Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Transl Stroke Res ; 15(1): 219-237, 2024 02.
Article in English | MEDLINE | ID: mdl-36631632

ABSTRACT

Subarachnoid hemorrhage (SAH) is a type of stroke with high morbidity and mortality. Netrin-1 (NTN-1) can alleviate early brain injury (EBI) following SAH by enhancing peroxisome proliferator-activated receptor gamma (PPARγ), which is an important transcriptional factor modulating lipid metabolism. Ferroptosis is a newly discovered type of cell death related to lipid metabolism. However, the specific function of ferroptosis in NTN-1-mediated neuroprotection following SAH is still unclear. This study aimed to evaluate the neuroprotective effects and the possible molecular basis of NTN-1 in SAH-induced EBI by modulating neuronal ferroptosis using the filament perforations model of SAH in mice and the hemin-stimulated neuron injury model in HT22 cells. NTN-1 or a vehicle was administered 2 h following SAH. We examined neuronal death, brain water content, neurological score, and mortality. NTN-1 treatment led to elevated survival probability, greater survival of neurons, and increased neurological score, indicating that NTN-1-inhibited ferroptosis ameliorated neuron death in vivo/in vitro in response to SAH. Furthermore, NTN-1 treatment enhanced the expression of PPARγ, nuclear factor erythroid 2-related factor 2 (Nrf2), and glutathione peroxidase 4 (GPX4), which are essential regulators of ferroptosis in EBI after SAH. The findings show that NTN-1 improves neurological outcomes in mice and protects neurons from death caused by neuronal ferroptosis. Furthermore, the mechanism underlying NTN-1 neuroprotection is correlated with the inhibition of ferroptosis, attenuating cell death via the PPARγ/Nrf2/GPX4 pathway and coenzyme Q10-ferroptosis suppressor protein 1 (CoQ10-FSP1) pathway.


Subject(s)
Brain Injuries , Ferroptosis , Subarachnoid Hemorrhage , Rats , Mice , Animals , NF-E2-Related Factor 2/metabolism , PPAR gamma , Rats, Sprague-Dawley , Subarachnoid Hemorrhage/complications , Netrin-1/pharmacology , Brain Injuries/drug therapy , Brain Injuries/etiology , Brain Injuries/metabolism , Signal Transduction
2.
Front Cell Dev Biol ; 11: 1154831, 2023.
Article in English | MEDLINE | ID: mdl-37009480

ABSTRACT

Sirtuin-3 (SIRT3) is responsible for maintaining mitochondrial homeostasis by deacetylating substrates in an NAD+-dependent manner. SIRT3, the primary deacetylase located in the mitochondria, controls cellular energy metabolism and the synthesis of essential biomolecules for cell survival. In recent years, increasing evidence has shown that SIRT3 is involved in several types of acute brain injury. In ischaemic stroke, subarachnoid haemorrhage, traumatic brain injury, and intracerebral haemorrhage, SIRT3 is closely related to mitochondrial homeostasis and with the mechanisms of pathophysiological processes such as neuroinflammation, oxidative stress, autophagy, and programmed cell death. As SIRT3 is the driver and regulator of a variety of pathophysiological processes, its molecular regulation is significant. In this paper, we review the role of SIRT3 in various types of brain injury and summarise SIRT3 molecular regulation. Numerous studies have demonstrated that SIRT3 plays a protective role in various types of brain injury. Here, we present the current research available on SIRT3 as a target for treating ischaemic stroke, subarachnoid haemorrhage, traumatic brain injury, thus highlighting the therapeutic potential of SIRT3 as a potent mediator of catastrophic brain injury. In addition, we have summarised the therapeutic drugs, compounds, natural extracts, peptides, physical stimuli, and other small molecules that may regulate SIRT3 to uncover additional brain-protective mechanisms of SIRT3, conduct further research, and provide more evidence for clinical transformation and drug development.

