Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 194
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell Mol Life Sci ; 80(9): 267, 2023 Aug 26.
Article in English | MEDLINE | ID: mdl-37626241

ABSTRACT

Previous studies show a woman's pregnancy is correlated with post-reproductive longevity, and nulliparity is associated with higher risk of incident heart failure, suggesting pregnancy likely exerts a cardioprotection. We previously reported a cardioprotective phenomenon termed myocardial hypertrophic preconditioning, but it is unknown whether pregnancy-induced physiological hypertrophic preconditioning (PHP) can also protect the heart against subsequent pathological hypertrophic stress. We aimed to clarify the phenomenon of PHP and its mechanisms. The pluripara mice whose pregnancy-induced physiological hypertrophy regressed and the nulliparous mice underwent angiotensin II (Ang II) infusion or transverse aortic constriction (TAC). Echocardiography, invasive left ventricular hemodynamic measurement and histological analysis were used to evaluate cardiac remodeling and function. Silencing or overexpression of Foxo3 by adeno-associated virus was used to investigate the role of FoxO3a involved in the antihypertrophic effect. Compared with nulliparous mice, pathological cardiac hypertrophy induced by Ang II infusion, or TAC was significantly attenuated and heart failure induced by TAC was markedly improved in mice with PHP. Activation of FoxO3a was significantly enhanced in the hearts of postpartum mice. FoxO3a inhibited myocardial hypertrophy by suppressing signaling pathway of phosphorylated glycogen synthase kinase-3ß (p-GSK3ß)/ß-catenin/Cyclin D1. Silencing or overexpression of Foxo3 attenuated or enhanced the anti-hypertrophic effect of PHP in mice with pathological stimulation. Our findings demonstrate that PHP confers resistance to subsequent hypertrophic stress and slows progression to heart failure through activation of FoxO3a/GSK3ß pathway.


Subject(s)
Aortic Valve Stenosis , Heart Failure , Peptide Hormones , Animals , Female , Mice , Pregnancy , Angiotensin II , Cardiomegaly/genetics , Glycogen Synthase Kinase 3 beta/genetics , Heart
2.
J Mol Cell Cardiol ; 174: 63-76, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36436251

ABSTRACT

Whether long noncoding RNAs participate in the formation of abdominal aortic aneurysms (AAAs) through the regulation of SMC phenotypic switching is unknown. lincRNA-p21 induced by reactive oxygen species (ROS) is likely functionally associated with SMC phenotypic switching. We thus investigated the role of lincRNA-p21 in SMC phenotypic switching-associated AAA formation and its underlying mechanisms. An analysis of human and mouse abdominal aortic samples revealed that the lincRNA-p21 levels were significantly higher in AAA tissue. Stimulation with hydrogen peroxide upregulated the expression of lincRNA-p21 in a dose-dependent manner and converted SMCs from a contractile phenotype to a synthetic, proteolytic, and proinflammatory phenotype in vitro. Moreover, lincRNA-p21 promoted fracture of elastic fibres, reconstruction of the vascular wall, and AAA formation in vivo by modulating SMC phenotypic switching in two mouse models of AAA induced by angiotensin II or porcine pancreatic elastase (PPE) perfusion. Using a bioinformatics prediction method and luciferase reporter gene assays, we further proved that lincRNA-p21 sponged miR-204-5p to release the transcriptional activity of Mekk3 and promoted the NF-κB pathway and thereby played a role in the SMC phenotypic switch and AAA formation. The ROS levels were positively correlated with the lincRNA-p21 levels in human and mouse AAA tissues. The knockdown of lincRNA-p21 in a PPE-induced mouse AAA model increased the miR-204-5p levels and reduced the expression of Mekk3, whereas lincRNA-p21 overexpression had the opposite effect. Collectively, the results indicated that ROS-induced lincRNA-p21 sponges miR-204-5p to accelerate synthetic and proinflammatory SMC phenotypes through the Mekk3/NF-κB pathway in AAA formation. Thus, lincRNA-p21 may have therapeutic potential for AAA formation.


Subject(s)
Aortic Aneurysm, Abdominal , MicroRNAs , RNA, Long Noncoding , Humans , Mice , Swine , Animals , Reactive Oxygen Species/metabolism , NF-kappa B/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Aortic Aneurysm, Abdominal/metabolism , Phenotype , Disease Models, Animal , MicroRNAs/genetics , MicroRNAs/metabolism , Myocytes, Smooth Muscle/metabolism
3.
Lab Invest ; 103(9): 100170, 2023 09.
Article in English | MEDLINE | ID: mdl-37150296

ABSTRACT

Epigenetic modification is involved in tumorigenesis and cancer progression. We developed an epigenetic modification-associated molecular classification of gastric cancer (GC) to identify signature genes that accurately predict prognosis and the efficacy of immunotherapy. Least absolute shrinkage and selection operator and multivariate Cox regression analysis were conducted to develop an epigenetic modification-associated molecular classification. We investigated the significance of PIP4P2, an independent prognostic factor of the classification system, in predicting the prognosis and immunotherapy efficacy of patients with GC. The epigenetic modification-associated molecular classification was highly associated with the clinicopathological characteristics of patients and the existing classification of GC. PIP4P2 was highly expressed in GC tissue and tumor-associated macrophages. High PIP4P2 expression in GC tissue-induced tumor progression by activating PI3K/AKT signal transduction had a negative impact on immunotherapy efficacy. High expression of PIP4P2 in macrophages was correlated with poor prognosis in patients with GC. PIP4P2 is an independent unfavorable prognostic factor of epigenetic modification-associated molecular classification, is involved in tumorigenic progression, and is essential for assessing the prognosis and immunotherapy efficacy of GC.


