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.
Mol Med Rep ; 16(6): 9627-9635, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29039533

ABSTRACT

Early brain injury is considered to be a major risk that is related to the prognosis of subarachnoid hemorrhage (SAH). In SAH model rats, brain edema and apoptosis have been closely related with death rate and neurological function. Sirtuin 1 (SIRT1) was reported to be involved in apoptosis in cerebral ischemia and brain tumor formation through p53 deacetylation. The present study aimed to evaluate the role of SIRT1 in a rat endovascular perforation model of SAH. The SIRT1 activator resveratrol (RES) was administered 48 h prior to SAH induction and the SIRT1 inhibitor Sirtinol (SIR) was used to reverse the effects of RES on SIRT1 expression. Mortality rate, neurological function and brain water content were measured 24 h post­SAH induction. Proteins associated with the blood brain barrier (BBB), apoptosis and SIRT1 in the cortex, such as zona occludens 1 (ZO­1), occludin, claudin­5, SIRT1, p53 and cleaved caspase3 were investigated. mRNA expression of the p53 downstream molecules including Bcl­associated X protein, P53 upregulated modulator of apoptosis, Noxa and BH3 interacting­domain death agonist were also investigated. Neuronal apoptosis was also investigated by immunofluorescence. RES pretreatment reduced the mortality rate and improved neurological function, which was consistent with reduced brain water content and neuronal apoptosis; these effects were partially reversed by co­treatment with SIR. SIRT1 may reduce the brain water content by improvement of dysfunctional BBB permeability, and protein analysis revealed that both ZO­1, occludin and claudin­5 may be involved, and these effects were reversed by SIRT1 inhibition. SIRT1 may also affect apoptosis post­SAH through p53 deacetylation, and the analysis of p53 related downstream pro­apoptotic molecules supported this hypothesis. Localization of neuron specific apoptosis revealed that SIRT1 may regulate neuronal apoptosis following SAH. SIRT1 may also ease brain edema and neuronal protection through BBB improvement and p53 deacetylation. SIRT1 activators such as RES may have the potential to improve the prognosis of patients with SAH and clinical research should be investigated further.


Subject(s)
Apoptosis/drug effects , Brain Edema/etiology , Brain Edema/metabolism , Neurons/drug effects , Neurons/metabolism , Sirtuin 1/metabolism , Stilbenes/pharmacology , Subarachnoid Hemorrhage/complications , Animals , Brain Edema/diagnosis , Brain Edema/drug therapy , Caspase 3/metabolism , Disease Models, Animal , Fluorescent Antibody Technique , Male , Mortality , Neuroprotection , Occludin/genetics , Occludin/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Resveratrol , Zonula Occludens-1 Protein/genetics , Zonula Occludens-1 Protein/metabolism
2.
Mol Neurobiol ; 53(4): 2668-78, 2016 May.
Article in English | MEDLINE | ID: mdl-26143258

ABSTRACT

Minocycline has beneficial effects in early brain injury (EBI) following subarachnoid hemorrhage (SAH); however, the molecular mechanisms underlying these effects have not been clearly identified. This study was undertaken to determine the influence of minocycline on inflammation and neural apoptosis and the possible mechanisms of these effects in early brain injury following subarachnoid hemorrhage. SAH was induced by the filament perforation model of SAH in male Sprague-Dawley rats. Minocycline or vehicle was given via an intraperitoneal injection 1 h after SAH induction. Minocycline treatment markedly attenuated brain edema secondary to blood-brain barrier (BBB) dysfunction by inhibiting NLRP3 inflammasome activation, which controls the maturation and release of pro-inflammatory cytokines, especially interleukin-1ß (IL-1ß). Minocycline treatment also markedly reduced the number of terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling (TUNEL)-positive cells. To further identify the potential mechanisms, we demonstrated that minocycline increased Bcl2 expression and reduced the protein expression of P53, Bax, and cleaved caspase-3. In addition, minocycline reduced the cortical levels of reactive oxygen species (ROS), which are closely related to both NLRP3 inflammasome and P53 expression. Minocycline protects against NLRP3 inflammasome-induced inflammation and P53-associated apoptosis in early brain injury following SAH. Minocycline's anti-inflammatory and anti-apoptotic effect may involve the reduction of ROS. Minocycline treatment may exhibit important clinical potentials in the management of SAH.


