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.
J Vasc Res ; 54(1): 22-32, 2017.
Article in English | MEDLINE | ID: mdl-28122380

ABSTRACT

BACKGROUND/AIMS: Endothelial colony-forming cells (ECFCs) have the potential to be used in regenerative medicine. Dysfunction of ECFCs is correlated with the onset of cardiovascular disorders, especially coronary artery disease (CAD). Binding of vascular endothelial growth factor A (VEGFA) to vascular endothelial growth factor receptor-2 (VEGFR2) triggers cell motility and angiogenesis of ECFCs, which are crucial to vascular repair. METHODS: To identify the miRNA-VEGFR2-dependent regulation of ECFC functions, ECFCs isolated from peripheral blood of disease-free and CAD individuals were subjected to small RNA sequencing for identification of anti-VEGFR2 miRNAs. The angiogenic activities of the miRNAs were determined in both in vitro and in vivo mice models. RESULTS: Three miRNAs, namely miR-410-3p, miR-497-5p, and miR-2355-5p, were identified to be upregulated in CAD-ECFCs, and VEGFR2 was their common target gene. Knockdown of these miRNAs not only restored the expression of VEGFR2 and increased angiogenic activities of CAD-ECFCs in vitro, but also promoted blood flow recovery in ischemic limbs in vivo. miR-410-3p, miR-497-5p, and miR-2355-5p could serve as potential biomarkers for CAD detection as they are highly expressed in the plasma of CAD patients. CONCLUSIONS: This modulation could help develop new therapeutic modalities for cardiovascular diseases and other vascular dysregulated diseases, especially tumor angiogenesis.


Subject(s)
Coronary Artery Disease/metabolism , Endothelial Progenitor Cells/metabolism , MicroRNAs/metabolism , Neovascularization, Physiologic , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Antagomirs/genetics , Antagomirs/metabolism , Case-Control Studies , Cell Movement , Cell Proliferation , Cells, Cultured , Computational Biology , Coronary Artery Disease/genetics , Coronary Artery Disease/pathology , Disease Models, Animal , Endothelial Progenitor Cells/pathology , Endothelial Progenitor Cells/transplantation , Gene Expression Profiling/methods , Gene Expression Regulation , Hindlimb , Humans , Ischemia/genetics , Ischemia/metabolism , Ischemia/physiopathology , Ischemia/surgery , Mice, Nude , MicroRNAs/genetics , Muscle, Skeletal/blood supply , Recovery of Function , Regional Blood Flow , Time Factors , Transfection , Vascular Endothelial Growth Factor Receptor-2/genetics
2.
PLoS One ; 11(1): e0147067, 2016.
Article in English | MEDLINE | ID: mdl-26799933

ABSTRACT

Diabetes mellitus (DM) is a metabolic disease that is increasing worldwide. Furthermore, it is associated with the deregulation of vascular-related functions, which can develop into major complications among DM patients. Endothelial colony forming cells (ECFCs) have the potential to bring about medical repairs because of their post-natal angiogenic activities; however, such activities are impaired by high glucose- (HG) and the DM-associated conditions. Far-infrared radiation (FIR) transfers energy as heat that is perceived by the thermoreceptors in human skin. Several studies have revealed that FIR improves vascular endothelial functioning and boost angiogenesis. FIR has been used as anti-inflammatory therapy and as a clinical treatment for peripheral circulation improvement. In addition to vascular repair, there is increasing evidence to show that FIR can be applied to a variety of diseases, including cardiovascular disorders, hypertension and arthritis. Yet mechanism of action of FIR and the biomarkers that indicate FIR effects remain unclear. MicroRNA-134 (miR-134-5p) was identified by small RNA sequencing as being increased in high glucose (HG) treated dfECFCs (HG-dfECFCs). Highly expressed miR-134 was also validated in dmECFCs by RT-qPCR and it is associated with impaired angiogenic activities of ECFCs. The functioning of ECFCs is improved by FIR treatment and this occurs via a reduction in the level of miR-134 and an increase in the NRIP1 transcript, a direct target of miR-134. Using a mouse ischemic hindlimb model, the recovery of impaired blood flow in the presence of HG-dfECFCs was improved by FIR pretreatment and this enhanced functionality was decreased when there was miR-134 overexpression in the FIR pretreated HG-dfECFCs. In conclusion, our results reveal that the deregulation of miR-134 is involved in angiogenic defects found in DM patients. FIR treatment improves the angiogenic activity of HG-dfECFCs and dmECFCs and FIR has potential as a treatment for DM. Detection of miR-134 expression in FIR-treated ECFCs should help us to explore further the effectiveness of FIR therapy.


