Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 19 de 19
1.
Sci Total Environ ; 922: 171291, 2024 Apr 20.
Article En | MEDLINE | ID: mdl-38423311

6-PPD quinone (6-PPDQ), an emerging environmental pollutant, is converted based on 6-PPD via ozonation. However, a systematic evaluation on possible neurotoxicity of long-term and low-dose 6-PPDQ exposure and the underlying mechanism remain unknown. In the present work, 0.1-10 µg/L 6-PPDQ was added to treat Caenorhabditis elegans for 4.5 days, with locomotion behavior, neuronal development, sensory perception behavior, neurotransmitter content, and levels of neurotransmission-related genes being the endpoints. 6-PPDQ exposure at 0.1-10 µg/L significantly reduced locomotion behavior, and that at 1-10 µg/L decreased sensory perception behavior in nematodes. Moreover, 6-PPDQ exposure at 10 µg/L notably induced damage to the development of dopaminergic, glutamatergic, serotonergic, and GABAergic neurons. Importantly, nematodes with chronic 6-PPDQ exposure at 10 µg/L were confirmed to suffer obviously decreased dopamine, serotonin, glutamate, dopamine, and GABA contents and altered neurotransmission-related gene expression. Meanwhile, the potential binding sites of 6-PPDQ and neurotransmitter synthesis-related proteins were further shown by molecular docking method. Lastly, Pearson's correlation analysis showed that locomotion behavior and sensory perception behavior were positively correlated with the dopaminergic, serotonergic, glutamatergic, and GABAergic neurotransmission. Consequently, 6-PPDQ exposure disturbed neurotransmitter transmission, while such changed molecular foundation for neurotransmitter transmission was related to 6-PPDQ toxicity induction. The present work sheds new lights on the mechanisms of 6-PPDQ and its possible neurotoxicity to organisms at environmentally relevant concentrations.


Caenorhabditis elegans , Dopamine , Animals , Molecular Docking Simulation , GABAergic Neurons/metabolism , Neurotransmitter Agents/metabolism , Benzoquinones/metabolism
2.
Biochem Biophys Res Commun ; 670: 36-46, 2023 08 30.
Article En | MEDLINE | ID: mdl-37271038

Physiological high shear stress (HSS), a frictional force generated by flowing blood, is essential for endothelial homeostasis under normal physiological conditions. HSS suppresses atherosclerosis by inhibiting endothelial inflammation. However, the molecular mechanisms underlying this process have not been fully elucidated. Here, we report that HSS downregulated the mRNA and protein levels of ras homolog family member J (RHOJ) in endothelial cells (ECs). Silencing endogenous RHOJ expression decreased the mRNA and protein levels of proinflammatory vascular cell adhesion molecule 1 (VCAM-1) and intercellular cell adhesion molecule 1 (ICAM-1) in ECs, leading to a reduction in monocyte adhesion to ECs. Conversely, the overexpression of RHOJ had the opposite effect. RNA-sequencing analysis uncovered several differentially expressed genes (such as yes-associated protein 1 (YAP1),heme oxygenase-1 (HO1), and monocyte chemoattractant protein-1 (MCP1)) and pathways (such as nuclear factor-kappa B (NF-κB), fluid shear stress and atherosclerosis, and cell adhesion pathways) as RHOJ targets. Additionally, HSS was observed to alleviate endothelial inflammation by inhibiting RHOJ expression. Finally, methylated RNA immunoprecipitation sequencing (MeRIP-seq) illustrated that fluid shear stress regulates RHOJ expression in an N6-methyladenosine (m6A)-dependent manner. Mechanistically, the RNA m6A writer, methyltransferase 3 (METTL3), and the RNA m6A readers, YTH N6-methyladenosine RNA-binding protein F 3 (YTHDF3) and YTH N6-methyladenosine RNA-binding protein C 1/2 (YTHDC1/2), are involved in this process. Taken together, our data demonstrate that HSS-induced downregulation of RHOJ contributes to endothelial homeostasis by suppressing endothelial inflammation and that RHOJ inhibition in ECs is a promising therapeutic strategy for endothelial dysfunction.