3.
Front Neurol ; 14: 1146106, 2023.
Article in English | MEDLINE | ID: mdl-37034089

ABSTRACT

Objective: Aneurysmal subarachnoid hemorrhage (aSAH) is a common and potentially fatal cerebrovascular disease. Poor-grade aSAH (Hunt-Hess grades IV and V) accounts for 20-30% of patients with aSAH, with most patients having a poor prognosis. This study aimed to develop a stable nomogram model for predicting adverse outcomes at 6 months in patients with aSAH, and thus, aid in improving the prognosis. Method: The clinical data and imaging findings of 150 patients with poor-grade aSAH treated with microsurgical clipping of intracranial aneurysms on admission from December 2015 to October 2021 were retrospectively analyzed. Least absolute shrinkage and selection operator (LASSO), logistic regression analyses, and a nomogram were used to develop the prognostic models. Receiver operating characteristic (ROC) curves and Hosmer-Lemeshow tests were used to assess discrimination and calibration. The bootstrap method (1,000 repetitions) was used for internal validation. Decision curve analysis (DCA) was performed to evaluate the clinical validity of the nomogram model. Result: LASSO regression analysis showed that age, Hunt-Hess grade, Glasgow Coma Scale (GCS), aneurysm size, and refractory hyperpyrexia were potential predictors for poor-grade aSAH. Logistic regression analyses revealed that age (OR: 1.107, 95% CI: 1.056-1.116, P < 0.001), Hunt-Hess grade (OR: 8.832, 95% CI: 2.312-33.736, P = 0.001), aneurysm size (OR: 6.871, 95% CI: 1.907-24.754, P = 0.003) and refractory fever (OR: 3.610, 95% CI: 1.301-10.018, P < 0.001) were independent predictors of poor outcome. The area under the ROC curve (AUC) was 0.909. The calibration curve and Hosmer-Lemeshow tests showed that the nomogram had good calibration ability. Furthermore, the DCA curve showed better clinical utilization of the nomogram. Conclusion: This study provides a reliable and valuable nomogram that can accurately predict the risk of poor prognosis in patients with poor-grade aSAH after microsurgical clipping. This tool is easy to use and can help physicians make appropriate clinical decisions to significantly improve patient prognosis.

4.
Brain Res Bull ; 193: 72-83, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36535306

ABSTRACT

As a subtype of stroke, subarachnoid hemorrhage (SAH) has a notoriously high rate of disability and mortality owing to the lack of effective intervention. Early brain injury (EBI) is the main factor responsible for the dismal prognosis of SAH patients. The current study intends to explore the molecular mechanism underlying the effect of MH on EBI after SAH from a novel perspective of pyroptosis, a highly specific inflammatory programmed cell death, in the SAH rat model. Sprague-Dawley (SD) rats were divided into different groups in accordance with various treatments. In the treatment group, the rats underwent mild hypothermia for 4 h after modeling; in the inhibitor group, Compound C (an inhibitor of AMPK) was administered intravenous injections (i.v.) 30 min before modeling. Neurological score, neuronal death, brain water content, inflammatory reaction, and expression levels of pyroptosis-related proteins were evaluated in the rats. Our results indicate that the MH therapy significantly increased the neurological score and assuaged brain edema, neuronal injury, and inflammatory reaction induced by SAH. Meanwhile, MH therapy upregulated the level of AMPK phosphorylation whereas downregulated the protein expressions of NLRP3, ASC, cleaved caspase-1, GSDMD, IL-1ß, and IL-18. The reversed effect of MH therapy by Compound C concretely indicated that MH therapy inhibited pyroptosis through an AMPK-dependent pathway. Our study also found that MH therapy potently curbed the increasing trend of brain temperature (BT), rectal temperature (RT), and ICP after SAH. Taken together, our data indicate that the neuroprotective effects of MH therapy were manifested by inhibiting pyroptosis via the AMPK/NLRP3 inflammasome pathway, which may serve as a promising therapy for the intervention of SAH.