Subject(s)
Apoptosis , Brain Injuries/drug therapy , Inflammasomes/metabolism , Inflammation/drug therapy , Minocycline/therapeutic use , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Subarachnoid Hemorrhage/complications , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/drug effects , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/pathology , Brain Injuries/etiology , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Inflammation/complications , Inflammation/pathology , Interleukin-1beta/metabolism , Male , Microglia/drug effects , Microglia/pathology , Minocycline/pharmacology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Protein Transport/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Transcription Factor RelA/metabolism , Water
3.
J Pineal Res ; 57(3): 340-7, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25187344

ABSTRACT

Melatonin is a strong anti-oxidant that has beneficial effects against early brain injury (EBI) following a subarachnoid hemorrhage (SAH) in rats; protection includes the reduction of both mortality and neurological deficits. The molecular mechanisms underlying these clinical effects in the SAH model have not been clearly identified. This study examined the influence of melatonin on brain edema secondary to disruption of the blood-brain barrier (BBB) and the relationship between these effects and pro-inflammatory cytokines in EBI following SAH using the filament perforation model of SAH in male Sprague-Dawley rats. Melatonin (150 mg/kg) or vehicle was given via an intraperitoneal injection 2 hr after SAH induction. Brain samples were extracted 24 hr after SAH. Melatonin treatment markedly attenuated brain edema secondary to BBB dysfunctions by preventing the disruption of tight junction protein expression (ZO-1, occludin, and claudin-5). Melatonin treatment also repressed cortical levels of pro-inflammatory cytokines (IL-1ß, IL-6, and TNF-α), which were increased in EBI 24 hr after SAH. To further identify the mechanism of this protection, we demonstrated that administration of melatonin attenuated matrix metallopeptidase 9 expression/activity and vascular endothelial growth factor expression, which are related to the inflammatory response and BBB disruption in EBI after SAH. Taken together, this report shows that melatonin prevents disruption of tight junction proteins which might play a role in attenuating brain edema secondary to BBB dysfunctions by repressing the inflammatory response in EBI after SAH, possibly associated with regulation of pro-inflammatory cytokines.


Subject(s)
Brain Edema/prevention & control , Brain Injuries/complications , Cytokines/metabolism , Inflammation Mediators/metabolism , Melatonin/administration & dosage , Subarachnoid Hemorrhage/complications , Animals , Brain Edema/etiology , Male , Melatonin/therapeutic use , Rats , Rats, Sprague-Dawley
4.
Biochem Biophys Res Commun ; 437(3): 469-74, 2013 Aug 02.
Article in English | MEDLINE | ID: mdl-23850688

ABSTRACT

Although hypoxia-inducible factor-1α (HIF-1α) has been extensively studied in brain injury following hypoxia-ischemia, the role of HIF-1α in early brain injury (EBI) after subarachnoid hemorrhage (SAH) remains unclear. The present study was under taken to investigate a potential role of HIF-1α in EBI after SAH. Rats (n=60) were randomly divided into sham+vehicle, SAH+2-methoxyestradiol (2ME2), and SAH+vehicle groups. The SAH model was induced by endovascular perforation and all the rats were subsequently sacrificed at 24h after SAH. We found that treatment with 2ME2 suppressed the expression of HIF-1α, BNIP3 and VEGF and reduced cell apoptosis, blood-brain barrier (BBB) permeability, brain edema, and neurologic scores. Double fluorescence labeling revealed that HIF-1α was expressed predominantly in the nuclei of neurons and TUNEL-positive cells. Our work demonstrated that HIF-1α may play a role in EBI after SAH, causing cell apoptosis, BBB disruption, and brain edema by up-regulating its downstream targets, BNIP3 and VEGF. These effects were blocked by the HIF-1α inhibitor, 2ME2.


Subject(s)
Brain Injuries/drug therapy , Brain Injuries/etiology , Estradiol/analogs & derivatives , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Subarachnoid Hemorrhage/complications , 2-Methoxyestradiol , Animals , Brain Injuries/metabolism , Disease Models, Animal , Endovascular Procedures/methods , Estradiol/administration & dosage , Estradiol/therapeutic use , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/biosynthesis , Mitochondrial Proteins , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/therapeutic use , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/biosynthesis , Rats , Rats, Sprague-Dawley , Subarachnoid Hemorrhage/drug therapy , Subarachnoid Hemorrhage/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...