Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 8.262
Filter
1.
Cell Commun Signal ; 22(1): 383, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39075489

ABSTRACT

BACKGROUND: Acute hypobaric hypoxia-induced brain injury has been a challenge in the health management of mountaineers; therefore, new neuroprotective agents are urgently required. Meldonium, a well-known cardioprotective drug, has been reported to have neuroprotective effects. However, the relevant mechanisms have not been elucidated. We hypothesized that meldonium may play a potentially novel role in hypobaric hypoxia cerebral injury. METHODS: We initially evaluated the neuroprotection efficacy of meldonium against acute hypoxia in mice and primary hippocampal neurons. The potential molecular targets of meldonium were screened using drug-target binding Huprot™ microarray chip and mass spectrometry analyses after which they were validated with surface plasmon resonance (SPR), molecular docking, and pull-down assay. The functional effects of such binding were explored through gene knockdown and overexpression. RESULTS: The study clearly shows that pretreatment with meldonium rapidly attenuates neuronal pathological damage, cerebral blood flow changes, and mitochondrial damage and its cascade response to oxidative stress injury, thereby improving survival rates in mice brain and primary hippocampal neurons, revealing the remarkable pharmacological efficacy of meldonium in acute high-altitude brain injury. On the one hand, we confirmed that meldonium directly interacts with phosphoglycerate kinase 1 (PGK1) to promote its activity, which improved glycolysis and pyruvate metabolism to promote ATP production. On the other hand, meldonium also ameliorates mitochondrial damage by PGK1 translocating to mitochondria under acute hypoxia to regulate the activity of TNF receptor-associated protein 1 (TRAP1) molecular chaperones. CONCLUSION: These results further explain the mechanism of meldonium as an energy optimizer and provide a strategy for preventing acute hypobaric hypoxia brain injury at high altitudes.


Subject(s)
Brain Injuries , Phosphoglycerate Kinase , Animals , Phosphoglycerate Kinase/metabolism , Phosphoglycerate Kinase/genetics , Mice , Brain Injuries/drug therapy , Brain Injuries/metabolism , Brain Injuries/pathology , Male , Hippocampus/drug effects , Hippocampus/pathology , Hippocampus/metabolism , Hypoxia/drug therapy , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Mice, Inbred C57BL , Oxidative Stress/drug effects , Mitochondria/drug effects , Mitochondria/metabolism
2.
Mol Brain ; 17(1): 47, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39075534

ABSTRACT

In this study, we examined how systemic inflammation affects repair of brain injury. To this end, we created a brain-injury model by stereotaxic injection of ATP, a damage-associated molecular pattern component, into the striatum of mice. Systemic inflammation was induced by intraperitoneal injection of lipopolysaccharide (LPS-ip). An analysis of magnetic resonance images showed that LPS-ip reduced the initial brain injury but slowed injury repair. An immunostaining analysis using the neuronal marker, NeuN, showed that LPS-ip delayed removal of dead/dying neurons, despite the fact that LPS-ip enhanced infiltration of monocytes, which serve to phagocytize dead cells/debris. Notably, infiltrating monocytes showed a widely scattered distribution. Bulk RNAseq analyses showed that LPS-ip decreased expression of genes associated with phagocytosis, with PCR and immunostaining of injured brains confirming reduced levels of Cd68 and Clec7a, markers of phagocytic activity, in monocytes. Collectively, these results suggest that systemic inflammation affects properties of blood monocytes as well as brain cells, resulting in delay in clearing damaged cells and activating repair processes.


Subject(s)
Brain , Inflammation , Lipopolysaccharides , Mice, Inbred C57BL , Monocytes , Phagocytosis , Animals , Phagocytosis/drug effects , Monocytes/metabolism , Inflammation/pathology , Brain/pathology , Male , Lipopolysaccharides/pharmacology , Brain Injuries/pathology , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Neurons/metabolism , Neurons/pathology , Neurons/drug effects , Lectins, C-Type/metabolism , Wound Healing , Mice , Adenosine Triphosphate/metabolism , CD68 Molecule
3.
Discov Med ; 36(186): 1453-1463, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39054716

