Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(11)2024 May 30.
Article in English | MEDLINE | ID: mdl-38892220

ABSTRACT

Despite the great progress in diagnosis, prevention, and treatment, cardiovascular diseases (CVDs) are still the most prominent cause of death worldwide [...].


Subject(s)
Cardiovascular Diseases , Cell Communication , RNA, Untranslated , Cardiovascular Diseases/genetics , Cardiovascular Diseases/metabolism , Humans , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Animals , MicroRNAs/genetics , MicroRNAs/metabolism
2.
Biomedicines ; 11(7)2023 Jul 06.
Article in English | MEDLINE | ID: mdl-37509546

ABSTRACT

Due to their different biological functions, extracellular vesicles (EVs) have great potential from a therapeutic point of view. They are released by all cell types, carrying and delivering different kinds of biologically functional cargo. Under pathological events, cells can increase their secretion of EVs and can release different amounts of cargo, thus making EVs great biomarkers as indicators of pathological progression. Moreover, EVs are also known to be able to transport and deliver cargo to different recipient cells, having an important role in cellular communication. Interestingly, EVs have recently been explored as biological alternatives for the delivery of therapeutics, being considered natural drug delivery carriers. Because cardiovascular disorders (CVDs) are the leading cause of death worldwide, in this review, we will discuss the up-to-date knowledge regarding the biophysical properties and biological components of EVs, focusing on myocardial infarction, diabetic cardiomyopathy, and sepsis-induced cardiomyopathy, three very different types of CVDs.

3.
Eur Heart J ; 43(43): 4562-4576, 2022 11 14.
Article in English | MEDLINE | ID: mdl-35292818

ABSTRACT

AIMS: Histone H3 dimethylation at lysine 79 is a key epigenetic mark uniquely induced by methyltransferase disruptor of telomeric silencing 1-like (DOT1L). We aimed to determine whether DOT1L modulates vascular smooth muscle cell (VSMC) phenotype and how it might affect atherosclerosis in vitro and in vivo, unravelling the related mechanism. METHODS AND RESULTS: Gene expression screening of VSMCs stimulated with the BB isoform of platelet-derived growth factor led us to identify Dot1l as an early up-regulated epigenetic factor. Mouse and human atherosclerotic lesions were assessed for Dot1l expression, which resulted specifically localized in the VSMC compartment. The relevance of Dot1l to atherosclerosis pathogenesis was assessed through deletion of its gene in the VSMCs via an inducible, tissue-specific knock-out mouse model crossed with the ApoE-/- high-fat diet model of atherosclerosis. We found that the inactivation of Dot1l significantly reduced the progression of the disease. By combining RNA- and H3K79me2-chromatin immunoprecipitation-sequencing, we found that DOT1L and its induced H3K79me2 mark directly regulate the transcription of Nf-κB-1 and -2, master modulators of inflammation, which in turn induce the expression of CCL5 and CXCL10, cytokines fundamentally involved in atherosclerosis development. Finally, a correlation between coronary artery disease and genetic variations in the DOT1L gene was found because specific polymorphisms are associated with increased mRNA expression. CONCLUSION: DOT1L plays a key role in the epigenetic control of VSMC gene expression, leading to atherosclerosis development. Results identify DOT1L as a potential therapeutic target for vascular diseases.


Subject(s)
Atherosclerosis , Muscle, Smooth, Vascular , Humans , Mice , Animals , Muscle, Smooth, Vascular/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Monocytes/metabolism , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Atherosclerosis/genetics , Atherosclerosis/prevention & control , Atherosclerosis/metabolism , Mice, Knockout , Gene Silencing , Cells, Cultured
4.
EMBO Mol Med ; 13(10): e14060, 2021 10 07.
Article in English | MEDLINE | ID: mdl-34551209

ABSTRACT

The role of single nucleotide polymorphisms (SNPs) in the etiopathogenesis of cardiovascular diseases is well known. The effect of SNPs on disease predisposition has been established not only for protein coding genes but also for genes encoding microRNAs (miRNAs). The miR-143/145 cluster is smooth muscle cell-specific and implicated in the pathogenesis of atherosclerosis. Whether SNPs within the genomic sequence of the miR-143/145 cluster are involved in cardiovascular disease development is not known. We thus searched annotated sequence databases for possible SNPs associated with miR-143/145. We identified one SNP, rs41291957 (G > A), located -91 bp from the mature miR-143 sequence, as the nearest genetic variation to this miRNA cluster, with a minor allele frequency > 10%. In silico and in vitro approaches determined that rs41291957 (A) upregulates miR-143 and miR-145, modulating phenotypic switching of vascular smooth cells towards a differentiated/contractile phenotype. Finally, we analysed association between rs41291957 and CAD in two cohorts of patients, finding that the SNP was a protective factor. In conclusion, our study links a genetic variation to a pathological outcome through involvement of miRNAs.