Subject(s)
Brain Injuries , Hypothermia , Subarachnoid Hemorrhage , Rats , Animals , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pyroptosis , Rats, Sprague-Dawley , AMP-Activated Protein Kinases/metabolism , Subarachnoid Hemorrhage/metabolism , Brain Injuries/metabolism , Inflammation
5.
ESC Heart Fail ; 10(2): 1422-1425, 2023 04.
Article in English | MEDLINE | ID: mdl-36347824

ABSTRACT

This case study aimed to identify rare viral myocarditis combined with encephalitis as a COVID-19 vaccine complication. A 59-year-old male patient with a 2 day history of headache and rapidly progressive cognitive decline, who had received the third dose of COVID-19 vaccine 12 days before, was admitted to our hospital. The patient had no underlying systemic conditions, no prior medical history, and no prior history of COVID-19 infection. The patient was diagnosed with viral myocarditis and encephalitis by two neurologists and two cardiologists after laboratory examination, head computed tomography, and magnetic resonance imaging evaluation. The patient experienced cardiogenic shock and developed severe arrhythmia, resulting in his death 10 h after admission. Clinically suspected lethal viral myocarditis combined with encephalitis in the patient could be related to COVID-19 vaccination. Adverse effects of the Chinese COVID-19 vaccine, especially serious complications, have been uncommon. This case study highlights a rare complication after COVID-19 vaccination that needs high attention.


Subject(s)
COVID-19 , Encephalitis , Myocarditis , Male , Humans , Middle Aged , COVID-19 Vaccines/adverse effects , COVID-19/complications , COVID-19/diagnosis , Myocarditis/diagnosis , Myocarditis/etiology , Asian People
6.
Front Cell Neurosci ; 16: 1025708, 2022.
Article in English | MEDLINE | ID: mdl-36582214

ABSTRACT

Subarachnoid haemorrhage (SAH) is a common cerebrovascular disease with high disability and mortality rates worldwide. The pathophysiological mechanisms involved in an aneurysm rupture in SAH are complex and can be divided into early brain injury and delayed brain injury. The initial mechanical insult results in brain tissue and vascular disruption with hemorrhages and neuronal necrosis. Following this, the secondary injury results in diffused cerebral damage in the peri-core area. However, the molecular mechanisms of neuronal death following an aneurysmal SAH are complex and currently unclear. Furthermore, multiple cell death pathways are stimulated during the pathogenesis of brain damage. Notably, particular attention should be devoted to necrosis, apoptosis, autophagy, necroptosis, pyroptosis and ferroptosis. Thus, this review discussed the mechanism of neuronal death and its influence on brain injury after SAH.

7.
Nat Commun ; 13(1): 7133, 2022 11 21.
Article in English | MEDLINE | ID: mdl-36414666

ABSTRACT

The rational design of PROTACs is difficult due to their obscure structure-activity relationship. This study introduces a deep neural network model - DeepPROTACs to help design potent PROTACs molecules. It can predict the degradation capacity of a proposed PROTAC molecule based on structures of given target protein and E3 ligase. The experimental dataset is mainly collected from PROTAC-DB and appropriately labeled according to the DC50 and Dmax values. In the model of DeepPROTACs, the ligands as well as the ligand binding pockets are generated and represented with graphs and fed into Graph Convolutional Networks for feature extraction. While SMILES representations of linkers are fed into a Bidirectional Long Short-Term Memory layer to generate the features. Experiments show that DeepPROTACs model achieves 77.95% average prediction accuracy and 0.8470 area under receiver operating characteristic curve on the test set. DeepPROTACs is available online at a web server ( https://bailab.siais.shanghaitech.edu.cn/services/deepprotacs/ ) and at github ( https://github.com/fenglei104/DeepPROTACs ).


Subject(s)
Deep Learning , Neural Networks, Computer , Proteins , Ubiquitin-Protein Ligases/metabolism
8.
Nat Commun ; 13(1): 6632, 2022 Nov 04.
Article in English | MEDLINE | ID: mdl-36333329

ABSTRACT

In this paper, we report a unique type of core-shell crystalline material that combines an inorganic zeolitic cage structure with a macrocyclic host arrangement and that can remove trace levels of iodine from water effectively. These unique assemblies are made up of an inorganic Archimedean truncatedhexahedron (tcu) polyhedron in the kernel which possesses six calixarene-like shell cavities. The cages have good adaptability to guests and can be assembled into a series of supramolecular structures in the crystalline state with different lattice pore shapes. Due to the unique core-shell porous structures, the compounds are not only stable in organic solvents but also in water. The characteristics of the cages enable rapid iodine capture from low concentration aqueous I2/KI solutions (down to 4 ppm concentration). We have studied the detailed process and mechanism of iodine capture and aggregation at the molecular level. The facile synthesis, considerable adsorption capacity, recyclability, and ß- and γ-radiation resistance of the cages should make these materials suitable for the extraction of iodine from aqueous effluent streams (most obviously, radioactive iodide produced by atomic power generation).

