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










Publication year range
1.
Neuromolecular Med ; 26(1): 8, 2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38546874

ABSTRACT

This study focuses on understanding the role of c-Myc, a cancer-associated transcription factor, in the penumbra following ischemic stroke. While its involvement in cell death and survival is recognized, its post-translational modifications, particularly acetylation, remain understudied in ischemia models. Investigating these modifications could have significant clinical implications for controlling c-Myc activity in the central nervous system. Although previous studies on c-Myc acetylation have been limited to non-neuronal cells, our research examines its expression in perifocal cells during stroke recovery to explore regulatory mechanisms via acetylation. We found that in peri-infarct neurons, c-Myc is upregulated with acetylation at K148 but not K323 during the acute phase of stroke, with SIRT2 deacetylase primarily affecting K148 acetylation. Molecular dynamics simulations suggest that lysine 148 plays a crucial role in stabilizing c-Myc spatial structure. Increased acetylation at K148 reduces c-Myc compaction, potentially limiting its nuclear penetration, promoting calpain-mediated cleavage, and decreasing nuclear localization. Additionally, cytoplasmic acetylation at K148 may alter c-Myc's interaction with unidentified proteins, potentially influencing its pro-apoptotic effects and promoting cytoplasmic accumulation. Targeting SIRT2 with selective inhibitors could be a promising avenue for future stroke therapy strategies.


Subject(s)
Sirtuin 2 , Stroke , Humans , Lysine/metabolism , Acetylation , Protein Processing, Post-Translational , Stroke/metabolism , Ischemia , Neurons/metabolism , Proto-Oncogene Proteins c-myc/metabolism
2.
Biophys Rev ; 15(5): 1279-1286, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37974996

ABSTRACT

The health significance of transient ischemic attacks (TIAs) is largely underestimated. Often, TIAs are not given significant importance, and in vain, because TIAs are a predictor of the development of serious cardiovascular diseases and even death. Because of this, and because of the difficulty in diagnosing the disease, TIAs and related microinfarcts are poorly investigated. Photothrombotic models of stroke and TIA allow reproducing the occlusion of small brain vessels, even single ones. When dosing the concentration of photosensitizer, intensity and irradiation time, it is possible to achieve occlusion of well-defined small vessels with high reproducibility, and with the help of modern methods of blood flow assessment it is possible to achieve spontaneous restoration of blood flow without vessel rupture. In this review, we discuss the features of microinfarcts and the contemporary experimental approaches used to model TIA and microinfarcts, with an emphasis on models using the principle of photothrombosis of brain vessels. We review modern techniques for in vivo detection of blood flow in small brain vessels, as well as biomarkers of microinfarcts.

3.
Transl Stroke Res ; 2023 Aug 15.
Article in English | MEDLINE | ID: mdl-37580538

ABSTRACT

p53 expression and acetylation are crucial for the survival and death of neurons in penumbra. At the same time, the outcome of ischemia for penumbra cells depends largely on the histone acetylation status, but the effect of histone acetyltransferases and deacetylases on non-histone proteins like p53 is largely understudied. With combined in silico and in vitro approach, we have identified enzymes capable of acetylation/deacetylation, distribution, stability, and pro-apoptotic activity of p53 in ischemic penumbra in the course of post-stroke recovery, and also detected involved loci of acetylation in p53. The dynamic regulation of the acetylation of p53 at lysine 320 is controlled by acetyltransferase PCAF and histone deacetylases HDAC1 and HDAC6. The in silico simulation have made it possible to suggest the acetylation of p53 at lysine 320 acetylation may facilitate the shuttling of p53 between the nucleus and cytoplasm in penumbra neurons. Acetylation of p53 at lysine 320 is more preferable than acetylation at lysine 373 and probably promotes survival and repair of penumbra neurons after stroke. Strategies to increase p53 acetylation at lysine 320 via increasing PCAF activity, inhibiting HDAC1 or HDAC6, inhibiting p53, or a combination of these interventions may have therapeutic benefits for stroke recovery and would be promising for neuroprotective therapy of stroke.

4.
Mol Neurobiol ; 57(7): 3219-3227, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32506381

ABSTRACT

Stroke is one of the leading reasons of human death. Ischemic penumbra that surrounds the stroke-induced infarction core is potentially salvageable, but molecular mechanisms of its formation are poorly known. Histone acetylation induces chromatin decondensation and stimulates gene expression. We studied the changes in the levels and localization of histone acetyltransferases HAT1 and PCAF in penumbra after photothrombotic stroke (PTS, a stroke model). In PTS, laser irradiation induces local occlusion of cerebral vessels after photosensitization by Rose Bengal. HAT1 and PCAF are poorly expressed in normal cortical neurons and astrocytes, but they are overexpressed 4-24 h after PTS. Their predominant localization in neuronal nuclei did not change after PTS, but their levels in the astrocyte nuclei significantly increased. Western blotting showed the increase of HAT1 and PCAF levels in the cytoplasmic fraction of the PTS-induced penumbra. In the nuclear fraction, PCAF level did not change, and HAT1 was overexpressed only at 24 h post-PTS. PTS-induced upregulation of HAT1 and PCAF in the penumbra was mainly associated with overexpression in the cytoplasm of neurons and especially astrocytes. HAT1 and PCAF did not co-localize with TUNEL-positive cells that indicated their nonparticipation in PTS-induced apoptosis.


