Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
FASEB J ; 38(3): e23459, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38329343

ABSTRACT

Wound healing is facilitated by neoangiogenesis, a complex process that is essential to tissue repair in response to injury. MicroRNAs are small, noncoding RNAs that can regulate the wound healing process including stimulation of impaired angiogenesis that is associated with type-2 diabetes (T2D). Expression of miR-409-3p was significantly increased in the nonhealing skin wounds of patients with T2D compared to the non-wounded normal skin, and in the skin of a murine model with T2D. In response to high glucose, neutralization of miR-409-3p markedly improved EC growth and migration in human umbilical vein endothelial cells (HUVECs), promoted wound closure and angiogenesis as measured by increased CD31 in human skin organoids, while overexpression attenuated EC angiogenic responses. Bulk mRNA-Seq transcriptomic profiling revealed BTG2 as a target of miR-409-3p, where overexpression of miR-409-3p significantly decreased BTG2 mRNA and protein expression. A 3' untranslated region (3'-UTR) luciferase assay of BTG2 revealed decreased luciferase activity with overexpression of miR-409-3p, while inhibition had opposite effects. Mechanistically, in response to high glucose, miR-409-3p deficiency in ECs resulted in increased mTOR phosphorylation, meanwhile BTG-anti-proliferation factor 2 (BTG2) silencing significantly decreased mTOR phosphorylation. Endothelial-specific and tamoxifen-inducible miR-409-3p knockout mice (MiR-409IndECKO ) with hyperglycemia that underwent dorsal skin wounding showed significant improvement of wound closure, increased blood flow, granulation tissue thickness (GTT), and CD31 that correlated with increased BTG2 expression. Taken together, our results show that miR-409-3p is a critical mediator of impaired angiogenesis in diabetic skin wound healing.


Subject(s)
Diabetes Mellitus, Type 2 , Immediate-Early Proteins , MicroRNAs , Tumor Suppressor Proteins , Animals , Humans , Mice , Angiogenesis , Cell Proliferation/physiology , Diabetes Mellitus, Type 2/genetics , Glucose , Human Umbilical Vein Endothelial Cells/metabolism , Immediate-Early Proteins/genetics , Luciferases , Mice, Obese , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Messenger , TOR Serine-Threonine Kinases , Tumor Suppressor Proteins/genetics , Wound Healing/genetics
2.
Mol Ther Nucleic Acids ; 32: 995-1009, 2023 Jun 13.
Article in English | MEDLINE | ID: mdl-37332476

ABSTRACT

Angiogenesis is critical for tissue repair following myocardial infarction (MI), which is exacerbated under insulin resistance or diabetes. MicroRNAs are regulators of angiogenesis. We examined the metabolic regulation of miR-409-3p in post-infarct angiogenesis. miR-409-3p was increased in patients with acute coronary syndrome (ACS) and in a mouse model of acute MI. In endothelial cells (ECs), miR-409-3p was induced by palmitate, while vascular endothelial growth factor (VEGF) and fibroblast growth factor (FGF) decreased its expression. Overexpression of miR-409-3p decreased EC proliferation and migration in the presence of palmitate, whereas inhibition had the opposite effects. RNA sequencing (RNA-seq) profiling in ECs identified DNAJ homolog subfamily B member 9 (DNAJB9) as a target of miR-409-3p. Overexpression of miR-409-3p decreased DNAJB9 mRNA and protein expression by 47% and 31% respectively, while enriching DNAJB9 mRNA by 1.9-fold after Argonaute2 microribonucleoprotein immunoprecipitation. These effects were mediated through p38 mitogen-activated protein kinase (MAPK). Ischemia-reperfusion (I/R) injury in EC-specific miR-409-3p knockout (KO) mice (miR-409ECKO) fed a high-fat, high-sucrose diet increased isolectin B4 (53.3%), CD31 (56%), and DNAJB9 (41.5%). The left ventricular ejection fraction (EF) was improved by 28%, and the infarct area was decreased by 33.8% in miR-409ECKO compared with control mice. These findings support an important role of miR-409-3p in the angiogenic EC response to myocardial ischemia.

3.
Cell Mol Life Sci ; 78(23): 7663-7679, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34698882

ABSTRACT

Endothelial cells (ECs) within the microvasculature of brown adipose tissue (BAT) are important in regulating the plasticity of adipocytes in response to increased metabolic demand by modulating the angiogenic response. However, the mechanism of EC-adipocyte crosstalk during this process is not completely understood. We used RNA sequencing to profile microRNAs derived from BAT ECs of obese mice and identified an anti-angiogenic microRNA, miR-409-3p. MiR-409-3p overexpression inhibited EC angiogenic properties; whereas, its inhibition had the opposite effects. Mechanistic studies revealed that miR-409-3p targets ZEB1 and MAP4K3. Knockdown of ZEB1/MAP4K3 phenocopied the angiogenic effects of miR-409-3p. Adipocytes co-cultured with conditioned media from ECs deficient in miR-409-3p showed increased expression of BAT markers, UCP1 and CIDEA. We identified a pro-angiogenic growth factor, placental growth factor (PLGF), released from ECs in response to miR-409-3p inhibition. Deficiency of ZEB1 or MAP4K3 blocked the release of PLGF from ECs and PLGF stimulation of 3T3-L1 adipocytes increased UCP1 expression in a miR-409-3p dependent manner. MiR-409-3p neutralization improved BAT angiogenesis, glucose and insulin tolerance, and energy expenditure in mice with diet-induced obesity. These findings establish miR-409-3p as a critical regulator of EC-BAT crosstalk by modulating a ZEB1-MAP4K3-PLGF signaling axis, providing new insights for therapeutic intervention in obesity.


