Subject(s)
Brain Injuries , Brain Ischemia , Nanofibers , Rats , Animals , Vascular Endothelial Growth Factor A/therapeutic use , Vascular Endothelial Growth Factor A/metabolism , Fibroblast Growth Factor 2/therapeutic use , Phosphatidylinositol 3-Kinases , Nanofibers/therapeutic use , Vascular Endothelial Growth Factors , Brain Ischemia/metabolism , Ischemia
2.
Nanomedicine ; 48: 102639, 2023 02.
Article in English | MEDLINE | ID: mdl-36549557

ABSTRACT

We investigated the potential effects and mechanisms of vascular endothelial growth factor (VEGF)-nanofiber membranes (NFMs) treatment in a rat model of chronic cerebral hypoperfusion (CCH). VEGF-NFMs treatment promoted angiogenesis in surgical temporal cortex and hippocampus, alleviating decreased CBF in these two cerebral regions. VEGF-NFMs application improved reduced NAA/Cr ratio, preventing neuronal loss. VEGF-NFMs sticking decreased the number of TUNEL-positive cells in surgical temporal cortex, ameliorated impaired synaptic plasticity, and inhibited the release of pro-inflammatory cytokines and the activation of microglia and astrocytes in surgical temporal cortex and hippocampus. Furthermore, BDNF-TrkB/PI3K/AKT, BDNF-TrkB/ERK and HIF-1a/VEGF/ERK pathways were involved in the treatment of VEGF-NFMs against CCH-induced neuronal injury. These results showed the neuroprotective effects of VEGF-NFMs sticking may initiate from neurovascular repairing followed by inhibition of neuronal apoptosis and neuronal and synaptic damage, eventually leading to the suppression of cognitive dysfunction, which provided theoretical foundation for further clinical transformation of VEGF-NFMs.


Subject(s)
Brain Ischemia , Cognitive Dysfunction , Nanofibers , Rats , Animals , Vascular Endothelial Growth Factor A/metabolism , Phosphatidylinositol 3-Kinases , Brain-Derived Neurotrophic Factor , Brain Ischemia/drug therapy
3.
Int J Nanomedicine ; 16: 3661-3678, 2021.
Article in English | MEDLINE | ID: mdl-34093011

ABSTRACT

INTRODUCTION: Brain ischemia is a common neurological disorder worldwide that activates a cascade of pathophysiological events involving decreases in oxygen and glucose levels. Despite substantial efforts to explore its pathogenesis, the management of ischemic neuronal injury remains an enormous challenge. Accumulating evidence suggests that VEGF modified nanofiber (NF) materials and the fatty-acid amide hydrolase (FAAH) inhibitor URB597 exert an influence on alleviating ischemic brain damage. We aimed to further investigate their effects on primary hippocampal neurons, as well as the underlying mechanisms following oxygen-glucose deprivation (OGD). METHODS: Different layers of VEGF-A loaded polycaprolactone (PCL) nanofibrous membranes were first synthesized by using layer-by-layer (LBL) self-assembly of electrospinning methods. The physicochemical and biological properties of VEGF-A NF membranes, and their morphology, hydrophilicity, and controlled-release of VEGF-A were then estimated. Furthermore, the effects of VEGF-A NF and URB597 on OGD-induced mitochondrial oxidative stress, inflammatory responses, neuronal apoptosis, and endocannabinoid signaling components were assessed. RESULTS: The VEGF-A NF membrane and URB597 can not only promote hippocampal neuron adhesion and viability following OGD but also exhibited antioxidant/anti-inflammatory and mitochondrial membrane potential protection. The VEGF-A NF membrane and URB597 also inhibited OGD-induced cellular apoptosis through activating CB1R signaling. These results indicate that VEGF-A could be controlled-released by LBL self-assembled NF membranes. DISCUSSION: The VEGF-A NF membrane and URB597 displayed positive synergistic neuroprotective effects through the inhibition of mitochondrial oxidative stress and activation of CB1R/PI3K/AKT/BDNF signaling, suggesting that a VEGF-A loaded NF membrane and the FAAH inhibitor URB597 could be of therapeutic value in ischemic cerebrovascular diseases.


Subject(s)
Benzamides/pharmacology , Carbamates/pharmacology , Nanofibers/chemistry , Neurons/drug effects , Neuroprotective Agents/pharmacology , Vascular Endothelial Growth Factor A/pharmacology , Amidohydrolases/antagonists & inhibitors , Amidohydrolases/metabolism , Animals , Apoptosis/drug effects , Apoptosis/physiology , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Brain Ischemia/pathology , Cells, Cultured , Endocannabinoids/metabolism , Glucose/metabolism , Hippocampus/drug effects , Hippocampus/pathology , Membranes, Artificial , Neurons/metabolism , Neurons/pathology , Oxygen/metabolism , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor A/chemistry
4.
Neurotox Res ; 37(4): 847-856, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31808139

ABSTRACT

Chronic cerebral hypoperfusion (CCH) is a major contributor to cognitive decline and degenerative processes leading to Alzheimer's disease, vascular dementia, and aging. However, the delicate mechanism of CCH-induced neuronal damage, and therefore proper treatment, remains unclear. WIN55,212-2 (WIN) is a nonselective cannabinoid receptor agonist that has been shown to have effects on hippocampal neuron survival. In this study, we investigated the potential roles of WIN, as well as its underlying mechanism in a rat CCH model of bilateral common carotid artery occlusion. Hippocampal morphological changes and mitochondrial ultrastructure were detected using hematoxylin and eosin staining and electron microscopy, respectively. Various biomarkers, such as reactive oxidative species (ROS), superoxide dismutase (SOD), catalase (CAT), and malondialdehyde (MDA) were used to assess the level of oxidative stress in the hippocampus. Furthermore, the expression levels of neuronal nuclei (NeuN), apoptosis signal-regulating kinase 1 (ASK1)-p38 signaling proteins, cleaved Caspase-9 and -3, and cytochrome-c (Cyt-C) were accessed by western blotting. CCH decreased the levels of NeuN, Cyt-C (mitochondrial), SOD, and CAT, and increased the levels of MDA, phosphorylated ASK1 and phosphorylated p38, cleaved Caspase-9 and -3, and Cyt-C (cytoplasm), which were reversed by WIN treatment. Chronic treatment with WIN also improved CCH-induced neuronal degeneration and mitochondrial fragmentation. These findings indicated that WIN may be a potential therapeutic agent for ischemic neuronal damage, involving a mechanism associated with the suppression of oxidative stress and ASK1-p38 signaling.


Subject(s)
Benzoxazines/therapeutic use , Brain Ischemia/metabolism , Cannabinoid Receptor Agonists/therapeutic use , Hippocampus/metabolism , MAP Kinase Kinase Kinase 5/metabolism , Morpholines/therapeutic use , Naphthalenes/therapeutic use , Oxidative Stress/physiology , Animals , Benzoxazines/pharmacology , Brain Ischemia/drug therapy , Cannabinoid Receptor Agonists/pharmacology , Hippocampus/drug effects , MAP Kinase Kinase Kinase 5/antagonists & inhibitors , MAP Kinase Signaling System/drug effects , Male , Morpholines/pharmacology , Naphthalenes/pharmacology , Neurons/drug effects , Neurons/metabolism , Oxidative Stress/drug effects , Random Allocation , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...