Subject(s)
Coronary Artery Disease , MicroRNAs , Coronary Artery Disease/genetics , Genetic Predisposition to Disease , Genome , Humans , MicroRNAs/genetics , Myocytes, Smooth Muscle , Polymorphism, Single Nucleotide
5.
Int J Mol Sci ; 21(12)2020 Jun 19.
Article in English | MEDLINE | ID: mdl-32575472

ABSTRACT

Reactive oxygen species (ROS) affect many cellular functions and the proper redox balance between ROS and antioxidants contributes substantially to the physiological welfare of the cell. During pathological conditions, an altered redox equilibrium leads to increased production of ROS that in turn may cause oxidative damage. MicroRNAs (miRNAs) regulate gene expression at the post-transcriptional level contributing to all major cellular processes, including oxidative stress and cell death. Several miRNAs are expressed in response to ROS to mediate oxidative stress. Conversely, oxidative stress may lead to the upregulation of miRNAs that control mechanisms to buffer the damage induced by ROS. This review focuses on the complex crosstalk between miRNAs and ROS in diseases of the cardiac (i.e., cardiac hypertrophy, heart failure, myocardial infarction, ischemia/reperfusion injury, diabetic cardiomyopathy) and pulmonary (i.e., idiopathic pulmonary fibrosis, acute lung injury/acute respiratory distress syndrome, asthma, chronic obstructive pulmonary disease, lung cancer) compartments. Of note, miR-34a, miR-144, miR-421, miR-129, miR-181c, miR-16, miR-31, miR-155, miR-21, and miR-1/206 were found to play a role during oxidative stress in both heart and lung pathologies. This review comprehensively summarizes current knowledge in the field.


Subject(s)
Heart Diseases/genetics , Lung Diseases/genetics , MicroRNAs/genetics , Reactive Oxygen Species/metabolism , Gene Expression Regulation , Heart Diseases/metabolism , Humans , Lung Diseases/metabolism , Oxidative Stress
6.
Circ Res ; 126(12): e120-e135, 2020 06 05.
Article in English | MEDLINE | ID: mdl-32216529

ABSTRACT

RATIONALE: MicroRNAs (miRNAs, miRs) are small noncoding RNAs that modulate gene expression by negatively regulating translation of target genes. Although the role of several miRNAs in vascular smooth muscle cells (VSMCs) has been extensively characterized, the function of miRNA-128-3p (miR-128) is still unknown. OBJECTIVE: To determine if miR-128 modulates VSMC phenotype and to define the underlying mechanisms. METHODS AND RESULTS: We screened for miRNAs whose expression is modulated by an altered DNA methylation status in VSMCs, and among the hits, we selected miR-128. We found that miR-128 was expressed in various tissues, primary murine cells, and pathological murine and human vascular specimens. Through gain- and loss-of-function approaches, we determined that miR-128 affects VSMC proliferation, migration, differentiation, and contractility. The alterations of those properties were dependent upon epigenetic regulation of key VSMC differentiation genes; notably, Kruppel-like factor 4 was found to be a direct target of miR-128 and able to modulate the methylation status of the pivotal VSMC gene myosin heavy chain 11 (Myh11). Finally, in vivo lentiviral delivery of miR-128 prevented intimal hyperplasia in a mouse model of carotid restenosis without modifying vital cardiovascular parameters. CONCLUSION: miR-128 is a critical modulator of VSMCs and is regulated by epigenetic modifications upon stress. Its modulation in the context of disease could be exploited for therapeutic purposes.


Subject(s)
Carotid Stenosis/metabolism , MicroRNAs/metabolism , Muscle, Smooth, Vascular/metabolism , Animals , Carotid Stenosis/genetics , Cell Differentiation , Cell Movement , Cell Proliferation , Cells, Cultured , DNA Methylation , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , MicroRNAs/genetics , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/physiology , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism
8.
Nanomedicine ; 19: 95-105, 2019 07.
Article in English | MEDLINE | ID: mdl-31028887

ABSTRACT

Functional RNAs, such as microRNAs, are emerging as innovative tools in the treatment of aggressive and incurable cancers. In this study, we explore the potential of silica dioxide nanoparticles (SiO2NPs) in the delivery of biologically active miRNAs. Focusing on the tumor-suppressor miR-34a, we evaluated miRNAs delivery by SiO2NPs into the mammary gland, using in vitro as well as in vivo model systems. We showed that silica nanoparticles can efficiently deliver miR-34a into normal and cancer epithelial cells grown in culture without major signs of toxicity. Delivered miRNA retained the ability to silence artificial as well endogenous targets and can reduce the growth of mammospheres in 3D culture. Finally, miR-34a delivery through intra-tumor administration of SiO2NPs leads to a reduced mammary tumor growth. In conclusion, our studies suggest that silica nanoparticles can mediate the delivery of miR-34a directly into mammary tumors while preserving its molecular and biological activity.