Subject(s)
Stomach Neoplasms , Humans , Stomach Neoplasms/genetics , Phosphatidylinositol 3-Kinases , Carcinogenesis , Epigenesis, Genetic , Immunotherapy , Prognosis
4.
Diabetes Obes Metab ; 25(3): 664-674, 2023 03.
Article in English | MEDLINE | ID: mdl-36314246

ABSTRACT

AIM: To assess the efficacy and safety of liraglutide to reduce visceral and ectopic fat in adults with or without type 2 diabetes mellitus (T2DM). METHODS: Four databases were searched up to 6 May 2022 for randomized clinical trials assessing the effect of liraglutide on visceral and ectopic fat. The mean and standard deviation of the values of visceral fat, ectopic fat and body mass index were calculated. Subgroup analyses were performed based on the type of disease (T2DM or non-T2DM), duration of intervention, dosage of liraglutide and whether life interventions were added to liraglutide therapy. We extracted and integrated the safety assessments reported in each article. RESULTS: Sixteen randomized clinical trials with, in total, 845 participants were included in the meta-analysis. Liraglutide could significantly decrease visceral fat [standard mean difference (SMD) = -0.72, 95% confidence interval (CI; -1.12, -0.33)], liver fat [SMD = -0.78, 95% CI (-1.24, -0.32)] and body mass index [weighted mean difference = -1.44, 95% CI (-1.95, -0.92)] in adult patients with or without T2DM when compared with the control group. However, reduction of epicardial fat by liraglutide [SMD = -0.74, 95% CI (-1.82, 0.34)] was not statistically significant. Subgroup analysis revealed that an adequate dosage (≥1.8 mg/day) and appropriate duration of treatment (ranging from 16 to 40 weeks) were the decisive factors for liraglutide to reduce visceral fat effectively. Mild gastrointestinal reactions were the main adverse event of liraglutide. CONCLUSIONS: Liraglutide significantly and safely reduces visceral and ectopic liver fat irrespective of T2DM status, and reduces visceral fat provided adequate dosage and duration of therapy are ensured.


Subject(s)
Diabetes Mellitus, Type 2 , Liraglutide , Adult , Humans , Liraglutide/adverse effects , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/chemically induced , Liver , Body Mass Index , Adipose Tissue , Hypoglycemic Agents/adverse effects , Randomized Controlled Trials as Topic
5.
BMC Cardiovasc Disord ; 23(1): 263, 2023 05 19.
Article in English | MEDLINE | ID: mdl-37208590

ABSTRACT

BACKGROUND: Preexisting impaired renal function (IRF) and contrast-induced nephropathy (CIN) after percutaneous coronary intervention (PCI) in patients with ST-segment elevation myocardial infarction (STEMI) are important prognostic parameters, but it is unknown whether delayed PCI is still beneficial for STEMI patients with IRF. METHODS: A retrospective single-center cohort study was performed in 164 patients who presented at least 12 h after symptom onset, and were diagnosed with STEMI and IRF. They were assigned to two groups to receive PCI plus optimal medical therapy (OMT) and OMT alone respectively. Clinical outcomes at 30 days and 1 year were compared between two groups, and hazard ratio for survival was analyzed using Cox regression model. A power analysis demanded 34 patients in each group to produce a power of 90% and a P value of 0.05. RESULTS: The 30-day mortality was significantly lower in PCI group (n = 126) than in non-PCI group (n = 38) (11.1% versus 28.9%, P = 0.018), while there was no significant difference in the 1-year mortality and incidence of cardiovascular comorbidities between the two groups. Cox regression analysis showed that patients with IRF didn't benefit from receiving PCI on survival rate (P = 0.267). CONCLUSIONS: Delayed PCI is not beneficial on one-year clinical outcomes for STEMI patients with IRF.


Subject(s)
Percutaneous Coronary Intervention , ST Elevation Myocardial Infarction , Humans , ST Elevation Myocardial Infarction/diagnostic imaging , ST Elevation Myocardial Infarction/therapy , Retrospective Studies , Cohort Studies , Percutaneous Coronary Intervention/adverse effects , Kidney/physiology , Treatment Outcome
6.
Mol Ther ; 30(11): 3477-3498, 2022 11 02.
Article in English | MEDLINE | ID: mdl-35791879

ABSTRACT

Reactive oxygen species (ROS) derived from oxygen-dependent mitochondrial metabolism are the essential drivers of cardiomyocyte (CM) cell-cycle arrest in adulthood. Mitochondria-localized circular RNAs (circRNAs) play important roles in regulating mitochondria-derived ROS production, but their functions in cardiac regeneration are still unknown. Herein, we investigated the functions and underlying mechanism of mitochondria-localized circSamd4 in cardiac regeneration. We found that circSamd4 was selectively expressed in fetal and neonatal CMs. The transcription factor Nrf2 controlled circSamd4 expression by binding to the promoter of circSamd4 host gene. CircSamd4 overexpression reduced while circSamd4 silenced increased mitochondrial oxidative stress and subsequent oxidative DNA damage. Moreover, circSamd4 overexpression induced CM proliferation and prevented CM apoptosis, which reduced the size of the fibrotic area and improved cardiac function after myocardial infarction (MI). Mechanistically, circSamd4 reduced oxidative stress generation and maintained mitochondrial dynamics by inducing the mitochondrial translocation of the Vcp protein, which downregulated Vdac1 expression and prevented the mitochondrial permeability transition pore (mPTP) from opening. Our findings suggest that circSamd4 is a novel therapeutic target for heart failure after MI.