Atherosclerosis , Endothelial Cells , Humans , Endothelial Cells/metabolism , Inflammation/genetics , Inflammation/metabolism , RNA/metabolism , RNA, Messenger/metabolism , Atherosclerosis/genetics , Atherosclerosis/metabolism , RNA-Binding Proteins/metabolism , Methyltransferases/metabolism , rho GTP-Binding Proteins/metabolism
3.
Pharmacol Ther ; 248: 108455, 2023 08.
Article En | MEDLINE | ID: mdl-37257760

BACKGROUND: Cancer is a major burden of disease worldwide and increasing evidence shows that inflammation contributes to cancer development and progression. Eicosanoids are derived from dietary polyunsaturated fatty acids, such as arachidonic acid (AA), and are mainly produced by a series of enzymatic pathways that include cyclooxygenase (COX), lipoxygenase (LOX), and cytochrome P-450 epoxygenase (CYP). Eicosanoids consist of at least several hundred individual molecules and play important roles in the inflammatory response and inflammation-related cancers. SCOPE AND APPROACH: Dietary sources of AA and biosynthesis of eicosanoids from AA through different metabolic pathways are summarized. The bioactivities of eicosanoids and their potential molecular mechanisms on inflammation and cancer are revealed. Additionally, current challenges and limitations in eicosanoid research on inflammation-related cancer are discussed. KEY FINDINGS AND CONCLUSIONS: Dietary AA generates a large variety of eicosanoids, including prostaglandins, thromboxane A2, leukotrienes, cysteinyl leukotrienes, lipoxins, hydroxyeicosatetraenoic acids (HETEs), and epoxyeicosatrienoic acids (EETs). Eicosanoids exert different bioactivities and mechanisms involved in the inflammation and related cancer developments. A deeper understanding of eicosanoid biology may be advantageous in cancer treatment and help to define cellular targets for further therapeutic development.


Eicosanoids , Neoplasms , Humans , Eicosanoids/metabolism , Arachidonic Acid/metabolism , Neoplasms/metabolism , Leukotrienes , Inflammation/metabolism , Cyclooxygenase 2
4.
Crit Rev Food Sci Nutr ; : 1-17, 2022 Oct 13.
Article En | MEDLINE | ID: mdl-36226711

Proanthocyanidins, widespread in natural plant sources, are bioactive substances that exhibit broad benefits to human health. Of note, proanthocyanidins have been reported to lower blood pressure and prevent hypertension, but a critical review of this is lacking. In this review, information on the basic structures and absorption of dietary proanthocyanidins as well as their bioactivities and related mechanisms on the lowering of blood pressure derived via in vivo and clinical studies are summarized. Clinical studies have shown that proanthocyanidins have a pronounced blood pressure-lowering effect, effectively preventing hypertension and reducing the occurrence of cardiovascular and cerebrovascular diseases. The potential mechanisms, which are herein reviewed in detail, involve the improvement of vascular function, reduction of oxidative stress and inflammation, and modulation of lipid metabolism. Taken together, this work provides information for a better understanding of the antihypertensive effects of proanthocyanidins, which may promote their use to reduce the risk of developing hypertension.

5.
Exp Cell Res ; 418(1): 113262, 2022 09 01.
Article En | MEDLINE | ID: mdl-35714940

Pathological cardiac hypertrophy is an independent risk factor for the development of heart failure. Long noncoding RNAs (lncRNAs), an emerging class of non-protein-coding transcripts, are involved in regulation of multiple cardiac diseases through diverse molecular mechanism, whereas the role of cytoplasmic lncRNAs in regulating cardiac hypertrophy remains unclear. In this study, we identified a novel and functional long noncoding RNA Gm17501, which was predominantly expressed in the cytoplasm of cardiomyocytes. The expression level of lncRNA Gm17501 was altered in cardiac hypertrophy induced by pressure overload and phenylephrine treatment. Moreover, lncRNA Gm17501 expression was decreased in the heart tissue of patients with heart failure. Silencing lncRNA Gm17501 aggravated cardiac hypertrophy under pathological stress. Inhibition of lncRNA Gm17501 did not alter the expression of nearby genes but decreased mRNA level of calcium handling proteins which were involved in cardiac contraction. Therefore, the cytoplasmic lncRNA Gm17501 might protect cardiomyocytes against hypertrophy, possibly by maintaining calcium signaling pathway.