Subject(s)
Epithelial Cells/metabolism , Mammary Neoplasms, Animal/metabolism , MicroRNAs/administration & dosage , Nanoparticles/chemistry , Animals , Cell Proliferation , Endocytosis , Female , Mice, Inbred C57BL , Nanoparticles/ultrastructure , Silicon Dioxide/chemistry
9.
Oncogene ; 38(3): 360-374, 2019 01.
Article in English | MEDLINE | ID: mdl-30093634

ABSTRACT

The role of the tumour-suppressor miR-34 family in breast physiology and in mammary stem cells (MaSCs) is largely unknown. Here, we revealed that miR-34 family, and miR-34a in particular, is implicated in mammary epithelium homoeostasis. Expression of miR-34a occurs upon luminal commitment and differentiation and serves to inhibit the expansion of the pool of MaSCs and early progenitor cells, likely in a p53-independent fashion. Mutant mice (miR34-KO) and loss-of-function approaches revealed two separate functions of miR-34a, controlling both proliferation and fate commitment in mammary progenitors by modulating several pathways involved in epithelial cell plasticity and luminal-to-basal conversion. In particular, miR-34a acts as endogenous inhibitor of the Wnt/beta-catenin signalling pathway, targeting up to nine upstream regulators at the same time, thus modulating the expansion of the MaSCs/early progenitor pool. These multiple roles of miR-34a are maintained in a model of human breast cancer, in which chronic expression of miR-34a in triple-negative mesenchymal-like cells (enriched in cancer stem cells-CSCs) could promote a luminal-like differentiation programme, restrict the CSC pool, and inhibit tumour propagation. Hence, activation of miR-34a-dependent programmes could provide a therapeutic opportunity for the subset of breast cancers, which are rich in CSCs and respond poorly to conventional therapies.


Subject(s)
Breast Neoplasms/pathology , Mammary Glands, Animal/cytology , MicroRNAs/physiology , RNA, Neoplasm/physiology , Animals , Breast Neoplasms/metabolism , Cell Differentiation , Cell Line, Tumor , Cell Proliferation/physiology , Cell Self Renewal/physiology , Epithelial Cells/metabolism , Female , Humans , Mammary Glands, Animal/abnormalities , Mammary Glands, Animal/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Mice, Knockout , MicroRNAs/genetics , Neoplastic Stem Cells/metabolism , Spheroids, Cellular , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Wnt Signaling Pathway
10.
Circ Res ; 124(4): 498-510, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30582454

ABSTRACT

RATIONALE: microRNAs (miRNAs) modulate gene expression by repressing translation of targeted genes. Previous work has established a role for miRNAs in regulating vascular smooth muscle cell (VSMC) activity. Whether circular RNAs are involved in the modulation of miRNA activity in VSMCs is unknown. OBJECTIVE: We aimed to identify circular RNAs interacting with miRNAs enriched in VSMCs and modulating the cells' activity. METHODS AND RESULTS: RNA sequencing and bioinformatics identified several circular RNAs enriched in VSMCs; however, only one, possessing multiple putative binding sites for miR-145, was highly conserved between mouse and man. This circular RNA gemmed from alternative splicing of Lrp6 (lipoprotein receptor 6), a gene highly expressed in vessels and implicated in vascular pathologies and was thus named circ_Lrp6. Its role as a miR-145 sponge was confirmed by determining reciprocal interaction through RNA immunoprecipitation, stimulated emission depletion microscopy, and competitive luciferase assays; functional inhibition of miR-145 was assessed by measuring expression of the target genes ITGß8 (integrin-ß8), FASCIN (fascin actin-bundling protein 1), KLF4 (Kruppel-like factor 4), Yes1 (YES proto-oncogene 1), and Lox (lysyl oxidase). The interaction was preferentially localized to P-bodies, sites of mRNA degradation. Using loss- and gain-of-function approaches, we found that circ_Lrp6 hindered miR-145-mediated regulation of VSMC migration, proliferation, and differentiation. Differential expression of miR-145 and circ_Lrp6 in murine and human vascular diseases suggests that the ratio of circ_Lrp6 bound to miR-145 versus unbound could play a role in vascular pathogenesis. Viral delivery of circ_Lrp6 shRNA prevented intimal hyperplasia in mouse carotids. CONCLUSIONS: circ_Lrp6 is an intracellular modulator and a natural sponge for miR-145, counterbalancing the functions of the miRNA in VSMCs.