ABSTRACT

BACKGROUND: Levosimendan (Levo) is a drug commonly used to treat heart failure. Recent studies have suggested that Levo may have neuroprotective effects, but it is still unknown how exactly it contributes to hypoxia-induced brain damage. Thus, the aim of this study was to investigate how Levo affects hypoxia-induced brain damage and to clarify any possible underlying mechanisms. METHODS: One group of rats (Levo group) was pretreated with Levo via oral force-feeding for four weeks. Another group (Ferrostatin-1 (Fer-1) group) was pretreated with intraperitoneal injections of Fer-1 for four weeks. A rat model of chronic hypoxia was created by treating rats with 13% O2 for 14 days in a closed hypoxia chamber. For each group (Control, Model, Levo, Fer-1), we evaluated learning and memory capacity and the morphology and structure of neurons in the rats' brain tissue. Other measurements included tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1ß), and interleukin-6 (IL-6); malondialdehyde (MDA), superoxide dismutase (SOD), and glutathione peroxidase (GSH-Px); Fe2+; apoptosis; cleaved caspase-3, caspase-3; phosphatase and tensin homolog (PTEN), protein kinase B (Akt), phosphorylated Akt (p-Akt); and ferroptosis-related proteins Nuclear factor erythroid 2-related factor 2 (Nrf2), glutathione peroxidase 4 (GPX4), and solute carrier family 7 member 11 (SLC7A11). RESULTS: The Model group rats had considerably fewer neurons than the Control group, with loosely arranged cells, and markedly impaired learning and memory abilities (p < 0.05). Oxidative damage and inflammation in brain tissues of the Model group were significantly intensified, accompanied by a substantial increase in neuronal apoptosis (p < 0.05). PTEN protein, Fe2+ concentration, and cleaved caspase-3 expression were all significantly upregulated, whereas p-Akt, Nrf2, GPX4, and SLC7A11 proteins were dramatically downregulated (p < 0.05). Both the Levo and Fer-1 groups demonstrated significantly more neurons and closely arranged cells than the Model group, along with a notable improvement in learning and memory abilities (p < 0.05). Oxidative damage and inflammation in brain tissues of the Levo and Fer-1 groups were markedly alleviated, and neuronal apoptosis was suppressed (p < 0.05). p-Akt, Nrf2, GPX4, and SLC7A11 proteins were dramatically upregulated, whereas the expression of cleaved caspase-3, PTEN protein, and Fe2+ content was considerably downregulated (p < 0.05). CONCLUSIONS: Levo effectively mitigates brain injury in rats with chronic hypoxia, likely by regulating ferroptosis via the PTEN/Akt signaling pathway.


Subject(s)
Ferroptosis , PTEN Phosphohydrolase , Proto-Oncogene Proteins c-akt , Signal Transduction , Simendan , Animals , PTEN Phosphohydrolase/metabolism , Rats , Ferroptosis/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Male , Simendan/pharmacology , Simendan/therapeutic use , Rats, Sprague-Dawley , Brain Injuries/drug therapy , Brain Injuries/metabolism , Brain Injuries/etiology , Brain Injuries/pathology , Disease Models, Animal , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects , Cyclohexylamines , Phenylenediamines
4.
Cancer Imaging ; 24(1): 95, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39026377

ABSTRACT

BACKGROUND: Radiotherapy is a major therapeutic approach in patients with brain tumors. However, it leads to cognitive impairments. To improve the management of radiation-induced brain sequalae, deformation-based morphometry (DBM) could be relevant. Here, we analyzed the significance of DBM using Jacobian determinants (JD) obtained by non-linear registration of MRI images to detect local vulnerability of healthy cerebral tissue in an animal model of brain irradiation. METHODS: Rats were exposed to fractionated whole-brain irradiation (WBI, 30 Gy). A multiparametric MRI (anatomical, diffusion and vascular) study was conducted longitudinally from 1 month up to 6 months after WBI. From the registration of MRI images, macroscopic changes were analyzed by DBM and microscopic changes at the cellular and vascular levels were evaluated by quantification of cerebral blood volume (CBV) and diffusion metrics including mean diffusivity (MD). Voxel-wise comparisons were performed on the entire brain and in specific brain areas identified by DBM. Immunohistology analyses were undertaken to visualize the vessels and astrocytes. RESULTS: DBM analysis evidenced time-course of local macrostructural changes; some of which were transient and some were long lasting after WBI. DBM revealed two vulnerable brain areas, namely the corpus callosum and the cortex. DBM changes were spatially associated to microstructural alterations as revealed by both diffusion metrics and CBV changes, and confirmed by immunohistology analyses. Finally, matrix correlations demonstrated correlations between JD/MD in the early phase after WBI and JD/CBV in the late phase both in the corpus callosum and the cortex. CONCLUSIONS: Brain irradiation induces local macrostructural changes detected by DBM which could be relevant to identify brain structures prone to radiation-induced tissue changes. The translation of these data in patients could represent an added value in imaging studies on brain radiotoxicity.


Subject(s)
Brain Injuries , Animals , Rats , Male , Brain Injuries/etiology , Brain Injuries/diagnostic imaging , Brain Injuries/pathology , Brain Neoplasms/radiotherapy , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/pathology , Radiation Injuries/diagnostic imaging , Radiation Injuries/pathology , Radiation Injuries/etiology , Brain/radiation effects , Brain/diagnostic imaging , Brain/pathology , Magnetic Resonance Imaging/methods , Radiation Injuries, Experimental/diagnostic imaging , Radiation Injuries, Experimental/pathology , Radiation Injuries, Experimental/etiology , Multiparametric Magnetic Resonance Imaging/methods
5.
J Biochem Mol Toxicol ; 38(7): e23760, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38953502

