Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Adv Tech Stand Neurosurg ; 49: 1-18, 2024.
Article in English | MEDLINE | ID: mdl-38700677

ABSTRACT

Although the pathogenetic pathway of moyamoya disease (MMD) remains unknown, studies have indicated that variations in the RING finger protein RNF 213 is the strongest susceptible gene of MMD. In addition to the polymorphism of this gene, many circulating angiogenetic factors such as growth factors, vascular progenitor cells, inflammatory and immune mediators, angiogenesis related cytokines, as well as circulating proteins promoting intimal hyperplasia, excessive collateral formation, smooth muscle migration and atypical migration may also play critical roles in producing this disease. Identification of these circulating molecules biomarkers may be used for the early detection of this disease. In this chapter, how the hypothesized pathophysiology of these factors affect MMD and the interactive modulation between them are summarized.


Subject(s)
Biomarkers , Moyamoya Disease , Ubiquitin-Protein Ligases , Moyamoya Disease/genetics , Moyamoya Disease/diagnosis , Humans , Biomarkers/metabolism , Biomarkers/blood , Ubiquitin-Protein Ligases/genetics , Adenosine Triphosphatases/genetics
2.
Pharmaceuticals (Basel) ; 16(5)2023 May 18.
Article in English | MEDLINE | ID: mdl-37242547

ABSTRACT

Studies of the neurobiological causes of anxiety disorders have suggested that the γ-aminobutyric acid (GABA) system increases synaptic concentrations and enhances the affinity of GABAA (type A) receptors for benzodiazepine ligands. Flumazenil antagonizes the benzodiazepine-binding site of the GABA/benzodiazepine receptor (BZR) complex in the central nervous system (CNS). The investigation of flumazenil metabolites using liquid chromatography (LC)-tandem mass spectrometry will provide a complete understanding of the in vivo metabolism of flumazenil and accelerate radiopharmaceutical inspection and registration. The main goal of this study was to investigate the use of reversed-phase high performance liquid chromatography (PR-HPLC), coupled with electrospray ionization triple-quadrupole tandem mass spectrometry (ESI-QqQ MS), to identify flumazenil and its metabolites in the hepatic matrix. Carrier-free nucleophilic fluorination with an automatic synthesizer for [18F]flumazenil, combined with nano-positron emission tomography (NanoPET)/computed tomography (CT) imaging, was used to predict the biodistribution in normal rats. The study showed that 50% of the flumazenil was biotransformed by the rat liver homogenate in 60 min, whereas one metabolite (M1) was a methyl transesterification product of flumazenil. In the rat liver microsomal system, two metabolites were identified (M2 and M3), as their carboxylic acid and hydroxylated ethyl ester forms between 10 and 120 min, respectively. A total of 10-30 min post-injection of [18F]flumazenil showed an immediate decreased in the distribution ratio observed in the plasma. Nevertheless, a higher ratio of the complete [18F]flumazenil compound could be used for subsequent animal studies. [18F] According to in vivo nanoPET/CT imaging and ex vivo biodistribution assays, flumazenil also showed significant effects on GABAA receptor availability in the amygdala, prefrontal cortex, cortex, and hippocampus in the rat brain, indicating the formation of metabolites. We reported the completion of the biotransformation of flumazenil by the hepatic system, as well as [18F]flumazenil's potential as an ideal ligand and PET agent for the determination of the GABAA/BZR complex for multiplex neurological syndromes at the clinical stage.