Heart Failure , RNA, Long Noncoding , Animals , Cardiomegaly/pathology , Gene Expression Regulation , Heart Failure/metabolism , Humans , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
6.
Arterioscler Thromb Vasc Biol ; 42(5): 644-658, 2022 05.
Article En | MEDLINE | ID: mdl-35296150

BACKGROUND: N6-methyladenosine (m6A) plays a critical role in various biological processes. However, no study has addressed the role of m6A modification in the statin-induced protection of endothelial cells (ECs). METHODS: Quantitative real-time polymerase chain reaction and Western blotting analyses were used to study the expression of m6A regulatory genes in atorvastatin-treated ECs. Gain- and loss-of-function assays, methylated RNA immunoprecipitation analysis, and dual-luciferase reporter assays were performed to clarify the function of FTO (fat mass and obesity-associated protein) in ECs. RESULTS: Atorvastatin decreased FTO protein expression in ECs. The knockdown of FTO enhanced the mRNA and protein expression of KLF2 (Kruppel-like factor 2) and eNOS (endothelial NO synthase) but attenuated TNFα (tumor necrosis factor alpha)-induced VCAM-1 (vascular cell adhesion molecule 1) and ICAM-1 (intercellular adhesion molecule 1) expression, as well as the adhesion of monocytes to ECs. Conversely, FTO overexpression significantly upregulated the mRNA and protein levels of VCAM-1 and ICAM-1, downregulated those of KLF2 and eNOS, and strongly attenuated the atorvastatin-mediated induction of KLF2 and eNOS expression. Subsequent investigations demonstrated that KLF2 and eNOS are functionally critical targets of FTO. Mechanistically, FTO interacted with KLF2 and eNOS transcripts and regulated their expression in an m6A-dependent manner. After FTO silencing, KLF2 and eNOS transcripts with higher levels of m6A modification in their 3' untranslated regions were captured by YTHDF3 (YT521-B homology m6A RNA-binding protein 3), resulting in mRNA stabilization and the induction of KLF2 and eNOS protein expression. CONCLUSIONS: FTO might serve as a novel molecular target to modulate endothelial function in vascular diseases.


Hydroxymethylglutaryl-CoA Reductase Inhibitors , Adenosine/analogs & derivatives , Adenosine/metabolism , Adenosine/pharmacology , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/genetics , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Atorvastatin/pharmacology , Endothelial Cells/metabolism , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Intercellular Adhesion Molecule-1 , Obesity/genetics , RNA, Messenger/genetics , Vascular Cell Adhesion Molecule-1
7.
J Cardiovasc Transl Res ; 15(3): 514-523, 2022 06.
Article En | MEDLINE | ID: mdl-35229250

Stem cell-based therapy for ischemic heart disease (IHD) has become a promising but controversial strategy during the past two decades. The fate and effects of stem cells engrafted into ischemia myocardium are still not fully understood. Stem cell-derived exosomes, a subcategory of extracellular vesicles with nano size, have been considered as an efficient and safe transporter for microRNAs (miRNAs) and a central mediator of the cardioprotective potentials of the parental cells. Hypoxia, pharmacological intervention, and gene manipulation could alter the exosomal miRNAs cargos from stem cells and promote therapeutic potential. Furthermore, several bioengineering methods were also successfully applied to modify miRNAs content and components of exosomal membrane proteins recently. In this review, we outline relevant results about exosomal miRNAs from stem cells and focus on the current strategies to promote their therapeutic efficiency in IHD.


Exosomes , Extracellular Vesicles , MicroRNAs , Myocardial Ischemia , Exosomes/genetics , Exosomes/metabolism , Extracellular Vesicles/metabolism , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Myocardial Ischemia/genetics , Myocardial Ischemia/metabolism , Myocardial Ischemia/therapy , Stem Cells/metabolism
8.
Bioengineered ; 13(3): 4702-4716, 2022 03.
Article En | MEDLINE | ID: mdl-35188876

Extracellular vesicles (EVs) are a promising new therapeutic platform. However, the low cargo-loading efficiency limits their clinical translation. In this study, we developed a high-yield EV cargo-loading device and explored its ability to encapsulate gene editing proteins. A series of fusion protein-based systems were constructed and their cargo loading efficiencies were compared by a NanoGlo luciferase assay. A myristoylated (Myr) peptide tag cloned from the N-terminal region of charged multivesicular body protein 6 (CHMP6), termed Myr(CHMP6), outcompeted CD9, ARRDC1, and other short polypeptides as an active packaging device. As determined by nanoparticle tracking analysis and transmission electron microscopy, the overexpression of Myr(CHMP6) increased small EV (sEV) production in Lenti-X 293T  cells without altering sEV morphology. The high passive packaging efficiency of Myr(CHMP6) was also elucidated for unmodified cargo loading. Western blotting revealed that Myr(CHMP6) facilitated the loading of Cre and Cas9 into sEVs without the generation of packaging device-cargo fusion proteins. Furthermore, Myr(CHMP6)-modified sEVs loaded with Cre or Cas9 promoted gene-editing in recipient cells, as observed using a fluorescence reporter system. Subsequent investigation demonstrated a dose-dependent effect of Myr(CHMP6) tag-induced cargo-loading. Mechanistically, N-myristoylation alone was necessary but not sufficient for the effective packaging of proteins into EVs. Thus, our results indicated that Myr(CHMP6) induces sEV production and may be effective in loading gene editing proteins into sEVs for therapeutic purposes.