ABSTRACT

Cyclophosphamide (CP) is an antineoplastic drug widely used in chemotherapy. Curcumin (CUR) and piperine (PP) show a protective effect on neurodegenerative and neurological diseases. This research was designed to measure several biochemical parameters in the brain tissue of CP-applied rats to investigate the impact of combined CUR-PP administration. The study evaluated six groups of eight rats: Group 1 was the control; Groups 2 and 3 were administered 200 or 300 mg/kg CUR-PP via oral gavage; Group 4 received only 200 mg/kg CP on day 1; Groups 5 and 6 received CP + CUR-PP for 7 days. Data from all parameters indicated that CP caused brain damage. Phosphorylated TAU (pTAU), amyloid-beta peptide 1-42 (Aß1-42), glutamate (GLU), and gamma amino butyric acid (GABA) parameters were the same in Groups 4, 5, and 6. On the other hand, 8-hydroxy-2-deoxyguanosine (8-OHdG), nitric oxide (NO), interleukin-6 (IL-6), nuclear factor kappa beta (NF-kß), malondialdehyde (MDA), and tumor necrosis factor-alpha (TNF-α) levels in the CP + CUR-PP groups were lower than those in the CP group (p < 0.05). However, superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), and reduced glutathione (GSH) parameters were higher in the CP + CUR-PP groups compared to the CP group (p < 0.05). It is thought that the similarity of Groups 5 and 6 with Group 4 in Aß1-42, pTAU, GLU, and GABA parameters hinder the determination of treatment protection however, they might have a therapeutic effect if the applied dose or study duration were changed. This study attempted to evaluate the effects of a CUR-PP combination on CP-induced brain damage in rats by measuring biochemical parameters and performing histopathological examinations. Based on the findings, this CUR-PP combination could be considered an alternative medicine option in cases with conditions similar to those evaluated in this study.


Subject(s)
Alkaloids , Benzodioxoles , Brain Injuries , Curcumin , Cyclophosphamide , Piperidines , Polyunsaturated Alkamides , Animals , Polyunsaturated Alkamides/pharmacology , Benzodioxoles/pharmacology , Curcumin/pharmacology , Piperidines/pharmacology , Alkaloids/pharmacology , Rats , Cyclophosphamide/toxicity , Cyclophosphamide/adverse effects , Male , Brain Injuries/chemically induced , Brain Injuries/drug therapy , Brain Injuries/metabolism , Brain Injuries/pathology , Brain Injuries/prevention & control , Rats, Wistar , Brain/metabolism , Brain/drug effects , Brain/pathology , Oxidative Stress/drug effects , Neuroprotective Agents/pharmacology
7.
Cells ; 13(11)2024 May 28.
Article in English | MEDLINE | ID: mdl-38891060

ABSTRACT

Mitochondrial aldehyde dehydrogenase-2 (ALDH2) metabolizes acetaldehyde to acetate. People with ALDH2 deficiency and Aldh2-knockout (KO) mice are more susceptible to alcohol-induced tissue damage. However, the underlying mechanisms behind ALDH2-related gut-associated brain damage remain unclear. Age-matched young female Aldh2-KO and C57BL/6J wild-type (WT) mice were gavaged with binge alcohol (4 g/kg/dose, three doses) or dextrose (control) at 12 h intervals. Tissues and sera were collected 1 h after the last ethanol dose and evaluated by histological and biochemical analyses of the gut and hippocampus and their extracts. For the mechanistic study, mouse neuroblast Neuro2A cells were exposed to ethanol with or without an Aldh2 inhibitor (Daidzin). Binge alcohol decreased intestinal tight/adherens junction proteins but increased oxidative stress-mediated post-translational modifications (PTMs) and enterocyte apoptosis, leading to elevated gut leakiness and endotoxemia in Aldh2-KO mice compared to corresponding WT mice. Alcohol-exposed Aldh2-KO mice also showed higher levels of hippocampal brain injury, oxidative stress-related PTMs, and neuronal apoptosis than the WT mice. Additionally, alcohol exposure reduced Neuro2A cell viability with elevated oxidative stress-related PTMs and apoptosis, all of which were exacerbated by Aldh2 inhibition. Our results show for the first time that ALDH2 plays a protective role in binge alcohol-induced brain injury partly through the gut-brain axis, suggesting that ALDH2 is a potential target for attenuating alcohol-induced tissue injury.


Subject(s)
Aldehyde Dehydrogenase, Mitochondrial , Binge Drinking , Brain Injuries , Gastrointestinal Tract , Animals , Female , Mice , Aldehyde Dehydrogenase, Mitochondrial/metabolism , Aldehyde Dehydrogenase, Mitochondrial/genetics , Apoptosis/drug effects , Binge Drinking/pathology , Brain Injuries/pathology , Brain Injuries/metabolism , Ethanol/pharmacology , Ethanol/toxicity , Hippocampus/pathology , Hippocampus/drug effects , Hippocampus/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Mitochondria/drug effects , Oxidative Stress/drug effects , Gastrointestinal Tract/injuries , Gastrointestinal Tract/metabolism
8.
Biomaterials ; 311: 122678, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38917705

ABSTRACT

Drug transmission through the blood-brain barrier (BBB) is considered an arduous challenge for brain injury treatment following the return of spontaneous circulation after cardiac arrest (CA-ROSC). Inspired by the propensity of melanoma metastasis to the brain, B16F10 cell membranes are camouflaged on 2-methoxyestradiol (2ME2)-loaded reactive oxygen species (ROS)-triggered "Padlock" nanoparticles that are constructed by phenylboronic acid pinacol esters conjugated D-a-tocopheryl polyethylene glycol succinate (TPGS-PBAP). The biomimetic nanoparticles (BM@TP/2ME2) can be internalized, mainly mediated by the mutual recognition and interaction between CD44v6 expressed on B16F10 cell membranes and hyaluronic acid on cerebral vascular endothelial cells, and they responsively release 2ME2 by the oxidative stress microenvironment. Notably, BM@TP/2ME2 can scavenge excessive ROS to reestablish redox balance, reverse neuroinflammation, and restore autophagic flux in damaged neurons, eventually exerting a remarkable neuroprotective effect after CA-ROSC in vitro and in vivo. This biomimetic drug delivery system is a novel and promising strategy for the treatment of cerebral ischemia-reperfusion injury after CA-ROSC.


