Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Commun Signal ; 18(1): 57, 2020 04 06.
Article in English | MEDLINE | ID: mdl-32252776

ABSTRACT

BACKGROUND: Chronic brain hypoperfusion (CBH) is closely related to Alzheimer's disease (AD) and vascular dementia (VaD). Meanwhile, synaptic pathology plays a prominent role in the initial stage of AD and VaD. However, whether and how CBH impairs presynaptic plasticity is currently unclear. METHODS: In the present study, we performed a battery of techniques, including primary neuronal culture, patch clamp, stereotaxic injection of the lentiviral vectors, morris water maze (MWM), dual luciferase reporter assay, FM1-43 fluorescence dye evaluation, qRT-PCR and western blot, to investigate the regulatory effect of miR-153 on hippocampal synaptic vesicle release both in vivo and in vitro. The CBH rat model was generated by bilateral common carotid artery ligation (2VO). RESULTS: Compared to sham rats, 2VO rats presented decreased field excitatory postsynaptic potential (fEPSP) amplitude and increased paired-pulse ratios (PPRs) in the CA3-CA1 pathway, as well as significantly decreased expression of multiple vesicle fusion-related proteins, including SNAP-25, VAMP-2, syntaxin-1A and synaptotagmin-1, in the hippocampi. The levels of microRNA-153 (miR-153) were upregulated in the hippocampi of rats following 2VO surgery, and in the plasma of dementia patients. The expression of the vesicle fusion-related proteins affected by 2VO was inhibited by miR-153, elevated by miR-153 inhibition, and unchanged by binding-site mutation or miR masks. FM1-43 fluorescence images showed that miR-153 blunted vesicle exocytosis, but this effect was prevented by either 2'-O-methyl antisense oligoribonucleotides to miR-153 (AMO-153) and miR-masking of the miR-153 binding site in the 3' untranslated region (3'UTR) of the Snap25, Vamp2, Stx1a and Syt1 genes. Overexpression of miR-153 by lentiviral vector-mediated miR-153 mimics (lenti-pre-miR-153) decreased the fEPSP amplitude and elevated the PPR in the rat hippocampus, whereas overexpression of the antisense molecule (lenti-AMO-153) reversed these changes triggered by 2VO. Furthermore, lenti-AMO-153 attenuated the cognitive decline of 2VO rats. CONCLUSIONS: Overexpression of miR-153 controls CBH-induced presynaptic vesicle release impairment by posttranscriptionally regulating the expression of four vesicle release-related proteins by targeting the 3'UTRs of the Stx1a, Snap25, Vamp2 and Syt1 genes. These findings identify a novel mechanism of presynaptic plasticity impairment during CBH, which may be a new drug target for prevention or treatment of AD and VaD. Video Abstract.


Subject(s)
Dementia, Vascular/metabolism , Hypoxia-Ischemia, Brain/metabolism , MicroRNAs/physiology , Synaptic Vesicles/metabolism , Aged , Animals , Humans , Male , Rats , Rats, Sprague-Dawley , Synaptosomal-Associated Protein 25/metabolism , Synaptotagmin I/metabolism , Syntaxin 1/metabolism , Vesicle-Associated Membrane Protein 2/metabolism
2.
Acta Pharmacol Sin ; 41(5): 620-628, 2020 May.
Article in English | MEDLINE | ID: mdl-31863057

ABSTRACT

Obstructive sleep apnea (OSA) is closely associated with central nervous system diseases and could lead to autonomic nerve dysfunction, which is often seen in neurodegenerative diseases. Previous studies have shown that metoprolol prevents several chronic OSA-induced cardiovascular diseases through inhibiting autonomic nerve hyperactivity. It remains unclear whether chronic OSA can lead to dendritic remodeling in the brain, and whether metoprolol affects the dendritic remodeling. In this study we investigated the effect of metoprolol on dendrite morphology in a canine model of chronic OSA, which was established in beagles through clamping and reopening the endotracheal tube for 4 h every other day for 12 weeks. OSA beagles were administered metoprolol (5 mg· kg-1· d-1). The dendritic number, length, crossings and spine density of neurons in hippocampi and prefrontal cortices were assessed by Golgi staining. And the protein levels of hypoxia-inducible factor-1α (HIF-1α) and brain-derived neurotrophic factor (BDNF) were measured by Western blotting. We showed that chronic OSA successfully induced significant brain hypoxia evidenced by increased HIF-1α levels in CA1 region and dentate gyrus of hippocampi, as well as in prefrontal cortex. Furthermore, OSA led to markedly decreased dendrite number, length and intersections, spine loss as well as reduced BDNF levels. Administration of metoprolol effectively prevented the dendritic remodeling and spine loss induced by chronic OSA. In addition, administration of metoprolol reversed the decreased BDNF, which might be associated with the metoprolol-induced neuronal protection. In conclusion, metoprolol protects against neuronal dendritic remodeling in hippocampi and prefrontal cortices induced by chronic OSA in canine.