3.
Int J Mol Sci ; 24(3)2023 Feb 02.
Article in English | MEDLINE | ID: mdl-36769234

ABSTRACT

The cerebral vascular system stringently regulates cerebral blood flow (CBF). The components of the blood-brain barrier (BBB) protect the brain from pathogenic infections and harmful substances, efflux waste, and exchange substances; however, diseases develop in cases of blood vessel injuries and BBB dysregulation. Vascular pathology is concurrent with the mechanisms underlying aging, Alzheimer's disease (AD), and vascular dementia (VaD), which suggests its involvement in these mechanisms. Therefore, in the present study, we reviewed the role of vascular dysfunction in aging and neurodegenerative diseases, particularly AD and VaD. During the development of the aforementioned diseases, changes occur in the cerebral blood vessel morphology and local cells, which, in turn, alter CBF, fluid dynamics, and vascular integrity. Chronic vascular inflammation and blood vessel dysregulation further exacerbate vascular dysfunction. Multitudinous pathogenic processes affect the cerebrovascular system, whose dysfunction causes cognitive impairment. Knowledge regarding the pathophysiology of vascular dysfunction in neurodegenerative diseases and the underlying molecular mechanisms may lead to the discovery of clinically relevant vascular biomarkers, which may facilitate vascular imaging for disease prevention and treatment.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Dementia, Vascular , Neurodegenerative Diseases , Vascular Diseases , Humans , Neurodegenerative Diseases/pathology , Alzheimer Disease/pathology , Brain/pathology , Dementia, Vascular/pathology , Blood-Brain Barrier/pathology , Vascular Diseases/pathology , Cognitive Dysfunction/pathology
4.
J Cell Mol Med ; 25(15): 7418-7425, 2021 08.
Article in English | MEDLINE | ID: mdl-34216182

ABSTRACT

We previously showed a hydroxamic acid-based histone deacetylase inhibitor (HDACi), compound 13, provides neuroprotection against chronic cerebral hypoperfusion (CCH) both in vitro under oxygen-glucose deprivation (OGD) conditions and in vivo under bilateral common carotid artery occlusion (BCCAO) conditions. Intriguingly, the protective effect of this HDACi is via H3K14 or H4K5 acetylation-mediated differential BDNF isoform activation. BDNF is involved in cell proliferation and differentiation in development, synaptic plasticity and in learning and memory related with receptors or synaptic proteins. B6 mice underwent BCCAO and were randomized into 4 groups; a sham without BCCAO (sham), BCCAO mice injected with DMSO (DMSO), mice injected with HDACi-compound 13 (compound 13) and mice injected with suberoylanilide hydroxamic acid (SAHA). The cortex and hippocampus of mice were harvested at 3 months after BCCAO, and levels of BDNF, AMPA receptor and dopamine receptors (D1, D2 and D3) were studied using Western blotting analysis or immunohistochemistry. We found that the AMPA receptor plays a key role in the molecular mechanism of this process by modulating HDAC. This protective effect of HDACi may be through BDNF; therefore, activation of this downstream signalling molecule, for example by AMPA receptors, could be a therapeutic target or intervention applied under CCH conditions.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Dementia, Vascular/drug therapy , Histone Deacetylase Inhibitors/therapeutic use , Neuroprotective Agents/therapeutic use , Receptors, AMPA/metabolism , Animals , Arterial Occlusive Diseases/complications , Carotid Arteries/pathology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Dementia, Vascular/etiology , Dementia, Vascular/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Histone Deacetylase Inhibitors/pharmacology , Mice , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology
5.
Int J Mol Sci ; 22(4)2021 Feb 08.
Article in English | MEDLINE | ID: mdl-33567654

ABSTRACT

Moyamoya disease (MMD) is a cerebrovascular disease that presents with vascular stenosis and a hazy network of collateral formations in angiography. However, the detailed pathogenic pathway remains unknown. Studies have indicated that in addition to variations in the of genetic factor RNF213, unusual circulating angiogenetic factors observed in patients with MMD may play a critical role in producing "Moyamoya vessels". Circulating angiogenetic factors, such as growth factors, vascular progenitor cells, cytokines, inflammatory factors, and other circulating proteins, could promote intimal hyperplasia in vessels and excessive collateral formation with defect structures through endothelial hyperplasia, smooth muscle migration, and atypical neovascularization. This study summarizes the hypothesized pathophysiology of how these circulating factors affect MMD and the interactive modulation between them.


