Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 36
1.
Biochem Biophys Res Commun ; 715: 150004, 2024 Jun 30.
Article En | MEDLINE | ID: mdl-38678784

Megakaryopoiesis and platelet production is a complex process that is underpotential regulation at multiple stages. Many long non-coding RNAs (lncRNAs) are distributed in hematopoietic stem cells and platelets. lncRNAs may play important roles as key epigenetic regulators in megakaryocyte differentiation and proplatelet formation. lncRNA NORAD can affect cell ploidy by sequestering PUMILIO proteins, although its direct effect on megakaryocyte differentiation and thrombopoiesis is still unknown. In this study, we demonstrate NORAD RNA is highly expressed in the cytoplasm during megakaryocyte differentiation. Interestingly, we identified for the first time that NORAD has a strong inhibitory effect on megakaryocyte differentiation and proplatelet formation from cultured megakaryocytes. DUSP6/ERK1/2 pathway is activated in response to NORAD knockdown during megakaryocytopoiesis, which is achieved by sequestering PUM2 proteins. Finally, compared with the wild-type control mice, NORAD knockout mice show a faster platelet recovery after severe thrombocytopenia induced by 6 Gy total body irradiation. These findings demonstrate lncRNA NORAD has a key role in regulating megakaryocyte differentiation and thrombopoiesis, which provides a promising molecular target for the treatment of platelet-related diseases such as severe thrombocytopenia.


Blood Platelets , Cell Differentiation , Dual Specificity Phosphatase 6 , Megakaryocytes , Mice, Knockout , RNA, Long Noncoding , Thrombopoiesis , Megakaryocytes/metabolism , Megakaryocytes/cytology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Animals , Thrombopoiesis/genetics , Blood Platelets/metabolism , Mice , Dual Specificity Phosphatase 6/metabolism , Dual Specificity Phosphatase 6/genetics , MAP Kinase Signaling System , Thrombocytopenia/genetics , Thrombocytopenia/metabolism , Thrombocytopenia/pathology , Humans , Mice, Inbred C57BL , Cells, Cultured
2.
Research (Wash D C) ; 6: 0187, 2023.
Article En | MEDLINE | ID: mdl-37426471

Monocyte-to-M0/M1 macrophage differentiation with unclear molecular mechanisms is a pivotal cellular event in many cardiovascular diseases including atherosclerosis. Long non-coding RNAs (lncRNAs) are a group of protein expression regulators; however, the roles of monocyte-lncRNAs in macrophage differentiation and its related vascular diseases are still unclear. The study aims to investigate whether the novel leukocyte-specific lncRNA Morrbid could regulate macrophage differentiation and atherogenesis. We identified that Morrbid was increased in monocytes and arterial walls from atherosclerotic mouse and from patients with atherosclerosis. In cultured monocytes, Morrbid expression was markedly increased during monocyte to M0 macrophage differentiation with an additional increase during M0 macrophage-to-M1 macrophage differentiation. The differentiation stimuli-induced monocyte-macrophage differentiation and the macrophage activity were inhibited by Morrbid knockdown. Moreover, overexpression of Morrbid alone was sufficient to elicit the monocyte-macrophage differentiation. The role of Morrbid in monocyte-macrophage differentiation was also identified in vivo in atherosclerotic mice and was verified in Morrbid knockout mice. We identified that PI3-kinase/Akt was involved in the up-regulation of Morrbid expression, whereas s100a10 was involved in Morrbid-mediated effect on macrophage differentiation. To provide a proof of concept of Morrbid in pathogenesis of monocyte/macrophage-related vascular disease, we applied an acute atherosclerosis model in mice. The results revealed that overexpression of Morrbid enhanced but monocyte/macrophage-specific Morrbid knockout inhibited the monocytes/macrophages recruitment and atherosclerotic lesion formation in mice. The results suggest that Morrbid is a novel biomarker and a modulator of monocyte-macrophage phenotypes, which is involved in atherogenesis.

3.
Sleep Med ; 109: 110-117, 2023 09.
Article En | MEDLINE | ID: mdl-37429109

OBJECTIVE: Although paradoxical insomnia is a prevalent subtype of chronic insomnia, the etiology of it is unclear. Contrary to complaints of little or no sleep, polysomnography (PSG) findings show that paradoxical insomnia patients have near normal sleep macrostructure. The purpose of this study is to determine the changes of microstructure and explore the etiology of paradoxical insomnia. METHODS: The PSG findings of 89 paradoxical insomnia patients were compared with those of 41 gender balanced healthy controls without sleep complaints. All subjects underwent nocturnal PSG recordings. Conventional PSG measures and microarousals were quantified and statistically analyzed. Receiver operating characteristic curve and correlation analysis were used to evaluate the potential of REM sleep microarousals and REM duration as indicators of paradoxical insomnia. RESULTS: Compared with the controls, paradoxical insomnia patients had no significant differences in sleep macrostructures. Statistical analysis showed that non-rapid eye movement (NREM) microarousals revealed no significant differences between paradoxical insomnia patients and controls. Noticeably, more spontaneous microarousals appeared in rapid eye movement (REM) stage for paradoxical insomnia patients. Based on receiver operating characteristic curve (ROC), the optimal cutoff value of REM sleep microarousals could predict paradoxical insomnia. Furthermore, a positive correlation between microarousals in REM sleep and the duration of REM sleep was presented in paradoxical insomnia patients. CONCLUSIONS: The frequency of REM microarousals and the duration of REM sleep could reflect the real sleep state of paradoxical insomnia patients. That suggested PSG investigation extended to microarousal could be helpful to understand the etiology in paradoxical insomnia.


Sleep Initiation and Maintenance Disorders , Sleep, REM , Humans , Sleep , Polysomnography , ROC Curve
4.
Toxicology ; 494: 153587, 2023 08 01.
Article En | MEDLINE | ID: mdl-37406984

In recent years, the role and mechanism of long non-coding RNA (LncRNA) in cardiovascular diseases have received increasing attention. The chemotherapy agent, doxorubicin (DOX), is one of the most effective drugs for various cancers, but its efficacy is limited by its cardiotoxicity. Therefore, further exploration is required for the molecular mechanism of DOX-induced cardiotoxicity. This study intended to investigate the role of LncRNA Non-coding RNA activated by DNA damage (NORAD) in DOX-induced cardiotoxicity, for which we adopted the AC16 human cardiomyocyte cell line for the exploration. The results showed that LncRNA NORAD knockdown could increase DOX-induced cardiomyocyte apoptosis and mitochondrial ROS level. LncRNA NORAD overexpression obtained reverse results, which further validated its role in DOX-induced cardiomyocyte apoptosis and mitochondrial ROS level. Moreover, cardiotoxicity was induced in both LncRNA NORAD-knockout and wild-type mice with DOX, showing that gene knockout aggravated pathologic lesions in the myocardial tissues of mice. Taken together, LncRNA NORAD affected DOX-induced cardiotoxicity via mitochondrial apoptosis, fission (PUM-MFF), and autophagy (p53-Parkin) pathways both in vivo and in vitro.


Cardiotoxicity , RNA, Long Noncoding , Mice , Humans , Animals , Cardiotoxicity/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Reactive Oxygen Species/metabolism , Doxorubicin/toxicity , Myocytes, Cardiac , Apoptosis
5.
Aging (Albany NY) ; 15(15): 7513-7532, 2023 07 29.
Article En | MEDLINE | ID: mdl-37517088

Long noncoding RNAs (lncRNAs) play important roles in the development of age-related macular degeneration (AMD). However, the effect of long non-coding RNA activated by DNA damage (NORAD) on AMD remains unknown. This study aimed to investigate the effect of NORAD on RPE cell senescence and degeneration. Irradiated adult retinal pigment epithelial cell line-19 (ARPE-19) and sodium iodate-treated mice were used as in vitro and in vivo AMD models. Results showed that irradiation-induced AMD characteristics of ARPE-19 and NORAD-knockdown aggravated cell cycle arrest in the G2/M phase, cell apoptosis and cell senescence along with the increased expression of phosphorylated P53 (p-P53) and P21. AMD factors C3, ICAM-1, APP, APOE, and VEGF-A were also increased by NORAD-knockdown. Moreover, NORAD-knockdown increased irradiation-induced reduction of mitochondrial homeostasis factors, (i.e., TFAM and POLG) and mitochondrial respiratory chain complex genes (i.e., ND1 and ND5) along with mitochondrial reactive oxygen species (ROS). We also identified a strong interaction of NORAD and PGC-1α and sirtuin 1 (SIRT1) in ARPE-19; that is, NORAD knockdown increases the acetylation of PGC-1α. In NORAD knockout mice, NORAD-knockout accelerated the sodium iodate-reduced retinal thickness reduction, function impairment and loss of retinal pigment in the fundus. Therefore, NORAD-knockdown accelerates retinal cell senescence, apoptosis, and AMD markers via PGC-1α acetylation, mitochondrial ROS, and the p-P53-P21signaling pathway, in which NORAD-mediated effect on PGC-1α acetylation might occur through the direct interaction with PGC-1α and SIRT1.


Macular Degeneration , RNA, Long Noncoding , Mice , Animals , Reactive Oxygen Species/metabolism , Oxidative Stress , RNA, Long Noncoding/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Sirtuin 1/genetics , Sirtuin 1/metabolism , Macular Degeneration/genetics , Macular Degeneration/metabolism , Retinal Pigments/metabolism , Retinal Pigments/pharmacology , Retinal Pigment Epithelium
6.
Front Immunol ; 14: 1157196, 2023.
Article En | MEDLINE | ID: mdl-37313407

Background: Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by loss of tolerance to self-antigen, autoantibody production, and abnormal immune response. Cuproptosis is a recently reported cell death form correlated with the initiation and development of multiple diseases. This study intended to probe cuproptosis-related molecular clusters in SLE and constructed a predictive model. Methods: We analyzed the expression profile and immune features of cuproptosis-related genes (CRGs) in SLE based on GSE61635 and GSE50772 datasets and identified core module genes associated with SLE occurrence using the weighted correlation network analysis (WGCNA). We selected the optimal machine-learning model by comparing the random forest (RF) model, support vector machine (SVM) model, generalized linear model (GLM), and the extreme gradient boosting (XGB) model. The predictive performance of the model was validated by nomogram, calibration curve, decision curve analysis (DCA), and external dataset GSE72326. Subsequently, a CeRNA network based on 5 core diagnostic markers was established. Drugs targeting core diagnostic markers were acquired using the CTD database, and Autodock vina software was employed to perform molecular docking. Results: Blue module genes identified using WGCNA were highly related to SLE initiation. Among the four machine-learning models, the SVM model presented the best discriminative performance with relatively low residual and root-mean-square error (RMSE) and high area under the curve (AUC = 0.998). An SVM model was constructed based on 5 genes and performed favorably in the GSE72326 dataset for validation (AUC = 0.943). The nomogram, calibration curve, and DCA validated the predictive accuracy of the model for SLE as well. The CeRNA regulatory network includes 166 nodes (5 core diagnostic markers, 61 miRNAs, and 100 lncRNAs) and 175 lines. Drug detection showed that D00156 (Benzo (a) pyrene), D016604 (Aflatoxin B1), D014212 (Tretinoin), and D009532 (Nickel) could simultaneously act on the 5 core diagnostic markers. Conclusion: We revealed the correlation between CRGs and immune cell infiltration in SLE patients. The SVM model using 5 genes was selected as the optimal machine learning model to accurately evaluate SLE patients. A CeRNA network based on 5 core diagnostic markers was constructed. Drugs targeting core diagnostic markers were retrieved with molecular docking performed.


Apoptosis , Autoimmune Diseases , Lupus Erythematosus, Systemic , MicroRNAs , Humans , Aflatoxin B1 , Benzo(a)pyrene , Lupus Erythematosus, Systemic/diagnosis , Lupus Erythematosus, Systemic/genetics , Molecular Docking Simulation , Copper
7.
Toxicol Lett ; 377: 29-37, 2023 Mar 15.
Article En | MEDLINE | ID: mdl-36739041

Recombinant human endostatin (rh-endostatin) is an anti-angiogenic drug, which is used for the treatment of advanced non-small-cell lung cancer (NSCLC) and other cancers. However, its side effects, especially the cardiotoxicity with unclear mechanisms limit its wide application in clinical practice. In this study, human cardiomyocyte cell line AC16 and human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) treated with different doses of rh-endostatin were used to analyze its effect on cardiac cell toxicity. The results revealed that rh-endostatin dose-dependently enhanced cardiomyocyte apoptosis through Apaf-1 apoptotic factor and apoptosis-related proteins such as p53. rh-endostatin-induced changes of mitochondrial function and mitophagy were involved in rh-endostatin-mediated cardiac cell toxicity. Rh-endostatin-induced cardiotoxicity was further verified in vivo in mice. Interestingly, Rh-endostatin-induced cardiotoxicity was inhibited by dihydromyricetin (DHM) both in cultured cells in vitro and in mouse hearts in vivo. The study provides new inside into rh-endostatin-induced cardiotoxicity and identified a novel potential medication DHM to overcome the serious adverse effect.


Carcinoma, Non-Small-Cell Lung , Drug-Related Side Effects and Adverse Reactions , Induced Pluripotent Stem Cells , Lung Neoplasms , Humans , Mice , Animals , Myocytes, Cardiac , Endostatins/toxicity , Endostatins/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Cardiotoxicity , Lung Neoplasms/metabolism , Mice, Inbred C57BL , Drug-Related Side Effects and Adverse Reactions/metabolism
8.
Stroke Vasc Neurol ; 8(3): 181-192, 2023 Jun.
Article En | MEDLINE | ID: mdl-36241224

BACKGROUND: Ischaemic stroke and other cardiovascular illnesses are characterised by abnormalities in the processes of thrombosis and haemostasis, which rely on platelet activity. In platelets, a wide variety of microRNAs (long non-coding RNA, lncRNAs) is found. Due to the absence of nuclear DNA in platelets, lncRNAs may serve as critical post-transcriptional regulators of platelet activities. However, research into the roles of lncRNAs in platelets is limited. OBJECTIVE: The purpose of this study is to learn more about the molecular mechanism by which MALAT1 affects platelet activity and thrombus formation. METHODS/RESULTS: The CD34+ megakaryocytes used in this research as an in vitro model for human megakaryocytes and platelets. Cell adhesion and spreading are enhanced in the absence and presence of agonists in CD34+ megakaryocytes subjected to MALAT1 knockdown (KD). The adhesion and activity of platelet-like particles produced by MALAT1 KD cells are significantly enhanced at rest and after thrombin activation. Thrombus development on a collagen matrix is also greatly enhanced in the microfluidic whole-blood perfusion model: platelets lacking MALAT1 exhibit elevated accumulation, distributing area and activity. In addition, MALAT1-deficient mice bleed less and form a stable occlusive thrombus more quickly than wild-type mice. PTEN and PDK1 regulated the activity of MALAT1 in platelets to carry out its PI3k/Akt/GSK-3ß signalling pathway-related function. CONCLUSION: The suppression of MALAT1 expression significantly increases platelet adhesion, spreading, platelet activity, and thrombus formation. lncRNAs may constitute a unique class of platelet function modulators.


Brain Ischemia , RNA, Long Noncoding , Stroke , Thrombosis , Animals , Humans , Mice , Blood Platelets/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Platelet Aggregation , Brain Ischemia/metabolism , Disease Models, Animal , Stroke/metabolism , Thrombosis/genetics
9.
Platelets ; 34(1): 2157382, 2023 Dec.
Article En | MEDLINE | ID: mdl-36550091

Noncoding RNAs (ncRNAs) are a group of RNA molecules that cannot encode proteins, and a better understanding of the complex interaction networks coordinated by ncRNAs will provide a theoretical basis for the development of therapeutics targeting the regulatory effects of ncRNAs. Platelets are produced upon the differentiation of hematopoietic stem cells into megakaryocytes, 1011 per day, and are renewed every 8-9 days. The process of thrombopoiesis is affected by multiple factors, in which ncRNAs also exert a significant regulatory role. This article reviewed the regulatory roles of ncRNAs, mainly microRNAs (miRNAs), circRNAs (circular RNAs), and long non-coding RNAs (lncRNAs), in thrombopoiesis in recent years as well as their roles in primary immune thrombocytopenia (ITP).


What is the context? Platelets are produced from progenitor cells named megakaryocytes (MKs) differentiated from bone marrow-derived hematopoietic stem cells (HSCs).Thrombopoiesis refers to the process by which platelet-producing MKs release platelet granules into peripheral blood under the shear force of blood flow for further development and maturation.The process of megakaryocytopoiesis and thrombopoiesis is affected by multiple factors, wherein some ncRNAs also exert a significant regulatory role.miRNAs/lncRNAs play a promising role in t primary immune thrombocytopenia (ITP).What is new? This article reviewed the regulatory roles of ncRNAs, mainly microRNAs (miRNAs), circRNAs (circular RNAs), and long non-coding RNAs (lncRNAs), in thrombopoiesis.This article also reviewed the roles of ncRNAs in ITP.What is the impact?Changes in ncRNA expression are associated with changes in the production of MKs, thrombopoiesis, and platelet function, which allows a new understanding of the pathogenesis of many congenital or acquired platelet-related diseases.


MicroRNAs , Thrombopoiesis , Humans , Thrombopoiesis/genetics , Blood Platelets/metabolism , Megakaryocytes/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Untranslated/metabolism , RNA, Untranslated/pharmacology
10.
Crit Rev Oncol Hematol ; 180: 103849, 2022 Dec.
Article En | MEDLINE | ID: mdl-36261116

Long non-coding RNA (LncRNA) is a class of non-coding RNA comprising more than 200 nucleotides. Several studies report that LncRNAs are widely involved in biological processes such as DNA methylation, histone modification, and chromatin remodeling. LncRNA interacts with DNA, RNA, and protein molecules thus regulating expression of target genes. LncRNAs are associated with occurrence and development of tumors, nervous systems, metabolic diseases, reproductive development, and cardiovascular diseases. Therefore, LncRNAs are implicated in progression of several diseases. Studies report that LncRNA plays a crucial role in regulation of gene expression during proliferation and differentiation stages of red blood cell development. LncRNA promotes development of hematopoietic stem cells and plays an important role in myeloid and lymphatic function. In this paper, studies on regulatory mechanism of LncRNA in blood cells and pathogenesis in hematological malignancies were reviewed.


Blood Cells , Hematologic Neoplasms , RNA, Long Noncoding , Humans , Blood Cells/metabolism , DNA Methylation , Hematologic Neoplasms/genetics , RNA, Long Noncoding/genetics
11.
Cells ; 11(14)2022 07 21.
Article En | MEDLINE | ID: mdl-35883699

Recently, long noncoding RNAs (lncRNAs) have been key regulators for both mRNAs and proteins in nucleated cells. However, the expression profiles of lncRNAs in non-nucleated cells such as platelets are currently unclear. In this study, we determined the expression profiles of lncRNAs in human platelets. We found that 6109 lncRNAs were expressed in human platelets. Interestingly, 338 lncRNAs were differentially expressed in hyperreactive and hyporeactive platelets. Bioinformatics' analysis revealed that these aberrantly expressed lncRNAs might be related to platelet activity and other platelet functions. To provide a proof of concept, we measured the expression levels of PARLncRNA-1, a down-regulated lncRNA of hyperreactive platelets, in platelets from 12 patients with acute myocardial infarction and their controls. We found that the lncRNA was also significantly down-regulated in platelets from patients, which was partially reversed by treatment with aspirin a known antiplatelet drug. LncRNAs may represent a novel class of modulators for platelet functions.


RNA, Long Noncoding , Gene Expression Profiling , Humans , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA, Messenger/genetics
12.
Stroke Vasc Neurol ; 7(6): 500-509, 2022 12.
Article En | MEDLINE | ID: mdl-35725243

BACKGROUND: Postsynaptic density protein-95 (PSD95) plays an important role in cerebral ischaemia injury, but its mechanism needs further research. This study aimed to explore the role of PSD95 in (Ang-(1-7))-Mas-mediated cerebral ischaemia protection and its regulatory mechanism. METHODS: Oxygen-glucose deprivation (OGD) neuron and rat middle cerebral artery occlusion (MCAO) models were used as in vitro and in vivo models, respectively. TAT-MAS9C was used to disrupt the interaction between PSD95 and Mas. The recombinant PSD95 adenovirus (Ad-PSD95) was used to overexpress PSD95 in neurons. RESULTS: Results showed that in OGD neurons, Ang-(1-7) could promote cell viability; reduce cell apoptosis; reduce the cell membrane localisation of Mas; upregulate the expression levels of pAKT, bcl-2 and I-κB; and downregulate the expression levels of Bax, pI-κB, tumour necrosis factor alpha and interleukin-1ß. TAT-MAS9C could enhance the aforementioned effects of Ang-(1-7). However, the PSD95 overexpression inhibited the aforementioned effects of Ang-(1-7). In the MCAO rat model, the 2,3,5-triphenyltetrazolium chloride (TTC) staining showed that Ang-(1-7) reduced the infarct volume. The Morris water maze test showed that the number of crossings over the platform area in the Ang-(1-7) group was significantly increased. TAT-MAS9C could promote the protective effect of Ang-(1-7). CONCLUSIONS: Results suggested that PSD95 alleviated the activation of AKT and the inhibition of nuclear factor kappa B signalling pathway mediated by the Ang-(1-7)-Mas complex, thereby reducing neuronal activity, increasing apoptosis and inhibiting the Ang-(1-7)-Mas-mediated cerebral ischaemia protection.


Brain Injuries , Brain Ischemia , Rats , Animals , Disks Large Homolog 4 Protein/metabolism , Brain Ischemia/metabolism , Signal Transduction , Apoptosis , Infarction, Middle Cerebral Artery/pathology
13.
Int Immunopharmacol ; 105: 108576, 2022 Apr.
Article En | MEDLINE | ID: mdl-35121224

INTRODUCTION: Obstructive sleep apnea-hypopnea syndrome (OSAHS) is a common sleep disorder that causes severe physiological disturbance. Evidence showed that OSAHS is an important associated comorbidity that can affect the survival of patients with pulmonary fibrosis. Until now, the potential mechanisms by which OSAHS accelerates the progression of lung fibrosis remain unclear. By constructing a pathological model of chronic intermittent hypoxia (CIH), the present study aimed to explore the pathological progress and potential mechanism of lung injury caused by OSAHS. Meanwhile, SMND-309 was given for treatment to evaluate its potential therapeutic role in CIH-induced lung injury. METHODS: Mice were randomly divided into (C57BL/6 wild-type) WT+(room air) RA, WT + CIH, SMND-309 + RA, and SMND-309 + CIH groups. The WT + CIH and SMND-309 + CIH groups were exposed to CIH condition for 12 weeks, while the other groups were processed in normal oxygen at the same time. The SMND-309 + RA and SMND-309 + CIH groups were intraperitoneally injected with SMND-309 at the last week of the modeling period. After 12 weeks of treatment, three mice from each group were perfused through the heart. Lung tissues were isolated, fixed, sectioned, and stained with H&E, Masson, and immunofluorescence stain. The rest of the lung tissues were harvested for Western blot and ELISA assays. RESULTS: CIH treatment increased the expression of pro-inflammatory factors (TNF-α and IL-6), resulting in lung tissue structure disorder, inflammatory cell infiltration, increased pulmonary capillary permeability, and pulmonary edema. The activation of the NF-κB signaling pathway played a crucial role in the process of inflammation. Noticeably, we observed M2 macrophage accumulation in the lung after CIH exposure, which promoted epithelial-mesenchymal transition (EMT) and pulmonary tissue fibrosis. ELISA assays showed the increased expression of TGF-ß, IL-10, and IL-4 in the CIH group. SMND-309 inhibited pulmonary inflammation, reduced the accumulation of M2 macrophage, alleviated collagen deposition andlung damage. CONCLUSION: CIH could induce chronic lung inflammation, promote the activation of M2 macrophages, trigger the occurrence of EMT, and accelerate the deposition of lung collagen, eventually leading to lung tissue damage. This study presents a possible explanation by which interstitial lung diseases, particularly idiopathic pulmonary fibrosis (IPF) with OSAHS, are usually associated with fast progress and poor prognosis. SMND-309 showed a good protective effect on CIH-induced lung damage.


Lung Injury , Animals , Mice , Caffeic Acids , Hypoxia/complications , Hypoxia/metabolism , Mice, Inbred C57BL
14.
Mol Med ; 28(1): 3, 2022 01 14.
Article En | MEDLINE | ID: mdl-35030992

The mechanisms of chronic intermittent hypoxia (CIH)-induced cognitive deficits remain unclear. Here, our study found that about 3 months CIH treatment induced lipid droplets (LDs) accumulation in hippocampal nerve and glia cells of C57BL/6 mice, and caused severe neuro damage including neuron lesions, neuroblast (NB) apoptosis and abnormal glial activation. Studies have shown that the neuronal metabolism disorders might contribute to the CIH induced-hippocampal impairment. Mechanistically, the results showed that pyruvate dehydrogenase complex E1ɑ subunit (PDHA1) and the pyruvate dehydrogenase complex (PDC) activator pyruvate dehydrogenase phosphatase 1 (PDP1) did not noticeable change after intermittent hypoxia. Consistent with those results, the level of Acetyl-CoA in hippocampus did not significantly change after CIH exposure. Interestingly, we found that CIH produced large quantities of ROS, which activated the JNK/SREBP/ACC pathway in nerve and glia cells. ACC catalyzed the carboxylation of Acetyl-CoA to malonyl-CoA and then more lipid acids were synthesized, which finally caused aberrant LDs accumulation. Therefore, the JNK/SREBP/ACC pathway played a crucial role in the cognitive deficits caused by LDs accumulation after CIH exposure. Additionally, LDs were peroxidized by the high level of ROS under CIH conditions. Together, lipid metabolic disorders contributed to nerve and glia cells damage, which ultimately caused behavioral dysfunction. An active component of Salvia miltiorrhiza, SMND-309, dramatically alleviated these injuries and improved cognitive deficits of CIH mice.


Acetyl-CoA Carboxylase/metabolism , Cognition , Eye Proteins/metabolism , GTP-Binding Protein Regulators/metabolism , Lipid Droplets/metabolism , Phosphoproteins/metabolism , Protein Phosphatase 2C/metabolism , Sleep Apnea, Obstructive/etiology , Sleep Apnea, Obstructive/metabolism , Sterol Regulatory Element Binding Proteins/metabolism , Animals , Caffeic Acids/pharmacology , Cognitive Dysfunction , Disease Models, Animal , Hippocampus/metabolism , Hypoxia/metabolism , Learning , Lipid Peroxidation , MAP Kinase Signaling System , Memory , Mice , Neurons , Reactive Oxygen Species/metabolism , Signal Transduction , Sleep Apnea, Obstructive/diagnosis , Sleep Apnea, Obstructive/drug therapy
15.
Front Genet ; 11: 571467, 2020.
Article En | MEDLINE | ID: mdl-33193674

Platelets are derived from megakaryocytes and play an important role in blood coagulation. By using high throughput sequencing, we have found that the long non-coding RNA (lncRNA) nuclear paraspeckle assembly transcript 1 (NEAT1) is abundant in platelets (GEO ID: 200097348). However, little is known about its role in regulating megakaryocyte differentiation and platelet activity. This study aims to clarify the effect of NEAT1 on MEG-01 differentiation and platelet-like particle (PLP) activity. NEAT1 in MEG-01 cells was knocked down by siRNA transfection. The adhesion of MEG-01 and PLP to collagen-coated coverslips was observed under a fluorescence microscope. Flow cytometry was used to investigate cell apoptosis, cell cycle, the levels of D41/CD42b on MEG-01 cells and CD62P on PLPs. Quantitative real-time polymerase chain reaction was used to detect NEAT1 and IL-8 expression levels. Western blot was used to measure the protein levels of Bcl-2, Bax, cleaved caspase-3, and IL-8. RNA-binding protein immunoprecipitation was used to detect the interaction of NEAT1 and splicing factor proline/glutamine-rich (SFPQ). Results showed that NEAT1 knockdown decreased the adhesion ability of thrombin-stimulated MEG-01 and PLP. The expression of CD62P on PLPs and CD41/CD42b on MEG-01 cells was inhibited by NEAT1 knockdown. In addition, NEAT1 knockdown inhibited cell apoptosis with increased Bcl2/Bax ratio and decreased cleaved caspase-3, and reduced the percentage of cells in the G0/G1 phase. Meanwhile, NEAT1 knockdown inhibited the expression of IL-8. A strong interaction of NEAT1 and SFPQ, a transcriptional repressor of IL-8, was identified. NEAT1 knockdown reduced the interaction between SFPQ and NEAT1.The results suggest that lncRNA NEAT1 knockdown decreases MEG-01 differentiation, PLP activity, and IL-8 level. The results also indicate that the regulation of NEAT1 on IL-8 may be realized via a direct interaction between NEAT1 and SFPQ.

16.
Oxid Med Cell Longev ; 2020: 4586839, 2020.
Article En | MEDLINE | ID: mdl-32566081

Stroke breaks the oxidative balance in the body and causes extra reactive oxygen species (ROS) generation, leading to oxidative stress damage. Long noncoding RNAs (lncRNAs) and microRNAs play pivotal roles in oxidative stress-mediated brain injury. Safflor yellow B (SYB) was able to effectively reduce ischemia-mediated brain damage by increasing antioxidant capacity and inhibiting cell apoptosis. In this study, we investigated the putative involvement of lncRNA AK046177 and microRNA-134 (miR-134) regulation in SYB against ischemia/reperfusion- (I/R-) induced neuronal injury. I/R and oxygen-glucose deprivation/reoxygenation (OGD/R) were established in vivo and in vitro. Cerebral infarct volume, neuronal apoptosis, and protein expression were detected. The effects of SYB on cell activity, cell respiration, nuclear factor erythroid 2-related factor 2 (Nrf2), antioxidant enzymes, and ROS were evaluated. I/R or OGD/R upregulated the expression of AK046177 and miR-134 and subsequently inhibited the activation and expression of CREB, which caused ROS generation and brain/cell injury. SYB attenuated the effects of AK046177, inhibited miR-134 expression, and promoted CREB activation, which in turn promoted Nrf2 expression, and then increased antioxidant capacities, improved cell respiration, and reduced apoptosis. We suggested that the antioxidant effects of SYB were driven by an AK046177/miR-134/CREB-dependent mechanism that inhibited this pathway, and that SYB has potential use in reducing or possibly preventing I/R-induced neuronal injury.


Brain Injuries/drug therapy , Brain Injuries/genetics , Brain Ischemia/drug therapy , Brain Ischemia/genetics , Chalcone/analogs & derivatives , Down-Regulation/genetics , RNA, Long Noncoding/genetics , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Brain Injuries/complications , Brain Injuries/physiopathology , Brain Ischemia/complications , Brain Ischemia/physiopathology , Caspase 3/metabolism , Cell Respiration/drug effects , Cell Survival/drug effects , Chalcone/pharmacology , Chalcone/therapeutic use , Cyclic AMP/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Glutathione Peroxidase/metabolism , Male , Malondialdehyde/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/ultrastructure , NADPH Oxidase 4/metabolism , NF-E2-Related Factor 2/metabolism , Phosphorylation , RNA, Long Noncoding/metabolism , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , bcl-2-Associated X Protein/metabolism
17.
Aging (Albany NY) ; 12(7): 6385-6400, 2020 04 08.
Article En | MEDLINE | ID: mdl-32267831

Long noncoding RNAs (lncRNAs) play important roles in the development of vascular diseases. However, the effect of lncRNA NORAD on atherosclerosis remains unknown. This study aimed to investigate the effect NORAD on endothelial cell injury and atherosclerosis. Ox-LDL-treated human umbilical vein endothelial cells (HUVECs) and high-fat-diet (HFD)-fed ApoE-/- mice were used as in vitro and in vivo models. Results showed that NORAD-knockdown induced cell cycle arrest in G0/G1 phase, aggravated ox-LDL-induced cell viability reduction, cell apoptosis, and cell senescence along with the increased expression of Bax, P53, P21 and cleaved caspase-3 and the decreased expression of Bcl-2. The effect of NORAD on cell viability was further verified via NORAD-overexpression. NORAD- knockdown increased ox-LDL-induced reactive oxygen species, malondialdehyde, p-IKBα expression levels and NF-κB nuclear translocation. Proinflammatory molecules ICAM, VCAM, and IL-8 were also increased by NORAD- knockdown. Additionally, we identified the strong interaction of NORAD and IL-8 transcription repressor SFPQ in HUVECs. In ApoE-/- mice, NORAD-knockdown increased the lipid disorder and atherosclerotic lesions. The results have suggested that lncRNA NORAD attenuates endothelial cell senescence, endothelial cell apoptosis, and atherosclerosis via NF-κB and p53-p21 signaling pathways and IL-8, in which NORAD-mediated effect on IL-8 might through the direct interaction with SFPQ.


Atherosclerosis/genetics , Endothelial Cells/metabolism , Interleukin-8/metabolism , Lipoproteins, LDL/metabolism , RNA, Long Noncoding/metabolism , Animals , Apoptosis , Cell Cycle Checkpoints , Cellular Senescence , Down-Regulation , Gene Knockdown Techniques , Human Umbilical Vein Endothelial Cells , Humans , Mice , NF-kappa B/metabolism , Oxidation-Reduction , Signal Transduction
18.
Int Immunopharmacol ; 81: 106284, 2020 Apr.
Article En | MEDLINE | ID: mdl-32058931

Chronic intermittent hypoxia (CIH) is the main symptom of obstructive sleep apnea syndrome (OSAS) and causes neural damage and cognitive deficits via neuroinflammation. Toll-like receptors (TLRs), especially TLR2, play an important role in neuroinflammation. However, the mechanisms by which TLR2 participates in CIH-induced cognitive deficits remain unclear. In this study, wild-type (WT) and TLR2 knock out (KO) mice were exposed to CIH for 8 weeks, and their social novelty discrimination, spatial learning and memory were severely compromised. Additionally, seriously damaged neurons and abnormally activated glia were observed in the CA1 and dentate gyrus (DG) areas of the hippocampus. Mechanistically, knocking out the TLR2 gene significantly alleviated these pathological changes and improved the behavioral performance. Together, these findings demonstrate that the TLR2-MyD88 signaling pathway might play an important role in CIH-induced cognitive deficits.


Cognitive Dysfunction/metabolism , Hypoxia/metabolism , Neurogenic Inflammation/metabolism , Neurons/pathology , Sleep Apnea, Obstructive/metabolism , Toll-Like Receptor 2/metabolism , Animals , Apoptosis , Behavior, Animal , Cognitive Dysfunction/immunology , Disease Models, Animal , Humans , Hypoxia/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurogenic Inflammation/immunology , Signal Transduction , Sleep Apnea, Obstructive/immunology , Toll-Like Receptor 2/genetics
19.
Pflugers Arch ; 472(3): 343-354, 2020 03.
Article En | MEDLINE | ID: mdl-32086614

Our previous study showed that the adipose afferent reflex (AAR) induced by chemical stimulation of white adipose tissue (WAT) increased sympathetic outflow and blood pressure. We also found that pro-inflammatory cytokines (PICs) in the hypothalamic paraventricular nucleus (PVN) potentiate the cardiac sympathetic afferent reflex in rats. However, the role of PICs in the PVN in regulating the AAR is still not clear. This study determined whether PICs in the PVN mediate the AAR in rats. The AAR was evaluated based on renal sympathetic nerve activity and mean arterial blood pressure in response to capsaicin injection into inguinal WAT (iWAT). PIC levels were measured by ELISA. PVN microinjection with the PICs tumor necrosis factor (TNF)-α or interleukin (IL)-1ß enhanced the AAR in a dose-dependent manner. Furthermore, pretreatment via the bilateral microinjection of the TNF-α-blocker etanercept or IL-1ß blocker IL-1ra into the PVN attenuated the AAR. In rats pretreated with TNF-α or IL-1ß, a sub-response dose of angiotensin II (Ang II) significantly enhanced the AAR. Moreover, delivery of the angiotensin II type 1(AT1) receptor antagonist losartan into the PVN attenuated the effects of TNF-α or IL-1ß on the AAR. In addition, stimulating either iWAT or retroperitoneal WAT with capsaicin increased TNF-α or IL-1ß levels in the PVN, but the injection of capsaicin into the jugular vein, skeletal muscle, and skin had no effects on TNF-α or IL-1ß levels in the PVN. These results suggest that TNF-α or IL-1ß and Ang II in the PVN synergistically enhance the AAR in rats.


Adipose Tissue, White/metabolism , Cytokines/metabolism , Inflammation/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Reflex/physiology , Adipose Tissue, White/drug effects , Angiotensin II/pharmacology , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Heart/drug effects , Heart/physiology , Interleukin-1beta/metabolism , Kidney/drug effects , Kidney/metabolism , Losartan/pharmacology , Paraventricular Hypothalamic Nucleus/drug effects , Rats , Rats, Sprague-Dawley , Reflex/drug effects , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/metabolism , Tumor Necrosis Factor-alpha/metabolism
20.
Biomolecules ; 10(2)2020 01 25.
Article En | MEDLINE | ID: mdl-31991759

Heart failure (HF) is a deadly disease that is difficult to accurately diagnose. Circular RNAs (circRNAs) are a novel class of noncoding RNAs that might play important roles in many cardiovascular diseases. However, their role in HF remains unclear. CircRNA microarrays were performed on plasma samples obtained from three patients with HF and three healthy controls. The profiling results were validated by quantitative reverse transcription polymerase chain reaction. The diagnostic value of circRNAs for HF was evaluated by receiver operating characteristic (ROC) curves. The expression profiles indicated that 477 circRNAs were upregulated and 219 were downregulated in the plasma of patients with HF compared with healthy controls. Among the dysregulated circRNAs, hsa_circ_0112085 (p = 0.0032), hsa_circ_0062960 (p = 0.0006), hsa_circ_0053919 (p = 0.0074) and hsa_circ_0014010 (p = 0.025) showed significantly higher expression in patients with HF compared with healthy controls. The area under the ROC curve for hsa_circ_0062960 for HF diagnosis was 0.838 (p < 0.0001). Correlation analysis showed that the expression of hsa_circ_0062960 was highly correlated with B-type natriuretic peptide (BNP) serum levels. Some differential circRNAs were found to be related to platelet activity by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses. The landscape of circRNA expression profiles may play a role in HF pathogenesis and improve our understanding of platelet function in HF. Moreover, hsa_circ_0062960 has potential as a novel diagnostic biomarker for HF.


Gene Expression Regulation/genetics , Heart Failure/blood , RNA, Circular/blood , Aged , Female , Heart Failure/genetics , Heart Failure/pathology , Humans , Male , Microarray Analysis , Middle Aged , Natriuretic Peptide, Brain/blood , Natriuretic Peptide, Brain/genetics , Platelet Activation/genetics
...