Subject(s)
MicroRNAs/metabolism , Muscle, Smooth, Vascular/metabolism , RNA, Circular/genetics , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Humans , Integrin beta Chains/genetics , Integrin beta Chains/metabolism , Kruppel-Like Factor 4 , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Protein-Lysine 6-Oxidase/genetics , Protein-Lysine 6-Oxidase/metabolism , Proto-Oncogene Mas , Proto-Oncogene Proteins c-yes/genetics , Proto-Oncogene Proteins c-yes/metabolism , RNA, Circular/metabolism
11.
Nat Commun ; 9(1): 3119, 2018 08 07.
Article in English | MEDLINE | ID: mdl-30087332

ABSTRACT

Little is known about miRNA decay. A target-directed miRNA degradation mechanism (TDMD) has been suggested, but further investigation on endogenous targets is necessary. Here, we identify hundreds of targets eligible for TDMD and show that an endogenous RNA (Serpine1) controls the degradation of two miRNAs (miR-30b-5p and miR-30c-5p) in mouse fibroblasts. In our study, TDMD occurs when the target is expressed at relatively low levels, similar in range to those of its miRNAs (100-200 copies per cell), and becomes more effective at high target:miRNA ratios (>10:1). We employ CRISPR/Cas9 to delete the miR-30 responsive element within Serpine1 3'UTR and interfere with TDMD. TDMD suppression increases miR-30b/c levels and boosts their activity towards other targets, modulating gene expression and cellular phenotypes (i.e., cell cycle re-entry and apoptosis). In conclusion, a sophisticated regulatory layer of miRNA and gene expression mediated by specific endogenous targets exists in mammalian cells.


Subject(s)
Fibroblasts/metabolism , MicroRNAs/genetics , Serpin E2/metabolism , 3' Untranslated Regions , Animals , Apoptosis , CRISPR-Cas Systems , Cell Cycle , Gene Deletion , Gene Expression , Gene Expression Profiling , Gene Expression Regulation , Mice , MicroRNAs/metabolism , Mutation , Phenotype , RNA Stability , Sequence Analysis, RNA , Serpin E2/genetics
12.
Circ Res ; 116(11): 1753-64, 2015 May 22.
Article in English | MEDLINE | ID: mdl-25801897

ABSTRACT

RATIONALE: The miR-143/145 cluster is highly expressed in smooth muscle cells (SMCs), where it regulates phenotypic switch and vascular homeostasis. Whether it plays a role in neighboring endothelial cells (ECs) is still unknown. OBJECTIVE: To determine whether SMCs control EC functions through passage of miR-143 and miR-145. METHODS AND RESULTS: We used cocultures of SMCs and ECs under different conditions, as well as intact vessels to assess the transfer of miR-143 and miR-145 from one cell type to another. Imaging of cocultured cells transduced with fluorescent miRNAs suggested that miRNA transfer involves membrane protrusions known as tunneling nanotubes. Furthermore, we show that miRNA passage is modulated by the transforming growth factor (TGF) ß pathway because both a specific transforming growth factor-ß (TGFß) inhibitor (SB431542) and an shRNA against TGFßRII suppressed the passage of miR-143/145 from SMCs to ECs. Moreover, miR-143 and miR-145 modulated angiogenesis by reducing the proliferation index of ECs and their capacity to form vessel-like structures when cultured on matrigel. We also identified hexokinase II (HKII) and integrin ß 8 (ITGß8)-2 genes essential for the angiogenic potential of ECs-as targets of miR-143 and miR-145, respectively. The inhibition of these genes modulated EC phenotype, similarly to miR-143 and miR-145 overexpression in ECs. These findings were confirmed by ex vivo and in vivo approaches, in which it was shown that TGFß and vessel stress, respectively, triggered miR-143/145 transfer from SMCs to ECs. CONCLUSIONS: Our results demonstrate that miR-143 and miR-145 act as communication molecules between SMCs and ECs to modulate the angiogenic and vessel stabilization properties of ECs.


Subject(s)
Human Umbilical Vein Endothelial Cells/metabolism , MicroRNAs/metabolism , Myocytes, Smooth Muscle/metabolism , Transforming Growth Factor beta/pharmacology , Animals , Benzamides/pharmacology , Biological Transport/drug effects , Blood Vessels/metabolism , Blood Vessels/physiology , Blotting, Western , Cell Communication/drug effects , Cell Communication/genetics , Cell Surface Extensions/drug effects , Cell Surface Extensions/metabolism , Cells, Cultured , Coculture Techniques , Dioxoles/pharmacology , Gene Expression Regulation , Hexokinase/genetics , Hexokinase/metabolism , Human Umbilical Vein Endothelial Cells/physiology , Human Umbilical Vein Endothelial Cells/ultrastructure , Humans , Integrin beta Chains/genetics , Integrin beta Chains/metabolism , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Microscopy, Confocal , Microscopy, Electron, Scanning , Myocytes, Smooth Muscle/ultrastructure , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...