Subject(s)
Biomarkers/blood , Moyamoya Disease/blood , Moyamoya Disease/pathology , Neovascularization, Pathologic/pathology , Animals , Humans
6.
J Cell Mol Med ; 24(12): 6966-6977, 2020 06.
Article in English | MEDLINE | ID: mdl-32374084

ABSTRACT

Vascular dementia (VaD) is the second most common cause of dementia, but the treatment is still lacking. Although many studies have reported that histone deacetylase inhibitors (HDACis) confer protective effects against ischemic and hypoxic injuries, their role in VaD is still uncertain. Previous studies shown, one HDACi protected against cognitive decline in animals with chronic cerebral hypoperfusion (CCH). However, the underlying mechanisms remain elusive. In this study, we tested several 10,11-dihydro-5H-dibenzo[b,f]azepine hydroxamates, which act as HDACis in the CCH model (in vivo), and SH-SY5Y (neuroblastoma cells) with oxygen-glucose deprivation (OGD, in vitro). We identified a compound 13, which exhibited the best cell viability under OGD. The compound 13 could increase, in part, the protein levels of brain-derived neurotrophic factor (BDNF). It increased acetylation status on lysine 14 residue of histone 3 (H3K14) and lysine 5 of histone 4 (H4K5). We further clarified which promoters (I, II, III, IV or IX) could be affected by histone acetylation altered by compound 13. The results of chromatin immunoprecipitation and Q-PCR analysis indicate that an increase in H3K14 acetylation leads to an increase in the expression of BDNF promoter II, while an increase in H4K5 acetylation results in an increase in the activity of BDNF promoter II and III. Afterwards, these cause an increase in the expression of BDNF exon II, III and coding exon IX. In summary, the HDACi compound 13 may increase BDNF specific isoforms expression to rescue the ischemic and hypoxic injuries through changes of acetylation on histones.


Subject(s)
Brain Ischemia/drug therapy , Brain-Derived Neurotrophic Factor/metabolism , Glucose/deficiency , Histone Deacetylase Inhibitors/therapeutic use , Lysine/metabolism , Neuroprotective Agents/therapeutic use , Oxygen/metabolism , Acetylation/drug effects , Animals , Brain-Derived Neurotrophic Factor/genetics , Cell Line, Tumor , Chronic Disease , Exons/genetics , Hippocampus/drug effects , Hippocampus/pathology , Histone Deacetylase Inhibitors/pharmacology , Histones/metabolism , Humans , Male , Mice, Inbred C57BL , Models, Biological , Neurons/drug effects , Neurons/pathology , Neuroprotective Agents/pharmacology , Promoter Regions, Genetic/genetics , Up-Regulation/drug effects
7.
Int J Mol Sci ; 21(6)2020 Mar 21.
Article in English | MEDLINE | ID: mdl-32245271

ABSTRACT

The pathophysiology of vascular cognitive impairment (VCI) is associated with chronic cerebral hypoperfusion (CCH). Increased high-mobility group box protein 1 (HMGB1), a nonhistone protein involved in injury and inflammation, has been established in the acute phase of CCH. However, the role of HMGB1 in the chronic phase of CCH remains unclear. We developed a novel animal model of CCH with a modified bilateral common carotid artery occlusion (BCCAO) in C57BL/6 mice. Cerebral blood flow (CBF) reduction, the expression of HMGB1 and its proinflammatory cytokines (tumor necrosis factor-alpha [TNF-α], interleukin [IL]-1ß, and IL-6), and brain pathology were assessed. Furthermore, we evaluated the effect of HMGB1 suppression through bilateral intrahippocampus injection with the CRISPR/Cas9 knockout plasmid. Three months after CCH induction, CBF decreased to 30-50% with significant cognitive decline in BCCAO mice. The 7T-aMRI showed hippocampal atrophy, but amyloid positron imaging tomography showed nonsignificant amyloid-beta accumulation. Increased levels of HMGB1, TNF-α, IL-1ß, and IL-6 were observed 3 months after BCCAO. HMGB1 suppression with CRISPR/Cas9 knockout plasmid restored TNF-α, IL-1ß, and IL-6 and attenuated hippocampal atrophy and cognitive decline. We believe that HMGB1 plays a pivotal role in CCH-induced VCI pathophysiology and can be a potential therapeutic target of VCI.


Subject(s)
Brain Ischemia/metabolism , Cerebrovascular Circulation , HMGB1 Protein/metabolism , Amyloid beta-Peptides/metabolism , Animals , Behavior Rating Scale , Brain Ischemia/diagnostic imaging , Brain Ischemia/genetics , Brain Ischemia/physiopathology , CRISPR-Cas Systems , Carotid Stenosis , Chronic Disease , Dementia, Vascular/physiopathology , Disease Models, Animal , Gene Knockout Techniques , HMGB1 Protein/genetics , Hippocampus/diagnostic imaging , Hippocampus/pathology , Hippocampus/physiopathology , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL , Psychomotor Performance , Tumor Necrosis Factor-alpha/metabolism
8.
Eur J Med Chem ; 187: 111915, 2020 Feb 01.
Article in English | MEDLINE | ID: mdl-31838329

ABSTRACT

A series of 10,11-dihydro-5H-dibenzo [b,f]azepine hydroxamates (4-15) were synthesized, behaving as histone deacetylase inhibitors, and examined for their influence on vascular cognitive impairment (VCI), which correlated with dementia. The results revealed that (E)-3-(4-(((3-(3-chloro-10,11-dihydro-5H-dibenzo [b,f]azepin-5-yl)propyl)amino)methyl)phenyl)-N-hydroxy-acrylamide (13) increases cerebral blood flow (CBF), attenuates cognitive impairment, and improves hippocampal atrophy in in vivo study. It is also able to increase the level of histone acetylation (H3K14 or H4K5) in the cortex and hippocampus of chronic cerebral hypoperfusion (CCH) mice; as a result, it could be a potential HDAC inhibitor for the treatment of vascular cognitive impairment.


Subject(s)
Azepines/pharmacology , Clomipramine/analogs & derivatives , Cognitive Dysfunction/drug therapy , Dementia, Vascular/drug therapy , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Hydroxamic Acids/pharmacology , Protective Agents/pharmacology , Animals , Azepines/chemistry , Cell Line, Tumor , Clomipramine/chemistry , Clomipramine/pharmacology , Cognitive Dysfunction/metabolism , Dementia, Vascular/metabolism , Dose-Response Relationship, Drug , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/chemistry , Humans , Hydroxamic Acids/chemical synthesis , Hydroxamic Acids/chemistry , Male , Mice , Mice, Inbred C57BL , Molecular Structure , Protective Agents/chemical synthesis , Protective Agents/chemistry , Structure-Activity Relationship
9.
Oxid Med Cell Longev ; 2017: 7968905, 2017.
Article in English | MEDLINE | ID: mdl-28491238

ABSTRACT

MicroRNAs play important roles in cell proliferation, differentiation, and apoptosis, and their expression influences cardiomyocyte apoptosis resulting from ischemia-induced myocardial infarction. Here, we determined the role of miR expression in cardiomyocyte apoptosis during hypoxia and reoxygenation. The rat cardiomyocyte cell line H9c2 was incubated for 3 h in normal or hypoxia medium, followed by reoxygenation for 24 h and transfection with a miR-302 mimic or antagomir. The effect of miR-302 on myeloid leukemia cell-differentiation protein-1 (Mcl-1) expression was determined by western blot, real-time polymerase chain reaction, and luciferase reporter assays, with cell viability assays. We observed that miR-302 expression was elevated by hypoxia/reoxygenation injury and increased further or decreased by transfection of the miR-302 mimic or miR-302 antagomir, respectively. Additionally, elevated miR-302 levels increased apoptosis-related protein levels and cardiomyocyte apoptosis, and luciferase reporter assays revealed miR-302 binding to the Mcl-1 mRNA 3' untranslated region. Our findings suggested that miR-302 overexpression aggravated hypoxia/reoxygenation-mediated cardiomyocyte apoptosis by inhibiting antiapoptotic Mcl-1 expression, thereby activating proapoptotic molecules. Furthermore, results indicating cardiomyocyte rescue from hypoxia/reoxygenation injury following treatment with miR-302 antagomir suggested that miR-302 inhibition might constitute a therapeutic strategy for protection against cardiomyocyte apoptosis during hypoxia/reoxygenation injury.