Subject(s)
2-Methoxyestradiol , Heart Arrest , Nanoparticles , Reactive Oxygen Species , Animals , Reactive Oxygen Species/metabolism , Nanoparticles/chemistry , Mice , 2-Methoxyestradiol/pharmacology , 2-Methoxyestradiol/chemistry , Heart Arrest/drug therapy , Brain Injuries/drug therapy , Brain Injuries/metabolism , Brain Injuries/pathology , Male , Mice, Inbred C57BL , Drug Delivery Systems , Cell Line, Tumor , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Oxidative Stress/drug effects
9.
J Neurosci ; 44(32)2024 Aug 07.
Article in English | MEDLINE | ID: mdl-38844342

ABSTRACT

Sleep slow waves are the hallmark of deeper non-rapid eye movement sleep. It is generally assumed that gray matter properties predict slow-wave density, morphology, and spectral power in healthy adults. Here, we tested the association between gray matter volume (GMV) and slow-wave characteristics in 27 patients with moderate-to-severe traumatic brain injury (TBI, 32.0 ± 12.2 years old, eight women) and compared that with 32 healthy controls (29.2 ± 11.5 years old, nine women). Participants underwent overnight polysomnography and cerebral MRI with a 3 Tesla scanner. A whole-brain voxel-wise analysis was performed to compare GMV between groups. Slow-wave density, morphology, and spectral power (0.4-6 Hz) were computed, and GMV was extracted from the thalamus, cingulate, insula, precuneus, and orbitofrontal cortex to test the relationship between slow waves and gray matter in regions implicated in the generation and/or propagation of slow waves. Compared with controls, TBI patients had significantly lower frontal and temporal GMV and exhibited a subtle decrease in slow-wave frequency. Moreover, higher GMV in the orbitofrontal cortex, insula, cingulate cortex, and precuneus was associated with higher slow-wave frequency and slope, but only in healthy controls. Higher orbitofrontal GMV was also associated with higher slow-wave density in healthy participants. While we observed the expected associations between GMV and slow-wave characteristics in healthy controls, no such associations were observed in the TBI group despite lower GMV. This finding challenges the presumed role of GMV in slow-wave generation and morphology.


Subject(s)
Brain Injuries, Traumatic , Gray Matter , Magnetic Resonance Imaging , Sleep, Slow-Wave , Humans , Female , Male , Gray Matter/diagnostic imaging , Gray Matter/pathology , Gray Matter/physiopathology , Adult , Sleep, Slow-Wave/physiology , Brain Injuries, Traumatic/physiopathology , Brain Injuries, Traumatic/diagnostic imaging , Brain Injuries, Traumatic/pathology , Young Adult , Polysomnography , Cerebral Cortex/physiopathology , Cerebral Cortex/diagnostic imaging , Middle Aged , Brain Injuries/physiopathology , Brain Injuries/diagnostic imaging , Brain Injuries/pathology
10.
Eur J Pharmacol ; 978: 176765, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-38906236

ABSTRACT

Subarachnoid hemorrhage (SAH) is a neurological condition with high mortality and poor prognosis, and there are currently no effective therapeutic drugs available. Poly (ADP-ribose) polymerase 1 (PARP-1) dependent cell death pathway-parthanatos is closely associated with stroke. We investigated improvements in neurological function, oxidative stress, blood-brain barrier and parthanatos-related protein expression in rats with SAH after intraperitoneal administration of PARP-1 inhibitor (AG14361). Our study found that the expression of parthanatos-related proteins was significantly increased after SAH. Immunofluorescence staining showed increased expression of apoptosis-inducing factor (AIF) in the nucleus after SAH. Administration of PARP-1 inhibitor significantly reduced malondialdehyde (MDA) level and the expression of parthanatos-related proteins. Immunofluorescence staining showed that PARP-1 inhibitor reduced the expression of 8-hydroxy-2' -deoxyguanosine (8-OHdG) and thus reduced oxidative stress. Moreover, PARP-1 inhibitor could inhibit inflammation-associated proteins level and neuronal apoptosis, protect the blood-brain barrier and significantly improve neurological function after SAH. These results suggest that PARP-1 inhibitor can significantly improve SAH, and the underlying mechanism may be through inhibiting parthanatos pathway.