9.
Front Pharmacol ; 13: 924826, 2022.
Article in English | MEDLINE | ID: mdl-36059982

ABSTRACT

Subarachnoid hemorrhage (SAH) is a severe type of stroke featuring exceptionally high rate of morbidity and mortality due to the lack of effective management. Ferroptosis can be defined as a novel iron-dependent programmed cell death in contrast to classical apoptosis and necrosis. Astragaloside IV (AS-IV) is an active ingredient extracted from Astragalus membranaceus with established therapeutic effect on CNS diseases. However, the exact role of ferroptosis in Astragaloside IV-mediated neuroprotection after SAH is yet to be demonstrated. In the present study, the SAH model of SD male rats with endovascular perforation was used to gauge the neuroprotective effect of AS-IV on SAH-induced early brain injury (EBI) and to clarify the potential molecular mechanism. We found that the induction of SAH reduced the levels of SLC7A11 and glutathione peroxidase 4 (GPX4) in the brain, exacerbated iron accumulation, enhanced lipid reactive oxygen species (ROS) level, and stimulated neuronal ferroptosis. However, the administration of AS-IV and the ferroptosis inhibitor Ferrostatin-1 (Fer-1) enhanced the antioxidant capacity after SAH and suppressed the accumulation of lipid peroxides. Meanwhile, AS-IV triggered Nrf2/HO-1 signaling pathway and alleviated ferroptosis due to the induction of SAH. The Nrf2 inhibitor ML385 blocked the beneficial effects of neuroprotection. These results consistently suggest that ferroptosis is profoundly implicated in facilitating EBI in SAH, and that AS-IV thwarts the process of ferroptosis in SAH by activating Nrf2/HO-1 pathway.

10.
Brain Res Bull ; 173: 132-140, 2021 08.
Article in English | MEDLINE | ID: mdl-34023434

ABSTRACT

The degree of early brain injury (EBI) is a significant factor that affects the prognosis of patients with subarachnoid hemorrhage (SAH). Evidence has shown that fibroblast growth factor-2 (FGF-2) may alleviate the serious consequences of EBI after SAH. The objective of the current study was to investigate the underlying mechanism that mediates the neuroprotective effects of FGF-2 in the SAH rat model. Sprague-Dawley (SD) rats that underwent different treatments were divided into various groups. FGF-2 was administered intranasally to rats in the treatment group within 30 min after modeling. Rapamycin (an autophagy activator) or LY294002 (a PI3K/Akt pathway inhibitor) was administered intracerebroventricularly (i.c.v.) 30 min before modeling. Neurological scale and brain water content were measured in the brain tissue of the rats. TUNEL staining, Western blot, and immunofluorescence staining were performed to examine and compare the diverse effects of FGF-2 treatment, activated autophagy, and inhibited the PI3K/Akt pathway. We found that FGF-2 treatment effectively reduced the number of TUNEL-positive cells, decreased the brain water content, and improved the neurological function of rats after SAH. Additionally, the expression levels of autophagy-related proteins (LC3 and Beclin-1) were obviously decreased in the FGF-2 treatment group compared with the SAH + vehicle group. The therapeutic effects of FGF-2 in the SAH + FGF-2+rapamycin group were weakened compared with that in the SAH + FGF-2+DMSO group. In the event of the PI3K/Akt pathway inhibition, the expression levels of LC3 and Beclin-1 were enhanced, and the therapeutic effects of FGF-2 were compromised. In summary, our data collectively demonstrated that FGF-2 may suppress autophagy levels to play a neuroprotective role, at least partially by activating the PI3K/Akt pathway. These results highlight FGF-2 as a promising solution to the clinical intervention of SAH.


Subject(s)
Autophagy/drug effects , Fibroblast Growth Factor 2/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Signal Transduction/drug effects , Subarachnoid Hemorrhage/metabolism , Animals , Brain/drug effects , Brain/metabolism , Male , Neurons/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...