Subject(s)
Myocardial Infarction , RNA, Circular , Humans , Infant, Newborn , Adult , RNA, Circular/genetics , Reactive Oxygen Species/metabolism , Mitochondrial Permeability Transition Pore , Myocytes, Cardiac/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/therapy , Myocardial Infarction/metabolism
7.
Mol Ther ; 30(2): 915-931, 2022 02 02.
Article in English | MEDLINE | ID: mdl-34547461

ABSTRACT

Macrophage polarization plays a crucial role in regulating abdominal aortic aneurysm (AAA) formation. Circular RNAs (circRNAs) are important regulators of macrophage polarization during the development of cardiovascular diseases. How-ever, the roles of circRNAs in regulating AAA formation through modulation of macrophage polarization remain unknown. In the present study, we compared circRNA microarray data under two distinct polarizing conditions (M1 and M2 macrophages) and identified an M1-enriched circRNA, circCdyl. Loss- and gain-of-function assay results demonstrated that circCdyl overexpression accelerated angiotensin II (Ang II)- and calcium chloride (CaCl2)-induced AAA formation by promoting M1 polarization and M1-type inflammation, while circCdyl deficiency showed the opposite effects. RNA pulldown, mass spectrometry analysis, and RNA immunoprecipitation (RIP) assays were conducted to elucidate the underlying mechanisms by which circCdyl regulates AAA formation and showed that circCdyl promotes vascular inflammation and M1 polarization by inhibiting interferon regulatory factor 4 (IRF4) entry into the nucleus, significantly inducing AAA formation. In addition, circCdyl was shown to act as a let-7c sponge, promoting C/EBP-δ expression in macrophages to induce M1 polarization. Our results indicate an important role for circCdyl-mediated macrophage polarization in AAA formation and provide a potent therapeutic target for AAA treatment.


Subject(s)
Aortic Aneurysm, Abdominal , RNA, Circular , Angiotensin II , Animals , Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/metabolism , Inflammation/genetics , Inflammation/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , RNA, Circular/genetics
8.
Acta Biochim Biophys Sin (Shanghai) ; 55(5): 831-841, 2023 May 25.
Article in English | MEDLINE | ID: mdl-37232575

ABSTRACT

Systemic and pulmonary arterial hypertension (PAH) can induce left and right ventricular hypertrophy, respectively, but common therapeutic targets for both left and right hypertrophy are limited. In this study, we attempt to explore potential common therapeutic targets and screen out potential target drugs for further study. Cardiac mRNA expression profiles in mice with transverse aortic constriction (TAC) and pulmonary arterial constriction (PAC) are obtained from online databases. After bioinformatics analyses, we generate TAC and PAC mouse models to validate the phenotypes of cardiac remodelling as well as the identified hub genes. Bioinformatics analyses show that there are 214 independent differentially expressed genes (DEGs) in GSE136308 (TAC related) and 2607 independent DEGs in GSE30922 (PAC related), while 547 shared DEGs are associated with the function of the extracellular matrix (ECM) or involved in the PI3K-Akt signaling pathway, cytokine-cytokine receptor interactions, and ECM-receptor interactions. We identifyd Fn1, Il6, Col1a1, Igf1, Col1a2, Timp1, Col3a1, Cd44, Ctgf and Postn as hub genes of the shared DEGs, and most of them are associated with myocardial fibrosis. Those hub genes and phenotypes of cardiac remodelling are validated in our TAC and PAC mouse models. Furthermore, we identify dehydroisoandrosterone (DHEA), iloprost and 4,5-dianilinophthalimide (DAPH) as potential therapeutic drugs targeting both left and right ventricular hypertrophy and validate the effect of DHEA. These findings suggest that DHEA could be an effective drug for pressure overload-induced left or right ventricular hypertrophy by regulating the shared hub differentially expressed genes associated with fibrosis.