Subject(s)
Apoptosis , Gene Expression Regulation , MicroRNAs/antagonists & inhibitors , Myeloid Cell Leukemia Sequence 1 Protein/biosynthesis , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/metabolism , Animals , Cell Hypoxia , Cell Line , MicroRNAs/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/pathology , Rats
10.
Toxicol Appl Pharmacol ; 266(3): 399-407, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23201462

ABSTRACT

Fissistigma bracteolatum is widely used in traditional medicine to treat inflammatory diseases. However, its active components and mechanisms of action remain unclear. In this study, (3Z)-6,7-dihydroxy-4-methoxy-3-(phenylmethylidene)-5-(3-phenylpropanoyl)-1-benzofuran-2(3H) (bractelactone), a novel chalcone from F. bracteolatum, showed potent inhibitory effects against superoxide anion (O2·â») production, elastase release, and CD11b expression in formyl-L-methionyl-L-leucyl-L-phenylalanine (FMLP)-induced human neutrophils. However, bractelactone showed only weak inhibition of phorbol myristate acetate-caused O2·â» production. The peak cytosolic calcium concentration ([Ca²âº](i)) was unaltered by bractelactone in FMLP-induced neutrophils, but the decay time of [Ca²âº](i) was significantly shortened. In a calcium-free solution, changes in [Ca²âº](i) caused by the addition of extracellular Ca²âº were inhibited by bractelactone in FMLP-activated cells. In addition, bractelactone did not alter the phosphorylation of p38 MAPK, ERK, JNK, or AKT or the concentration of cAMP. These results suggest that bractelactone selectively inhibits store-operated calcium entry (SOCE). In agreement with this concept, bractelactone suppressed sustained [Ca²âº](i) changes in thapsigargin-activated neutrophils. Furthermore, bractelactone did not alter FMLP-induced formation of inositol 1,4,5-triphosphate. Taken together, our results demonstrate that the anti-inflammatory effects of bractelactone, an active ingredient of F. bracteolatum, in human neutrophils are through the selective inhibition of SOCE.


Subject(s)
Annonaceae/chemistry , Anti-Inflammatory Agents/pharmacology , Chalcones/pharmacology , Neutrophil Activation/drug effects , Adult , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/isolation & purification , CD11b Antigen/metabolism , Calcium Signaling/drug effects , Chalcones/chemistry , Chalcones/isolation & purification , Humans , Immunoblotting , Inhibitory Concentration 50 , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Mitogen-Activated Protein Kinases/metabolism , Neutrophils/drug effects , Neutrophils/metabolism , Neutrophils/physiology , Pancreatic Elastase/metabolism , Phosphorylation/drug effects , Plant Stems/chemistry , Reactive Oxygen Species/metabolism , Superoxides/metabolism , Young Adult
11.
Toxicol Appl Pharmacol ; 260(2): 193-200, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22381622

