Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
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
3.
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
4.
J Colloid Interface Sci ; 385(1): 1-7, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22885113

ABSTRACT

A complex nanostructure of rutile TiO(2) microspheres with ultrasmall nanorods on surfaces was prepared by a simple solvothermal method. This complex nanostructure is different from the hierarchical structure of microspheres composed of nanorods. The obtained complex nanostructure possesses an epitaxy-like interface between the nanorod-shell and the sphere-core, which often provides superior physical and chemical properties. The size and morphology of the obtained rutile TiO(2) complex nanostructure were observed by scanning electron microscopy and transmission electron microscopy (TEM). Their intrinsic crystallography was characterized by X-ray diffraction, high-resolution TEM, and selected area electron diffraction. Controlled experiments were designed using varied temperatures and assistant reagent compositions to study their influences on the crystal phase and morphology of TiO(2). The formation process of this complex nanostructure was determined via time-dependent experiments. Its photoluminescence spectra showed the strongest emission at about 400 nm with a blue-shift. The photocatalytic experiments demonstrated the obtained complex nanostructure had the highest catalytic efficiency in the five TiO(2) samples with different morphologies.

SELECTION OF CITATIONS
SEARCH DETAIL
...