Extracellular Vesicles , Gene Editing , Extracellular Vesicles/metabolism , Multivesicular Bodies , Peptides/metabolism
9.
Front Cardiovasc Med ; 9: 774627, 2022.
Article En | MEDLINE | ID: mdl-35224032

Cardiac remodeling is the critical process in heart failure due to many cardiovascular diseases including myocardial infarction, hypertension, cardiovascular disease and cardiomyopathy. However, treatments for heart failure focusing on cardiac remodeling show relatively limited effectiveness. In recent decades, epitranscriptomic modifications were found abundantly present throughout the progression of cardiac remodeling, and numerous types of biochemical modifications were identified. m6A modification is the methylation of the adenosine base at the nitrogen-6 position, and dysregulation of m6A modification has been implicated in a wide range of diseases. However, function of m6A modifications still remain largely unknown in cardiac diseases, especially cardiac remodeling. LncRNAs are also shown to play a vital role in the pathophysiology of cardiac remodeling and heart failure. The crosstalk between lncRNAs and m6A modification provides a novel prospective for exploring possible regulatory mechanism and therapeutic targets of cardiac remodeling. This review summarizes the role of m6A modification in cardiac remodeling in the current researches.

10.
Front Cardiovasc Med ; 7: 582890, 2020.
Article En | MEDLINE | ID: mdl-33324687

Cardiac hypertrophy is a pathophysiological response to harmful stimuli. The continued presence of cardiac hypertrophy will ultimately develop into heart failure. The mitochondrion is the primary organelle of energy production, and its dysfunction plays a crucial role in the progressive development of heart failure from cardiac hypertrophy. Hispidulin, a natural flavonoid, has been substantiated to improve energy metabolism and inhibit oxidative stress. However, how hispidulin regulates cardiac hypertrophy and its underlying mechanism remains unknown. We found that hispidulin significantly inhibited pressure overload-induced cardiac hypertrophy and improved cardiac function in vivo and blocked phenylephrine (PE)-induced cardiomyocyte hypertrophy in vitro. We further proved that hispidulin remarkably improved mitochondrial function, manifested by increased electron transport chain (ETC) subunits expression, elevated ATP production, increased oxygen consumption rates (OCR), normalized mitochondrial morphology, and reduced oxidative stress. Furthermore, we discovered that Sirt1, a well-recognized regulator of mitochondrial function, might be a target of hispidulin, as evidenced by its upregulation after hispidulin treatment. Cotreatment with EX527 (a Sirt1-specific inhibitor) and hispidulin nearly completely abolished the antihypertrophic and protective effects of hispidulin on mitochondrial function, providing further evidence that Sirt1 could be the pivotal downstream effector of hispidulin in regulating cardiac hypertrophy.

11.
Hypertension ; 75(1): 79-90, 2020 01.
Article En | MEDLINE | ID: mdl-31735087

Transcribed ultraconserved regions (T-UCRs) are a novel class of long noncoding RNAs transcribed from UCRs, which exhibit 100% DNA sequence conservation among humans, mice, and rats. However, whether T-UCRs regulate cardiac hypertrophy remains unclear. We aimed to explore the effects of T-UCRs on cardiac hypertrophy. First, we performed long noncoding RNA microarray analysis on hearts of mice subjected to sham surgery or aortic banding and found that the T-UCR uc.323 was decreased significantly in mice with aortic banding-induced cardiac hypertrophy. In vitro loss- and gain-of-function experiments demonstrated that uc.323 protected cardiomyocytes against hypertrophy induced by phenylephrine. Additionally, we discovered that mammalian target of rapamycin 1 contributed to phenylephrine-induced uc.323 downregulation and uc.323-mediated cardiomyocyte hypertrophy. We further mapped the possible target genes of uc.323 through global microarray mRNA expression analysis after uc.323 knockdown and found that uc.323 regulated the expression of cardiac hypertrophy-related genes such as CPT1b (Carnitine Palmitoyl transferase 1b). Then, chromatin immunoprecipitation proved that EZH2 (enhancer of zeste homolog 2) bound to the promoter of CPT1b via H3K27me3 (trimethylation of lysine 27 of histone H3) to induce CPT1b downregulation. And overexpression of CPT1b could block uc.323-mediated cardiomyocyte hypertrophy. Finally, we found that uc.323 deficiency induced cardiac hypertrophy. Our results reveal that uc.323 is a conserved T-UCR that inhibits cardiac hypertrophy, potentially by regulating the transcription of CPT1b via interaction with EZH2.


Cardiomegaly/genetics , Carnitine O-Palmitoyltransferase/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Myocardium/metabolism , RNA, Long Noncoding/genetics , Animals , Cardiomegaly/metabolism , Carnitine O-Palmitoyltransferase/metabolism , Conserved Sequence , Enhancer of Zeste Homolog 2 Protein/metabolism , Gene Expression Regulation , Male , Mice , Myocytes, Cardiac/metabolism , RNA, Long Noncoding/metabolism , Rats , Rats, Sprague-Dawley , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Transcription, Genetic
12.
Theranostics ; 9(24): 7268-7281, 2019.
Article En | MEDLINE | ID: mdl-31695767

Rationale: An imbalance between protein synthesis and degradation is one of the mechanisms of cardiac hypertrophy. Increased transcription in cardiomyocytes can lead to excessive protein synthesis and cardiac hypertrophy. Maf1 is an RNA polymerase III (RNA pol III) inhibitor that plays a pivotal role in regulating transcription. However, whether Maf1 regulates of cardiac hypertrophy remains unclear. Methods: Cardiac hypertrophy was induced in vivo by thoracic aortic banding (AB) surgery. Both the in vivo and in vitro gain- and loss-of-function experiments by Maf1 knockout (KO) mice and adenoviral transfection were used to verify the role of Maf1 in cardiac hypertrophy. RNA pol III and ERK1/2 inhibitor were utilized to identify the effects of RNA pol III and ERK1/2. The possible interaction between Maf1 and ERK1/2 was clarified by immunoprecipitation (IP) analysis. Results: Four weeks after surgery, Maf1 KO mice exhibited significantly exacerbated AB-induced cardiac hypertrophy characterized by increased heart size, cardiomyocyte surface area, and atrial natriuretic peptide (ANP) expression and by exacerbated pulmonary edema. Also, the deficiency of Maf1 causes more severe cardiac dilation and dysfunction than wild type (WT) mice after pressure overload. In contrast, compared with adenoviral-GFP injected mice, mice injected with adenoviral-Maf1 showed significantly ameliorated AB-induced cardiac hypertrophy. In vitro study has demonstrated that Maf1 could significantly block phenylephrine (PE)-induced cardiomyocyte hypertrophy by inhibiting RNA pol III transcription. However, application of an RNA pol III inhibitor markedly improved Maf1 knockdown-promoted cardiac hypertrophy. Moreover, ERK1/2 was identified as a regulator of RNA pol III, and ERK1/2 inhibition by U0126 significantly repressed Maf1 knockdown-promoted cardiac hypertrophy accompanied by suppressed RNA pol III transcription. Additionally, IP analysis demonstrated that Maf1 could directly bind ERK1/2, suggesting Maf1 could interact with ERK1/2 and then inhibit RNA pol III transcription so as to attenuate the development of cardiac hypertrophy. Conclusions: Maf1 ameliorates PE- and AB-induced cardiac hypertrophy by inhibiting RNA pol III transcription via ERK1/2 signaling suppression.


Cardiomegaly/metabolism , RNA Polymerase III/metabolism , Repressor Proteins/metabolism , Animals , Cardiomegaly/etiology , Cardiomegaly/genetics , Humans , MAP Kinase Signaling System , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenylephrine/adverse effects , RNA Polymerase III/antagonists & inhibitors , RNA Polymerase III/genetics , Rats , Rats, Sprague-Dawley , Repressor Proteins/genetics
13.
J Nutr Biochem ; 62: 221-229, 2018 12.
Article En | MEDLINE | ID: mdl-30312797