ABSTRACT

Alveolar macrophages play significant roles in the pathogenesis of several inflammatory lung diseases. Increases in exhaled nitric oxide (NO) are well documented to reflect disease severity in the airway. In this study, we investigated the effect of 3-(5'-hydroxymethyl-2'-furyl)-1-benzyl indazole (YC-1), a known activator of soluble guanylyl cyclase, on prostaglandin (PG)E1 (a stable PGE2 analogue) and forskolin (a adenylate cyclase activator) induced NO production and inducible NO synthase (iNOS) expression in rat alveolar macrophages (NR8383). YC-1 did not directly cause NO production or iNOS expression, but drastically potentiated PGE1- or forskolin-induced NO production and iNOS expression in NR8383 alveolar macrophages. Combination treatment with YC-1 and PGE1 significantly increased phosphorylation of the cAMP response element-binding protein (CREB), but not nuclear factor (NF)-κB activation. The combined effect on NO production, iNOS expression, and CREB phosphorylation was reversed by a protein kinase (PK)A inhibitor (H89), suggesting that the potentiating functions were mediated through a cAMP/PKA signaling pathway. Consistent with this, cAMP analogues, but not the cGMP analogue, caused NO release, iNOS expression, and CREB activation. YC-1 treatment induced an increase in PGE1-induced cAMP formation, which occurred through the inhibition of cAMP-specific phosphodiesterase (PDE) activity. Furthermore, the combination of rolipram (an inhibitor of PDE4), but not milronone (an inhibitor of PDE3), and PGE1 also triggered NO production and iNOS expression. In summary, YC-1 potentiates PGE1-induced NO production and iNOS expression in alveolar macrophages through inhibition of cAMP PDE activity and activation of the cAMP/PKA/CREB signaling pathway.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Enzyme Activators/pharmacology , Indazoles/pharmacology , Lung Diseases/immunology , Macrophages, Alveolar/drug effects , Animals , Colforsin/pharmacology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Dinoprostone/pharmacology , Guanylate Cyclase/metabolism , Immunoblotting , Isoquinolines/pharmacology , Macrophages, Alveolar/metabolism , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type II/metabolism , Phosphoric Diester Hydrolases/metabolism , Rats , Receptors, Cytoplasmic and Nuclear/metabolism , Soluble Guanylyl Cyclase , Sulfonamides/pharmacology
12.
J Cereb Blood Flow Metab ; 26(4): 491-501, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16094316

ABSTRACT

Prostacyclin (PGI2), a potent vasodilator and inhibitor of platelet aggregation and leukocyte activation, is crucial in vascular diseases such as stroke. Prostacyclin synthase (PGIS) is the key enzyme for PGI2 synthesis. Although expression of PGIS was noted in the brain, its role in ischemic insult remains unclear. Here we reported the temporal and spatial expression of PGIS mRNA and protein after 60-min transient ischemia. Northern blot and in situ hybridization revealed a delayed increase of PGIS mRNA in the ischemic cortex at 24- to 72-h after ischemia; PGIS was detected mainly in the ipsilateral penumbra area, pyriform cortex, hippocampus, and leptomeninges. Western blot and immunohistochemical analysis revealed that PGIS proteins were expressed temporally and spatially similar to PGIS mRNA. PGIS was heavily colocalized with PECAM-1 to endothelial cells at the leptomeninges, large and small vessels, and localized to neuronal cells, largely at the penumbra area. A substantial amount of PGIS was also detected in the macrophage and glial cells. To evaluate its role against ischemic infarct, we overexpressed PGIS by adenoviral gene transfer. When infused 72 h before ischemia (- 72 h), Adv-PGIS reduced infarct volume by approximately 50%. However, it had no effect on infarct volume when infused immediately after ischemia (0 h). Eicosanoid analysis revealed selective elevation of PGI2 at - 72 h while PGI2 and TXB2 were both elevated at 0 h, altering the PGI2/thromboxane A2 (TXA2) ratio from 10 to 4. These findings indicate that PGIS protects the brain by enhancing PGI2 synthesis and creating a favorable PGI2/TXA2 ratio.


Subject(s)
Brain Ischemia/enzymology , Cytochrome P-450 Enzyme System/genetics , Epoprostenol/biosynthesis , Gene Expression Regulation , Intramolecular Oxidoreductases/genetics , Reperfusion Injury/enzymology , Animals , Brain/cytology , Brain/enzymology , Cytochrome P-450 Enzyme System/analysis , Intramolecular Oxidoreductases/analysis , Kinetics , RNA, Messenger , Rats , Thromboxane A2/analysis , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...