Subject(s)
Adipose Tissue, Brown/pathology , Insulin Resistance , MicroRNAs/genetics , Neovascularization, Pathologic/pathology , Placenta Growth Factor/metabolism , Protein Serine-Threonine Kinases/metabolism , Zinc Finger E-box-Binding Homeobox 1/metabolism , Adipose Tissue, Brown/metabolism , Animals , Endothelial Cells/metabolism , Endothelial Cells/pathology , Male , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/metabolism , Placenta Growth Factor/genetics , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Zinc Finger E-box-Binding Homeobox 1/genetics
4.
Genes (Basel) ; 11(11)2020 11 16.
Article in English | MEDLINE | ID: mdl-33207533

ABSTRACT

Rapid spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus responsible for coronavirus disease 2019 (COVID-19), has led to a global pandemic, failures of local health care systems, and global economic recession. MicroRNAs (miRNAs) have recently emerged as important regulators of viral pathogenesis, particularly among RNA viruses, but the impact of host miRNAs on SARS-CoV-2 infectivity remains unknown. In this study, we utilize the combination of powerful bioinformatic prediction algorithms and miRNA profiling to predict endogenous host miRNAs that may play important roles in regulating SARS-CoV-2 infectivity. We provide a collection of high-probability miRNA binding sites within the SARS-CoV-2 genome as well as within mRNA transcripts of critical viral entry proteins ACE2 and TMPRSS2 and their upstream modulators, the interferons (IFN). By utilizing miRNA profiling datasets of SARS-CoV-2-resistant and -susceptible cell lines, we verify the biological plausibility of the predicted miRNA-target RNA interactions. Finally, we utilize miRNA profiling of SARS-CoV-2-infected cells to identify predicted miRNAs that are differentially regulated in infected cells. In particular, we identify predicted miRNA binders to SARS-CoV-2 ORFs (miR-23a (1ab), miR-29a, -29c (1ab, N), miR-151a, -151b (S), miR-4707-3p (S), miR-298 (5'-UTR), miR-7851-3p (5'-UTR), miR-8075 (5'-UTR)), ACE2 3'-UTR (miR-9-5p, miR-218-5p), TMPRSS2 3'-UTR (let-7d-5p, -7e-5p, miR-494-3p, miR-382-3p, miR-181c-5p), and IFN-α 3'-UTR (miR-361-5p, miR-410-3p). Overall, this study provides insight into potential novel regulatory mechanisms of SARS-CoV-2 by host miRNAs and lays the foundation for future investigation of these miRNAs as potential therapeutic targets or biomarkers.


Subject(s)
Angiotensin-Converting Enzyme 2/genetics , Genome, Viral , Interferons/genetics , MicroRNAs/genetics , RNA, Messenger/genetics , SARS-CoV-2/genetics , Serine Endopeptidases/genetics , Angiotensin-Converting Enzyme 2/metabolism , Computational Biology/methods , Gene Silencing , Humans , Interferons/metabolism , MicroRNAs/chemistry , MicroRNAs/metabolism , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Serine Endopeptidases/metabolism , Transcriptome , Viral Proteins/genetics
5.
Am J Physiol Cell Physiol ; 318(3): C524-C535, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31913696

ABSTRACT

Neoangiogenesis is critical for tissue repair in response to injury such as myocardial ischemia or dermal wound healing. MicroRNAs are small noncoding RNAs and important regulators of angiogenesis under physiological and pathological disease states. Therefore, identification of microRNAs that may restore impaired angiogenesis in response to tissue injury may provide new targets for therapy. Using a microRNA microarray profiling approach, we identified a human-specific microRNA, miR-4674, that was significantly decreased in patients after myocardial tissue injury and had an endothelial cell (EC)-enriched expression pattern. Functionally, overexpression of miR-4674 markedly attenuated EC proliferation, migration, network tube formation, and spheroid sprouting, whereas blockade of miR-4674 had the opposite effects. Transcriptomic profiling, gene set enrichment analyses, bioinformatics, 3'-untranslated region (3'-UTR) reporter and microribonucleoprotein immunoprecipitation (miRNP-IP) assays, and small interfering RNA dependency studies revealed that miR-4674 regulates VEGF stimulated-p38 mitogen-activated protein kinase (MAPK) signaling and targets interleukin 1 receptor-associated kinase 1 (Irak1) and BICD cargo adaptor 2 (Bicd2) in ECs. Furthermore, Irak1 and Bicd2 were necessary for miR-4674-driven EC proliferation and migration. Finally, neutralization of miR-4674 increased angiogenesis, Irak1 and Bicd2 expression, and p38 phosphorylation in human skin organoids as a model of tissue injury. Collectively, targeting miR-4674 may provide a novel therapeutic target for tissue repair in pathological disease states associated with impaired angiogenesis.


