Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Phytomedicine ; 114: 154793, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37011420

ABSTRACT

BACKGROUND: Aloe-emodin (AE), a natural anthraquinone extract from traditional Chinese medicinal plants, has been certified to protect against acute myocardial ischemia. However, its effect on cardiac remodeling after chronic myocardial infarction (MI) and the possible mechanism remain unclear. PURPOSE: This study investigated the effect of AE on cardiac remodeling and oxidative damage induced by myocardial infarction (MI) in vitro and explored the underlying mechanisms. METHODS: Echocardiography and Masson staining were used to demonstrate myocardial dysfunction and fibrosis. Cell apoptosis was detected by TUNEL staining. The expressions of fibrosis-related factors such as type I collagen, α-smooth muscle actin (α-SMA) and connective tissue growth factor (CTGF) were detected by Western blot. RESULTS: Our data demonstrated that AE treatment significantly improved cardiac function, reduced structural remodeling, and reduced cardiac apoptosis and oxidative stress in mice with myocardial infarction. In vitro, AE could protect neonatal mouse cardiomyocytes (NMCM) from angiotensin II (Ang II)-induced cardiomyocyte hypertrophy and apoptosis, and significantly inhibited (p < 0.05) Ang II-induced reactive oxygen species (ROS) increase. Furthermore, AE treatment significantly reversed the Ang ii-induced upregulation. CONCLUSION: In summary, our work reveals for the first time that AE activates the TGF-ß signaling pathway by up-regulating Smad7 expression, which in turn regulates the expression of fibrosis-related genes, ultimately improving cardiac function, inhibiting the development of cardiac fibrosis and hypertrophy in rats with chronic MI.


Subject(s)
Aloe , Cardiomyopathies , Emodin , Myocardial Infarction , Mice , Rats , Animals , Emodin/pharmacology , Ventricular Remodeling , Signal Transduction , Transforming Growth Factor beta1/metabolism , Myocardial Infarction/drug therapy , Myocytes, Cardiac , Cardiomyopathies/metabolism , Hypertrophy/pathology , Fibrosis , Myocardium/metabolism , Angiotensin II/pharmacology , Smad7 Protein/metabolism
2.
Mol Immunol ; 157: 8-17, 2023 05.
Article in English | MEDLINE | ID: mdl-36958140

ABSTRACT

TBK1-IRF3 complex plays vital roles in antiviral immune responses, its regulatory mechanisms are currently incompletely understood. p120-catenin (p120), an armadillo-repeat protein, mainly regulates the stability of classical cadherins and the development of epithelial-to-mesenchymal transitions (EMTs). Here we report that p120 is a positive regulator of type I IFN production. Ectopic expression of p120 enhanced Vesicular stomatitis virus and Sendai-virus-induced type I IFN production, whereas knockdown of p120 expression suppressed type I IFN production. Mechanistically, p120 promoted phosphorylation of IRF3 via stabilizing the TBK1-IRF3 complex. Consistently, p120 knock down mice are more susceptible to VSV infection as indicated by higher tissue viral titers, less IFN-I production and greater infiltration of immune cells. This study reveals p120 as an important positive regulator in innate immunity and identifies that p120 facilitates host antiviral response through stabilizing TBK1-IRF3 complex.


Subject(s)
Interferon Type I , Protein Serine-Threonine Kinases , Animals , Mice , Protein Serine-Threonine Kinases/metabolism , Delta Catenin , Antiviral Agents , Immunity, Innate , Phosphorylation , Interferon Regulatory Factor-3/metabolism
3.
Cell Death Dis ; 10(12): 908, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31787746

ABSTRACT

Non-coding RNAs participate in many cardiac pathophysiological processes, including myocardial infarction (MI). Here we showed the interplay between long non-coding RNA taurine-upregulated gene 1 (lncR-TUG1), miR-9a-5p (miR-9) and Krüppel-like factor 5 (KLF5). LncR-TUG1 was upregulated in ischemic heart and in cultured cardiomyocytes exposed to H2O2. Knockdown of lncR-TUG1 markedly ameliorated impaired cardiac function of MI mice. Further study showed that lncR-TUG1 acted as a competitive endogenous RNA of miR-9, and silencing of lncR-TUG1 inhibited cardiomyocyte apoptosis by upregulating miR-9 expression. Furthermore, the miR-9 overexpression obviously prevented ischemia injury and significantly inhibited H2O2-induced cardiomyocyte apoptosis via inhibition of mitochondrial apoptotic pathway. KLF5, as a target gene of miR-9 by dual-luciferase reporter assay, was involved in the process of miR-9 in regulating cardiomyocyte apoptosis. Our data identified the KLF5 was downregulated by miR-9 overexpression and knockdown of KLF5 inhibited cardiomyocyte apoptosis induced by H2O2. MiR-9 exerts anti-cardiomyocyte apoptotic affects by targeting KLF5. Collectively, our data identify a novel function of lncR-TUG1/miR-9/KLF5 axis in regulating cardiomyocyte apoptosis that affects myocardial infarction progression.


Subject(s)
Apoptosis/genetics , Kruppel-Like Transcription Factors/metabolism , MicroRNAs/metabolism , Myocardial Ischemia/genetics , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , RNA, Long Noncoding/metabolism , 3' Untranslated Regions/genetics , Animals , Apoptosis/drug effects , Base Sequence , Cell Survival/drug effects , Down-Regulation/drug effects , HEK293 Cells , Humans , Hydrogen Peroxide/toxicity , Kruppel-Like Transcription Factors/genetics , Male , MicroRNAs/genetics , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Ischemia/pathology , Myocytes, Cardiac/drug effects , RNA, Long Noncoding/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats
4.
Free Radic Biol Med ; 145: 187-197, 2019 12.
Article in English | MEDLINE | ID: mdl-31574344

ABSTRACT

Brain-derived neurotrophic factor (BDNF)/tropomyosin-related kinase B (TrkB) pathway is associated with ischemic heart diseases (IHD). 7,8-dihydroxyflavone (7,8-DHF), BDNF mimetic, is a potent agonist of TrkB. We aimed to investigate the effects and the underlying mechanisms of 7,8-DHF on cardiac ischemia. Myocardial ischemic mouse model was induced by ligation of left anterior descending coronary artery. 7,8-DHF (5 mg/kg) was administered intraperitoneally two days after ischemia for four weeks. Echocardiography, HE staining and transmission electron microscope were used to examine the function, histology and ultrastructure of the heart. H9c2 cells were treated with hydrogen peroxide (H2O2), 7,8-DHF or TrkB inhibitor ANA-12. The effects of 7,8-DHF on cell viability, mitochondrial membrane potential (MMP) and mitochondrial superoxide generation were examined. Furthermore, mitochondrial fission and protein expression of mitochondrial dynamics (Mfn2 [mitofusin 2], OPA1 [optic atrophy 1], Drp1 [dynamin-related protein 1] and Fis-1 [fission 1]) was detected by mitotracker green staining and western blot, respectively. 7,8-DHF attenuated cardiac dysfunction and cardiomyocyte abnormality of myocardial ischemic mice. Moreover, 7,8-DHF increased cell viability and reduced cell death accompanied by improving MMP, inhibiting mitochondrial superoxide and preventing excessive mitochondrial fission of H2O2-treated H9c2 cells. The cytoprotective effects of 7,8-DHF were antagonized by ANA-12. Mechanistically, 7,8-DHF repressed OMA1-dependent conversion of L-OPA1 into S-OPA1, which was abolished by Akt inhibitor. In conclusion, 7,8-DHF protects against cardiac ischemic injury by inhibiting the proteolytic cleavage of OPA1. These findings provide a novel pharmacological effect of 7,8-DHF on mitochondrial dynamics and a new potential target for IHD.


Subject(s)
Brain-Derived Neurotrophic Factor/pharmacology , GTP Phosphohydrolases/genetics , Membrane Glycoproteins/genetics , Myocardial Ischemia/drug therapy , Optic Atrophy, Autosomal Dominant/drug therapy , Protein-Tyrosine Kinases/genetics , Animals , Brain-Derived Neurotrophic Factor/genetics , Cell Death/drug effects , Cell Survival/drug effects , Flavones/pharmacology , GTP Phosphohydrolases/chemistry , Heart/drug effects , Heart/physiopathology , Humans , Hydrogen Peroxide/metabolism , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Dynamics , Myocardial Ischemia/genetics , Myocardial Ischemia/pathology , Neuroprotective Agents/pharmacology , Optic Atrophy, Autosomal Dominant/genetics , Optic Atrophy, Autosomal Dominant/pathology
5.
Pharmacol Res ; 146: 104315, 2019 08.
Article in English | MEDLINE | ID: mdl-31207343

ABSTRACT

Aloe-emodin (AE) is an anthraquinone derived from rhubarb and has a variety of pharmacological actions. However, the role of AE in regulating ischemic heart diseases is still unclear. The present study investigated the effect of AE on cardiac injuries induced by myocardial infarction (MI) in vivo and oxidative insults in vitro and explored the mechanisms involved. TUNEL and Flow cytometry were performed to measure cell apoptosis. Western blot analysis was employed to detect expression of Bcl-2, Bax and Caspase-3 proteins. Real-time PCR was used to quantify the microRNAs levels. Our data showed that AE protected neonatal rat ventricular myocytes (NRVMs) from hydrogen peroxide (H2O2) induced apoptosis and significantly inhibited H2O2-induced reactive oxygen species (ROS) elevation. Furthermore, AE treatment significantly reversed H2O2-induced upregulation of Bax/Bcl-2 and the loss of mitochondrial membrane potential. In vivo, AE treatment significantly reduced infarct size, ameliorated impaired cardiac function and obviously decreased cardiac apoptosis and oxidative stress in MI mice heart. Meanwhile, AE restored H2O2-induced downregulation of miR-133, and transfection with miR-133 inhibitor abolished the anti-apoptotic and anti-oxidative effects of AE. Moreover, AE prevented H2O2-induced increase in caspase-3 activity, which was diminished by application of miR-133 inhibitor. Our results indicate that AE protectes against myocardial infarction via the upregulation of miR-133, inhibition of ROS production and suppression of caspase-3 apoptotic signaling pathway.


Subject(s)
Aloe/chemistry , Apoptosis/drug effects , Emodin/pharmacology , MicroRNAs/metabolism , Myocardial Infarction/drug therapy , Up-Regulation/drug effects , Animals , Antioxidants/metabolism , Cells, Cultured , Down-Regulation/drug effects , Heart/drug effects , Hydrogen Peroxide/pharmacology , Male , Mice , Myocardial Infarction/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
6.
Oxid Med Cell Longev ; 2019: 4579806, 2019.
Article in English | MEDLINE | ID: mdl-31191799

ABSTRACT

Acute myocardial infarction (AMI) is the leading cause of sudden death worldwide. MicroRNA-155 (miR-155) has been reported to target antiapoptotic genes in various diseases models, but the functional role of miR-155 in response to MI injury needs further investigations. This study investigated the role of miR-155 in myocardial ischemia injury. TUNEL and flow cytometry were performed to measure cell apoptosis. Western blot analysis was employed to detect protein expressions of Bcl-2, XIAP, Bax, and caspase-3. qRT-PCR was used to quantify miRNA levels. We showed that miR-155 was dynamically elevated in murine hearts subjected to MI and in neonatal rat ventricular cardiomyocyte (NRVM) injury induced by hydrogen peroxide (H2O2). In response to H2O2, the silencing of miR-155 using AMO-155 (antisense inhibitor oligodeoxyribonucleotides) significantly increased cell viability and reduced cell apoptosis. Moreover, AMO-155 reversed the H2O2-induced downregulation of Bcl-2 and XIAP and upregulation of Bax and cleaved-caspase-3. Further study revealed that AMO-155 resulted in a decrease of H2O2-induced JC-1-labelled monomeric cell number. In addition, AMO-155 markedly decreased infarct size, ameliorated impaired cardiac function, and significantly reduced apoptotic cell percentages in MI mice heart. The RNA-binding protein Quaking (QKI) was predicted as a target gene of miR-155 through bioinformatic analysis, and AMO-155 attenuated the downregulation of QKI in H2O2-treated cardiomyocytes and MI mice heart. Knockdown of QKI by siRNA abolished the antiapoptotic effects of AMO-155. Taken together, miR-155 is upregulated in the MI heart and NRVMs in response to H2O2 stress, and downregulating of miR-155 protects cardiomyocytes against apoptosis. Mechanistically, it is probably due to the repression of QKI signaling pathway.


Subject(s)
MicroRNAs/metabolism , Myocardial Infarction/metabolism , RNA-Binding Proteins/metabolism , Animals , Apoptosis/drug effects , Blotting, Western , Caspase 3/metabolism , Cells, Cultured , Hydrogen Peroxide/pharmacology , In Situ Nick-End Labeling , Male , MicroRNAs/genetics , Myocardial Infarction/genetics , Myocytes, Cardiac/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA-Binding Proteins/genetics , Rats , Real-Time Polymerase Chain Reaction , Signal Transduction/drug effects , X-Linked Inhibitor of Apoptosis Protein/metabolism
7.
Cell Physiol Biochem ; 51(1): 186-200, 2018.
Article in English | MEDLINE | ID: mdl-30439699

ABSTRACT

BACKGROUND/AIMS: MicroRNAs play an important role in regulating myocardial infarction (MI)-induced cardiac injury. MicroRNA-124 (miR-124) plays a vital role in regulating cellular proliferation, differentiation and apoptosis. Although the alteration of miR-124 was confirmed in peripheral blood of MI patients, little is known regarding the biological functions of miR-124 in cardiomyocytes. This study was designed to explore the role of miR-124 in MI and its underlying mechanisms. METHODS: Real-time PCR was used to quantify the microRNAs levels. TUNEL and Flow cytometry were performed to measure cell apoptosis. Western blot analysis was employed to detect expression of Bcl-2, Bax, Caspase-3 and STAT3 proteins. RESULTS: We revealed that miR-124 was significantly up-regulated in a mice model of MI and in neonatal rat ventricular myocytes (NRVMs) with H2O2 treatment. H2O2 treatment induced cardiomyocyte injury with reduced cell viability and enhanced apoptotic cell death, whereas silencing expression of miR-124 by AMO-124 (antisense inhibitor oligodeoxyribonucleotides) alleviated these deleterious changes. AMO-124 decreased the expression of Bax and cleaved-caspase-3 and upregulated the expression of Bcl-2 in H2O2-treated NRVMs. Besides, AMO-124 improved mitochondrial dysfunction of NRVMs induced by H2O2 treatment. Moreover, antagomir-124 markedly decreased the infarct area and apoptotic cardiomyocytes and improved cardiac function in MI mice. Furthermore, we identified STAT3 as a direct target of miR-124, and downregulation of miR-124 ameliorated the diminished levels of STAT3 and p-STAT3 (Tyr705) in response to H2O2 or MI. STAT3 inhibitor, stattic, was shown to attenuate the elevation of p-STAT3 in NRVMs with AMO-124 transfection. Inhibiting of STAT3 activity by stattic abrogated protective effects of AMO-124 on H2O2-induced cardiomyocytes apoptosis. CONCLUSION: Taken together, our data demonstrate that downregulation of miR-124 inhibits MI-induced apoptosis through upregulating STAT3, which suggests the therapeutic potential of miR-124 for myocardial infarction.


Subject(s)
Apoptosis , MicroRNAs/metabolism , STAT3 Transcription Factor/metabolism , 3' Untranslated Regions , Animals , Antagomirs/metabolism , Antagomirs/therapeutic use , Apoptosis/drug effects , Caspase 3/metabolism , Cyclic S-Oxides/pharmacology , Down-Regulation/drug effects , Hydrogen Peroxide/pharmacology , Male , Membrane Potential, Mitochondrial/drug effects , Mice , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Myocardial Infarction/drug therapy , Myocardial Infarction/pathology , Myocardial Infarction/veterinary , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Sprague-Dawley , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/genetics
8.
Mol Med Rep ; 16(5): 5793-5798, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28849060

ABSTRACT

Systemic lupus erythematosus is a connective tissue disease characterized by autoimmune inflammation, which leads to specific and nonspecific immune disorders with the formation of various autoantibodies by activated B cells. B­cell­activating factor (BAFF) is secreted by macrophages and activated T cells, and is responsible for the proliferation, maturation and differentiation of B cells. However, the mechanism of BAFF involvement in lupus nephritis (LN) remains unclear. The aim of the present study was to investigate the association between BAFF and phosphoinositide 3­kinase/protein kinase B/mammalian target of rapamycin (PI3K/Akt/mTOR) signaling in order to elucidate the pathogenesis of LN. In the present study, 18 patients with LN and 20 controls were included. The clinical data were analyzed and plasma levels of BAFF were measured using an ELISA. The mRNA and protein levels of BAFF, phosphorylated (p)­PI3K, p­Akt and p­mTOR in kidney tissues were measured using reverse transcription­quantitative polymerase chain reaction (RT­qPCR) and western blotting. Plasma BAFF levels were significantly increased in patients with LN compared with the controls (P<0.001). mRNA and protein levels of BAFF, p­PI3K, p­Akt and p­mTOR in kidney tissue were significantly increased in patients with LN compared with the controls (all P<0.001). mRNA and protein levels of BAFF in the kidney tissues of patients with LN were positively correlated with the levels of p­PI3K, p­Akt and p­mTOR. The results of the present study revealed a correlation between BAFF and the PI3K/Akt/mTOR signaling pathway, and it is hypothesized that they are involved in the pathogenesis of LN.


Subject(s)
B-Cell Activating Factor/blood , Lupus Nephritis/blood , Lymphocyte Activation/genetics , Proto-Oncogene Proteins c-akt/genetics , TOR Serine-Threonine Kinases/genetics , Adult , Aged , Autoantibodies/genetics , Autoantibodies/immunology , Cell Differentiation/genetics , Cell Proliferation/genetics , Female , Humans , Kidney/metabolism , Kidney/pathology , Lupus Nephritis/immunology , Lupus Nephritis/pathology , Lymphocyte Activation/immunology , Male , Middle Aged , Phosphatidylinositol 3-Kinases/genetics , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...