Subject(s)
Dendrites/drug effects , Disease Models, Animal , Metoprolol/pharmacology , Neurons/drug effects , Sleep Apnea, Obstructive/drug therapy , Animals , Chronic Disease , Dendrites/metabolism , Dogs , Dose-Response Relationship, Drug , Male , Metoprolol/administration & dosage , Neurons/metabolism , Sleep Apnea, Obstructive/metabolism
3.
Cell Commun Signal ; 16(1): 91, 2018 11 29.
Article in English | MEDLINE | ID: mdl-30497498

ABSTRACT

BACKGROUND: The link between cardiac diseases and cognitive deterioration has been accepted from the concept of "cardiogenic dementia", which was proposed in the late 1970s. However, the molecular mechanism is unclarified. METHODS: The two animal models used in this study were cardiac-specific overexpression of microRNA-1-2 transgenic (Tg) mice and a myocardial infarction mouse model generated by left coronary artery ligation (LCA). First, we observed the microRNA-1 (miR-1) level and synaptic vesicles (SV) distribution in the hippocampus using in situ hybridization and transmission electron microscopy (TEM) and evaluated the expression of vesicle exocytosis related proteins by western blotting. Second, we used dual luciferase reporter assay as well as antagonist and miRNA-masking techniques to identify the posttranscriptional regulatory effect of miR-1 on the Snap25 gene. Third, FM1-43 staining was performed to investigate the effect of miR-1 on synaptic vesicle exocytosis. Lastly, we used GW4869 to inhibit the biogenesis and secretion of exosomes to determine the transportation effect of exosomes for miR-1 from the heart to the brain. RESULTS: Compared with the levels in age-matched WT mice, miR-1 levels were increased in both the hearts and hippocampi of Tg mice, accompanied by the redistribution of SVs and the reduction in SV exocytosis-related protein SNAP-25 expression. In vitro studies showed that SNAP-25 protein expression was down- or upregulated by miR-1 overexpression or inhibition, respectively, however, unchanged by miRNA-masking the 3'UTR of the Snap25 gene. SV exocytosis was inhibited by miR-1 overexpression, which could be prevented by co-transfection with an anti-miR-1 oligonucleotide fragment (AMO-1). The knockdown of miR-1 by hippocampal stereotaxic injection of AMO-1 carried by a lentivirus vector (lenti-pre-AMO-1) led to the upregulation of SNAP-25 expression and prevented SV concentration in the synapses in the hippocampi of Tg mice. The application of GW4869 significantly reversed the increased miR-1 level in the blood and hippocampi as well as reduced the SNAP-25 protein levels in the hippocampi of both Tg and LCA mice. CONCLUSION: The overexpression of miR-1 in the heart attenuated SV exocytosis in the hippocampus by posttranscriptionally regulating SNAP-25 through the transportation of exosomes. This study contributes to the understanding of the relationship between cardiovascular disease and brain dysfunction.


Subject(s)
Exocytosis , Exosomes/metabolism , Gene Expression Regulation , MicroRNAs/genetics , Myocardium/metabolism , Synaptic Vesicles/metabolism , Synaptosomal-Associated Protein 25/metabolism , Animals , Base Sequence , Hippocampus/cytology , Humans , Male , Mice , Mice, Inbred C57BL , Myocardium/cytology , Synaptosomal-Associated Protein 25/genetics , Transcription, Genetic
4.
Cell Physiol Biochem ; 50(4): 1601-1616, 2018.
Article in English | MEDLINE | ID: mdl-30359966

ABSTRACT

BACKGROUNDS/AIMS: It has been reported that myocardial infarction (MI) is a risk factor for vascular dementia. However, the molecular mechanism remains largely unknown. METHODS: MI mice were generated by ligation of the left coronary artery (LCA) for 4 weeks. Passive and active avoidance tests were performed to evaluate the cognitive ability of MI mice. A theta-burst stimulation (TBS) protocol was applied to elicit long-term potentiation (LTP) of the perforant pathway-dentate gyrus synapse (PP-DG). Western blot analysis was employed to assess protein levels. RESULTS: In this study, we demonstrated that after 4 weeks of MI, C57BL/6 mice had significantly impaired memory. Compared with the sham group, in vivo physiological recording in the MI group revealed significantly decreased amplitude of population spikes (PS) with no effect on the latency and duration of the stimulus-response curve. The amplitude of LTP was markedly decreased in the MI group compared with the sham group. Further examination showed that the expression of the TBS-LTP-related proteins BDNF, GluA1 and phosphorylated GluA1 were all decreased in the MI group compared with those in the sham group. Strikingly, all these changes were prevented by hippocampal stereotaxic injection of an anti-miR-1 oligonucleotide fragment carried by a lentivirus vector (lenti-pre-AMO-1). CONCLUSION: MI induced cognitive decline and TBS-LTP impairment, and decreased BDNF and GluA1 phosphorylation levels from overexpression of miR-1ated were involved in this process.


Subject(s)
Long-Term Potentiation/physiology , MicroRNAs/metabolism , Myocardial Infarction/pathology , Animals , Antagomirs/metabolism , Behavior, Animal , Brain-Derived Neurotrophic Factor/metabolism , Cells, Cultured , Dentate Gyrus/physiology , Disease Models, Animal , Electric Stimulation , Electrodes, Implanted , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Myocardial Infarction/metabolism , Neurons/cytology , Neurons/metabolism , Protein Interaction Maps , Receptors, AMPA/metabolism , Synapses/metabolism
5.
J Mol Cell Cardiol ; 120: 12-27, 2018 07.
Article in English | MEDLINE | ID: mdl-29775643

ABSTRACT

Cardiovascular diseases are risk factors for dementia, but the mechanisms remain elusive. Here, we report that myocardial infarction (MI) generated by the ligation of the left coronary artery (LCA) could lead to increased miR-1 levels in the hippocampus and blood with neuronal microtubule damage and decreased TPPP/p25 protein expression in the hippocampus. These changes could be prevented by a knockdown of miR-1 using hippocampal stereotaxic injections of anti-miR-1 oligonucleotide fragments carried by a lentivirus vector (lenti-pre-AMO-miR-1). TPPP/p25 protein was downregulated by miR-1 overexpression, upregulated by miR-1 inhibition, and unchanged by binding-site mutations or miR-masks, indicating that the TPPP/p25 gene was a potential target for miR-1. Additionally, the pharmacological inhibition of sphingomyelinase by GW4869 to inhibit exosome generation in the heart significantly attenuated the increased miR-1 levels in the hippocampi of transgenic (Tg) and MI mice. Collectively, the present study demonstrates that MI could directly lead to neuronal microtubule damage independent of MI-induced chronic brain hypoperfusion but involving the overexpression of miR-1 in the hippocampus that was transported by exosomes from infarcted hearts. This study reveals a novel insight into the molecular mechanisms of heart-to-brain communication at the miRNA level.


Subject(s)
Hippocampus/pathology , MicroRNAs/metabolism , Microtubules/metabolism , Myocardial Infarction/complications , Myocardial Infarction/metabolism , Analysis of Variance , Aniline Compounds/pharmacology , Animals , Benzylidene Compounds/pharmacology , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Exosomes/metabolism , Genetic Vectors/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/antagonists & inhibitors , Myocytes, Cardiac/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Phosphotransferases/metabolism , Rats , Rats, Sprague-Dawley , Transfection
6.
Cell Death Dis ; 8(6): e2850, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28569780

ABSTRACT

Impaired synaptic plasticity and neuron loss are hallmarks of Alzheimer's disease and vascular dementia. Here, we found that chronic brain hypoperfusion (CBH) by bilateral common carotid artery occlusion (2VO) decreased the total length, numbers and crossings of dendrites and caused neuron death in rat hippocampi and cortices. It also led to increase in N-terminal ß-amyloid precursor protein (N-APP) and death receptor-6 (DR6) protein levels and in the activation of caspase-3 and caspase-6. Further study showed that DR6 protein was downregulated by miR-195 overexpression, upregulated by miR-195 inhibition, and unchanged by binding-site mutation and miR-masks. Knockdown of endogenous miR-195 by lentiviral vector-mediated overexpression of its antisense molecule (lenti-pre-AMO-miR-195) decreased the total length, numbers and crossings of dendrites and neuron death, upregulated N-APP and DR6 levels, and elevated cleaved caspase-3 and caspase-6 levels. Overexpression of miR-195 using lenti-pre-miR-195 prevented these changes triggered by 2VO. We conclude that miR-195 is involved in CBH-induced dendritic degeneration and neuron death through activation of the N-APP/DR6/caspase pathway.


Subject(s)
Amyloid beta-Protein Precursor/genetics , Brain Ischemia/genetics , MicroRNAs/genetics , Neurons/metabolism , Receptors, Death Domain/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Base Sequence , Binding Sites , Brain Ischemia/metabolism , Brain Ischemia/pathology , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/pathology , Carotid Arteries/surgery , Caspase 3/genetics , Caspase 3/metabolism , Caspase 6/genetics , Caspase 6/metabolism , Cell Death , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cerebrovascular Disorders/pathology , Cerebrovascular Disorders/surgery , Dentate Gyrus/metabolism , Dentate Gyrus/pathology , Gene Expression Regulation , Male , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Neurons/pathology , Oligoribonucleotides, Antisense/genetics , Oligoribonucleotides, Antisense/metabolism , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Receptors, Death Domain/metabolism , Signal Transduction
7.
Int J Biol Sci ; 11(4): 390-403, 2015.
Article in English | MEDLINE | ID: mdl-25798059

ABSTRACT

Increased incidence of arrhythmias in women after menopause has been widely documented, which is considered to be related to estrogen (E2) deficiency induced cardiac electrophysiological abnormalities. However, its molecular mechanism remains incompletely clear. In the present study, we found cardiac conduction blockage in post-menopausal rats. Thereafter, the results showed that cardiac gap junctions were impaired and Connexin43 (Cx43) expression was reduced in the myocardium of post-menopausal rats. The phenomenon was also observed in ovariectomized (OVX) rats, which was attenuated by E2 supplement. Further study displayed that microRNA-23a (miR-23a) level was significantly increased in both post-menopausal and OVX rats, which was reversed by daily E2 treatment after OVX. Importantly, forced overexpression of miR-23a led to gap junction impairment and Cx43 downregulation in cultured cardiomyocytes, which was rescued by suppressing miR-23a by transfection of miR-23a specific inhibitory oligonucleotide (AMO-23a). GJA1 was identified as the target gene of miR-23a by luciferase assay and miRNA-masking antisense ODN (miR-Mask) assay. We also found that E2 supplement could reverse cardiac conduction blockage, Cx43 downregulation, gap junction remodeling and miR-23a upregulation in post-menopausal rats. These findings provide the evidence that miR-23a mediated repression of Cx43 participates in estrogen deficiency induced damages of cardiac gap junction, and highlights a new insight into molecular mechanism of post-menopause related arrhythmia at the microRNA level.


Subject(s)
Connexin 43/metabolism , Estrogens/metabolism , Gap Junctions/metabolism , MicroRNAs/metabolism , Animals , Arrhythmias, Cardiac/metabolism , Blotting, Western , Cells, Cultured , Connexin 43/genetics , Estrogens/deficiency , Estrogens/pharmacology , Female , Fluorescent Antibody Technique , Gap Junctions/drug effects , MicroRNAs/genetics , Microscopy, Electron, Transmission , Myocardium/metabolism , Postmenopause , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...