Subject(s)
Endothelial Cells/metabolism , MAP Kinase Signaling System/physiology , MicroRNAs/biosynthesis , Neovascularization, Physiologic/physiology , Signal Transduction/physiology , Cell Proliferation/physiology , Female , Human Umbilical Vein Endothelial Cells , Humans , Male , MicroRNAs/genetics , Organ Culture Techniques
6.
Arterioscler Thromb Vasc Biol ; 39(7): 1458-1474, 2019 07.
Article in English | MEDLINE | ID: mdl-31092013

ABSTRACT

Objective- In response to tissue injury, the appropriate progression of events in angiogenesis is controlled by a careful balance between pro and antiangiogenic factors. We aimed to identify and characterize microRNAs that regulate angiogenesis in response to tissue injury. Approach and Results- We show that in response to tissue injury, microRNA-615-5p (miR-615-5p) is rapidly induced and serves as an antiangiogenic microRNA by targeting endothelial cell VEGF (vascular endothelial growth factor)-AKT (protein kinase B)/eNOS (endothelial nitric oxide synthase) signaling in vitro and in vivo. MiR-615-5p expression is increased in wounds of diabetic db/db mice, in plasma of human subjects with acute coronary syndromes, and in plasma and skin of human subjects with diabetes mellitus. Ectopic expression of miR-615-5p markedly inhibited endothelial cell proliferation, migration, network tube formation in Matrigel, and the release of nitric oxide, whereas miR-615-5p neutralization had the opposite effects. Mechanistic studies using transcriptomic profiling, bioinformatics, 3' untranslated region reporter and microribonucleoprotein immunoprecipitation assays, and small interfering RNA dependency studies demonstrate that miR-615-5p inhibits the VEGF-AKT/eNOS signaling pathway in endothelial cells by targeting IGF2 (insulin-like growth factor 2) and RASSF2 (Ras-associating domain family member 2). Local delivery of miR-615-5p inhibitors, markedly increased angiogenesis, granulation tissue thickness, and wound closure rates in db/db mice, whereas miR-615-5p mimics impaired these effects. Systemic miR-615-5p neutralization improved skeletal muscle perfusion and angiogenesis after hindlimb ischemia in db/db mice. Finally, modulation of miR-615-5p expression dynamically regulated VEGF-induced AKT signaling and angiogenesis in human skin organoids as a model of tissue injury. Conclusions- These findings establish miR-615-5p as an inhibitor of VEGF-AKT/eNOS-mediated endothelial cell angiogenic responses and that manipulating miR-615-5p expression could provide a new target for angiogenic therapy in response to tissue injury. Visual Overview- An online visual overview is available for this article.


Subject(s)
Endothelial Cells/physiology , MicroRNAs/physiology , Neovascularization, Physiologic , Nitric Oxide Synthase Type III/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Animals , Humans , Male , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type III/physiology , Phosphorylation , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction/physiology , Tumor Suppressor Proteins/physiology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/physiology
7.
FASEB J ; 33(4): 5599-5614, 2019 04.
Article in English | MEDLINE | ID: mdl-30668922

ABSTRACT

Angiogenesis is a critical process in repair of tissue injury that is regulated by a delicate balance between pro- and antiangiogenic factors. In disease states associated with impaired angiogenesis, we identified that miR-135a-3p is rapidly induced and serves as an antiangiogenic microRNA (miRNA) by targeting endothelial cell (EC) p38 signaling in vitro and in vivo. MiR-135a-3p overexpression significantly inhibited EC proliferation, migration, and network tube formation in matrigel, whereas miR-135-3p neutralization had the opposite effects. Mechanistic studies using transcriptomic profiling, bioinformatics, 3'-UTR reporter and miRNA ribonucleoprotein complex -immunoprecipitation assays, and small interfering RNA dependency studies revealed that miR-135a-3p inhibits the p38 signaling pathway in ECs by targeting huntingtin-interacting protein 1 (HIP1). Local delivery of miR-135a-3p inhibitors to wounds of diabetic db/db mice markedly increased angiogenesis, granulation tissue thickness, and wound closure rates, whereas local delivery of miR-135a-3p mimics impaired these effects. Finally, through gain- and loss-of-function studies in human skin organoids as a model of tissue injury, we demonstrated that miR-135a-3p potently modulated p38 signaling and angiogenesis in response to VEGF stimulation by targeting HIP1. These findings establish miR-135a-3p as a pivotal regulator of pathophysiological angiogenesis and tissue repair by targeting a VEGF-HIP1-p38K signaling axis, providing new targets for angiogenic therapy to promote tissue repair.-Icli, B., Wu, W., Ozdemir, D., Li, H., Haemmig, S., Liu, X., Giatsidis, G., Cheng, H. S., Avci, S. N., Kurt, M., Lee, N., Guimaraes, R. B., Manica, A., Marchini, J. F., Rynning, S. E., Risnes, I., Hollan, I., Croce, K., Orgill, D. P., Feinberg, M. W. MicroRNA-135a-3p regulates angiogenesis and tissue repair by targeting p38 signaling in endothelial cells.


Subject(s)
Endothelial Cells/pathology , MicroRNAs/genetics , Neovascularization, Pathologic/genetics , Signal Transduction/genetics , Wound Healing/genetics , p38 Mitogen-Activated Protein Kinases/genetics , Animals , Cell Line , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Mice, Inbred NOD/genetics , Vascular Endothelial Growth Factor A/genetics
8.
Cardiovasc Res ; 113(9): 1024-1034, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28505257

ABSTRACT

In this review, we focus on the emerging role of microRNAs, non-coding RNAs that regulate gene expression and signaling pathways, in dysfunctional adipose tissue. We highlight current paradigms of microRNAs involved in adipose differentiation and function in depots such as white, brown, and beige adipose tissues and potential implications of microRNA dysregulation in human disease such as obesity, inflammation, microvasculature dysfunction, and related cardiovascular diseases. We highlight accumulating studies indicating that adipocyte-derived microRNAs may not only serve as biomarkers of cardiometabolic disease, but also may directly regulate gene expression of other tissues. Finally, we discuss the future prospects, challenges, and emerging strategies for microRNA delivery and targeting for therapeutic applications in cardiovascular disease states associated with adipocyte dysfunction.