Subject(s)
Hypertrophy, Left Ventricular , Pulmonary Arterial Hypertension , Mice , Animals , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Right Ventricular/genetics , Pulmonary Arterial Hypertension/etiology , Pulmonary Arterial Hypertension/genetics , Ventricular Remodeling , Phosphatidylinositol 3-Kinases , Cardiomegaly , Computational Biology , Dehydroepiandrosterone , Fibrosis , Mice, Inbred C57BL
9.
Circulation ; 143(23): 2277-2292, 2021 06 08.
Article in English | MEDLINE | ID: mdl-33757294

ABSTRACT

BACKGROUND: Exercise can induce physiological myocardial hypertrophy (PMH), and former athletes can live 5 to 6 years longer than nonathletic controls, suggesting a benefit after regression of PMH. We previously reported that regression of pathological myocardial hypertrophy has antihypertrophic effects. Accordingly, we hypothesized that antihypertrophic memory exists even after PMH has regressed, increasing myocardial resistance to subsequent pathological hypertrophic stress. METHODS: C57BL/6 mice were submitted to 21 days of swimming training to develop PMH. After termination of exercise, PMH regressed within 1 week. PMH regression mice (exercise hypertrophic preconditioning [EHP] group) and sedentary mice (control group) then underwent transverse aortic constriction or a sham operation for 4 weeks. Cardiac remodeling and function were evaluated with echocardiography, invasive left ventricular hemodynamic measurement, and histological analysis. LncRNA sequencing, chromatin immunoprecipitation assay, and comprehensive identification of RNA-binding proteins by mass spectrometry and Western blot were used to investigate the role of Mhrt779 involved in the antihypertrophic effect induced by EHP. RESULTS: At 1 and 4 weeks after transverse aortic constriction, the EHP group showed less increase in myocardial hypertrophy and lower expression of the Nppa and Myh7 genes than the sedentary group. At 4 weeks after transverse aortic constriction, EHP mice had less pulmonary congestion, smaller left ventricular dimensions and end-diastolic pressure, and a larger left ventricular ejection fraction and maximum pressure change rate than sedentary mice. Quantitative polymerase chain reaction revealed that the long noncoding myosin heavy chain-associated RNA transcript Mhrt779 was one of the markedly upregulated lncRNAs in the EHP group. Silencing of Mhrt779 attenuated the antihypertrophic effect of EHP in mice with transverse aortic constriction and in cultured cardiomyocytes treated with angiotensin II, and overexpression enhanced the antihypertrophic effect. Using chromatin immunoprecipitation assay and quantitative polymerase chain reaction, we found that EHP increased histone 3 trimethylation (H3K4me3 and H3K36me3) at the a4 promoter of Mhrt779. Comprehensive identification of RNA-binding proteins by mass spectrometry and Western blot showed that Mhrt779 can bind SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily a, member 4 (Brg1) to inhibit the activation of the histone deacetylase 2 (Hdac2)/phosphorylated serine/threonine kinase (Akt)/phosphorylated glycogen synthase kinase 3ß(p-GSK3ß) pathway induced by pressure overload. CONCLUSIONS: Myocardial hypertrophy preconditioning evoked by exercise increases resistance to pathological stress via an antihypertrophic effect mediated by a signal pathway of Mhrt779/Brg1/Hdac2/p-Akt/p-GSK3ß.


Subject(s)
Cardiomegaly/therapy , Physical Conditioning, Animal , RNA, Long Noncoding/metabolism , Animals , Atrial Natriuretic Factor/genetics , Atrial Natriuretic Factor/metabolism , Cardiomegaly/genetics , Disease Models, Animal , Echocardiography , Glycogen Synthase Kinase 3 beta/metabolism , Hemodynamics , Histone Deacetylase 2/metabolism , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Up-Regulation , Ventricular Function, Left/physiology , Ventricular Remodeling
10.
J Transl Med ; 19(1): 381, 2021 09 08.
Article in English | MEDLINE | ID: mdl-34496872

ABSTRACT

BACKGROUND: Tumor-derived exosomes (TEXs) are involved in tumor progression and the immune modulation process and mediate intercellular communication in the tumor microenvironment. Although exosomes are considered promising liquid biomarkers for disease diagnosis, it is difficult to discriminate TEXs and to develop TEX-based predictive biomarkers. METHODS: In this study, the gene expression profiles and clinical information were collected from The Cancer Genome Atlas (TCGA) database, IMvigor210 cohorts, and six independent Gene Expression Omnibus datasets. A TEXs-associated signature named TEXscore was established to predict overall survival in multiple cancer types and in patients undergoing immune checkpoint blockade therapies. RESULTS: Based on exosome-associated genes, we first constructed a tumor-derived exosome signature named TEXscore using a principal component analysis algorithm. In single-cell RNA-sequencing data analysis, ascending TEXscore was associated with disease progression and poor clinical outcomes. In the TCGA Pan-Cancer cohort, TEXscore was elevated in tumor samples rather than in normal tissues, thereby serving as a reliable biomarker to distinguish cancer from non-cancer sources. Moreover, high TEXscore was associated with shorter overall survival across 12 cancer types. TEXscore showed great potential in predicting immunotherapy response in melanoma, urothelial cancer, and renal cancer. The immunosuppressive microenvironment characterized by macrophages, cancer-associated fibroblasts, and myeloid-derived suppressor cells was associated with high TEXscore in the TCGA and immunotherapy cohorts. Besides, TEXscore-associated miRNAs and gene mutations were also identified. Further experimental research will facilitate the extending of TEXscore in tumor-associated exosomes. CONCLUSIONS: TEXscore capturing tumor-derived exosome features might be a robust biomarker for prognosis and treatment responses in independent cohorts.


