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.
Entropy (Basel) ; 25(12)2023 Nov 24.
Article in English | MEDLINE | ID: mdl-38136462

ABSTRACT

Wearable technologies face challenges due to signal instability, hindering their usage. Thus, it is crucial to comprehend the connection between dynamic patterns in photoplethysmography (PPG) signals and cardiovascular health. In our study, we collected 401 multimodal recordings from two public databases, evaluating hemodynamic conditions like blood pressure (BP), cardiac output (CO), vascular compliance (C), and peripheral resistance (R). Using irregular-resampling auto-spectral analysis (IRASA), we quantified chaotic components in PPG signals and employed different methods to measure the fractal dimension (FD) and entropy. Our findings revealed that in surgery patients, the power of chaotic components increased with vascular stiffness. As the intensity of CO fluctuations increased, there was a notable strengthening in the correlation between most complexity measures of PPG and these parameters. Interestingly, some conventional morphological features displayed a significant decrease in correlation, indicating a shift from a static to dynamic scenario. Healthy subjects exhibited a higher percentage of chaotic components, and the correlation between complexity measures and hemodynamics in this group tended to be more pronounced. Causal analysis showed that hemodynamic fluctuations are main influencers for FD changes, with observed feedback in most cases. In conclusion, understanding chaotic patterns in PPG signals is vital for assessing cardiovascular health, especially in individuals with unstable hemodynamics or during ambulatory testing. These insights can help overcome the challenges faced by wearable technologies and enhance their usage in real-world scenarios.

2.
Neural Regen Res ; 18(3): 609-617, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36018185

ABSTRACT

Postoperative cognitive dysfunction (POCD) is a common surgical complication. Diabetes mellitus (DM) increases risk of developing POCD after surgery. DM patients with POCD seriously threaten the quality of patients' life, however, the intrinsic mechanism is unclear, and the effective treatment is deficiency. Previous studies have demonstrated neuronal loss and reduced neurogenesis in the hippocampus in mouse models of POCD. In this study, we constructed a mouse model of DM by intraperitoneal injection of streptozotocin, and then induced postoperative cognitive dysfunction by transient bilateral common carotid artery occlusion. We found that mouse models of DM-POCD exhibited the most serious cognitive impairment, as well as the most hippocampal neural stem cells (H-NSCs) loss and neurogenesis decline. Subsequently, we hypothesized that small extracellular vesicles secreted by induced pluripotent stem cell-derived mesenchymal stem cells (iMSC-sEVs) might promote neurogenesis and restore cognitive function in patients with DM-POCD. iMSC-sEVs were administered via the tail vein beginning on day 2 after surgery, and then once every 3 days for 1 month thereafter. Our results showed that iMSC-sEVs treatment significantly recovered compromised proliferation and neuronal-differentiation capacity in H-NSCs, and reversed cognitive impairment in mouse models of DM-POCD. Furthermore, miRNA sequencing and qPCR showed miR-21-5p and miR-486-5p were the highest expression in iMSC-sEVs. We found iMSC-sEVs mainly transferred miR-21-5p and miR-486-5p to promote H-NSCs proliferation and neurogenesis. As miR-21-5p was demonstrated to directly targete Epha4 and CDKN2C, while miR-486-5p can inhibit FoxO1 in NSCs. We then demonstrated iMSC-sEVs can transfer miR-21-5p and miR-486-5p to inhibit EphA4, CDKN2C, and FoxO1 expression in H-NSCs. Collectively, these results indicate significant H-NSC loss and neurogenesis reduction lead to DM-POCD, the application of iMSC-sEVs may represent a novel cell-free therapeutic tool for diabetic patients with postoperative cognitive dysfunction.

3.
Bioengineered ; 13(5): 12726-12737, 2022 05.
Article in English | MEDLINE | ID: mdl-35611768

ABSTRACT

Evidence suggests circRBMS1 regulates mRNA to mediate cell apoptosis, inflammation, and oxidative stress in different diseases. MST1 is reported to be the target and activator of apoptosis-related molecules and signaling pathways. Hence, the present study aims to investigate the role of circ-RBMS1/miR-2355-3p/MST1 in the development of I/R injury. In vitro experiments showed increased circ-RBMS1 and decreased miR-2355-3p in H/R-induced HCMs. CircRBMS1 served as a sponge for miR-2355-3p and miR-2355-3p targeted MST1. Furthermore, knockout of circRBMS1 attenuated cell apoptosis, oxidized stress, and inflammation in H/R-induced HCMs. In vivo experiments indicated circRBMS1 knockdown attenuated cardiac function damage, cell apoptosis, oxidative stress injury and inflammatory response through miR-2355-3p/MST1 axis in mice. In summary, these results demonstrated circRBMS1 played a protective role in myocardial I/R injury though inhibition of miR-2355-3p/MST1 axis. It might provide a new therapeutic target for cardiac I/R injury.


Subject(s)
MicroRNAs , Myocardial Reperfusion Injury , Protein Serine-Threonine Kinases , RNA, Circular , Animals , Apoptosis/genetics , Inflammation/metabolism , Mice , Mice, Knockout , MicroRNAs/genetics , MicroRNAs/metabolism , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/metabolism , Protein Serine-Threonine Kinases/genetics , RNA, Circular/genetics
4.
Lab Invest ; 102(5): 505-514, 2022 05.
Article in English | MEDLINE | ID: mdl-35066566

ABSTRACT

LncRNAs and miRNAs are correlated with the pathogenesis of myocardial ischemia-reperfusion injury (MIRI). Whether lncRNA ROR or miR-185-5p plays a crucial role in MIRI is still unclear. In in-vitro, human cardiac myocytes (HCMs) were treated with hypoxia/reoxygenation (H/R). Wistar rats were used to set up an in-vitro I/R model by means of recanalization after ligation. Evaluation of the myocardial injury marker lactate dehydrogenase (LDH) in HCMs cells was performed. The expression of miR-185-5p and ROR, IL-1ß, and IL-18 were detected by qRT-PCR. ELISA was also performed to evaluate the secretion of IL-1ß and IL-18. Western blotting was carried out to determine CDK6, NLRP3, GSDMD-N, ASC, and cleaved-caspase1 protein expression. The relationship between miR-185-5p and CDK6 or ROR was confirmed by a dual-luciferase reporter assay. Our findings revealed that H/R treated HCMs showed a significantly decreased miR-185-5p expression and increased expression of CDK6 and ROR. ROR knockdown reduced H/R induced pyroptosis and inflammation, while knockdown of miR-185-5p accelerated the effect. Furthermore, miR-185-5p was negatively regulated and absorbed by ROR in HCMs. Overexpression of miR-185-5p reversed the H/R-induced cell pyroptosis and upregulation of LDH, IL-1ß, and IL-18. In HCMs, miR-185-5p was also negatively regulated and related to CDK6 expression. Moreover, overexpression of CDK6 significantly inhibited the effects of miR-185-5p mimics on the inflammatory response and pyroptosis of HCMs. Knockdown of ROR alleviated H/R-induced myocardial injury by elevating miR-185-5p and inhibiting CDK6 expression. Taken together, our results show that the ROR/miR-185-5p/CDK6 axis modulates cell pyroptosis induced by H/R and the inflammatory response of HCMs.


Subject(s)
MicroRNAs , Myocardial Reperfusion Injury , RNA, Long Noncoding , Animals , Hypoxia , Interleukin-18 , MicroRNAs/genetics , MicroRNAs/metabolism , Myocardial Reperfusion Injury/genetics , RNA, Long Noncoding/genetics , Rats , Rats, Wistar
5.
Eur J Neurosci ; 54(3): 5000-5015, 2021 08.
Article in English | MEDLINE | ID: mdl-34192824

ABSTRACT

Ligustilide exerts potential neuroprotective effects against various cerebral ischaemic insults and neurodegenerative disorders. However, the function and mechanisms of LIG-mediated hippocampal neural stem cells (H-NSCs) activation as well as cognitive recovery in the context of post-operative cognitive dysfunction (POCD) remain elusive and need to be explored. Mice were subjected to transient global cerebral ischaemia and reperfusion (tGCI/R) injury and treated with LIG (80 mg/kg) or vehicle for 1 month. Morris water maze test and western blot were employed to assess cognitive function. Nissl staining and immunofluorescence (IF) staining were used to detect H-NSCs proliferation and neurogenesis in hippocampus. Subsequently, primary H-NSCs were treated with LIG, and the level of H-NSCs proliferation and neuronal-differentiation was examined by IF staining for Edu and ß-Tubulin III. The protein levels of ERK1/2, ß-catenin, NICD, TLR4, Akt and FoxO1 were examined using western blotting. Finally, pretreatment with the ERK agonist SCH772984 was performed to observe the change in ERK expression. LIG treatment promoted H-NSCs proliferation and neurogenesis, increased the number of neurons in the hippocampal subfields, and ultimately reversed cognitive impairment in tGCI/R injury. Furthermore, LIG also promoted primary H-NSCs proliferation and neuronal-differentiation, as well as ERK1/2 phosphorylation. Pretreatment with SCH772984 effectively reversed the ability of LIG to induce ERK1/2 phosphorylation and promote H-NSCs proliferation and neuronal-differentiation. LIG can promote cognitive recovery after tGCI/R injury by activating ERK1/2 in H-NSCs to promote their proliferation and neurogenesis in the hippocampus. Therefore, LIG has potential for use in the prevention and/or treatment of POCD.


Subject(s)
Neural Stem Cells , Postoperative Cognitive Complications , 4-Butyrolactone/analogs & derivatives , Animals , Cell Proliferation , Cognition , Hippocampus , Mice , Neurogenesis
6.
Neurosci Lett ; 729: 134991, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32330488

ABSTRACT

Hippocampal neural stem cells (H-NSCs) self-renewal and neurogenesis decrease with aging, but the intrinsic mechanism is unclear. In the current study, we detected the expression level of 8 conserved long intergenic noncoding RNAs (lincRNAs) in H-NSCs during aging, and investigated the function and mechanism of lincRNAs in regulating of H-NSCs. We found the proliferation and neuronal-differentiation capacities of H-NSCs reduced with aging and that this effect was accompanied by an increase in linc-FOXD3. Linc-FOXD3 knockdown improved H-NSCs proliferation and neuronal-differentiation capacities. Further mechanistic studies revealed that the effect of linc-FOXD3 knockdown on H-NSCs phenotypes was partially mediated by the up-regulation of Wnt/ß-catenin pathway. Thus, our study provides evidence that linc-FOXD3 could be a promising therapeutic target for the recovery of H-NSCs function during aging.


Subject(s)
Forkhead Transcription Factors/genetics , Hippocampus/drug effects , Neural Stem Cells/drug effects , RNA, Long Noncoding/pharmacology , Repressor Proteins/genetics , Wnt Signaling Pathway/drug effects , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Hippocampus/metabolism , Male , Mice, Inbred C57BL , Neural Stem Cells/metabolism , Neurogenesis/drug effects , RNA, Long Noncoding/genetics , Up-Regulation , Wnt Proteins/metabolism , Wnt Signaling Pathway/genetics , beta Catenin/drug effects , beta Catenin/metabolism
7.
Cell Biol Int ; 39(7): 816-23, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25689847

ABSTRACT

NAF-1 (nutrient-deprivation autophagy factor-1), an autophagy-related gene-related (ATG) protein, has been implicated in the autophagic pro-survival response. However, its role in autophagy has not been examined in the cardiomyocytes. In this study, we found that nutritional stress (NS) induced by glucose deprivation strongly induced autophagy in cultured neonatal rat cardiomyocytes, which was associated with NAF-1 down-regulation in cardiomyocytes under NS conditions. Furthermore, we demonstrate that ectopic expression of NAF-1 was sufficient to inhibit autophagy in cardiomyocytes under glucose deprivation conditions. Moreover, results of the co-immunoprecipitation assay indicate that NAF-1 antagonized autophagy by promoting the interaction between Beclin1 and Bcl-2 in NS-induced cardiomyocytes. Importantly, our results indicate that overexpression of NAF-1 significantly inhibited AMPK activity and protected cardiomyocytes from NS-induced cell death. Taken together, these data show that ectopic expression of NAF-1 antagonizes the degree of autophagy in cardiomyocytes and enhances cell survival during starvation conditions.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Autophagy , Membrane Proteins/metabolism , Myocytes, Cardiac/cytology , Signal Transduction , Starvation/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Beclin-1 , Myocytes, Cardiac/metabolism , Protein Interaction Maps , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , TOR Serine-Threonine Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...