Subject(s)
Adipose Tissue/metabolism , Cardiovascular Diseases/genetics , Cardiovascular System/metabolism , MicroRNAs/genetics , Obesity/genetics , Adipose Tissue/pathology , Adipose Tissue/physiopathology , Animals , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Cardiovascular Diseases/physiopathology , Cardiovascular System/pathology , Cardiovascular System/physiopathology , Gene Expression Regulation , Humans , MicroRNAs/metabolism , Obesity/metabolism , Obesity/pathology , Obesity/physiopathology , Phenotype , Signal Transduction
9.
Endocr Rev ; 38(2): 145-168, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28323921

ABSTRACT

Chronic, low-grade systemic inflammation and impaired microvascular function are critical hallmarks in the development of insulin resistance. Accordingly, insulin resistance is a major risk factor for type 2 diabetes and cardiovascular disease. Accumulating studies demonstrate that restoration of impaired function of the diabetic macro- and microvasculature may ameliorate a range of cardiovascular disease states and diabetes-associated complications. In this review, we focus on the emerging role of microRNAs (miRNAs), noncoding RNAs that fine-tune target gene expression and signaling pathways, in insulin-responsive tissues and cell types important for maintaining optimal vascular homeostasis and preventing the sequelae of diabetes-induced end organ injury. We highlight current pathophysiological paradigms of miRNAs and their targets involved in regulating the diabetic microvasculature in a range of diabetes-associated complications such as retinopathy, nephropathy, wound healing, and myocardial injury. We provide an update of the potential use of circulating miRNAs diagnostically in type I or type II diabetes. Finally, we discuss emerging delivery platforms for manipulating miRNA expression or function as the next frontier in therapeutic intervention to improve diabetes-associated microvascular dysfunction and its attendant clinical consequences.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Diabetic Angiopathies/metabolism , Diabetic Cardiomyopathies/metabolism , Diabetic Nephropathies/metabolism , MicroRNAs/metabolism , Diabetes Mellitus, Type 2/therapy , Diabetic Angiopathies/therapy , Diabetic Cardiomyopathies/therapy , Diabetic Nephropathies/therapy , Humans , MicroRNAs/therapeutic use
11.
FASEB J ; 30(9): 3216-26, 2016 09.
Article in English | MEDLINE | ID: mdl-27297585

ABSTRACT

Thrombogenic and inflammatory mediators, such as thrombin, induce NF-κB-mediated endothelial cell (EC) activation and dysfunction, which contribute to pathogenesis of arterial thrombosis. The role of anti-inflammatory microRNA-181b (miR-181b) on thrombosis remains unknown. Our previous study demonstrated that miR-181b inhibits downstream NF-κB signaling in response to TNF-α. Here, we demonstrate that miR-181b uniquely inhibits upstream NF-κB signaling in response to thrombin. Overexpression of miR-181b inhibited thrombin-induced activation of NF-κB signaling, demonstrated by reduction of phospho-IKK-ß, -IκB-α, and p65 nuclear translocation in ECs. MiR-181b also reduced expression of NF-κB target genes VCAM-1, intercellular adhesion molecule-1, E-selectin, and tissue factor. Mechanistically, miR-181b targets caspase recruitment domain family member 10 (Card10), an adaptor protein that participates in activation of the IKK complex in response to signals transduced from protease-activated receptor-1. miR-181b reduced expression of Card10 mRNA and protein, but not protease-activated receptor-1. 3'-Untranslated region reporter assays, argonaute-2 microribonucleoprotein immunoprecipitation studies, and Card10 rescue studies revealed that Card10 is a bona fide direct miR-181b target. Small interfering RNA-mediated knockdown of Card10 expression phenocopied effects of miR-181b on NF-κB signaling and targets. Card10 deficiency did not affect TNF-α-induced activation of NF-κB signaling, which suggested stimulus-specific regulation of NF-κB signaling and endothelial responses by miR-181b in ECs. Finally, in response to photochemical injury-induced arterial thrombosis, systemic delivery of miR-181b reduced thrombus formation by 73% in carotid arteries and prolonged time to occlusion by 1.6-fold, effects recapitulated by Card10 small interfering RNA. These data demonstrate that miR-181b and Card10 are important regulators of thrombin-induced EC activation and arterial thrombosis. These studies highlight the relevance of microRNA-dependent targets in response to ligand-specific signaling in ECs.-Lin, J., He, S., Sun, X., Franck, G., Deng, Y., Yang, D., Haemmig, S., Wara, A. K. M., Icli, B., Li, D., Feinberg, M. W. MicroRNA-181b inhibits thrombin-mediated endothelial activation and arterial thrombosis by targeting caspase recruitment domain family member 10.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , MicroRNAs/metabolism , Thrombin/metabolism , Animals , CARD Signaling Adaptor Proteins/genetics , Endothelial Cells , Endothelium, Vascular , Gene Expression , Gene Knockdown Techniques , Humans , Inflammation/metabolism , Mice , MicroRNAs/genetics , NF-kappa B/genetics , NF-kappa B/metabolism , Phosphorylation , RNA Interference , Signal Transduction/physiology , Thoracic Outlet Syndrome , Thrombin/genetics , Thrombosis/etiology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
12.
Circ Res ; 118(5): 810-21, 2016 Mar 04.
Article in English | MEDLINE | ID: mdl-26830849

ABSTRACT

RATIONALE: The pathogenesis of insulin resistance involves dysregulated gene expression and function in multiple cell types, including endothelial cells (ECs). Post-transcriptional mechanisms such as microRNA-mediated regulation of gene expression could affect insulin action by modulating EC function. OBJECTIVE: To determine whether microRNA-181b (miR-181b) affects the pathogenesis of insulin resistance by regulating EC function in white adipose tissue during obesity. METHODS AND RESULTS: MiR-181b expression was reduced in adipose tissue ECs of obese mice, and rescue of miR-181b expression improved glucose homeostasis and insulin sensitivity. Systemic intravenous delivery of miR-181b robustly accumulated in adipose tissue ECs, enhanced insulin-mediated Akt phosphorylation at Ser473, and reduced endothelial dysfunction, an effect that shifted macrophage polarization toward an M2 anti-inflammatory phenotype in epididymal white adipose tissue. These effects were associated with increased endothelial nitric oxide synthase and FoxO1 phosphorylation as well as nitric oxide activity in epididymal white adipose tissue. In contrast, miR-181b did not affect insulin-stimulated Akt phosphorylation in liver and skeletal muscle. Bioinformatics and gene profiling approaches revealed that Pleckstrin homology domain leucine-rich repeat protein phosphatase, a phosphatase that dephosphorylates Akt at Ser473, is a novel target of miR-181b. Knockdown of Pleckstrin homology domain leucine-rich repeat protein phosphatase increased Akt phosphorylation at Ser473 in ECs, and phenocopied miR-181b's effects on glucose homeostasis, insulin sensitivity, and inflammation of epididymal white adipose tissue in vivo. Finally, ECs from diabetic subjects exhibited increased Pleckstrin homology domain leucine-rich repeat protein phosphatase expression. CONCLUSIONS: Our data underscore the importance of adipose tissue EC function in controlling the development of insulin resistance. Delivery of miR-181b or Pleckstrin homology domain leucine-rich repeat protein phosphatase inhibitors may represent a new therapeutic approach to ameliorate insulin resistance by improving adipose tissue endothelial Akt-endothelial nitric oxide synthase-nitric oxide signaling.


Subject(s)
Adipose Tissue, White/metabolism , Blood Glucose/metabolism , Endothelial Cells/metabolism , Homeostasis/physiology , Insulin Resistance/physiology , MicroRNAs/biosynthesis , Animals , Cells, Cultured , Diet, High-Fat/adverse effects , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Obese
13.
J Mol Cell Cardiol ; 91: 151-9, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26776318

ABSTRACT

Wound healing is a physiological reparative response to injury and a well-orchestrated process that involves hemostasis, cellular migration, proliferation, angiogenesis, extracellular matrix deposition, and wound contraction and re-epithelialization. However, patients with type 2 diabetes mellitus (T2D) are frequently afflicted with impaired wound healing that progresses into chronic wounds or diabetic ulcers, and may lead to complications including limb amputation. Herein, we investigate the potential role of microRNA-26a (miR-26a) in a diabetic model of wound healing. Expression of miR-26a is rapidly induced in response to high glucose in endothelial cells (ECs). Punch skin biopsy wounding of db/db mice revealed increased expression of miR-26a (~3.5-fold) four days post-wounding compared to that of WT mice. Local administration of a miR-26a inhibitor, LNA-anti-miR-26a, induced angiogenesis (up to ~80%), increased granulation tissue thickness (by 2.5-fold) and accelerated wound closure (53% after nine days) compared to scrambled anti-miR controls in db/db mice. These effects were independent of altered M1/M2 macrophage ratios. Mechanistically, inhibition of miR-26a increased its target gene SMAD1 in ECs nine days post-wounding of diabetic mice. In addition, high glucose reduced activity of the SMAD1-3'-UTR. Diabetic dermal wounds treated with LNA-anti-miR-26a had increased expression of ID1, a downstream modulator or SMAD1, and decreased expression of the cell cycle inhibitor p27. These findings establish miR-26a as an important regulator on the progression of skin wounds of diabetic mice by specifically regulating the angiogenic response after injury, and demonstrate that neutralization of miR-26a may serve as a novel approach for therapy.


Subject(s)
Diabetes Mellitus, Experimental/genetics , MicroRNAs/genetics , Neovascularization, Pathologic/genetics , Oligonucleotides, Antisense/genetics , Smad1 Protein/genetics , Wounds, Nonpenetrating/genetics , Animals , Cell Movement , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Dermis/drug effects , Dermis/metabolism , Dermis/pathology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation , Glucose/pharmacology , Humans , Inhibitor of Differentiation Protein 1/genetics , Inhibitor of Differentiation Protein 1/metabolism , Macrophages/metabolism , Macrophages/pathology , Male , Mast Cells/metabolism , Mast Cells/pathology , Mice , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/therapy , Oligonucleotides, Antisense/metabolism , Re-Epithelialization , Signal Transduction , Smad1 Protein/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Wounds, Nonpenetrating/metabolism , Wounds, Nonpenetrating/pathology , Wounds, Nonpenetrating/therapy
14.
Trends Cardiovasc Med ; 24(6): 241-8, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25066487

ABSTRACT

In response to acute myocardial infarction (MI), a complex series of cellular and molecular signaling events orchestrate the myocardial remodeling that ensues weeks to months after injury. Clinical, epidemiological, and pathological studies demonstrate that inadequate or impaired angiogenesis after myocardial injury is often associated with decreased left ventricular (LV) function and clinical outcomes. The microRNA family, miR-26, plays diverse roles in regulating key aspects of cellular growth, development, and activation. Recent evidence supports a central role for the miR-26 family in cardiovascular disease by controlling critical signaling pathways, such as BMP/SMAD1 signaling, and targets relevant to endothelial cell growth, angiogenesis, and LV function post-MI. Emerging studies of the miR-26 family in other cell types including vascular smooth muscle cells, cardiac fibroblasts, and cardiomyocytes suggest that miR-26 may bear important implications for a range of cardiovascular repair mechanisms. This review examines the current knowledge of the miR-26 family's role in key cell types that critically control cardiovascular disease under pathological and physiological stimuli.


Subject(s)
Endothelial Cells/physiology , MicroRNAs/genetics , Myocardial Infarction , Ventricular Dysfunction, Left , Ventricular Remodeling/physiology , Cell Proliferation , Fibroblasts/physiology , Gene Expression Regulation , Humans , Myocardial Infarction/complications , Myocardial Infarction/genetics , Myocytes, Cardiac/physiology , Neovascularization, Pathologic/genetics , Signal Transduction , Ventricular Dysfunction, Left/etiology , Ventricular Dysfunction, Left/genetics
15.
Circ Res ; 114(1): 32-40, 2014 Jan 03.
Article in English | MEDLINE | ID: mdl-24084690

ABSTRACT

RATIONALE: Activated nuclear factor (NF)-κB signaling in the vascular endothelium promotes the initiation and progression of atherosclerosis. Targeting endothelial NF-κB may provide a novel strategy to limit chronic inflammation. OBJECTIVE: To examine the role of microRNA-181b (miR-181b) in endothelial NF-κB signaling and effects on atherosclerosis. METHODS AND RESULTS: MiR-181b expression was reduced in the aortic intima and plasma in apolipoprotein E-deficient mice fed a high-fat diet. Correspondingly, circulating miR-181b in the plasma was markedly reduced in human subjects with coronary artery disease. Systemic delivery of miR-181b resulted in a 2.3-fold overexpression of miR-181b in the aortic intima of apolipoprotein E-deficient mice and suppressed NF-κB signaling revealed by bioluminescence imaging and reduced target gene expression in the aortic arch in apolipoprotein E-deficient/NF-κB-luciferase transgenic mice. MiR-181b significantly inhibited atherosclerotic lesion formation, proinflammatory gene expression and the influx of lesional macrophages and CD4+ T cells in the vessel wall. Mechanistically, miR-181b inhibited the expression of the target gene importin-α3, an effect that reduced NF-κB nuclear translocation specifically in the vascular endothelium of lesions, whereas surprisingly leukocyte NF-κB signaling was unaffected despite a 7-fold overexpression of miR-181b. Our findings uncover that NF-κB nuclear translocation in leukocytes does not involve importin-α3, but rather importin-α5, which miR-181b does not target, highlighting that inhibition of NF-κB signaling in the endothelium is sufficient to mediate miR-181b's protective effects. CONCLUSIONS: Systemic delivery of miR-181b inhibits the activation of NF-κB and atherosclerosis through cell-specific mechanisms in the vascular endothelium. These findings support the rationale that delivery of miR-181b may provide a novel therapeutic approach to treat chronic inflammatory diseases such as atherosclerosis.


Subject(s)
Aorta/metabolism , Atherosclerosis/therapy , MicroRNAs/therapeutic use , NF-kappa B/metabolism , Tunica Intima/metabolism , Animals , Aorta/pathology , Apolipoproteins E/genetics , Atherosclerosis/etiology , Atherosclerosis/metabolism , Atherosclerosis/pathology , CD4-Positive T-Lymphocytes/metabolism , Diet, High-Fat/adverse effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Inflammation/metabolism , Karyopherins/genetics , Karyopherins/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , MicroRNAs/administration & dosage , MicroRNAs/blood , MicroRNAs/metabolism , NF-kappa B/antagonists & inhibitors , Tunica Intima/pathology
16.
Circ Res ; 113(11): 1231-41, 2013 Nov 08.
Article in English | MEDLINE | ID: mdl-24047927

ABSTRACT

RATIONALE: The rapid induction and orchestration of new blood vessels are critical for tissue repair in response to injury, such as myocardial infarction, and for physiological angiogenic responses, such as embryonic development and exercise. OBJECTIVE: We aimed to identify and characterize microRNAs (miR) that regulate pathological and physiological angiogenesis. METHODS AND RESULTS: We show that miR-26a regulates pathological and physiological angiogenesis by targeting endothelial cell (EC) bone morphogenic protein/SMAD1 signaling in vitro and in vivo. MiR-26a expression is increased in a model of acute myocardial infarction in mice and in human subjects with acute coronary syndromes. Ectopic expression of miR-26a markedly induced EC cycle arrest and inhibited EC migration, sprouting angiogenesis, and network tube formation in matrigel, whereas blockade of miR-26a had the opposite effects. Mechanistic studies demonstrate that miR-26a inhibits the bone morphogenic protein/SMAD1 signaling pathway in ECs by binding to the SMAD1 3'-untranslated region, an effect that decreased expression of Id1 and increased p21(WAF/CIP) and p27. In zebrafish, miR-26a overexpression inhibited formation of the caudal vein plexus, a bone morphogenic protein-responsive process, an effect rescued by ectopic SMAD1 expression. In mice, miR-26a overexpression inhibited EC SMAD1 expression and exercise-induced angiogenesis. Furthermore, systemic intravenous administration of an miR-26a inhibitor, locked nucleic acid-anti-miR-26a, increased SMAD1 expression and rapidly induced robust angiogenesis within 2 days, an effect associated with reduced myocardial infarct size and improved heart function. CONCLUSIONS: These findings establish miR-26a as a regulator of bone morphogenic protein/SMAD1-mediated EC angiogenic responses, and that manipulating miR-26a expression could provide a new target for rapid angiogenic therapy in ischemic disease states.


Subject(s)
Bone Morphogenetic Proteins/physiology , MicroRNAs/physiology , Neovascularization, Pathologic/physiopathology , Neovascularization, Physiologic/physiology , Signal Transduction/physiology , Smad1 Protein/physiology , Acute Coronary Syndrome/blood , Acute Coronary Syndrome/pathology , Acute Coronary Syndrome/physiopathology , Animals , Biomarkers/blood , Cell Proliferation , Disease Models, Animal , Embryonic Development/physiology , Endothelium, Vascular/pathology , Endothelium, Vascular/physiology , Endothelium, Vascular/physiopathology , Humans , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , MicroRNAs/blood , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Ventricular Dysfunction, Left/physiopathology , Ventricular Function, Left/physiology , Zebrafish
17.
Arterioscler Thromb Vasc Biol ; 33(7): 1552-60, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23685559

ABSTRACT

OBJECTIVE: The objective of this study was to investigate the role of Kruppel-like factor (KLF) 10, a zinc-finger transcription factor, in bone marrow (BM)-derived cell responses to arterial endothelial injury. Accumulating evidence indicates that BM-derived progenitors are recruited to sites of vascular injury and contribute to endothelial repair. APPROACH AND RESULTS: In response to carotid artery endothelial denudation, KLF10 mRNA expression was markedly increased in both BM and circulating lin(-) progenitor cells. To examine the specific role of KLF10 in arterial reendothelialization, we used 2 models of endothelial denudation (wire- and thermal-induced injury) of the carotid artery in wild-type (WT) and KLF10(-/-) mice. WT mice displayed higher areas of reendothelialization compared with KLF10(-/-) mice after endothelial injury using either method. BM transplant studies revealed that reconstitution of KLF10(-/-) mice with WT BM fully rescued the defect in reendothelialization and increased lin(-)CD34(+)kinase insert domain receptor(+) progenitors in the blood and injured carotid arteries. Conversely, reconstitution of WT mice with KLF10(-/-) BM recapitulated the defects in reendothelialization and peripheral cell progenitors. The media from cultured KLF10(-)/(-) BM progenitors was markedly inefficient in promoting endothelial cell growth and migration compared with the media from WT progenitors, indicative of defective paracrine trophic effects from KLF10(-)/(-) BM progenitors. Finally, BM-derived KLF10(-/-) lin(-) progenitors from reconstituted mice had reduced CXC-chemokine receptor 4 expression and impaired migratory responses. CONCLUSIONS: Collectively, these observations demonstrate a protective role for BM-derived KLF10 in paracrine and homing responses important for arterial endothelial injury and highlight KLF10 as a possible therapeutic target to promote endothelial repair in vascular disease states.


Subject(s)
Bone Marrow Cells/metabolism , Carotid Artery Injuries/metabolism , Cell Proliferation , Early Growth Response Transcription Factors/metabolism , Endothelial Cells/metabolism , Kruppel-Like Transcription Factors/metabolism , Paracrine Communication , Stem Cells/metabolism , Vascular System Injuries/metabolism , Animals , Antigens, CD34/metabolism , Biomarkers/metabolism , Bone Marrow Transplantation , Carotid Artery Injuries/genetics , Carotid Artery Injuries/pathology , Chemotaxis , Culture Media, Conditioned/metabolism , Disease Models, Animal , Early Growth Response Transcription Factors/deficiency , Early Growth Response Transcription Factors/genetics , Endothelial Cells/pathology , Female , Gene Expression Regulation , Kruppel-Like Transcription Factors/deficiency , Kruppel-Like Transcription Factors/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/metabolism , Receptors, CCR4/metabolism , Signal Transduction , Stem Cell Transplantation , Time Factors , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vascular System Injuries/genetics , Vascular System Injuries/pathology
18.
J Clin Invest ; 122(6): 1973-90, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22622040

ABSTRACT

EC activation and dysfunction have been linked to a variety of vascular inflammatory disease states. The function of microRNAs (miRNAs) in vascular EC activation and inflammation remains poorly understood. Herein, we report that microRNA-181b (miR-181b) serves as a potent regulator of downstream NF-κB signaling in the vascular endothelium by targeting importin-α3, a protein that is required for nuclear translocation of NF-κB. Overexpression of miR-181b inhibited importin-α3 expression and an enriched set of NF-κB-responsive genes such as adhesion molecules VCAM-1 and E-selectin in ECs in vitro and in vivo. In addition, treatment of mice with proinflammatory stimuli reduced miR-181b expression. Rescue of miR-181b levels by systemic administration of miR-181b "mimics" reduced downstream NF-κB signaling and leukocyte influx in the vascular endothelium and decreased lung injury and mortality in endotoxemic mice. In contrast, miR-181b inhibition exacerbated endotoxin-induced NF-κB activity, leukocyte influx, and lung injury. Finally, we observed that critically ill patients with sepsis had reduced levels of miR-181b compared with control intensive care unit (ICU) subjects. Collectively, these findings demonstrate that miR-181b regulates NF-κB-mediated EC activation and vascular inflammation in response to proinflammatory stimuli and that rescue of miR-181b expression could provide a new target for antiinflammatory therapy and critical illness.


Subject(s)
Endothelial Cells/metabolism , MicroRNAs/metabolism , Vasculitis/metabolism , Acute Lung Injury/genetics , Acute Lung Injury/immunology , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Acute Lung Injury/therapy , Animals , Critical Illness , E-Selectin/genetics , E-Selectin/immunology , E-Selectin/metabolism , Endothelial Cells/immunology , Endothelial Cells/pathology , Endotoxemia/genetics , Endotoxemia/immunology , Endotoxemia/metabolism , Endotoxemia/pathology , Endotoxemia/therapy , Humans , Leukocytes/immunology , Leukocytes/metabolism , Leukocytes/pathology , Lipopolysaccharides/toxicity , Mice , MicroRNAs/genetics , MicroRNAs/immunology , NF-kappa B/genetics , NF-kappa B/immunology , NF-kappa B/metabolism , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/immunology , Vascular Cell Adhesion Molecule-1/metabolism , Vasculitis/genetics , Vasculitis/immunology , Vasculitis/therapy , alpha Karyopherins/genetics , alpha Karyopherins/immunology , alpha Karyopherins/metabolism
19.
Biochem Biophys Res Commun ; 418(1): 116-21, 2012 Feb 03.
Article in English | MEDLINE | ID: mdl-22244893

ABSTRACT

The intracellular domain of ErbB4 receptor tyrosine kinase is known to translocate to the nucleus of cells where it can regulate p53 transcriptional activity. The purpose of this study was to examine whether ErbB4 can localize to the nucleus of adult rat ventricular myocytes (ARVM), and regulate p53 in these cells. We demonstrate that ErbB4 does locate to the nucleus of cardiac myocytes as a full-length protein, although nuclear location occurs as a full-length protein that does not require Protein Kinase C or γ-secretase activity. Consistent with this we found that only the non-cleavable JM-b isoform of ErbB4 is expressed in ARVM. Doxorubicin was used to examine ErbB4 role in regulation of a DNA damage response in ARVM. Doxorubicin induced p53 and p21 was suppressed by treatment with AG1478, an EGFR and ErbB4 kinase inhibitor, or suppression of ErbB4 expression with small interfering RNA. Thus ErbB4 localizes to the nucleus as a full-length protein, and plays a role in the DNA damage response induced by doxorubicin in cardiac myocytes.


Subject(s)
Cell Nucleus/enzymology , DNA Damage , ErbB Receptors/metabolism , Myocytes, Cardiac/enzymology , Animals , Cell Nucleus/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Doxorubicin/pharmacology , ErbB Receptors/antagonists & inhibitors , Heart Ventricles/cytology , Mice , Myocytes, Cardiac/drug effects , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Rats , Receptor, ErbB-4 , Tumor Suppressor Protein p53/metabolism , Tyrphostins/pharmacology
20.
Blood ; 118(24): 6461-4, 2011 Dec 08.
Article in English | MEDLINE | ID: mdl-21828132

ABSTRACT

Clinical studies using bone marrow-derived proangiogenic cells (PACs) have demonstrated modest improvements of function and/or perfusion of ischemic myocardium or skeletal muscle. Because the identities of these PACs and their functional ability to promote neovascularization remain poorly understood, it is possible that a subset of robust PACs exists but is obscured by the heterogeneous nature of this cell population. Herein, we found that common myeloid progenitors (CMPs) and granulocyte-macrophage progenitors (GMPs) preferentially differentiate into PACs compared with megakaryocyte-erythrocyte progenitors, hematopoietic stem cells, and common lymphoid progenitors. In vivo hindlimb ischemia studies and Matrigel plug assays verified the enhanced neovascularization properties uniquely associated with PACs derived from CMPs and GMPs. Taken together, these observations identify CMPs and GMPs as key bone marrow progenitors for optimal PAC function in vitro and in vivo and provide a foundation for novel therapeutic approaches to modulate angiogenesis.


Subject(s)
Granulocyte-Macrophage Progenitor Cells/physiology , Ischemia/physiopathology , Myeloid Progenitor Cells/physiology , Neovascularization, Physiologic , Animals , Biomarkers/metabolism , Bone Marrow Cells/physiology , Cells, Cultured , Coculture Techniques , Hindlimb , Human Umbilical Vein Endothelial Cells/physiology , Ischemia/metabolism , Ischemia/pathology , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/blood supply , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myocardial Ischemia/physiopathology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...