Subject(s)
Blood-Brain Barrier , Brain Injuries , Cell Death , Parthanatos , Poly (ADP-Ribose) Polymerase-1 , Subarachnoid Hemorrhage , Animals , Male , Rats , Apoptosis Inducing Factor/metabolism , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Brain Injuries/metabolism , Brain Injuries/etiology , Brain Injuries/drug therapy , Brain Injuries/pathology , Cell Death/drug effects , Oxidative Stress/drug effects , Parthanatos/drug effects , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Rats, Sprague-Dawley , Subarachnoid Hemorrhage/metabolism , Subarachnoid Hemorrhage/complications , Subarachnoid Hemorrhage/drug therapy , Subarachnoid Hemorrhage/pathology
11.
Biomed Pharmacother ; 177: 116978, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38906028

ABSTRACT

Radiation-induced brain injury (RIBI) is a significant challenge in radiotherapy for head and neck tumors, impacting patients' quality of life. In exploring potential treatments, this study focuses on memantine hydrochloride and hydrogen-rich water, hypothesized to mitigate RIBI through inhibiting the NLRP3/NLRC4/Caspase-1 pathway. In a controlled study involving 40 Sprague-Dawley rats, divided into five groups including a control and various treatment groups, we assessed the effects of these treatments on RIBI. Post-irradiation, all irradiated groups displayed symptoms like weight loss and salivation, with notable variations among different treatment approaches. Particularly, hydrogen-rich water showed a promising reduction in these symptoms. Histopathological analysis indicated substantial hippocampal damage in the radiation-only group, while the groups receiving memantine and/or hydrogen-rich water exhibited significant mitigation of such damage. Molecular studies, revealed a decrease in oxidative stress markers and an attenuated inflammatory response in the treatment groups. Immunohistochemistry further confirmed these molecular changes, suggesting the effectiveness of these agents. Echoing recent scientific inquiries into the protective roles of specific compounds against radiation-induced damages, our study adds to the growing body of evidence on the potential of memantine and hydrogen-rich water as novel therapeutic strategies for RIBI.


Subject(s)
Caspase 1 , Hydrogen , Memantine , NLR Family, Pyrin Domain-Containing 3 Protein , Pyroptosis , Rats, Sprague-Dawley , Water , Animals , Memantine/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Hydrogen/pharmacology , Pyroptosis/drug effects , Rats , Caspase 1/metabolism , Male , Signal Transduction/drug effects , Brain Injuries/etiology , Brain Injuries/metabolism , Brain Injuries/drug therapy , Brain Injuries/prevention & control , Brain Injuries/pathology , Radiation Injuries/drug therapy , Radiation Injuries/metabolism , Radiation Injuries/pathology , Oxidative Stress/drug effects , Radiation Injuries, Experimental/drug therapy , Radiation Injuries, Experimental/metabolism , Radiation Injuries, Experimental/pathology , Radiation Injuries, Experimental/prevention & control
12.
Neuropathol Appl Neurobiol ; 50(3): e12992, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38831600

ABSTRACT

PURPOSE: Radiation-induced brain injury, one of the side effects of cranial radiotherapy in tumour patients, usually results in durable and serious cognitive disorders. Microglia are important innate immune-effector cells in the central nervous system. However, the interaction between microglia and neurons in radiation-induced brain injury remains uncharacterised. METHODS AND MATERIALS: We established a microglia-neuron indirect co-culture model to assess the interaction between them. Microglia exposed to radiation were examined for pyroptosis using lactate dehydrogenase (LDH) release, Annexin V/PI staining, SYTOX staining and western blot. The role of nucleotide-binding oligomerisation domain-like receptor family pyrin domain containing 3 (NLRP3) was investigated in microglia exposed to radiation and in mouse radiation brain injury model through siRNA or inhibitor. Mini-mental state examination and cytokines in blood were performed in 23 patients who had experienced cranial irradiation. RESULTS: Microglia exerted neurotoxic features after radiation in the co-culture model. NLRP3 was up-regulated in microglia exposed to radiation, and then caspase-1 was activated. Thus, the gasdermin D protein was cleaved, and it triggered pyroptosis in microglia, which released inflammatory cytokines. Meanwhile, treatment with siRNA NLRP3 in vitro and NLRP3 inhibitor in vivo attenuated the damaged neuron cell and cognitive impairment, respectively. What is more, we found that the patients after radiation with higher IL-6 were observed to have a decreased MMSE score. CONCLUSIONS: These findings indicate that radiation-induced pyroptosis in microglia may promote radiation-induced brain injury via the secretion of neurotoxic cytokines. NLRP3 was evaluated as an important mediator in radiation-induced pyroptosis and a promising therapeutic target for radiation-induced brain injury.


Subject(s)
Brain Injuries , Microglia , NLR Family, Pyrin Domain-Containing 3 Protein , Pyroptosis , Pyroptosis/radiation effects , Pyroptosis/physiology , Microglia/metabolism , Microglia/radiation effects , Microglia/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Mice , Humans , Brain Injuries/metabolism , Brain Injuries/pathology , Brain Injuries/etiology , Male , Neurons/metabolism , Neurons/pathology , Neurons/radiation effects , Coculture Techniques , Radiation Injuries/pathology , Radiation Injuries/metabolism , Female , Mice, Inbred C57BL , Middle Aged
13.
Bratisl Lek Listy ; 125(7): 414-418, 2024.
Article in English | MEDLINE | ID: mdl-38943501