Subject(s)
Astrocytes/metabolism , Cerebral Cortex/metabolism , Histone Acetyltransferases/metabolism , Neurons/metabolism , Thrombotic Stroke/metabolism , p300-CBP Transcription Factors/metabolism , Animals , Astrocytes/pathology , Cell Nucleus/metabolism , Cerebral Cortex/pathology , Cytoplasm/metabolism , Male , Neurons/pathology , Rats , Rats, Wistar , Thrombotic Stroke/pathology
5.
Brain Res Bull ; 162: 151-165, 2020 09.
Article in English | MEDLINE | ID: mdl-32592806

ABSTRACT

Epigenetic processes play important roles in brain responses to ischemic injury. We studied effects of photothrombotic stroke (PTS, a model of ischemic stroke) on the intracellular level and cellular localization of histone deacetylases HDAC3, HDAC4 and HDAC6 in the rat brain cortex, and tested the potential neuroprotector ability of their inhibitors. The background level of HDAC3, HDAC4 and HDAC6 in the rat cerebral cortex was relatively low. HDAC3 localized in the nuclei of some neurons and few astrocytes. HDAC4 was found in the neuronal cytoplasm. After PTS, their levels in penumbra did not change, but HDAC4 appeared in the nuclei of some cells. Its level in the cytoplasmic, but not nuclear fraction of penumbra decreased at 24, but not 4 h after PTS. HDAC6 was upregulated in neurons and astrocytes in the PTS-induced penumbra, especially in the nuclear fraction. Unlike HDAC3 and HDAC4, HDAC6 co-localized with TUNEL-positive apoptotic cells. Inhibitory analysis confirmed the involvement of HDAC6, but not HDAC3 and HDAC4 in neurodegeneration. HDAC6 inhibitor HPOB, HDAC2/8 inhibitor α-phenyl tropolone, and non-specific histone deacetylase inhibitor sodium valproate, but not HDAC3 inhibitor BRD3308, or HDAC4 inhibitor LMK235, decreased PTS-induced infarction volume in the mouse brain, reduced apoptosis, and recovered the motor behavior. HPOB also restored PTS-impaired acetylation of α-tubulin. α-phenyl tropolone restored acetylation of histone H4 in penumbra cells. These results suggest that histone deacetylases HDAC6 and HDAC2 are the possible molecular targets for anti-ischemic therapy, and their inhibitors α-phenyl tropolone, HBOP and sodium valproate can be considered as promising neuroprotectors.


Subject(s)
Cerebral Cortex/enzymology , Histone Deacetylase 6/biosynthesis , Histone Deacetylase Inhibitors/pharmacology , Intracranial Thrombosis/enzymology , Stroke/enzymology , Tropolone/pharmacology , Valproic Acid/pharmacology , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Gene Expression , Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylases/biosynthesis , Intracranial Thrombosis/pathology , Intracranial Thrombosis/prevention & control , Lasers, Semiconductor/adverse effects , Male , Mice , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Rats , Stroke/pathology , Stroke/prevention & control , Tropolone/therapeutic use , Valproic Acid/therapeutic use
6.
Mol Neurobiol ; 57(1): 226-238, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31493239

ABSTRACT

In ischemic stroke, vascular occlusion rapidly induces tissue infarct. Over the ensuing hours, damage spreads to adjacent tissue and forms transition zone (penumbra), which is potentially salvageable. Epigenetic regulation of chromatin structure controls gene expression and protein synthesis. We studied the expression of histone deacetylases HDAC1 and HDAC2 in the penumbra at 4 or 24 h after photothrombotic stroke (PTS) in the rat brain cortex. PTS increased the expression of HDAC1 and HDAC2 in penumbra and caused the redistribution of HDAC1 but not HDAC2 from the neuronal nuclei to cytoplasm. In astrocytes, HDAC1 expression and localization did not change. In neurons, HDAC2 localized exclusively in nuclei, but in astrocytes, it was also observed in processes. PTS induced neuronal apoptosis in the penumbra. TUNEL-stained apoptotic neurons co-localized with HDAC2 but not HDAC1. These data suggest that HDAC2 may represent the potential target for anti-stroke therapy and its selective inhibition may be a promising strategy for the protection of the penumbra tissue after ischemic stroke.


Subject(s)
Apoptosis , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/metabolism , Light , Stroke/complications , Stroke/enzymology , Thrombosis/complications , Thrombosis/enzymology , Animals , Astrocytes/metabolism , Biomarkers/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Glial Fibrillary Acidic Protein/metabolism , Male , Rats, Wistar
7.
Mol Neurobiol ; 57(1): 239-248, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31512115

ABSTRACT

Unilateral photothrombotic stroke caused tissue infarct in the mouse cerebral cortex. The injury of the cerebral cortex impaired the mouse motor activity, in particular the functional asymmetry in forelimb use. In the peri-infarct cortical tissue outside the infarct core cell apoptosis occurred at 4 and 7 days after PTS. The downregulation of acetylated α-tubulin, a marker of stable microtubules, showed the destruction of neurites, axons, and dendrites in injured neurons. However, the upregulation of GAP43 indicates the stimulation of neurite growth that was possibly aimed at the recovery of the cortical tissue in the damaged cerebral hemisphere. Application of MI-192, an inhibitor of histone deacetylases HDAC2 and HDAC3, demonstrated the neuroprotective activity in the mouse brain subjected to photothrombotic stroke. It reduced the volume of the PTS-induced infarction core in the mouse brain, partly restored the functional symmetry in the forelimb use, decreased the level of PTS-induced apoptosis and acetylation of α-tubulin characteristic for stable microtubules, and increased the expression of GAP-43 in the cerebral cortex of the damaged hemisphere. These data point to the involvement of HDAC2 and HDAC3 in the photothrombotic injury of the mouse brain not only in the infarction core but also outside it. The application of MI192 after PTS reduced or eliminated these negative effects and exerted the neuroprotective effect on the mouse brain. It may be a promising neuroprotector agent for anti-stroke therapy.


Subject(s)
Benzamides/therapeutic use , Brain/pathology , Histone Deacetylase Inhibitors/therapeutic use , Light , Neuroprotective Agents/therapeutic use , Stroke/drug therapy , Thrombosis/drug therapy , Acetylation/drug effects , Animals , Apoptosis/drug effects , Behavior, Animal/drug effects , Benzamides/chemistry , Benzamides/pharmacology , Brain/drug effects , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , GAP-43 Protein/metabolism , Histone Deacetylase 2/antagonists & inhibitors , Histone Deacetylase 2/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Histones/metabolism , Male , Mice , Neuroprotective Agents/pharmacology , Stroke/complications , Thrombosis/complications , Tubulin/metabolism
8.
Biochemistry (Mosc) ; 80(6): 790-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26531025

ABSTRACT

Photodynamic impact on animal cerebral cortex using water-soluble Bengal Rose as a photosensitizer, which does not cross the blood-brain barrier and remains in blood vessels, induces platelet aggregation, vessel occlusion, and brain tissue infarction. This reproduces ischemic stroke. Irreversible cell damage within the infarction core propagates to adjacent tissue and forms a transition zone - the penumbra. Tissue necrosis in the infarction core is too fast (minutes) to be prevented, but much slower penumbral injury (hours) can be limited. We studied the changes in morphology and protein expression profile in penumbra 1 h after local photothrombotic infarction induced by laser irradiation of the cerebral cortex after Bengal Rose administration. Morphological study using standard hematoxylin/eosin staining showed a 3-mm infarct core surrounded by 1.5-2.0 mm penumbra. Morphological changes in the penumbra were lesser and decreased towards its periphery. Antibody microarrays against 224 neuronal and signaling proteins were used for proteomic study. The observed upregulation of penumbra proteins involved in maintaining neurite integrity and guidance (NAV3, MAP1, CRMP2, PMP22); intercellular interactions (N-cadherin); synaptic transmission (glutamate decarboxylase, tryptophan hydroxylase, Munc-18-1, Munc-18-3, and synphilin-1); mitochondria quality control and mitophagy (PINK1 and Parkin); ubiquitin-mediated proteolysis and tissue clearance (UCHL1, PINK1, Parkin, synphilin-1); and signaling proteins (PKBα and ERK5) could be associated with tissue recovery. Downregulation of PKC, PKCß1/2, and TDP-43 could also reduce tissue injury. These changes in expression of some neuronal proteins were directed mainly to protection and tissue recovery in the penumbra. Some upregulated proteins might serve as markers of protection processes in a penumbra.


Subject(s)
Brain Ischemia/metabolism , Intracranial Thrombosis/metabolism , Proteomics/methods , Animals , Blood-Brain Barrier/metabolism , Brain/metabolism , Cadherins/metabolism , Cerebral Cortex/blood supply , Cerebral Cortex/metabolism , Cerebral Infarction/metabolism , DNA-Binding Proteins/metabolism , Disease Models, Animal , Male , Rats , Signal Transduction
9.
Photodiagnosis Photodyn Ther ; 11(3): 357-64, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24981884

ABSTRACT

BACKGROUND: Radachlorin, a chlorine-derived photosensitizer, is used currently in photodynamic therapy (PDT) of skin cancer. In this work we studied Radachlorin-PDT effect on peripheral nerve and glial cells that are damaged along with tumor tissue. METHODS: We used simple model objects - a crayfish stretch receptor that consists of a single sensory neuron surrounded by glial cells and crayfish nerve cord consisting of nerve fibers and ganglia. Radachlorin absorption and emission spectra were registered using spectrophotometer and spectrofluorimeter. Radachlorin accumulation and intracellular localization were studied using the fluorescence microscope. Necrotic and apoptotic cells were visualized using propidium iodide and Hoechst 33342. Neuronal activity was registered using standard electrophysiological methods. RESULTS: Radachlorin absorption spectrum in the physiological van Harreveld saline (pH 7.3) contained maximums at 420 and 654nm. Its fluorescence band 620-700nm had a maximum at 664nm. In the crayfish stretch receptor Radachlorin localized predominantly to the glial envelope and penetrated slightly into the neuron body and axon. Radachlorin rapidly accumulated in the crayfish nerve cord tissue within 30min. Its elimination in the dye-free solution occurred slower: 11% loss for 2h. Radachlorin-PDT inactivated the neuron and induced necrosis of neurons and glial cells and glial apoptosis at concentrations as low as 10(-10)-10(-9)M. CONCLUSIONS: Radachlorin rapidly accumulates in the nervous tissue, mainly in glial cells, and demonstrates very high photodynamic efficacy that characterize it as a promising photosensitizer.


Subject(s)
Neuroglia/physiology , Neurons/physiology , Photochemotherapy/methods , Porphyrins/administration & dosage , Porphyrins/pharmacokinetics , Animals , Apoptosis/drug effects , Apoptosis/physiology , Astacoidea , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Drug Combinations , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neuroglia/cytology , Neuroglia/drug effects , Neurons/cytology , Neurons/drug effects , Photosensitizing Agents/administration & dosage , Photosensitizing Agents/pharmacokinetics
10.
Biochim Biophys Acta ; 1840(1): 262-70, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24055374

ABSTRACT

BACKGROUND: Photodynamic therapy (PDT) is used for cancer treatment including brain tumors. But the role of epigenetic processes in photodynamic injury of normal brain tissue is unknown. METHODS: 5-Aminolevulinic acid (ALA), a precursor of protoporphyrin IX (PpIX), was used to photosensitize mouse cerebral cortex. PpIX accumulation in cortical tissue was measured spectrofluorometrically. Hematoxylin/eosin, gallocyanin-chromalum and immunohistochemical staining were used to study morphological changes in PDT-treated cerebral cortex. Proteomic antibody microarrays were used to evaluate expression of 112 proteins involved in epigenetic regulation. RESULTS: ALA administration induced 2.5-fold increase in the PpIX accumulation in the mouse brain cortex compared to untreated mice. Histological study demonstrated PDT-induced injury of some neurons and cortical vessels. ALA-PDT induced dimethylation of histone H3, upregulation of histone deacetylases HDAC-1 and HDAC-11, and DNA methylation-dependent protein Kaiso that suppressed transcriptional activity. Upregulation of HDAC-1 and H3K9me2 was confirmed immunohistochemically. Down-regulation of transcription factor FOXC2, PABP, and hBrm/hsnf2a negatively regulated transcription. Overexpression of phosphorylated histone H2AX indicated activation of DNA repair, but down-regulation of MTA1/MTA1L1 and PML - impairment of DNA repair. Overexpression of arginine methyltransferase PRMT5 correlated with up-regulation of transcription factor E2F4 and importin α5/7. CONCLUSION: ALA-PDT injures and kills some but not all neurons and caused limited microvascular alterations in the mouse cerebral cortex. It alters expression of some proteins involved in epigenetic regulation of transcription, histone modification, DNA repair, nuclear protein import, and proliferation. GENERAL SIGNIFICANCE: These data indicate epigenetic markers of photo-oxidative injury of normal brain tissue.


Subject(s)
Aminolevulinic Acid/pharmacology , Cerebral Cortex/drug effects , Epigenesis, Genetic/drug effects , Gene Expression Regulation , Photochemotherapy , Photosensitizing Agents/pharmacology , Proteome/analysis , Animals , Cerebral Cortex/pathology , Cerebral Cortex/radiation effects , Epigenesis, Genetic/genetics , Epigenesis, Genetic/radiation effects , Epigenomics , Gene Expression Regulation/drug effects , Gene Expression Regulation/radiation effects , Histones/metabolism , Immunoenzyme Techniques , Male , Mice , Protein Array Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...