Subject(s)
Exosomes , Melanoma , Biomarkers, Tumor/genetics , Humans , Immunotherapy , Melanoma/genetics , Melanoma/therapy , Prognosis , Single-Cell Analysis , Tumor Microenvironment
11.
Clin Sci (Lond) ; 135(6): 811-828, 2021 03 26.
Article in English | MEDLINE | ID: mdl-33687053

ABSTRACT

Post-translational modification (PTM) by small ubiquitin-like modifier (SUMO) is a key regulator of cell proliferation and can be readily reversed by a family of SUMO-specific proteases (SENPs), making SUMOylation an ideal regulatory mechanism for developing novel therapeutic strategies for promoting a cardiac regenerative response. However, the role of SUMOylation in cardiac regeneration remains unknown. In the present study, we assessed whether targeting protein kinase B (Akt) SUMOylation can promote cardiac regeneration. Quantitative PCR and Western blotting results showed that small ubiquitin-like modifier-specific protease 2 (SENP2) is up-regulated during postnatal heart development. SENP2 deficiency promoted P7 and adult cardiomyocyte (CM) dedifferentiation and proliferation both in vitro and in vivo. Mice with SENP2 deficiency exhibited improved cardiac function after MI due to CM proliferation and angiogenesis. Mechanistically, the loss of SENP2 up-regulated Akt SUMOylation levels and increased Akt kinase activity, leading to a decrease in GSK3ß levels and subsequently promoting CM proliferation and angiogenesis. In summary, inhibition of SENP2-mediated Akt deSUMOylation promotes CM differentiation and proliferation by activating the Akt pathway. Our results provide new insights into the role of SUMOylation in cardiac regeneration.


Subject(s)
Cysteine Endopeptidases/metabolism , Heart/growth & development , Myocytes, Cardiac/metabolism , Sumoylation , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Cysteine Endopeptidases/deficiency , Cysteine Endopeptidases/genetics , Disease Models, Animal , Genetic Vectors/administration & dosage , Mice, Inbred C57BL , Myocardial Infarction , Myocytes, Cardiac/cytology , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/metabolism , Regeneration/physiology
12.
J Cell Mol Med ; 24(5): 3229-3241, 2020 03.
Article in English | MEDLINE | ID: mdl-31989761

ABSTRACT

The heterogeneity in prognoses and chemotherapeutic responses of colon cancer patients with similar clinical features emphasized the necessity for new biomarkers that help to improve the survival prediction and tailor therapies more rationally and precisely. In the present study, we established a stroma-related lncRNA signature (SLS) based on 52 lncRNAs to comprehensively predict clinical outcome. The SLS model could not only distinguish patients with different recurrence and mortality risks through univariate analysis, but also served as an independent factor for relapse-free and overall survival. Compared with the conventionally used TNM stage system, the SLS model clearly possessed higher predictive accuracy. Moreover, the SLS model also effectively screened chemotherapy-responsive patients, as only patients in the low-SLS group could benefit from adjuvant chemotherapy. The following cell infiltration and competing endogenous RNA (ceRNA) network functional analyses further confirmed the association between the SLS model and stromal activation-related biological processes. Additionally, this study also identified three phenotypically distinct colon cancer subtypes that varied in clinical outcome and chemotherapy benefits. In conclusion, our SLS model may be a significant determinant of survival and chemotherapeutic decision-making in colon cancer and may have a strong clinical transformation value.


Subject(s)
Biomarkers, Tumor/genetics , Colonic Neoplasms/drug therapy , Neoplasm Recurrence, Local/drug therapy , RNA, Long Noncoding/genetics , Chemotherapy, Adjuvant/adverse effects , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Regulatory Networks/drug effects , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Prognosis , Stromal Cells/drug effects , Stromal Cells/metabolism , Stromal Cells/pathology , Transcriptome , Tumor Microenvironment/drug effects
13.
Circulation ; 139(25): 2857-2876, 2019 06 18.
Article in English | MEDLINE | ID: mdl-30947518

ABSTRACT

BACKGROUND: circRNAs (circular RNAs) are emerging as powerful regulators of cardiac development and disease, but their roles in cardiac regeneration are still unknown. This study used superenhancers to distinguish key circRNAs in the regulation of cardiac regeneration and explored the mechanisms underlying circRNA functions. METHODS: We used integrated bioinformatics analysis of RNA sequencing data and superenhancer catalogs to identify superenhancer-associated circRNAs. Quantitative polymerase chain reactions and in situ hybridization were performed to determine the circRNA expression patterns in hearts. Gain- and loss-of-function assays were conducted to detect the role of circRNAs in cardiomyocyte proliferation and cardiac repair after myocardial infarction. Chromatin immunoprecipitation (ChIP) and electrophoretic mobility shift assays were used to determine the binding of Meis1 (Meis homeobox 1) on circNfix-associated superenhancers. RNA pulldown and luciferase reporter assays were used to study circRNA interactions with proteins and miRNAs (micro RNAs). RESULTS: We identified a circRNA, Nfix circRNA (circNfix), that was regulated by a superenhancer and overexpressed in the adult heart in humans, rats, and mice. The transcription factor Meis1 bound to the superenhancer at the circNfix locus, and increased its expression. In vitro and in vivo, cardiomyocyte proliferation was increased by knockdown of circNfix, whereas it was inhibited by circNfix overexpression. Moreover, circNfix downregulation promoted cardiomyocyte proliferation and angiogenesis and inhibited cardiomyocyte apoptosis after myocardial infarction, attenuating cardiac dysfunction and improving the prognosis. Mechanistically, circNfix reinforced the interaction of Ybx1 (Y-box binding protein 1) with Nedd4l (an E3 ubiquitin ligase), and induced Ybx1 degradation through ubiquitination, repressing cyclin A2 and cyclin B1 expression. In addition, circNfix acted as a sponge for miR-214 to promote Gsk3ß (glycogen synthase kinase 3 ß) expression and repress ß-catenin activity. CONCLUSIONS: Loss of superenhancer-regulated circNfix promotes cardiac regenerative repair and functional recovery after myocardial infarction by suppressing Ybx1 ubiquitin-dependent degradation and increasing miR-214 activity and thus may be a promising strategy for improving the prognosis after MI.