ABSTRACT

OBJECTIVE: Astrocytes undergo morphological and molecular changes in response to numerous pathological conditions. BACKROUND: Increased expression of glial fibrillary acidic protein (GFAP) has been reported as a characteristic feature of reactive astrocytes. However, GFAP-positive cells occur rarely in adult human brain cultures. These cultures are mostly composed of flat GFAP-negative "glia-like" cells, which remain poorly characterized in relation to reactive astrogliosis. METHODS: We examined the cultures from macroscopically injured and normal brain tissue from patients with brain trauma, gliomas, or brain metastases. Immunofluorescence and immunohistochemical methods were used for reactive astrocytes detection. RESULTS: The intensity of GFAP-positive staining was higher in reactive astrocytes in the brain tissue surrounding gliomas or metastases and lower in brain tissue damaged by traumatic injury. We did not observe any correlation between GFAP-positive reactive astrocytes in cultures and brain tissue. However, we found rapidly proliferating spindle-shaped cells in cultures prepared from injured brain tissue. CONCLUSION: Present data demonstrate the unexplained phenomenon of disparate cell morphologies in cultures when prepared either from macroscopically normal or injured human brain tissue. While normal cultures are mainly comprised of flat cells, the cultures from severely damaged brain tissue may be entirely composed of spindle-shaped cells usually classified as fibroblasts. We suggest that this spindle-shaped cellular morphology is not specific for fibroblasts, but it rather can be interpreted as the most favorable shape for rapid cell proliferation under culture conditions. After brain trauma, unknown processes may be triggered, such as induced cell proliferation which can be revealed under culture condition. Accordingly, we conclude that spindle-shaped cells are activated precursors of glial cells (Fig. 3, Ref. 15).


Subject(s)
Astrocytes , Fibroblasts , Glial Fibrillary Acidic Protein , Humans , Fibroblasts/pathology , Fibroblasts/metabolism , Glial Fibrillary Acidic Protein/metabolism , Astrocytes/pathology , Astrocytes/metabolism , Brain Injuries/pathology , Brain Injuries/metabolism , Female , Middle Aged , Male , Adult , Cells, Cultured , Aged , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Brain/pathology , Brain/cytology , Glioma/pathology , Glioma/metabolism , Neuroglia/pathology , Neuroglia/metabolism
14.
Methods Cell Biol ; 188: 237-254, 2024.
Article in English | MEDLINE | ID: mdl-38880526

ABSTRACT

The prevalence of central nervous system (CNS) dysfunction as a result of disease or trauma remains a clinically unsolved problem which is raising increased awareness in our aging society. Human Dental Pulp Stem Cells (hDPSCs) are excellent candidates to be used in tissue engineering and regenerative therapies of the CNS due to their neural differentiation ability and lack of tumorigenicity. Accordingly, they have been successfully used in animal models of spinal cord injury, stroke and peripheral neuropathies. The ideal therapy in brain injury should combine strategies aiming to protect the damaged lesion and, at the same time, accelerate brain tissue regeneration, thus promoting fast recovery while minimizing side or long-term effects. The use of bioresorbable nanopatterned poly(lactide-co-ɛ-caprolactone) (PLCL) polymeric scaffolds as hDPCSs carriers can represent an advantage for tissue regeneration. In this chapter, we describe the surgical procedures to implant functionalized bioresorbable scaffolds loaded with hDPSCs to improve the brain lesion microenvironment in an intracranial stab wound injury model severing the rostral migratory stream (RMS) that connects the brain subventricular zone (SVZ) and the olfactory bulb in nude mice. Additionally, we also describe the technical steps after animal sacrifice for histological tissue observation and characterization.


Subject(s)
Dental Pulp , Disease Models, Animal , Mice, Nude , Stem Cells , Tissue Scaffolds , Dental Pulp/cytology , Animals , Humans , Tissue Scaffolds/chemistry , Mice , Stem Cells/cytology , Stem Cell Transplantation/methods , Wounds, Stab/therapy , Absorbable Implants , Brain Injuries/therapy , Brain Injuries/pathology , Tissue Engineering/methods
15.
J Cell Mol Med ; 28(11): e18366, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38856956

ABSTRACT

Ischemic stroke is one of the main causes of disability and death. However, recanalization of occluded cerebral arteries is effective only within a very narrow time window. Therefore, it is particularly important to find neuroprotective biological targets for cerebral artery recanalization. Here, gene expression profiles of datasets GSE160500 and GSE97537 were downloaded from the GEO database, which were related to ischemic stroke in rats. Olfactory receptor 78 (Olfr78) was screened, and which highly associated with Calcium signalling pathway and MAPK pathway. Interacting protein of Olfr78, Prkaca, was predicted by STRING, and their interaction was validated by Co-IP analysis. Then, a rat model of middle cerebral artery occlusion/reperfusion (MCAO/R) and a neuronal cell model stimulated by oxygen-glucose deprivation/reoxygenation (OGD/R) were constructed, and the results showed that expression of Olfr78 and Prkaca was downregulated in MCAO rats and OGD/R-stimulated neurons. Overexpression of Olfr78 or Prkaca inhibited the secretion of inflammatory factors, Ca2+ overload, and OGD/R-induced neuronal apoptosis. Moreover, Overexpression of Prkaca increased protein levels of cAMP, PKA and phosphorylated p38 in OGD/R-stimulated neurons, while SB203580, a p38 inhibitor, treatment inhibited activation of the cAMP/PKA-MAPK pathway and counteracted the effect of Olfr78 overexpression on improvement of neuronal functions. Meanwhile, overexpression of Olfr78 or Prkaca markedly inhibited neuronal apoptosis and improved brain injury in MCAO/R rats. In conclusion, overexpression of Olfr78 inhibited Ca2+ overload and reduced neuronal apoptosis in MCAO/R rats by promoting Prkaca-mediated activation of the cAMP/PKA-MAPK pathway, thereby improving brain injury in cerebral ischaemia-reperfusion.


Subject(s)
Apoptosis , Cyclic AMP , Rats, Sprague-Dawley , Receptors, Odorant , Reperfusion Injury , Animals , Reperfusion Injury/metabolism , Reperfusion Injury/genetics , Rats , Male , Cyclic AMP/metabolism , Receptors, Odorant/metabolism , Receptors, Odorant/genetics , Brain Ischemia/metabolism , Brain Ischemia/genetics , Brain Ischemia/pathology , MAP Kinase Signaling System/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Brain Injuries/metabolism , Brain Injuries/etiology , Brain Injuries/pathology , Neurons/metabolism , Disease Models, Animal , Infarction, Middle Cerebral Artery/metabolism , Signal Transduction
16.
Int J Biol Macromol ; 273(Pt 1): 132887, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38851621

ABSTRACT

Acute alcohol intoxication is a harmful clinical condition characterized by behavioral and neurological symptoms, for which few effective therapies are available at present. Dysfunction of microglial BV-2 cells has been reported to be associated with acute alcohol-induced brain injuries. In the present study, the protective effects of Eucommia ulmoides Oliv. leaves polysaccharides (EULP) on acute alcoholic brain injury and microglial dysfunction were investigated. 14-day pretreatment of EULP significantly attenuated neurobehavioral deficit and neurotransmitter damage in the brain tissue of mice caused by acute alcohol exposure. Additionally, EULP regulated the metabolic disorder of brain tissue. Consistently, it was shown that EULP pretreatment significantly improved alcohol-induced phagocytosis decrease, oxidative stress and inflammation in BV-2 cells. Therefore, EULP may be proposed and employed as a potential therapeutic agent for alcohol-induced brain damage.


Subject(s)
Eucommiaceae , Microglia , Oxidative Stress , Plant Leaves , Polysaccharides , Animals , Polysaccharides/pharmacology , Polysaccharides/chemistry , Microglia/drug effects , Microglia/metabolism , Mice , Eucommiaceae/chemistry , Plant Leaves/chemistry , Oxidative Stress/drug effects , Male , Ethanol , Brain Injuries/drug therapy , Brain Injuries/chemically induced , Brain Injuries/metabolism , Brain Injuries/etiology , Brain Injuries/pathology , Cell Line , Phagocytosis/drug effects , Brain/drug effects , Brain/metabolism , Brain/pathology , Plant Extracts/pharmacology , Plant Extracts/chemistry
17.
Nat Neurosci ; 27(8): 1522-1533, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38862791

ABSTRACT

Injuries to the brain result in tunable cell responses paired with stimulus properties, suggesting the existence of intrinsic processes that encode and transmit injury information; however, the molecular mechanism of injury information encoding is unclear. Here, using ATP fluorescent indicators, we identify injury-evoked spatiotemporally selective ATP dynamics, Inflares, in adult mice of both sexes. Inflares are actively released from astrocytes and act as the internal representations of injury. Inflares encode injury intensity and position at their population level through frequency changes and are further decoded by microglia, driving changes in their activation state. Mismatches between Inflares and injury severity lead to microglia dysfunction and worsening of injury outcome. Blocking Inflares in ischemic stroke in mice reduces secondary damage and improves recovery of function. Our results suggest that astrocytic ATP dynamics encode injury information and are sensed by microglia.


Subject(s)
Adenosine Triphosphate , Astrocytes , Brain Injuries , Microglia , Animals , Microglia/metabolism , Astrocytes/metabolism , Adenosine Triphosphate/metabolism , Mice , Brain Injuries/metabolism , Brain Injuries/pathology , Male , Female , Mice, Inbred C57BL
18.
J Reprod Immunol ; 164: 104260, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38761507

ABSTRACT

INTRODUCTION: Recent studies have suggested the involvement of ferroptosis in preterm birth. Despite compelling evidence, the underlying mechanism remains unknown. This investigation aimed to determine the therapeutic effects of Ferrostatin-1 (Fer-1), an inhibitor of ferroptosis, in preterm birth and fetal brain injury. METHODS: Human placenta samples and clinical data of participants were collected to ascertain whether placental ferroptosis was associated with preterm birth. Lipopolysaccharide (LPS)-induced preterm birth mouse model was used to examine the protective effects of Fer-1 on preterm birth. Fetal brain tissues and offspring mice at 5 and 8 weeks were studied to determine the effects of Fer-1 on the cognitive function of offspring. RESULTS: We examined the mechanism of spontaneous preterm birth and discovered that placental ferroptosis was associated with preterm birth. Fer-1 inhibited preterm birth by ameliorating placental ferroptosis and maternal inflammation, thus improving LPS-induced intrauterine inflammation to maintain pregnancy. Antenatal administration of Fer-1 prevented LPS-induced fetal brain damage in the acute phase and improved long-term neurodevelopmental impairments by improving placental neuroendocrine signaling and maintaining placental function. CONCLUSION: Fer-1 inhibited preterm birth and fetal brain injury by inhibiting maternal inflammation and improving placental function. Our findings provide a novel therapeutic strategy for preterm birth.


Subject(s)
Brain Injuries , Cyclohexylamines , Ferroptosis , Lipopolysaccharides , Phenylenediamines , Placenta , Premature Birth , Animals , Female , Pregnancy , Mice , Premature Birth/immunology , Premature Birth/prevention & control , Humans , Cyclohexylamines/pharmacology , Cyclohexylamines/administration & dosage , Placenta/pathology , Placenta/immunology , Placenta/drug effects , Brain Injuries/prevention & control , Brain Injuries/immunology , Brain Injuries/etiology , Brain Injuries/drug therapy , Brain Injuries/pathology , Phenylenediamines/pharmacology , Phenylenediamines/administration & dosage , Lipopolysaccharides/immunology , Ferroptosis/drug effects , Inflammation/immunology , Inflammation/drug therapy , Disease Models, Animal , Mice, Inbred C57BL
19.
J Neuroinflammation ; 21(1): 116, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702778

ABSTRACT

BACKGROUND: Subarachnoid hemorrhage (SAH), a severe subtype of stroke, is characterized by notably high mortality and morbidity, largely due to the lack of effective therapeutic options. Although the neuroprotective potential of PPARg and Nrf2 has been recognized, investigative efforts into oroxin A (OA), remain limited in preclinical studies. METHODS: SAH was modeled in vivo through filament perforation in male C57BL/6 mice and in vitro by exposing HT22 cells to hemin to induce neuronal damage. Following the administration of OA, a series of methods were employed to assess neurological behaviors, brain water content, neuronal damage, cell ferroptosis, and the extent of neuroinflammation. RESULTS: The findings indicated that OA treatment markedly improved survival rates, enhanced neurological functions, mitigated neuronal death and brain edema, and attenuated the inflammatory response. These effects of OA were linked to the suppression of microglial activation. Moreover, OA administration was found to diminish ferroptosis in neuronal cells, a critical factor in early brain injury (EBI) following SAH. Further mechanistic investigations uncovered that OA facilitated the translocation of nuclear factor erythroid 2-related factor 2 (Nrf-2) from the cytoplasm to the nucleus, thereby activating the Nrf2/GPX4 pathway. Importantly, OA also upregulated the expression of FSP1, suggesting a significant and parallel protective effect against ferroptosis in EBI following SAH in synergy with GPX4. CONCLUSION: In summary, this research indicated that the PPARg activator OA augmented the neurological results in rodent models and diminished neuronal death. This neuroprotection was achieved primarily by suppressing neuronal ferroptosis. The underlying mechanism was associated with the alleviation of cellular death through the Nrf2/GPX4 and FSP1/CoQ10 pathways.


Subject(s)
Ferroptosis , Mice, Inbred C57BL , Neuroinflammatory Diseases , Subarachnoid Hemorrhage , Animals , Subarachnoid Hemorrhage/metabolism , Subarachnoid Hemorrhage/pathology , Subarachnoid Hemorrhage/complications , Ferroptosis/drug effects , Ferroptosis/physiology , Mice , Male , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/etiology , Brain Injuries/metabolism , Brain Injuries/pathology , Brain Injuries/drug therapy , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Neurons/metabolism , Neurons/drug effects , Neurons/pathology
20.
Proc Natl Acad Sci U S A ; 121(22): e2400648121, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38781210

ABSTRACT

After central nervous system injury, a rapid cellular and molecular response is induced. This response can be both beneficial and detrimental to neuronal survival in the first few days and increases the risk for neurodegeneration if persistent. Semaphorin4B (Sema4B), a transmembrane protein primarily expressed by cortical astrocytes, has been shown to play a role in neuronal cell death following injury. Our study shows that after cortical stab wound injury, cytokine expression is attenuated in Sema4B-/- mice, and microglia/macrophage reactivity is altered. In vitro, Sema4B enhances the reactivity of microglia following injury, suggesting astrocytic Sema4B functions as a ligand. Moreover, injury-induced microglia reactivity is attenuated in the presence of Sema4B-/- astrocytes compared to Sema4B+/- astrocytes. In vitro experiments indicate that Plexin-B2 is the Sema4B receptor on microglia. Consistent with this, in microglia/macrophage-specific Plexin-B2-/- mice, similar to Sema4B-/- mice, microglial/macrophage reactivity and neuronal cell death are attenuated after cortical injury. Finally, in Sema4B/Plexin-B2 double heterozygous mice, microglial/macrophage reactivity is also reduced after injury, supporting the idea that both Sema4B and Plexin-B2 are part of the same signaling pathway. Taken together, we propose a model in which following injury, astrocytic Sema4B enhances the response of microglia/macrophages via Plexin-B2, leading to increased reactivity.


Subject(s)
Astrocytes , Mice, Knockout , Microglia , Nerve Tissue Proteins , Semaphorins , Animals , Mice , Astrocytes/metabolism , Brain Injuries/metabolism , Brain Injuries/pathology , Brain Injuries/genetics , Cell Communication , Macrophages/metabolism , Mice, Inbred C57BL , Microglia/metabolism , Microglia/pathology , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Semaphorins/metabolism , Semaphorins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL