Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Stem Cell Reports ; 18(11): 2138-2153, 2023 11 14.
Article in English | MEDLINE | ID: mdl-37863045

ABSTRACT

Congenital heart disease often arises from perturbations of transcription factors (TFs) that guide cardiac development. ISLET1 (ISL1) is a TF that influences early cardiac cell fate, as well as differentiation of other cell types including motor neuron progenitors (MNPs) and pancreatic islet cells. While lineage specificity of ISL1 function is likely achieved through combinatorial interactions, its essential cardiac interacting partners are unknown. By assaying ISL1 genomic occupancy in human induced pluripotent stem cell-derived cardiac progenitors (CPs) or MNPs and leveraging the deep learning approach BPNet, we identified motifs of other TFs that predicted ISL1 occupancy in each lineage, with NKX2.5 and GATA motifs being most closely associated to ISL1 in CPs. Experimentally, nearly two-thirds of ISL1-bound loci were co-occupied by NKX2.5 and/or GATA4. Removal of NKX2.5 from CPs led to widespread ISL1 redistribution, and overexpression of NKX2.5 in MNPs led to ISL1 occupancy of CP-specific loci. These results reveal how ISL1 guides lineage choices through a combinatorial code that dictates genomic occupancy and transcription.


Subject(s)
Induced Pluripotent Stem Cells , Transcription Factors , Humans , Transcription Factors/metabolism , Myocytes, Cardiac , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Induced Pluripotent Stem Cells/metabolism , Cell Differentiation/genetics , Homeobox Protein Nkx-2.5/genetics , Homeobox Protein Nkx-2.5/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism
2.
Life Sci Alliance ; 6(8)2023 08.
Article in English | MEDLINE | ID: mdl-37263777

ABSTRACT

Nonalcoholic steatohepatitis (NASH) can lead to cirrhosis and hepatocellular carcinoma in their advanced stages; however, there are currently no approved therapies. Here, we show that microRNA (miR)-33b in hepatocytes is critical for the development of NASH. miR-33b is located in the intron of sterol regulatory element-binding transcription factor 1 and is abundantly expressed in humans, but absent in rodents. miR-33b knock-in (KI) mice, which have a miR-33b sequence in the same intron of sterol regulatory element-binding transcription factor 1 as humans and express miR-33b similar to humans, exhibit NASH under high-fat diet feeding. This condition is ameliorated by hepatocyte-specific miR-33b deficiency but unaffected by macrophage-specific miR-33b deficiency. Anti-miR-33b oligonucleotide improves the phenotype of NASH in miR-33b KI mice fed a Gubra Amylin NASH diet, which induces miR-33b and worsens NASH more than a high-fat diet. Anti-miR-33b treatment reduces hepatic free cholesterol and triglyceride accumulation through up-regulation of the lipid metabolism-related target genes. Furthermore, it decreases the expression of fibrosis marker genes in cultured hepatic stellate cells. Thus, inhibition of miR-33b using nucleic acid medicine is a promising treatment for NASH.


Subject(s)
Liver Neoplasms , MicroRNAs , Non-alcoholic Fatty Liver Disease , Mice , Humans , Animals , Non-alcoholic Fatty Liver Disease/genetics , Antagomirs , MicroRNAs/genetics , MicroRNAs/metabolism , Cholesterol , Liver Neoplasms/pathology , Transcription Factors
3.
Sci Rep ; 13(1): 8104, 2023 05 18.
Article in English | MEDLINE | ID: mdl-37202449

ABSTRACT

The human gaze is directed at various locations from moment to moment in acquiring information necessary to recognize the external environment at the fine resolution of foveal vision. Previous studies showed that the human gaze is attracted to particular locations in the visual field at a particular time, but it remains unclear what visual features produce such spatiotemporal bias. In this study, we used a deep convolutional neural network model to extract hierarchical visual features from natural scene images and evaluated how much the human gaze is attracted to the visual features in space and time. Eye movement measurement and visual feature analysis using the deep convolutional neural network model showed that the gaze was more strongly attracted to spatial locations containing higher-order visual features than to locations containing lower-order visual features or to locations predicted by conventional saliency. Analysis of the time course of gaze attraction revealed that the bias to higher-order visual features was prominent within a short period after the beginning of observation of the natural scene images. These results demonstrate that higher-order visual features are a strong gaze attractor in both space and time, suggesting that the human visual system uses foveal vision resources to extract information from higher-order visual features with higher spatiotemporal priority.


Subject(s)
Attention , Fixation, Ocular , Humans , Vision, Ocular , Visual Fields , Fovea Centralis , Visual Perception
4.
bioRxiv ; 2023 Jan 07.
Article in English | MEDLINE | ID: mdl-36711864

ABSTRACT

Chronic inflammation and tissue fibrosis are common stress responses that worsen organ function, yet the molecular mechanisms governing their crosstalk are poorly understood. In diseased organs, stress-induced changes in gene expression fuel maladaptive cell state transitions and pathological interaction between diverse cellular compartments. Although chronic fibroblast activation worsens dysfunction of lung, liver, kidney, and heart, and exacerbates many cancers, the stress-sensing mechanisms initiating the transcriptional activation of fibroblasts are not well understood. Here, we show that conditional deletion of the transcription co-activator Brd4 in Cx3cr1-positive myeloid cells ameliorates heart failure and is associated with a dramatic reduction in fibroblast activation. Analysis of single-cell chromatin accessibility and BRD4 occupancy in vivo in Cx3cr1-positive cells identified a large enhancer proximal to Interleukin-1 beta (Il1b), and a series of CRISPR deletions revealed the precise stress-dependent regulatory element that controlled expression of Il1b in disease. Secreted IL1B functioned non-cell autonomously to activate a p65/RELA-dependent enhancer near the transcription factor MEOX1, resulting in a profibrotic response in human cardiac fibroblasts. In vivo, antibody-mediated IL1B neutralization prevented stress-induced expression of MEOX1, inhibited fibroblast activation, and improved cardiac function in heart failure. The elucidation of BRD4-dependent crosstalk between a specific immune cell subset and fibroblasts through IL1B provides new therapeutic strategies for heart disease and other disorders of chronic inflammation and maladaptive tissue remodeling.

5.
Sci Transl Med ; 14(662): eabj8670, 2022 09 14.
Article in English | MEDLINE | ID: mdl-36103516

ABSTRACT

The low-density lipoprotein receptor (LDLR) controls cellular delivery of cholesterol and clears LDL from the bloodstream, protecting against atherosclerotic heart disease, the leading cause of death in the United States. We therefore sought to identify regulators of the LDLR beyond the targets of current therapies and known causes of familial hypercholesterolemia. We found that cold shock domain-containing protein E1 (CSDE1) enhanced hepatic LDLR messenger RNA (mRNA) decay via its 3' untranslated region and regulated atherogenic lipoproteins in vivo. Using parallel phenotypic genome-wide CRISPR interference screens in a tissue culture model, we identified 40 specific regulators of the LDLR that were not previously identified by observational human genetic studies. Among these, we demonstrated that, in HepG2 cells, CSDE1 regulated the LDLR at least as strongly as statins and proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors. In addition, we showed that hepatic gene silencing of Csde1 treated diet-induced dyslipidemia in mice to a similar degree as Pcsk9 silencing. These results suggest the therapeutic potential of targeting CSDE1 to manipulate the posttranscriptional regulation of the LDLR mRNA for the prevention of cardiovascular disease. Our approach of modeling a clinically relevant phenotype in a forward genetic screen, followed by mechanistic pharmacologic dissection and in vivo validation, may serve as a generalizable template for the identification of therapeutic targets in other human disease states.


Subject(s)
Cold-Shock Response , DNA-Binding Proteins/metabolism , Proprotein Convertase 9 , RNA-Binding Proteins/metabolism , Animals , Humans , Mice , Proprotein Convertase 9/genetics , Proprotein Convertase 9/metabolism , RNA, Messenger/genetics , Receptors, LDL/genetics , Receptors, LDL/metabolism , Transcription, Genetic
6.
Nat Commun ; 12(1): 843, 2021 02 16.
Article in English | MEDLINE | ID: mdl-33594062

ABSTRACT

Adaptive thermogenesis is essential for survival, and therefore is tightly regulated by a central neural circuit. Here, we show that microRNA (miR)-33 in the brain is indispensable for adaptive thermogenesis. Cold stress increases miR-33 levels in the hypothalamus and miR-33-/- mice are unable to maintain body temperature in cold environments due to reduced sympathetic nerve activity and impaired brown adipose tissue (BAT) thermogenesis. Analysis of miR-33f/f dopamine-ß-hydroxylase (DBH)-Cre mice indicates the importance of miR-33 in Dbh-positive cells. Mechanistically, miR-33 deficiency upregulates gamma-aminobutyric acid (GABA)A receptor subunit genes such as Gabrb2 and Gabra4. Knock-down of these genes in Dbh-positive neurons rescues the impaired cold-induced thermogenesis in miR-33f/f DBH-Cre mice. Conversely, increased gene dosage of miR-33 in mice enhances thermogenesis. Thus, miR-33 in the brain contributes to maintenance of BAT thermogenesis and whole-body metabolism via enhanced sympathetic nerve tone through suppressing GABAergic inhibitory neurotransmission. This miR-33-mediated neural mechanism may serve as a physiological adaptive defense mechanism for several stresses including cold stress.


Subject(s)
MicroRNAs/metabolism , Sympathetic Nervous System/physiology , Thermogenesis/genetics , Adipose Tissue, Brown/physiology , Animals , Body Temperature/physiology , Body Weight , Brain/metabolism , Cell Line , Cold Temperature , Diet, High-Fat , Endoplasmic Reticulum Stress , Humans , Integrases/metabolism , Male , Mice , Mice, Obese , MicroRNAs/genetics , Oxygen Consumption/physiology , Phenotype , Protein Subunits/genetics , Protein Subunits/metabolism , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism
7.
Science ; 371(6530)2021 02 12.
Article in English | MEDLINE | ID: mdl-33303684

ABSTRACT

Mapping the gene-regulatory networks dysregulated in human disease would allow the design of network-correcting therapies that treat the core disease mechanism. However, small molecules are traditionally screened for their effects on one to several outputs at most, biasing discovery and limiting the likelihood of true disease-modifying drug candidates. Here, we developed a machine-learning approach to identify small molecules that broadly correct gene networks dysregulated in a human induced pluripotent stem cell (iPSC) disease model of a common form of heart disease involving the aortic valve (AV). Gene network correction by the most efficacious therapeutic candidate, XCT790, generalized to patient-derived primary AV cells and was sufficient to prevent and treat AV disease in vivo in a mouse model. This strategy, made feasible by human iPSC technology, network analysis, and machine learning, may represent an effective path for drug discovery.


Subject(s)
Aortic Valve Disease/drug therapy , Aortic Valve Stenosis/drug therapy , Aortic Valve/pathology , Calcinosis/drug therapy , Gene Regulatory Networks/drug effects , Machine Learning , Nitriles/pharmacology , Nitriles/therapeutic use , Thiazoles/pharmacology , Thiazoles/therapeutic use , Algorithms , Animals , Aortic Valve/drug effects , Aortic Valve/metabolism , Aortic Valve/physiopathology , Aortic Valve Disease/genetics , Aortic Valve Disease/physiopathology , Aortic Valve Stenosis/genetics , Aortic Valve Stenosis/physiopathology , Calcinosis/genetics , Calcinosis/physiopathology , Disease Models, Animal , Drug Discovery , Drug Evaluation, Preclinical , Gene Expression Regulation/drug effects , Haploinsufficiency , Humans , Induced Pluripotent Stem Cells , Mice, Inbred C57BL , RNA-Seq , Receptor, Notch1/genetics , Small Molecule Libraries
8.
Commun Biol ; 3(1): 434, 2020 08 13.
Article in English | MEDLINE | ID: mdl-32792557

ABSTRACT

Recent high-throughput approaches have revealed a vast number of transcripts with unknown functions. Many of these transcripts are long noncoding RNAs (lncRNAs), and intergenic region-derived lncRNAs are classified as long intergenic noncoding RNAs (lincRNAs). Although Myosin heavy chain 6 (Myh6) encoding primary contractile protein is down-regulated in stressed hearts, the underlying mechanisms are not fully clarified especially in terms of lincRNAs. Here, we screen upregulated lincRNAs in pressure overloaded hearts and identify a muscle-abundant lincRNA termed Lionheart. Compared with controls, deletion of the Lionheart in mice leads to decreased systolic function and a reduction in MYH6 protein levels following pressure overload. We reveal decreased MYH6 results from an interaction between Lionheart and Purine-rich element-binding protein A after pressure overload. Furthermore, human LIONHEART levels in left ventricular biopsy specimens positively correlate with cardiac systolic function. Our results demonstrate Lionheart plays a pivotal role in cardiac remodeling via regulation of MYH6.


Subject(s)
Heart/physiopathology , Pressure , RNA, Long Noncoding/genetics , Systole/genetics , Animals , Biopsy , Dependovirus/metabolism , Heart Ventricles/ultrastructure , Humans , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Promoter Regions, Genetic/genetics , RNA, Long Noncoding/metabolism , Rats , Up-Regulation/genetics
9.
JACC Basic Transl Sci ; 4(6): 701-714, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31709319

ABSTRACT

No effective treatment is yet available to reduce infarct size and improve clinical outcomes after acute myocardial infarction by enhancing early reperfusion therapy using primary percutaneous coronary intervention. The study showed that Kyoto University Substance 121 (KUS121) reduced endoplasmic reticulum stress, maintained adenosine triphosphate levels, and ameliorated the infarct size in a murine cardiac ischemia and reperfusion injury model. The study confirmed the cardioprotective effect of KUS121 in a porcine ischemia and reperfusion injury model. These findings confirmed that KUS121 is a promising novel therapeutic agent for myocardial infarction in conjunction with primary percutaneous coronary intervention.

10.
J Am Heart Assoc ; 8(13): e012609, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31242815

ABSTRACT

Background Micro RNA (miR)-33 targets cholesterol transporter ATP -binding cassette protein A1 and other antiatherogenic targets and contributes to atherogenic progression. Its inhibition or deletion is known to result in the amelioration of atherosclerosis in mice. However, mice lack the other member of the miR-33 family, miR-33b, which exists in humans and other large mammals. Thus, precise evaluation and comparison of the responsibilities of these 2 miRs during the progression of atherosclerosis has not been reported, although they are essential. Methods and Results In this study, we performed a comprehensive analysis of the difference between the function of miR-33a and miR-33b using genetically modified mice. We generated 4 strains with or without miR-33a and miR-33b. Comparison between mice with only miR-33a (wild-type mice) and mice with only miR-33b (miR-33a-/-/miR-33b+/+) revealed the dominant expression of miR-33b in the liver. To evaluate the whole body atherogenic potency of miR-33a and miR-33b, we developed apolipoprotein E-deficient/miR-33a+/+/miR-33b-/- mice and apolipoprotein E-deficient/miR-33a-/-/miR-33b+/+ mice. With a high-fat and high-cholesterol diet, the apolipoprotein E-deficient/miR-33a-/-/miR-33b+/+ mice developed increased atherosclerotic plaque versus apolipoprotein E-deficient/miR-33a+/+/miR-33b-/- mice, in line with the predominant expression of miR-33b in the liver and worsened serum cholesterol profile. By contrast, a bone marrow transplantation study showed no significant difference, which was consistent with the relevant expression levels of miR-33a and miR-33b in bone marrow cells. Conclusions The miR-33 family exhibits differences in distribution and regulation and particularly in the progression of atherosclerosis; miR-33b would be more potent than miR-33a.


Subject(s)
Atherosclerosis/genetics , Hepatocytes/metabolism , Liver/metabolism , MicroRNAs/genetics , Plaque, Atherosclerotic/genetics , Animals , Apolipoproteins B/metabolism , Bone Marrow Transplantation , Cholesterol/metabolism , Cholesterol, Dietary , Diet, High-Fat , Disease Progression , Gene Expression Profiling , Gene Knock-In Techniques , Macrophages, Peritoneal/metabolism , Mice , Mice, Knockout , Mice, Knockout, ApoE , Mice, Transgenic , MicroRNAs/metabolism , Triglycerides/metabolism
11.
Clin Sci (Lond) ; 133(4): 583-595, 2019 02 28.
Article in English | MEDLINE | ID: mdl-30777884

ABSTRACT

Recent reports, including ours, have indicated that microRNA (miR)-33 located within the intron of sterol regulatory element binding protein (SREBP) 2 controls cholesterol homeostasis and can be a potential therapeutic target for the treatment of atherosclerosis. Here, we show that SPAST, which encodes a microtubule-severing protein called SPASTIN, was a novel target gene of miR-33 in human. Actually, the miR-33 binding site in the SPAST 3'-UTR is conserved not in mice but in mid to large mammals, and it is impossible to clarify the role of miR-33 on SPAST in mice. We demonstrated that inhibition of miR-33a, a major form of miR-33 in human neurons, via locked nucleic acid (LNA)-anti-miR ameliorated the pathological phenotype in hereditary spastic paraplegia (HSP)-SPG4 patient induced pluripotent stem cell (iPSC)-derived cortical neurons. Thus, miR-33a can be a potential therapeutic target for the treatment of HSP-SPG4.


Subject(s)
Genetic Therapy/methods , Induced Pluripotent Stem Cells/metabolism , MicroRNAs/genetics , Neural Stem Cells/metabolism , Neurites/metabolism , Oligonucleotides/genetics , Spastic Paraplegia, Hereditary/therapy , Spastin/genetics , 3' Untranslated Regions , Binding Sites , Cells, Cultured , Gene Expression Regulation , Humans , Induced Pluripotent Stem Cells/pathology , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Neural Stem Cells/pathology , Neurites/pathology , Neurogenesis , Oligonucleotides/metabolism , Phenotype , Spastic Paraplegia, Hereditary/genetics , Spastic Paraplegia, Hereditary/metabolism , Spastic Paraplegia, Hereditary/pathology , Spastin/metabolism
12.
Sci Rep ; 8(1): 16749, 2018 11 13.
Article in English | MEDLINE | ID: mdl-30425314

ABSTRACT

Acute cardiac rupture and adverse left ventricular (LV) remodeling causing heart failure are serious complications of acute myocardial infarction (MI). While cardio-hepatic interactions have been recognized, their role in MI remains unknown. We treated cultured cardiomyocytes with conditioned media from various cell types and analyzed the media by mass spectrometry to identify α1-microglobulin (AM) as an Akt-activating hepatokine. In mouse MI model, AM protein transiently distributed in the infarct and border zones during the acute phase, reflecting infiltration of AM-bound macrophages. AM stimulation activated Akt, NFκB, and ERK signaling and enhanced inflammation as well as macrophage migration and polarization, while inhibited fibrogenesis-related mRNA expression in cultured macrophages and cardiac fibroblasts. Intramyocardial AM administration exacerbated macrophage infiltration, inflammation, and matrix metalloproteinase 9 mRNA expression in the infarct and border zones, whereas disturbed fibrotic repair, then provoked acute cardiac rupture in MI. Shotgun proteomics and lipid pull-down analysis found that AM partly binds to phosphatidic acid (PA) for its signaling and function. Furthermore, systemic delivery of a selective inhibitor of diacylglycerol kinase α-mediated PA synthesis notably reduced macrophage infiltration, inflammation, matrix metalloproteinase activity, and adverse LV remodeling in MI. Therefore, targeting AM signaling could be a novel pharmacological option to mitigate adverse LV remodeling in MI.


Subject(s)
Alpha-Globulins/metabolism , Hormones/metabolism , Myocardial Infarction/pathology , Signal Transduction , Animals , Cell Membrane/metabolism , Cell Movement , Enzyme Activation , Fibrosis , Inflammation/metabolism , Liver/metabolism , Macrophages/cytology , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Phosphatidic Acids/biosynthesis , Proto-Oncogene Proteins c-akt/metabolism , Ventricular Remodeling
13.
Arterioscler Thromb Vasc Biol ; 38(10): 2460-2473, 2018 10.
Article in English | MEDLINE | ID: mdl-30354203

ABSTRACT

Objective- Atherosclerosis is a common disease caused by a variety of metabolic and inflammatory disturbances. MicroRNA (miR)-33a within SREBF2 (sterol regulatory element-binding factor 2) is a potent target for treatment of atherosclerosis through regulating both aspects; however, the involvement of miR-33b within SREBF1 remains largely unknown. Although their host genes difference could lead to functional divergence of miR-33a/b, we cannot dissect the roles of miR-33a/b in vivo because of lack of miR-33b sequences in mice, unlike human. Approach and Results- Here, we analyzed the development of atherosclerosis using miR-33b knock-in humanized mice under apolipoprotein E-deficient background. MiR-33b is prominent both in human and mice on atheroprone condition. MiR-33b reduced serum high-density lipoprotein cholesterol levels and systemic reverse cholesterol transport. MiR-33b knock-in macrophages showed less cholesterol efflux capacity and higher inflammatory state via regulating lipid rafts. Thus, miR-33b promotes vulnerable atherosclerotic plaque formation. Furthermore, bone marrow transplantation experiments strengthen proatherogenic roles of macrophage miR-33b. Conclusions- Our data demonstrated critical roles of SREBF1-miR-33b axis on both lipid profiles and macrophage phenotype remodeling and indicate that miR-33b is a promising target for treating atherosclerosis.


Subject(s)
Atherosclerosis/metabolism , MicroRNAs/metabolism , Plaque, Atherosclerotic , Sterol Regulatory Element Binding Protein 1/metabolism , Aged , Aged, 80 and over , Animals , Apoptosis , Atherosclerosis/genetics , Atherosclerosis/pathology , Bone Marrow Transplantation , Case-Control Studies , Cholesterol, HDL/blood , Disease Models, Animal , Female , Gene Expression Regulation , Humans , Intestinal Absorption , Macrophages/metabolism , Macrophages/pathology , Male , Membrane Microdomains/metabolism , Mice, Inbred C57BL , Mice, Knockout, ApoE , MicroRNAs/genetics , Middle Aged , Phenotype , Signal Transduction , Sterol Regulatory Element Binding Protein 1/genetics , Triglycerides/blood
14.
Sci Rep ; 8(1): 8553, 2018 06 04.
Article in English | MEDLINE | ID: mdl-29867212

ABSTRACT

Recent evidence suggests that the accumulation of macrophages as a result of obesity-induced adipose tissue hypoxia is crucial for the regulation of tissue fibrosis, but the molecular mechanisms underlying adipose tissue fibrosis are still unknown. In this study, we revealed that periostin (Postn) is produced at extraordinary levels by adipose tissue after feeding with a high-fat diet (HFD). Postn was secreted at least from macrophages in visceral adipose tissue during the development of obesity, possibly due to hypoxia. Postn-/- mice had lower levels of crown-like structure formation and fibrosis in adipose tissue and were protected from liver steatosis. These mice also showed amelioration in systemic insulin resistance compared with HFD-fed WT littermates. Mice deficient in Postn in their hematopoietic compartment also had lower levels of inflammation in adipose tissue, in parallel with a reduction in ectopic lipid accumulation compared with the controls. Our data indicated that the regulation of Postn in visceral fat could be beneficial for the maintenance of healthy adipose tissue in obesity.


Subject(s)
Cell Adhesion Molecules/deficiency , Cellulitis/metabolism , Dietary Fats/adverse effects , Insulin Resistance , Intra-Abdominal Fat/metabolism , Obesity/metabolism , Animals , Cellulitis/chemically induced , Cellulitis/genetics , Cellulitis/pathology , Dietary Fats/pharmacology , Fibrosis , Intra-Abdominal Fat/pathology , Mice , Mice, Knockout , Obesity/chemically induced , Obesity/genetics , Obesity/pathology
15.
Mol Cell Biol ; 38(14)2018 07 15.
Article in English | MEDLINE | ID: mdl-29712758

ABSTRACT

MicroRNA 33 (miR-33) targets ATP-binding cassette transporter A1 (ABCA1), and its deficiency increases serum high-density lipoprotein (HDL)-cholesterol (HDL-C) and ameliorates atherosclerosis. Although we previously reported that miR-33 deficiency increased peripheral Ly6Chigh monocytes on an ApoE-deficient background, the effect of miR-33 on the monocyte population has not been fully elucidated, especially in a wild-type (WT) background. We found that Ly6Chigh monocytes in miR-33-/- mice were decreased in peripheral blood and increased in bone marrow (BM). Expansion of myeloid progenitors and decreased apoptosis in Lin- Sca1+ c-Kit+ (LSK) cells were observed in miR-33-/- mice. A BM transplantation study and competitive repopulation assay revealed that hematopoietic miR-33 deficiency caused myeloid expansion and increased peripheral Ly6Chigh monocytes and that nonhematopoietic miR-33 deficiency caused reduced peripheral Ly6Chigh monocytes. Expression of high-mobility group AT-hook 2 (HMGA2) targeted by miR-33 increased in miR-33-deficient LSK cells, and its knockdown abolished the reduction of apoptosis. Transduction of human apolipoprotein A1 and ABCA1 in WT mouse liver increased HDL-C and reduced peripheral Ly6Chigh monocytes. These data indicate that miR-33 deficiency affects distribution of inflammatory monocytes through dual pathways. One pathway involves the enhancement of Hmga2 expression in hematopoietic stem cells to increase Ly6Chigh monocytes, and the other involves the elevation of HDL-C to decrease peripheral Ly6Chigh monocytes.


Subject(s)
Antigens, Ly/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Monocytes/metabolism , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter 1/metabolism , Animals , Apolipoprotein A-I/genetics , Apolipoprotein A-I/metabolism , Apolipoproteins E/metabolism , Apoptosis , Atherosclerosis/genetics , Atherosclerosis/metabolism , Cholesterol, HDL/blood , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Knockout, ApoE , Monocytes/classification , Monocytes/cytology , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transduction, Genetic
16.
Arterioscler Thromb Vasc Biol ; 37(11): 2161-2170, 2017 11.
Article in English | MEDLINE | ID: mdl-28882868

ABSTRACT

OBJECTIVE: Abdominal aortic aneurysm (AAA) is an increasingly prevalent and ultimately fatal disease with no effective pharmacological treatment. Because matrix degradation induced by vascular inflammation is the major pathophysiology of AAA, attenuation of this inflammation may improve its outcome. Previous studies suggested that miR-33 (microRNA-33) inhibition and genetic ablation of miR-33 increased serum high-density lipoprotein cholesterol and attenuated atherosclerosis. APPROACH AND RESULTS: MiR-33a-5p expression in central zone of human AAA was higher than marginal zone. MiR-33 deletion attenuated AAA formation in both mouse models of angiotensin II- and calcium chloride-induced AAA. Reduced macrophage accumulation and monocyte chemotactic protein-1 expression were observed in calcium chloride-induced AAA walls in miR-33-/- mice. In vitro experiments revealed that peritoneal macrophages from miR-33-/- mice showed reduced matrix metalloproteinase 9 expression levels via c-Jun N-terminal kinase inactivation. Primary aortic vascular smooth muscle cells from miR-33-/- mice showed reduced monocyte chemotactic protein-1 expression by p38 mitogen-activated protein kinase attenuation. Both of the inactivation of c-Jun N-terminal kinase and p38 mitogen-activated protein kinase were possibly because of the increase of ATP-binding cassette transporter A1 that is a well-known target of miR-33. Moreover, high-density lipoprotein cholesterol derived from miR-33-/- mice reduced expression of matrix metalloproteinase 9 in macrophages and monocyte chemotactic protein-1 in vascular smooth muscle cells. Bone marrow transplantation experiments indicated that miR-33-deficient bone marrow cells ameliorated AAA formation in wild-type recipients. MiR-33 deficiency in recipient mice was also shown to contribute the inhibition of AAA formation. CONCLUSIONS: These data strongly suggest that inhibition of miR-33 will be effective as a novel strategy for treating AAA.


Subject(s)
Aorta, Abdominal/metabolism , Aortic Aneurysm, Abdominal/prevention & control , Aortitis/prevention & control , Inflammation Mediators/metabolism , MicroRNAs/metabolism , Angiotensin II , Animals , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/metabolism , Aortitis/chemically induced , Aortitis/genetics , Aortitis/metabolism , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Bone Marrow Transplantation , Calcium Chloride , Cell Line , Chemokine CCL2/metabolism , Cholesterol, HDL/blood , Dilatation, Pathologic , Disease Models, Animal , Female , Genetic Predisposition to Disease , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/pathology , Male , Matrix Metalloproteinase 9/metabolism , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Phenotype , Signal Transduction , Time Factors , Transfection , Vascular Remodeling , p38 Mitogen-Activated Protein Kinases/metabolism
17.
ESC Heart Fail ; 4(2): 112-121, 2017 May.
Article in English | MEDLINE | ID: mdl-28451447

ABSTRACT

AIMS: Recent studies have shown that serum microRNA (miR) abundance is informative for the diagnosis or prognosis of heart failure. However, the dynamics and kinetics of miRs in acute heart failure are largely unknown. Serial measurement and analysis of serum miRs changes in individuals along their therapeutic course could reduce inter-individual variation and should detect potentially important serum miRs related to disease mechanisms. Based on this concept, we profiled serum miR signatures of blood samples that were obtained sequentially on the day of admission and on hospital Day 7. METHODS AND RESULTS: This prospective, observational study included 42 consecutive acute heart failure patients (74 ± 1 years old, 24 male). From admission to Day 7, most of the patients showed clinical improvement. In such a cohort, we detected several fluctuations of serum miRs by two distinct screening methods (quantitative PCR and high-throughput sequencing). One of these fluctuating serum miRs, miR-122-5p, decreased significantly from Day 1 to Day 7 [median arbitrary unit (1st:3rd quantile value); 4.62 [2.39:12.3] to 3.07 [1.67:5.39], P = 0.007]. This fluctuation was significantly correlated with changes in serum liver function markers (estimated coefficient and 95% confidence interval; vs change in aspartate aminotransferase 1.69, 0.890-2.484, P < 0.001 and r = 0.560, vs change in alanine aminotransferase 1.09, 0.406-1.771, P = 0.007 and r = 0.428). CONCLUSIONS: The serum miR signature of patients with acute heart failure might indicate the severity of the disease or patients' response to therapeutic intervention. Notably, serum miR-122-5p levels reflect liver damage in this condition.

18.
PLoS One ; 12(3): e0172798, 2017.
Article in English | MEDLINE | ID: mdl-28253326

ABSTRACT

BACKGROUND: Despite recent progress with drug-eluting stents, restenosis and thrombosis after endovascular intervention are still major limitations in the treatment of cardiovascular diseases. These problems are possibly caused by inappropriate inhibition of neointimal formation and retardation of re-endothelialization on the surface of the stents. miR-126 has been shown to have the potential to enhance vascular endothelial cell proliferation. METHODS AND RESULTS: We designed and constructed a 27-nt double strand RNA (dsRNA) conjugated to cholesterol, which has high membrane permeability, and formed mature miR-126 after transfection. For site-specific induction of miR-126, we utilized poly (DL-lactide-co-glycolide) nanoparticles (NPs). miR-126-dsRNA-containing NPs (miR-126 NPs) significantly reduced the protein expression of a previously identified miR-126 target, SPRED1, in human umbilical vascular endothelial cells (HUVECs), and miR-126 NPs enhanced the proliferation and migration of HUVECs. On the other hand, miR-126 NPs reduced the proliferation and migration of vascular smooth muscle cells, via the suppression of IRS-1. Finally, we developed a stent system that eluted miR-126. This delivery system exhibited significant inhibition of neointimal formation in a rabbit model of restenosis. CONCLUSIONS: miR-126 NP-conjugated stents significantly inhibited the development of neointimal hyperplasia in rabbits. The present study may indicate the possibility of a novel therapeutic option to prevent restenosis after angioplasty.


Subject(s)
Drug Carriers/chemistry , Drug-Eluting Stents , MicroRNAs/chemistry , MicroRNAs/genetics , Nanoparticles/chemistry , Neointima/prevention & control , Animals , Base Sequence , Cell Movement , Cell Proliferation , Cholesterol/metabolism , Human Umbilical Vein Endothelial Cells/cytology , Humans , Lactic Acid/chemistry , MicroRNAs/metabolism , Muscle, Smooth, Vascular/cytology , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , RNA, Double-Stranded/chemistry , RNA, Double-Stranded/genetics , RNA, Double-Stranded/metabolism , Rabbits
19.
Circ Res ; 120(5): 835-847, 2017 Mar 03.
Article in English | MEDLINE | ID: mdl-27920122

ABSTRACT

RATIONALE: Heart failure and atherosclerosis share the underlying mechanisms of chronic inflammation followed by fibrosis. A highly conserved microRNA (miR), miR-33, is considered as a potential therapeutic target for atherosclerosis because it regulates lipid metabolism and inflammation. However, the role of miR-33 in heart failure remains to be elucidated. OBJECTIVE: To clarify the role of miR-33 involved in heart failure. METHODS AND RESULTS: We first investigated the expression levels of miR-33a/b in human cardiac tissue samples with dilated cardiomyopathy. Increased expression of miR-33a was associated with improving hemodynamic parameters. To clarify the role of miR-33 in remodeling hearts, we investigated the responses to pressure overload by transverse aortic constriction in miR-33-deficient (knockout [KO]) mice. When mice were subjected to transverse aortic constriction, miR-33 expression levels were significantly upregulated in wild-type left ventricles. There was no difference in hypertrophic responses between wild-type and miR-33KO hearts, whereas cardiac fibrosis was ameliorated in miR-33KO hearts compared with wild-type hearts. Despite the ameliorated cardiac fibrosis, miR-33KO mice showed impaired systolic function after transverse aortic constriction. We also found that cardiac fibroblasts were mainly responsible for miR-33 expression in the heart. Deficiency of miR-33 impaired cardiac fibroblast proliferation, which was considered to be caused by altered lipid raft cholesterol content. Moreover, cardiac fibroblast-specific miR-33-deficient mice also showed decreased cardiac fibrosis induced by transverse aortic constriction as systemic miR-33KO mice. CONCLUSION: Our results demonstrate that miR-33 is involved in cardiac remodeling, and it preserves lipid raft cholesterol content in fibroblasts and maintains adaptive fibrotic responses in the remodeling heart.


Subject(s)
Cholesterol/metabolism , Membrane Microdomains/metabolism , MicroRNAs/metabolism , Myocardium/metabolism , Myocardium/pathology , Ventricular Remodeling/physiology , Adult , Aged , Animals , Cells, Cultured , Female , Fibroblasts/metabolism , Fibroblasts/physiology , Fibrosis/metabolism , Fibrosis/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Middle Aged , Rats , Rats, Sprague-Dawley
20.
PLoS One ; 10(11): e0142904, 2015.
Article in English | MEDLINE | ID: mdl-26562412

ABSTRACT

BACKGROUND: Recently, it has been reported that specific microRNA (miRNA) levels are elevated in serum and can be used as biomarkers in patients with cardiovascular diseases. However, miRNAs expression profiles and their sources in pericardial fluid (PF) are unclear. METHODS AND RESULTS: The purpose of this study was to identify the levels of miRNAs in PF in relation to those in the serum in patients undergoing cardiac surgery. Serum (S) and PF from patients undergoing coronary artery bypass graft (CABG) due to stable angina pectoris (sAP) and unstable AP (uAP) and aortic valve replacement due to aortic stenosis (AS) were analyzed for the detection of miRNAs. We named these samples S-sAP, S-uAP, S-AS, PF-sAP, PF-uAP, and PF-AS, respectively. We first measured the levels of miR-423-5p, which was recognized previously as a biomarker for heart failure. miR-423-5p levels were significantly higher in PF than serum. Although there was no difference in miR-423-5p levels among the PF-AS, PF-sAP, and PF-uAP, its levels were significantly elevated in S-uAP compared with those in S-AS and S-sAP. In order to clarify the source of miR-423-5p in PF, we measured the levels of muscle-enriched miR-133a and vascular-enriched miR-126 and miR-92a in the same samples. miR-133a levels were significantly higher in serum than in PF, and it was elevated in S-uAP compared with S-AS. miR-126 level was significantly increased in serum compared with PF, and the level of miR-92a the similar tendency. miR-423-5p is located in the first intron of NSRP1. There is another miRNA, miR-3184, encoded in the opposite direction in the same region. In vitro experiments indicated that the duplex of miR-423-5p and miR-3184-3p was more resistant to RNase than the duplex of miR-423-5p and miR-133-3p, which may help to stabilize miR-423-5p in the PF. CONCLUSIONS: Our results suggested that miR-423-5p is enriched in PF, and serum miR-423-5p may be associate with uAP. Its expression pattern was different to that of muscle- and vascular-enriched miRNAs, miR-133a, miR-126, and miR-92a.


Subject(s)
Angina Pectoris/genetics , Aortic Valve Stenosis/genetics , MicroRNAs/blood , MicroRNAs/genetics , Pericardial Fluid/metabolism , Aged , Aged, 80 and over , Angina Pectoris/blood , Angina Pectoris/surgery , Aortic Valve Stenosis/blood , Aortic Valve Stenosis/surgery , Base Sequence , Coronary Artery Bypass , Female , Gene Expression Profiling , Heart Valve Prosthesis Implantation , Humans , Introns , Male , MicroRNAs/analysis , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...