Subject(s)
Cell Proliferation , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , RNA, Circular/metabolism , Regeneration , Animals , Apoptosis , Cells, Cultured , Disease Models, Animal , Down-Regulation , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Myeloid Ecotropic Viral Integration Site 1 Protein/genetics , Myeloid Ecotropic Viral Integration Site 1 Protein/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/pathology , Nedd4 Ubiquitin Protein Ligases/genetics , Nedd4 Ubiquitin Protein Ligases/metabolism , Neovascularization, Physiologic , RNA, Circular/genetics , Rats, Sprague-Dawley , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , beta Catenin/genetics , beta Catenin/metabolism
14.
Arterioscler Thromb Vasc Biol ; 39(1): e10-e25, 2019 01.
Article in English | MEDLINE | ID: mdl-30580562

ABSTRACT

Objective- Vascular smooth muscle cell phenotypic transition plays a critical role in the formation of abdominal aortic aneurysms (AAAs). SM22α (smooth muscle 22α) has a vital role in maintaining the smooth muscle cell phenotype and is downregulated in AAA. However, whether manipulation of the SM22α gene influences the pathogenesis of AAA is unclear. Here, we investigated whether SM22α prevents AAA formation and explored the underlying mechanisms. Approach and Results- In both human and animal AAA tissues, a smooth muscle cell phenotypic switch was confirmed, as manifested by the downregulation of SM22α and α-SMA (α-smooth muscle actin) proteins. The methylation level of the SM22α gene promoter was dramatically higher in mouse AAA tissues than in control tissues. SM22α knockdown in ApoE-/- (apolipoprotein E-deficient) mice treated with Ang II (angiotensin II) accelerated the formation of AAAs, as evidenced by a larger maximal aortic diameter and more medial elastin degradation than those found in control mice, whereas SM22α overexpression exerted opposite effects. Similar results were obtained in a calcium chloride-induced mouse AAA model. Mechanistically, SM22α deficiency significantly increased reactive oxygen species production and NF-κB (nuclear factor-κB) activation in AAA tissues, whereas SM22α overexpression produced opposite effects. NF-κB antagonist SN50 or antioxidant N-acetyl-L-cysteine partially abrogated the exacerbating effects of SM22α silencing on AAA formation. Conclusions- SM22α reduction in AAAs because of the SM22α promoter hypermethylation accelerates AAA formation through the reactive oxygen species/NF-κB pathway, and therapeutic approaches to increase SM22α expression are potentially beneficial for preventing AAA formation.


Subject(s)
Aortic Aneurysm, Abdominal/prevention & control , Microfilament Proteins/physiology , Muscle Proteins/physiology , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/physiology , NF-kappa B/physiology , Reactive Oxygen Species/metabolism , Animals , Cells, Cultured , DNA Methylation , Humans , Male , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/cytology , NADPH Oxidases/metabolism , Phenotype , Phosphorylation , Promoter Regions, Genetic
15.
Mol Ther ; 27(6): 1166-1182, 2019 06 05.
Article in English | MEDLINE | ID: mdl-30879950

ABSTRACT

Transforming growth factor ß (TGF-ß) drives epithelial-mesenchymal transition (EMT), playing vital roles in cancer metastasis. The crosstalk between microRNAs (miRNAs) and TGF-ß are frequently observed and involved in TGF-ß-induced EMT. Here, we determine that miR-577 is significantly upregulated in gastric cancer (GC). miR-577 expression is positively correlated with GC metastasis status and poor patient prognosis. Functional assays demonstrate that miR-577 promotes metastasis and chemoresistance by inducing EMT and stemness-like properties. Moreover, TGF-ß promotes the expression of miR-577, and miR-577 participates TGF-ß-mediated cancer metastasis. Mechanistically, TGF-ß activates miR-577 via NF-κB-mediated transcription, and miR-577 enhances TGF-ß signaling by targeting the serum deprivation protein response (SDPR), which directly interacts with ERK to inactivate the ERK-NF-κB pathway, hence forming a feedback loop to drive tumor metastasis. A plausible mechanism of EMT induction by the TGF-ß network is elucidated. Our findings suggest that the TGF-ß-miR-577-SDPR axis may be a potential prognostic marker and therapeutic target against cancer metastasis in GC.