Subject(s)
Endothelium, Vascular/physiopathology , Glucose/metabolism , Infrared Rays , MicroRNAs/physiology , Animals , Endothelium, Vascular/pathology , Extremities/blood supply , Humans , Ischemia/pathology , Mice , MicroRNAs/genetics
3.
BMC Genomics ; 16 Suppl 2: S2, 2015.
Article in English | MEDLINE | ID: mdl-25708300

ABSTRACT

BACKGROUND: Identification of genes with ascending or descending monotonic expression patterns over time or stages of stem cells is an important issue in time-series microarray data analysis. We propose a method named Monotonic Feature Selector (MFSelector) based on a concept of total discriminating error (DEtotal) to identify monotonic genes. MFSelector considers various time stages in stage order (i.e., Stage One vs. other stages, Stages One and Two vs. remaining stages and so on) and computes DEtotal of each gene. MFSelector can successfully identify genes with monotonic characteristics. RESULTS: We have demonstrated the effectiveness of MFSelector on two synthetic data sets and two stem cell differentiation data sets: embryonic stem cell neurogenesis (ESCN) and embryonic stem cell vasculogenesis (ESCV) data sets. We have also performed extensive quantitative comparisons of the three monotonic gene selection approaches. Some of the monotonic marker genes such as OCT4, NANOG, BLBP, discovered from the ESCN dataset exhibit consistent behavior with that reported in other studies. The role of monotonic genes found by MFSelector in either stemness or differentiation is validated using information obtained from Gene Ontology analysis and other literature. We justify and demonstrate that descending genes are involved in the proliferation or self-renewal activity of stem cells, while ascending genes are involved in differentiation of stem cells into variant cell lineages. CONCLUSIONS: We have developed a novel system, easy to use even with no pre-existing knowledge, to identify gene sets with monotonic expression patterns in multi-stage as well as in time-series genomics matrices. The case studies on ESCN and ESCV have helped to get a better understanding of stemness and differentiation. The novel monotonic marker genes discovered from a data set are found to exhibit consistent behavior in another independent data set, demonstrating the utility of the proposed method. The MFSelector R function and data sets can be downloaded from: http://microarray.ym.edu.tw/tools/MFSelector/.


Subject(s)
Computational Biology/methods , Gene Expression Profiling/methods , Oligonucleotide Array Sequence Analysis/methods , Stem Cells/metabolism , Algorithms , Cell Differentiation/genetics , Cell Lineage/genetics , Cluster Analysis , Homeodomain Proteins/genetics , Humans , Internet , Nanog Homeobox Protein , Neovascularization, Physiologic/genetics , Neurogenesis/genetics , Octamer Transcription Factor-3/genetics , Stem Cells/cytology , Time Factors
4.
mBio ; 5(5): e01469-14, 2014 Sep 30.
Article in English | MEDLINE | ID: mdl-25271282

ABSTRACT

UNLABELLED: Pairing high-throughput sequencing technologies with high-throughput mutagenesis enables genome-wide investigations of pathogenic organisms. Knowledge of the specific functions of protein domains encoded by the genome of the hepatitis C virus (HCV), a major human pathogen that contributes to liver disease worldwide, remains limited to insight from small-scale studies. To enhance the capabilities of HCV researchers, we have obtained a high-resolution functional map of the entire viral genome by combining transposon-based insertional mutagenesis with next-generation sequencing. We generated a library of 8,398 mutagenized HCV clones, each containing one 15-nucleotide sequence inserted at a unique genomic position. We passaged this library in hepatic cells, recovered virus pools, and simultaneously assayed the abundance of mutant viruses in each pool by next-generation sequencing. To illustrate the validity of the functional profile, we compared the genetic footprints of viral proteins with previously solved protein structures. Moreover, we show the utility of these genetic footprints in the identification of candidate regions for epitope tag insertion. In a second application, we screened the genetic footprints for phenotypes that reflected defects in later steps of the viral life cycle. We confirmed that viruses with insertions in a region of the nonstructural protein NS4B had a defect in infectivity while maintaining genome replication. Overall, our genome-wide HCV mutant library and the genetic footprints obtained by high-resolution profiling represent valuable new resources for the research community that can direct the attention of investigators toward unidentified roles of individual protein domains. IMPORTANCE: Our insertional mutagenesis library provides a resource that illustrates the effects of relatively small insertions on local protein structure and HCV viability. We have also generated complementary resources, including a website (http://hangfei.bol.ucla.edu) and a panel of epitope-tagged mutant viruses that should enhance the research capabilities of investigators studying HCV. Researchers can now detect epitope-tagged viral proteins by established antibodies, which will allow biochemical studies of HCV proteins for which antibodies are not readily available. Furthermore, researchers can now quickly look up genotype-phenotype relationships and base further mechanistic studies on the residue-by-residue information from the functional profile. More broadly, this approach offers a general strategy for the systematic functional characterization of viruses on the genome scale.


Subject(s)
Genome, Viral , Hepacivirus/genetics , Viral Proteins/genetics , Cell Line , Chromosome Mapping , Cloning, Molecular , DNA Transposable Elements/genetics , DNA, Viral/genetics , Gene Library , Hepacivirus/physiology , High-Throughput Nucleotide Sequencing , Humans , Mutagenesis, Insertional , Plasmids , Sequence Analysis, DNA , Transcription, Genetic , Transfection , Viral Proteins/metabolism , Virus Replication
5.
BMC Genomics ; 15: 802, 2014 Sep 18.
Article in English | MEDLINE | ID: mdl-25236949

ABSTRACT

BACKGROUND: Endothelial progenitor cells (EPCs) play a fundamental role in not only blood vessel development but also post-natal vascular repair. Currently EPCs are defined as early and late EPCs based on their biological properties and their time of appearance during in vitro culture. Both EPC types assist angiogenesis and have been linked to ischemia-related disorders, including coronary artery disease (CAD). RESULTS: We found late EPCs are more mobile than early EPCs and matured endothelial cells (ECs). To pinpoint the mechanism, microRNA profiles of early EPCs late EPCs, and ECs were deciphered by small RNA sequencing. Obtained signatures made up of both novel and known microRNAs, in which anti-angiogenic microRNAs such as miR-221 and miR-222 are more abundant in matured ECs than in late EPCs. Overexpression of miR-221 and miR-222 resulted in the reduction of genes involved in hypoxia response, metabolism, TGF-beta signalling, and cell motion. Not only hamper late EPC activities in vitro, both microRNAs (especially miR-222) also hindered in vivo vasculogenesis in a zebrafish model. Reporter assays showed that miR-222, but not miR-221, targets the angiogenic factor ETS1. In contrast, PIK3R1 is the target of miR-221, but not miR-222 in late EPCs. Clinically, both miR-221-PIK3R1 and miR-222-ETS1 pairs are deregulated in late EPCs of CAD patients. CONCLUSIONS: Our results illustrate EPCs and ECs exploit unique miRNA modalities to regulate angiogenic features, and explain why late EPC levels and activities are reduced in CAD patients. These data will further help to develop new plasma biomarkers and therapeutic approaches for ischemia-related diseases or tumor angiogenesis.


Subject(s)
Biomarkers/blood , Coronary Artery Disease/genetics , Endothelial Cells/metabolism , Fetal Blood/cytology , MicroRNAs/genetics , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Protein c-ets-1/genetics , Animals , Cells, Cultured , Class Ia Phosphatidylinositol 3-Kinase , Coronary Artery Disease/blood , Endothelial Progenitor Cells/metabolism , Female , Fetal Blood/metabolism , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells , Humans , In Vitro Techniques , MicroRNAs/blood , Neovascularization, Physiologic , Pregnancy , Sequence Analysis, RNA , Zebrafish
6.
PLoS One ; 9(5): e98070, 2014.
Article in English | MEDLINE | ID: mdl-24865854

ABSTRACT

Dysfunction and reduction of circulating endothelial progenitor cell (EPC) is correlated with the onset of cardiovascular disorders including coronary artery disease (CAD). VEGF is a known mitogen for EPC to migrate out of bone marrow to possess angiogenic activities, and the plasma levels of VEGF are inversely correlated to the progression of CAD. Circulating microRNAs (miRNAs) in patient body fluids have recently been considered to hold the potential of being novel disease biomarkers and drug targets. However, how miRNAs and VEGF cooperate to regulate CAD progression is still unclear. Through the small RNA sequencing (smRNA-seq), we deciphered the miRNome patterns of EPCs with different angiogenic activities, hypothesizing that miRNAs targeting VEGF must be more abundant in EPCs with lower angiogenic activities. Candidates of anti-VEGF miRNAs, including miR-361-5p and miR-484, were enriched in not only diseased EPCs but also the plasma of CAD patients. However, we found out only miR-361-5p, but not miR-484, was able to suppress VEGF expression and EPC activities. Reporter assays confirmed the direct binding and repression of miR-361-5p to the 3'-UTR of VEGF mRNA. Knock down of miR-361-5p not only restored VEGF levels and angiogenic activities of diseased EPCs in vitro, but further promoted blood flow recovery in ischemic limbs of mice. Collectively, we discovered a miR-361-5p/VEGF-dependent regulation that could help to develop new therapeutic modalities not only for ischemia-related diseases but also for tumor angiogenesis.


Subject(s)
Coronary Artery Disease/pathology , Endothelial Progenitor Cells/pathology , Ischemia/pathology , MicroRNAs/genetics , Neovascularization, Physiologic , Vascular Endothelial Growth Factor A/metabolism , Animals , Apoptosis , Case-Control Studies , Cell Movement , Cell Proliferation , Cells, Cultured , Coronary Artery Disease/blood , Coronary Artery Disease/genetics , Endothelial Progenitor Cells/metabolism , Humans , Immunoenzyme Techniques , Ischemia/etiology , Ischemia/metabolism , Mice , Mice, Nude , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor A/genetics
7.
Arterioscler Thromb Vasc Biol ; 34(4): 857-69, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24558106

ABSTRACT

OBJECTIVE: Defects in angiogenesis/vasculogenesis or vessel repair are major complications of coronary artery disease (CAD). Endothelial progenitor cells (EPCs) play a fundamental role in postnatal vascular repair and CAD. The role of microRNAs in CAD pathogenesis and their potential as biomarkers remain to be elucidated. APPROACH AND RESULTS: MicroRNA-31 (miR-31) level in both the plasma and EPCs of patients with CAD is found lower. miR-31 regulates EPC activities by targeting FAT atypical cadherin 4 and thromboxane A2 receptor, which show increased expression in CAD EPCs. Overexpressing miR-31 in CAD EPCs rescued their angiogenic and vasculogenic abilities both in vitro and in vivo. When exploring approaches to restore endogenous miR-31, we found that far-infrared treatment enhanced the expression of not only miR-31, but also miR-720 in CAD EPCs. miR-720, which was also decreased in EPCs and the plasma of patients with CAD, stimulated EPC activity by targeting vasohibin 1. The miR720-vasohibin 1 pair was shown to be downstream of FAT atypical cadherin 4, but not of thromboxane A2 receptor. FAT atypical cadherin 4 inhibited miR-720 expression via repression of the planar cell polarity signaling gene four-jointed box 1 (FJX1), which was required for miR-720 expression through a hypoxia-inducible factor 1, α subunit-dependent mechanism. Restoring miR-720 level strengthened activity of CAD EPCs. The miR-31-miR-720 pathway is shown critical to EPC activation and that downregulation of this pathway contributes to CAD pathogenesis. Circulating levels of miR-31, miR-720, and vasohibin 1 have the potential to allow early diagnosis of CAD and to act as prognosis biomarkers for CAD and other EPC-related diseases. CONCLUSIONS: Manipulating the expression of the miR-31-miR-720 pathway in malfunction EPCs should help develop novel therapeutic modalities.


Subject(s)
Coronary Artery Disease/blood , Endothelial Cells/metabolism , MicroRNAs/blood , Muscle, Skeletal/blood supply , Stem Cells/metabolism , Animals , Cadherins/metabolism , Case-Control Studies , Cell Cycle Proteins/metabolism , Cells, Cultured , Coronary Artery Disease/genetics , Coronary Artery Disease/physiopathology , Disease Models, Animal , Down-Regulation , Endothelial Cells/radiation effects , Endothelial Cells/transplantation , Genetic Markers , Hindlimb , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Infrared Rays , Ischemia/genetics , Ischemia/metabolism , Ischemia/physiopathology , Ischemia/surgery , Mice , Mice, Nude , Neovascularization, Physiologic , Oligonucleotides/metabolism , Receptors, Thromboxane A2, Prostaglandin H2/metabolism , Recovery of Function , Regional Blood Flow , Signal Transduction , Stem Cell Transplantation , Stem Cells/radiation effects , Time Factors , Transfection , Tumor Suppressor Proteins/metabolism
8.
BMC Genomics ; 13: 447, 2012 Sep 03.
Article in English | MEDLINE | ID: mdl-22943456

ABSTRACT

BACKGROUND: Endothelial progenitor cells (EPCs) play a fundamental role in post-natal vascular repair, yet EPCs from different anatomic locations possess unique biological properties. The underlying mechanisms are unclear. RESULTS: EPCs from CB expressed abundant genes involved in cell cycle, hypoxia signalling and blood vessel development, correlating with the phenotypes that CB-EPCs proliferated more rapidly, migrated faster, and formed tubule structure more efficiently. smRNA-seq further deciphered miRNome patterns in EPCs isolated from CB or PB: 54 miRNAs were enriched in CB-EPCs, while another 50 in PB-EPCs. Specifically, CB-EPCs expressed more angiogenic miRNAs such as miR-31, while PB-EPCs possessed more tumor suppressive miRNAs including miR-10a. Knocking down miR-31 levels in CB-EPCs suppressed cell migration and microtubule formation, while overexpressing miR-31 in PB-EPCs helped to recapitulate some of CB-EPC functions. CONCLUSIONS: Our results show the foundation for a more detailed understanding of EPCs from different anatomic sources. Stimulating the expression of angiogenic microRNAs or genes in EPCs of low activity (such as those from patients with cardiovascular diseases) might allow the development of novel therapeutic strategies.


Subject(s)
Endothelial Cells/metabolism , MicroRNAs/metabolism , Stem Cells/metabolism , Cell Movement , Cells, Cultured , Computational Biology , Endothelial Cells/cytology , Fetal Blood/cytology , Gene Expression Profiling , Humans , Leukocytes, Mononuclear/cytology , MicroRNAs/antagonists & inhibitors , Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...