Cardiac hypertrophy is a pathophysiological response to various pathological stresses and ultimately leads to heart failure. Oxidative stress is one of the critical processes involved in hypertrophy development. Fisetin, a small molecular flavonoid, has been shown to have anti-oxidative, anti-proliferative and anti-inflammatory properties. However, the effect of fisetin on cardiac hypertrophy remains unknown. In our present study, we showed that fisetin inhibited pressure overload-induced cardiac hypertrophy, improved cardiac function in vivo and suppressed phenylephrine (PE)-induced cardiomyocyte hypertrophy in vitro. Reactive oxygen species (ROS) levels were markedly decreased by fisetin treatment in both hypertrophic hearts and cardiomyocytes. Moreover, fisetin significantly up-regulated the expression of antioxidative genes, including catalase (CAT), superoxide dismutase 1 (SOD1) and heme oxygenase 1 (HO-1). Furthermore, co-treatment with N-acetylcysteine (NAC; ROS scavenger) and fisetin did not have synergistic inhibitory effects on PE-induced cardiomyocyte hypertrophy, indicating that the anti-hypertrophic effects of fisetin are mainly associated with the blockade of oxidative stress. Finally, the pro-hypertrophic signaling pathways, mitogen-activated protein kinase (MAPK) and mammalian target of rapamycin (mTOR) kinase, were found to be suppressed by fisetin after pressure overload and PE treatment. In conclusion, our study revealed that fisetin protects against cardiac hypertrophy and that oxidative stress inhibition may be one of the pivotal mechanisms involved.


Cardiomegaly/drug therapy , Flavonoids/pharmacology , Oxidative Stress/drug effects , Acetylcysteine/pharmacology , Animals , Cardiomegaly/etiology , Drug Synergism , Enzymes/genetics , Enzymes/metabolism , Flavonols , Gene Expression Regulation/drug effects , MAP Kinase Signaling System/drug effects , Male , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Phenylephrine/adverse effects , TOR Serine-Threonine Kinases/metabolism
14.
Nanoscale ; 10(39): 18795-18804, 2018 Oct 21.
Article En | MEDLINE | ID: mdl-30277241

Flake-shaped nanohybrids based on black phosphorus (BP) have been developed as multifunctional theranostic nanoplatforms for drug delivery, phototherapy and bioimaging. In this work, we report a facile strategy for fabrication of black phosphorus-Au nanoparticle hybrids (BP-AuNPs), which reveal an extraordinary near-infrared (NIR) photothermal transduction efficiency and drug delivery capacity. The applications of the nanocomposites as therapeutic agents for high-performance chemo-photothermal tumor therapy are accomplished in vitro and in vivo. BP-AuNPs also exhibit wonderful surface-enhanced Raman scattering (SERS) activity under NIR laser excitation with a low Raman background, allowing BP-AuNPs to be used as a promising two-dimensional (2D) fingerprint nanoprobe for bio-SERS analysis. The cellular component identification and label-free live-cell bioimaging based on this type of 2D SERS substrate are generally investigated, which open up promising new perspectives in nanomedicine, including diagnosis, imaging and therapy.


Gold , Metal Nanoparticles , Molecular Probes , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/drug therapy , Photochemotherapy , Animals , Gold/chemistry , Gold/pharmacology , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Mice , Molecular Probes/chemistry , Molecular Probes/pharmacology
15.
Noise Health ; 19(91): 245-253, 2017.
Article En | MEDLINE | ID: mdl-29319008

INTRODUCTION: The aim of the present study was to evaluate whether there are changes in cardiovascular risk factors among noise-exposed workers and to explore the possible mechanisms of a long-term noise exposure leading to cardiovascular disease and the sex differences of cardiovascular risk factors in this population. MATERIALS AND METHODS: Two hundred workers engaged in noise-related work, and a control group of 200 nonnoise-exposed workers hospitalized for occupational health examination were assigned into the study. All workers underwent a medical examination, electrocardiogram recording, blood pressure test, other blood tests, and audiometry. The collected blood was used to detect homocysteine (HCY), renin, angiotensin II, and other markers of cardiovascular risk factors. RESULTS: Our study suggests that the type of work with long-term exposure to noise might pose a cardiovascular risk, as evidenced by associated increases in plasma HCY levels, incidence of type 2 diabetes, and incidence of hypertension. DISCUSSION: Our research also reveals that among male workers, the levels of triglycerides, uric acid, HCY, renin activity, and the incidence of hypertension are higher than female, while high-density lipoprotein cholesterol is lower than female workers had. Additionally, the study emphasizes again the importance of weight control for reducing cardiovascular risk. CONCLUSION: Our study suggests that noise is a cardiovascular risk factor. Interventions in the work environment could be a preventable and controllable manner for reducing the incidence of cardiovascular disease.


Cardiovascular Diseases/etiology , Noise, Occupational/adverse effects , Occupational Diseases/etiology , Occupational Exposure/adverse effects , Adult , Angiotensin II/blood , China/epidemiology , Cholesterol/blood , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/epidemiology , Female , Homocysteine/blood , Humans , Hypertension/blood , Hypertension/complications , Hypertension/epidemiology , Incidence , Male , Middle Aged , Occupational Diseases/blood , Occupational Diseases/epidemiology , Renin/blood , Risk Factors , Sex Factors , Small-Area Analysis
16.
J Cell Sci ; 128(1): 70-80, 2015 Jan 01.
Article En | MEDLINE | ID: mdl-25395581

Although many studies have described the roles of microRNAs (miRNAs) in the modulation of the endothelial response to shear stress, the mechanisms remain incompletely understood. Here, we demonstrate that miR-34a expression in endothelial cells was downregulated by atheroprotective physiological high shear stress (HSS), whereas it was upregulated by atheroprone oscillatory shear stress (OSS). Blockade of endogenous miR-34a dramatically decreased basal vascular cell adhesion molecule-1 (VCAM-1) and intercellular adhesion molecule-1 (ICAM-1) protein expression levels. Conversely, miR-34a overexpression increased the protein levels of VCAM-1 and ICAM-1, consequently promoting monocyte adhesion to endothelial cells. Furthermore, miR-34a overexpression attenuated HSS-mediated suppression of VCAM-1 protein expression on endothelial cells, but promoted HSS-induced ICAM-1 expression. In addition, the OSS induction of endothelial cell VCAM-1 and ICAM-1 was suppressed by using an miR-34a inhibitor, which led to a reduction of monocyte adhesion to endothelial cells. Mechanistically, sirtuin 1 overexpression partially prevented miR-34a-induced VCAM-1 and ICAM-1 expression. Subsequent investigation demonstrated that miR-34a increased nuclear factor κB (NF-κB) p65 subunit (also known as RelA) acetylation (on residue Lys310), and silencing NF-κB signaling reduced miR-34a-induced VCAM-1 and ICAM-1 protein expression. These results demonstrate that miR-34a is involved in the flow-dependent regulation of endothelial inflammation.


Human Umbilical Vein Endothelial Cells/metabolism , MicroRNAs/metabolism , Signal Transduction , Cell Adhesion/genetics , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells/pathology , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Intercellular Adhesion Molecule-1/biosynthesis , Intercellular Adhesion Molecule-1/genetics , MicroRNAs/genetics , Monocytes/metabolism , Monocytes/pathology , Shear Strength , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Vascular Cell Adhesion Molecule-1/biosynthesis , Vascular Cell Adhesion Molecule-1/genetics
17.
J Cell Biochem ; 115(11): 1928-36, 2014 Nov.
Article En | MEDLINE | ID: mdl-24905663

Increasing evidences have illuminated the fundamental role of inflammation in mediating all stages of atherosclerosis. miR-155, a typical multi-functional miRNA, has recently emerged as a novel component of inflammatory signal transduction in the pathogenesis of atherosclerosis. However, little is known about whether endothelial highly expressed miR-155 can regulate endothelial inflammation-related transcription factors and the predicted role of miR-155 as a negative feedback regulator in endothelial inflammation involved in atherosclerosis. Bioinformatics analysis showed that RELA (nuclear factor-κB p65) is a potential target gene of miR-155 and this was confirmed by a luciferase reporter assay. Our results show that microRNA-155 mediate endothelial inflammation and decrease NFкB p65 and adhesion molecule expression in TNFα-stimulated endothelial cells. Transfection with miR-155 significantly inhibited TNFα-induced monocyte adhesion to endothelium. Inhibition of miR-155 enhanced p65 level and endothelial inflammatory response which was counteracted through the depletion of P65 by Si-P65. On the other hand, knockdown of eNOS, another target of miR-155, while transfecting with miR-155 inhibitor resulted in more significant inflammatory response. miR-155 is highly expressed in TNFα treated HUVECs, deprived of endogenous p65 could reverse TNFα-induced upregulation of miR-155. Thus, TNFα induced miR-155 may serve as a negative feedback regulator in endothelial inflammation involved in atherosclerosis by targeting nuclear transcription factor P65. These results provide a rationale for intervention of intracellular microRNA as possible anti-atherosclerotic targets.


Inflammation/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Transcription Factor RelA/genetics , Tumor Necrosis Factor-alpha/pharmacology , 3' Untranslated Regions , Cytokines/metabolism , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Inflammation/metabolism , Models, Biological , Transcription Factor RelA/metabolism
18.
Stem Cells ; 32(7): 1843-54, 2014 Jul.
Article En | MEDLINE | ID: mdl-24578274

The LIM-homeobox transcription factor islet-1 (ISL1) has been proposed to mark a cardiovascular progenitor cell lineage that gives rise to cardiomyocytes, endothelial cells, and smooth muscle cells. The aim of this study was to investigate whether forced expression of ISL1 in human mesenchymal stem cells (hMSCs) influenced the differentiation capacity and angiogenic properties of hMSCs. The lentiviral vector, EF1α-ISL1, was constructed using the Multisite Gateway System and used to transduce hMSCs. We found that ISL1 overexpression did not alter the proliferation, migration, or survival of hMSCs or affect their ability to differentiate into osteoblasts, adipocytes, cardiomyocytes, or endotheliocytes. However, ISL1-hMSCs differentiated into smooth muscle cells more efficiently than control hMSCs. Furthermore, conditioned medium from ISL1-hMSCs greatly enhanced the survival, migration, and tube-formation ability of human umbilical vein endothelial cells (HUVECs) in vitro. In vivo angiogenesis assays also showed much more vascular-like structures in the group cotransplanted with ISL1-hMSCs and HUVECs than in the group cotransplanted with control hMSCs and HUVECs. Quantitative RT-PCR and antibody arrays detected monocyte chemoattractant protein-3 (MCP3) at a higher level in conditioned medium from ISL1-hMSCs cultures than in conditioned medium from control hMSCs. Neutralization assays showed that addition of an anti-MCP3 antibody to ISL1-hMSCs-conditioned medium efficiently abolished the angiogenesis-promoting effect of ISL1-hMSCs. Our data suggest that overexpression of ISL1 in hMSCs promotes angiogenesis in vitro and in vivo through increasing secretion of paracrine factors, smooth muscle differentiation ability, and enhancing the survival of HUVECs.


Chemokine CCL7/metabolism , Human Umbilical Vein Endothelial Cells/physiology , LIM-Homeodomain Proteins/genetics , Mesenchymal Stem Cells/metabolism , Neovascularization, Physiologic , Transcription Factors/genetics , Animals , Cell Differentiation , Cells, Cultured , Chemokine CCL7/genetics , Gene Expression , Humans , LIM-Homeodomain Proteins/metabolism , Mice, SCID , Myocytes, Smooth Muscle/physiology , Transcription Factors/metabolism
19.
Biochem Biophys Res Commun ; 427(1): 138-42, 2012 Oct 12.
Article En | MEDLINE | ID: mdl-22989749

Shear stress associated with blood flow plays an important role in regulating gene expression and cell function in endothelial cells (ECs). MicroRNAs (miRNAs) are highly conserved, small non-coding RNAs that negatively regulate the expression of target genes by binding to the mRNA 3'-untranslated region (3'UTR) at the posttranscriptional level involved in diverse cellular processes. This study demonstrates that microRNA-101 in response to laminar shear stress (LSS) is involved in the flow regulation of gene expression in ECs. qRT-PCR analysis showed that miR-101 expression was significantly upregulated in human umbilical vein endothelial cells (HUVECs) exposed to 12 dyn/cm(2) laminar shear stress for 12h. We found that transfection of miR-101 significantly decreased the luciferase activity of plasmid reporter containing the 3'UTR of mammalian target of rapamycin (mTOR) gene. Western analysis revealed that the protein level of mTOR was significantly reduced in ECs transfected with miR-101. Furthermore, miR-101 overexpression induced cell cycle arrest at the G1/S transition and suppressed endothelial cell proliferation. Finally, transfection of miR-101 inhibitors attenuated the suppressive effects of LSS on mTOR expression, which identified the efficacy of loss-of-function of miR-101 in laminar flow-treated ECs. In conclusion, we have demonstrated that upregulation of miR-101 in response to LSS contributes to the suppressive effects of LSS on mTOR expression and EC proliferation. These studies advance our understanding of the posttranscriptional mechanisms by which shear stress modulates endothelial homeostasis.


Human Umbilical Vein Endothelial Cells/physiology , MicroRNAs/metabolism , Shear Strength/physiology , Stress, Mechanical , TOR Serine-Threonine Kinases/biosynthesis , Cell Proliferation , Cells, Cultured , Human Umbilical Vein Endothelial Cells/metabolism , Humans , MicroRNAs/biosynthesis , TOR Serine-Threonine Kinases/genetics , Up-Regulation
...