Subject(s)
Disease Progression , MAP Kinase Signaling System , MicroRNAs/metabolism , NF-kappa B/metabolism , Phosphate-Binding Proteins/metabolism , Stomach Neoplasms/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Feedback, Physiological , HEK293 Cells , Heterografts , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Plasmids/genetics , Prognosis , RNA, Small Interfering/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Survival Rate , Transcriptional Activation/genetics , Transfection , Tumor Burden/genetics , Up-Regulation
16.
Mol Ther ; 27(1): 29-45, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30528086

ABSTRACT

Reactivating post-natal myocardial regeneration potential may be a feasible strategy to regenerate the injured adult heart. Long non-coding RNAs (lncRNAs) have been implicated in regulating cellular differentiation, but whether they can elicit a regenerative response in the post-natal heart remains unknown. In this study, by characterizing the lncRNA transcriptome in human hearts during the fetal-to-adult transition, we found that 3,092 lncRNAs were differentially expressed, and we further identified a novel upregulated fetal lncRNA that we called endogenous cardiac regeneration-associated regulator (ECRAR), which promoted DNA synthesis, mitosis, and cytokinesis in post-natal day 7 and adult rat cardiomyocytes (CMs). Overexpression of ECRAR markedly stimulated myocardial regeneration and induced recovery of cardiac function after myocardial infarction (MI). Knockdown of ECRAR inhibited post-natal day 1 CM proliferation and prevented post-MI recovery. ECRAR was transcriptionally upregulated by E2F transcription factor 1 (E2F1). In addition, ECRAR directly bound to and promoted the phosphorylation of extracellular signal-regulated kinases 1 and 2 (ERK1/2), resulting in downstream targets of cyclin D1 and cyclin E1 activation, which, in turn, activated E2F1. The E2F1-ECRAR-ERK1/2 signaling formed a positive feedback loop to drive cell cycle progression, and, therefore, it promoted CM proliferation. These findings indicated that our newly discovered ECRAR may be a valuable therapeutic target for heart failure.


Subject(s)
MAP Kinase Signaling System/physiology , Myocardium/cytology , Myocardium/metabolism , RNA, Long Noncoding/metabolism , Regeneration/physiology , Animals , Cell Cycle/genetics , Cell Cycle/physiology , Cell Proliferation/genetics , Cell Proliferation/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Fetal Heart/cytology , Fetal Heart/metabolism , Humans , MAP Kinase Signaling System/genetics , Male , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , RNA, Long Noncoding/genetics , Rats , Rats, Wistar , Regeneration/genetics , Signal Transduction/genetics , Signal Transduction/physiology
17.
J Mol Cell Cardiol ; 131: 66-81, 2019 06.
Article in English | MEDLINE | ID: mdl-30991034

ABSTRACT

Abdominal aortic aneurysm (AAA) is accepted as a chronic vascular inflammatory disease. However, how the inflammatory response is regulated during AAA formation is not fully understood. This study was undertaken to determine whether the long noncoding RNA (lncRNA) H19 (H19) promotes AAA formation by enhancing aortic inflammation. qRT-PCR detected the upregulation of H19 in human and mouse AAA tissue samples. Co-staining for H19 and the macrophage marker MAC-2 showed that H19 was located in vascular smooth muscle cells (VSMCs) and infiltrating aortic macrophages. In vivo overexpression of H19 increased vascular inflammation and induced AAA formation, which was supported by exacerbated aortic morphology, maximum aortic diameter values, elastin degradation, expression of interleukin-6 (IL-6) and macrophage chemoattractant protein-1 (MCP-1), and macrophage infiltration. H19 suppression resulted in the opposite effects. A rescue experiment indicated that IL-6 neutralization significantly mitigated the aortic inflammation and AAA formation evoked by H19 overexpression. Luciferase reporter assays and ex vivo experiments using VSMCs and macrophages confirmed that H19 induced aneurysm formation in part via endogenous competition with the let-7a microRNA to induce the transcription of its target gene, IL-6. This mechanism was further validated by in vivo experiments using a mutant H19 that could not effectively bind let-7a. Collectively, our study revealed a pathogenic H19/let-7a/IL-6 inflammatory pathway in AAA formation, which offers a new potential therapeutic strategy for AAA.


Subject(s)
Aortic Aneurysm, Abdominal/genetics , Inflammation/genetics , RNA, Long Noncoding/genetics , Angiotensin II/genetics , Animals , Cells, Cultured , Disease Models, Animal , Humans , Interleukin-6/genetics , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Signal Transduction/genetics , Transcription, Genetic/genetics , Up-Regulation/genetics
18.
Cancer Immunol Immunother ; 68(3): 433-442, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30564892

ABSTRACT

Tumour-infiltrating immune cells are a source of important prognostic information for patients with resectable colon cancer. We developed a novel immune model based on systematic assessments of the immune landscape inferred from bulk tumor transcriptomes of stage I-III colon cancer patients. The "Cell type Identification By Estimating Relative Subsets Of RNA Transcripts (CIBERSORT)" algorithm was used to estimate the fraction of 22 immune cell types from six microarray public datasets. The random forest method and least absolute shrinkage and selection operator model were then used to establish immunoscores for diagnosis and prognosis. By comparing immune cell compositions in samples of 870 colon cancer patients and 70 normal controls, we constructed a diagnostic model, designated the diagnostic immune risk score (dIRS), that showed high specificity and sensitivity in both the training [area under the curve (AUC) = 0.98, p < 0.001] and validation (AUC 0.96, p < 0.001) sets. We also established a prognostic immune risk score (pIRS) that was found to be an independent prognostic factor for relapse-free survival in every series (training: HR 2.23; validation: HR 1.65; entire: HR 2.01; p < 0.001 for all), which showed better prognostic value than TNM stage. In addition, integration of the pIRS with clinical characteristics in a composite nomogram showed improved accuracy of relapse risk prediction, providing a higher net benefit than TNM stage, with well-fitted calibration curves. The proposed dIRS and pIRS models represent promising novel signatures for the diagnosis and prognosis prediction of colon cancer.


Subject(s)
Colonic Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor , Colonic Neoplasms/diagnosis , Colonic Neoplasms/mortality , Colonic Neoplasms/pathology , Female , Humans , Male , Microsatellite Instability , Middle Aged , Neoplasm Staging , Nomograms , Prognosis
19.
Clin Sci (Lond) ; 133(3): 425-441, 2019 02 14.
Article in English | MEDLINE | ID: mdl-30679264

ABSTRACT

MiRNAs regulate the cardiomyocyte (CM) cell cycle at the post-transcriptional level, affect cell proliferation, and intervene in harmed CM repair post-injury. The present study was undertaken to characterize the role of let-7i-5p in the processes of CM cell cycle and proliferation and to reveal the mechanisms thereof. In the present study, we used real-time qPCR (RT-qPCR) to determine the up-regulated let-7i-5p in CMs during the postnatal switch from proliferation to terminal differentiation and further validated the role of let-7i-5p by loss- and gain-of-function of let-7i-5p in CMs in vitro and in vivo We found that the overexpression of let-7i-5p inhibited CM proliferation, whereas the suppression of let-7i-5p significantly facilitated CM proliferation. E2F2 and CCND2 were identified as the targets of let-7i-5p, mediating its effect in regulating the cell cycle of CMs. Supperession of let-7i-5p promoted the recovery of heart function post-myocardial infarction by enhancing E2F2 and CCND2. Collectively, our results revealed that let-7i-5p is involved in the regulation of the CM cell cycle and further impacts proliferation, which may offer a new potential therapeutic strategy for cardiac repair after ischemic injury.


Subject(s)
Cell Proliferation , Cyclin D2/metabolism , E2F2 Transcription Factor/metabolism , MicroRNAs/genetics , Myocytes, Cardiac/metabolism , Animals , Cell Cycle , Cells, Cultured , Cyclin D2/genetics , E2F2 Transcription Factor/genetics , Male , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/cytology
20.
Clin Sci (Lond) ; 133(13): 1439-1455, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31235554

ABSTRACT

The long non-coding RNA (lncRNA) PTENP1 is a pseudogene of phosphatase and tensin homologue deleted on chromosome ten (PTEN), has been implicated in smooth muscle cell (SMC) proliferation and apoptosis. PTENP1 is the pseudogene of PTEN. However, it is unclear whether and how PTENP1 functions in the proliferation and apoptosis of human aortic SMCs (HASMCs). Here, we hypothesised that PTENP1 inhibits HASMC proliferation and enhances apoptosis by promoting PTEN expression. PCR analysis and Western blot assays respectively showed that both PTENP1 and PTEN were up-regulated in human aortic dissection (AD) samples. PTENP1 overexpression significantly increased the protein expression of PTEN, promoted apoptosis and inhibited the proliferation of HASMCs. PTENP1 silencing exhibited the opposite effects and mitigated H2O2-induced apoptosis of HASMCs. In an angiotensin II (Ang II)-induced mouse aortic aneurysm (AA) model, PTENP1 overexpression potentiated aortic SMC apoptosis, exacerbated aneurysm formation. Mechanistically, RNA pull-down assay and a series of luciferase reporter assays using miR-21 mimics or inhibitors identified PTENP1 as a molecular sponge for miR-21 to endogenously compete for the binding between miR-21 and the PTEN transcript, releasing PTEN expression. This finding was further supported by in vitro immunofluorescent evidence showing decreased cell apoptosis upon miR-21 mimic administration under baseline PTENP1 overexpression. Ex vivo rescue of PTEN significantly mitigated the SMC apoptosis induced by PTENP1 overexpression. Finally, Western blot assays showed substantially reduced Akt phosphorylation and cyclin D1 and cyclin E levels with up-regulated PTENP1 in HASMCs. Our study identified PTENP1 as a mediator of HASMC homeostasis and suggests that PTENP1 is a potential target in AD or AA intervention.


Subject(s)
Aortic Aneurysm/metabolism , Aortic Dissection/metabolism , Apoptosis , Cell Proliferation , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Pseudogenes , RNA, Long Noncoding/metabolism , Aortic Dissection/genetics , Aortic Dissection/pathology , Animals , Aorta/metabolism , Aorta/pathology , Aortic Aneurysm/genetics , Aortic Aneurysm/pathology , Cell Cycle , Cells, Cultured , Disease Models, Animal , Gene Expression Regulation , Humans , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA, Long Noncoding/genetics , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL