Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-33004402

ABSTRACT

INTRODUCTION: We previously demonstrated in primary cultures of human subcutaneous adipocytes and in a mouse model of diet-induced obesity that specific microRNA-22-3p antagomirs produce a significant reduction of fat mass and an improvement of several metabolic parameters. These effects are related to the activation of target genes such as KDM3A, KDM6B, PPARA, PPARGC1B and SIRT1 involved in lipid catabolism, thermogenesis, insulin sensitivity and glucose homeostasis. RESEARCH DESIGN AND METHODS: We now report a dedicated study exploring over the course of 3 months the metabolic and energetic effects of subcutaneous administration of our first miR-22-3p antagomir drug candidate (APT-110) in adult C57BL/6 male mice. Body composition, various blood parameters and energy expenditure were measured at several timepoints between week 12 and week 27 of age. RESULTS: Weekly subcutaneous injections of APT-110 for 12 weeks produced a sustained increase of energy expenditure as early as day 11 of treatment, a significant fat mass reduction, but no change of appetite nor physical activity. Insulin sensitivity as well as circulating glucose, cholesterol and leptin were improved. There was a dramatic reduction of liver steatosis after 3 months of active treatment. RNA sequencing revealed an activation of lipid metabolism pathways in a tissue-specific manner. CONCLUSIONS: These original findings suggest that microRNA-22-3p inhibition could lead to a potent treatment of fat accumulation, insulin resistance, and related complex metabolic disorders such as obesity, type 2 diabetes mellitus and non-alcoholic fatty liver disease.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , MicroRNAs , Animals , Insulin Resistance/genetics , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Obesity/drug therapy , Obesity/genetics
2.
Nucleic Acid Ther ; 30(2): 104-116, 2020 04.
Article in English | MEDLINE | ID: mdl-31873061

ABSTRACT

Diabesity is a growing pandemic with substantial health and financial consequences. We are developing microRNA (miRNA)-based drug candidates that transform fat storing adipocytes into fat burning adipocytes (browning effect) to treat metabolic diseases characterized by lipotoxicity. Through phenotypic screening in primary cultures of human subcutaneous adipocytes, we discovered that inhibition of miRNA-22-3p by several complementary antagomirs resulted in increased lipid oxidation, mitochondrial activity, and energy expenditure (EE). These effects may be mediated through activation of target genes like KDM3A, KDM6B, PPARA, PPARGC1B, and SIRT1 involved in lipid catabolism, thermogenesis, and glucose homeostasis. In the model of Diet-Induced Obesity in mice of various ages, weekly subcutaneous injections of various miRNA-22-3p antagomirs produced a significant fat mass reduction, but no change of appetite or body temperature. Insulin sensitivity, as well as circulating glucose and cholesterol levels, was also improved. These original findings suggest that miRNA-22-3p inhibition could become a potent treatment of human obesity and type 2 diabetes mellitus, the so-called diabesity characterized by lipotoxicity and insulin resistance.


Subject(s)
Adipocytes/drug effects , Diabetes Mellitus/therapy , Energy Metabolism/drug effects , MicroRNAs/genetics , Adipocytes/metabolism , Animals , Antagomirs/pharmacology , Diabetes Mellitus/genetics , Diabetes Mellitus/pathology , Humans , Insulin Resistance/genetics , Jumonji Domain-Containing Histone Demethylases/genetics , Lipid Peroxidation/drug effects , Mice , MicroRNAs/antagonists & inhibitors , PPAR alpha/genetics , RNA-Binding Proteins/genetics , Sirtuin 1/genetics
3.
J Neurosci ; 35(44): 14717-26, 2015 Nov 04.
Article in English | MEDLINE | ID: mdl-26538644

ABSTRACT

Dysregulation of amyloid-ß (Aß) metabolism is critical for Alzheimer's disease (AD) pathogenesis. Mounting evidence suggests that apolipoprotein E (ApoE) is involved in Aß metabolism. ATP-binding cassette transporter A1 (ABCA1) is a key regulator of ApoE lipidation, which affects Aß levels. Therefore, identifying regulatory mechanisms of ABCA1 expression in the brain may provide new therapeutic targets for AD. Here, we demonstrate that microRNA-33 (miR-33) regulates ABCA1 and Aß levels in the brain. Overexpression of miR-33 impaired cellular cholesterol efflux and dramatically increased extracellular Aß levels by promoting Aß secretion and impairing Aß clearance in neural cells. In contrast, genetic deletion of mir-33 in mice dramatically increased ABCA1 levels and ApoE lipidation, but it decreased endogenous Aß levels in cortex. Most importantly, pharmacological inhibition of miR-33 via antisense oligonucleotide specifically in the brain markedly decreased Aß levels in cortex of APP/PS1 mice, representing a potential therapeutic strategy for AD. SIGNIFICANCE STATEMENT: Brain lipid metabolism, in particular Apolipoprotein E (ApoE) lipidation, is critical to Aß metabolism and Alzheimer's disease (AD). Brain lipid metabolism is largely separated from the periphery due to blood-brain barrier and different repertoire of lipoproteins. Therefore, identifying the novel regulatory mechanism of brain lipid metabolism may provide a new therapeutic strategy for AD. Although there have been studies on brain lipid metabolism, its regulation, in particular by microRNAs, is relatively unknown. Here, we demonstrate that inhibition of microRNA-33 increases lipidation of brain ApoE and reduces Aß levels by inducing ABCA1. We provide a unique approach for AD therapeutics to increase ApoE lipidation and reduce Aß levels via pharmacological inhibition of microRNA in vivo.


Subject(s)
Amyloid beta-Peptides/metabolism , Apolipoproteins E/metabolism , Brain/metabolism , Lipid Metabolism/physiology , MicroRNAs/physiology , Amyloid beta-Peptides/genetics , Animals , Base Sequence , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data
4.
Hepatology ; 61(1): 141-52, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25131933

ABSTRACT

UNLABELLED: Identification of microRNAs (miRNAs) that regulate lipid metabolism is important to advance the understanding and treatment of some of the most common human diseases. In the liver, a few key miRNAs have been reported that regulate lipid metabolism, but since many genes contribute to hepatic lipid metabolism, we hypothesized that other such miRNAs exist. To identify genes repressed by miRNAs in mature hepatocytes in vivo, we injected adult mice carrying floxed Dicer1 alleles with an adenoassociated viral vector expressing Cre recombinase specifically in hepatocytes. By inactivating Dicer in adult quiescent hepatocytes we avoided the hepatocyte injury and regeneration observed in previous mouse models of global miRNA deficiency in hepatocytes. Next, we combined gene and miRNA expression profiling to identify candidate gene/miRNA interactions involved in hepatic lipid metabolism and validated their function in vivo using antisense oligonucleotides. A candidate gene that emerged from our screen was lipoprotein lipase (Lpl), which encodes an enzyme that facilitates cellular uptake of lipids from the circulation. Unlike in energy-dependent cells like myocytes, LPL is normally repressed in adult hepatocytes. We identified miR-29a as the miRNA responsible for repressing LPL in hepatocytes, and found that decreasing hepatic miR-29a levels causes lipids to accumulate in mouse livers. CONCLUSION: Our screen suggests several new miRNAs are regulators of hepatic lipid metabolism. We show that one of these, miR-29a, contributes to physiological lipid distribution away from the liver and protects hepatocytes from steatosis. Our results, together with miR-29a's known antifibrotic effect, suggest miR-29a is a therapeutic target in fatty liver disease.


Subject(s)
Lipid Metabolism , Lipoprotein Lipase/biosynthesis , Liver/metabolism , MicroRNAs/metabolism , Animals , Enzyme Repression , Fatty Liver/etiology , Hepatocytes/metabolism , Male , Mice, Inbred C57BL
5.
Cell Metab ; 20(4): 626-38, 2014 Oct 07.
Article in English | MEDLINE | ID: mdl-25295787

ABSTRACT

Macroautophagy (hereafter autophagy) is the major pathway by which macromolecules and organelles are degraded. Autophagy is regulated by the mTOR signaling pathway-the focal point for integration of metabolic information, with mTORC1 playing a central role in balancing biosynthesis and catabolism. Of the various inputs to mTORC1, the amino acid sensing pathway is among the most potent. Based upon transcriptome analysis of neurons subjected to nutrient deprivation, we identified let-7 microRNA as capable of promoting neuronal autophagy. We found that let-7 activates autophagy by coordinately downregulating the amino acid sensing pathway to prevent mTORC1 activation. Let-7 induced autophagy in the brain to eliminate protein aggregates, establishing its physiological relevance for in vivo autophagy modulation. Moreover, peripheral delivery of let-7 anti-miR repressed autophagy in muscle and white fat, suggesting that let-7 autophagy regulation extends beyond CNS. Hence, let-7 plays a central role in nutrient homeostasis and proteostasis regulation in higher organisms.


Subject(s)
Amino Acids/metabolism , Autophagy , MicroRNAs/metabolism , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , Adipose Tissue, White/metabolism , Animals , Base Sequence , Brain/metabolism , Cells, Cultured , HEK293 Cells , Humans , Insulin/metabolism , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/antagonists & inhibitors , Monomeric GTP-Binding Proteins/antagonists & inhibitors , Monomeric GTP-Binding Proteins/genetics , Monomeric GTP-Binding Proteins/metabolism , Muscle, Skeletal/metabolism , Neurons/cytology , Neurons/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Sequence Alignment , Signal Transduction
6.
EMBO Mol Med ; 6(9): 1133-41, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25038053

ABSTRACT

Plasma high-density lipoprotein (HDL) levels show a strong inverse correlation with atherosclerotic vascular disease. Previous studies have demonstrated that antagonism of miR-33 in vivo increases circulating HDL and reverse cholesterol transport (RCT), thereby reducing the progression and enhancing the regression of atherosclerosis. While the efficacy of short-term anti-miR-33 treatment has been previously studied, the long-term effect of miR-33 antagonism in vivo remains to be elucidated. Here, we show that long-term therapeutic silencing of miR-33 increases circulating triglyceride (TG) levels and lipid accumulation in the liver. These adverse effects were only found when mice were fed a high-fat diet (HFD). Mechanistically, we demonstrate that chronic inhibition of miR-33 increases the expression of genes involved in fatty acid synthesis such as acetyl-CoA carboxylase (ACC) and fatty acid synthase (FAS) in the livers of mice treated with miR-33 antisense oligonucleotides. We also report that anti-miR-33 therapy enhances the expression of nuclear transcription Y subunit gamma (NFYC), a transcriptional regulator required for DNA binding and full transcriptional activation of SREBP-responsive genes, including ACC and FAS. Taken together, these results suggest that persistent inhibition of miR-33 when mice are fed a high-fat diet (HFD) might cause deleterious effects such as moderate hepatic steatosis and hypertriglyceridemia. These unexpected findings highlight the importance of assessing the effect of chronic inhibition of miR-33 in non-human primates before we can translate this therapy to humans.


Subject(s)
Gene Silencing , Lipid Metabolism/genetics , MicroRNAs/genetics , Animals , Diet, High-Fat , Fatty Liver , Liver/drug effects , Liver/metabolism , Male , Mice, Inbred C57BL , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/pharmacology , Oligonucleotides, Antisense/therapeutic use , Triglycerides/blood
7.
Cell Metab ; 18(1): 51-61, 2013 Jul 02.
Article in English | MEDLINE | ID: mdl-23823476

ABSTRACT

Sterol regulatory element-binding proteins (SREBPs) have evolved as a focal point for linking lipid synthesis with other pathways that regulate cell growth and survival. Here, we have uncovered a polycistrionic microRNA (miRNA) locus that is activated directly by SREBP-2. Two of the encoded miRNAs, miR-182 and miR-96, negatively regulate the expression of Fbxw7 and Insig-2, respectively, and both are known to negatively affect nuclear SREBP accumulation. Direct manipulation of this miRNA pathway alters nuclear SREBP levels and endogenous lipid synthesis. Thus, we have uncovered a mechanism for the regulation of intracellular lipid metabolism mediated by the concerted action of a pair of miRNAs that are expressed from the same SREBP-2-regulated miRNA locus, and each targets a different protein of the multistep pathway that regulates SREBP function. These studies reveal an miRNA "operon" analogous to the classic model for genetic control in bacterial regulatory systems.


Subject(s)
Genes, Regulator/genetics , Homeostasis/genetics , Lipid Metabolism/genetics , MicroRNAs/genetics , Operon/genetics , Sterol Regulatory Element Binding Protein 2/genetics , Animals , Cells, Cultured , F-Box Proteins/genetics , F-Box Proteins/physiology , F-Box-WD Repeat-Containing Protein 7 , Genes, Regulator/physiology , Homeostasis/physiology , Lipid Metabolism/physiology , Liver/cytology , Liver/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , MicroRNAs/physiology , Models, Animal , Operon/physiology , Sterol Regulatory Element Binding Protein 2/physiology , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/physiology
8.
Arterioscler Thromb Vasc Biol ; 33(8): 1973-7, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23702658

ABSTRACT

OBJECTIVE: To study the efficacy of anti-miRNA-33 therapy on the progression of atherosclerosis. APPROACH AND RESULTS: Ldlr(-/-) mice were injected subcutaneously with PBS, control, or anti-miR-33 oligonucleotides weekly and fed a Western diet for 12 weeks. At the end of treatment, the expression of miR-33 target genes was increased in the liver and aorta, demonstrating effective inhibition of miR-33 function. Interestingly, plasma high-density lipoprotein (HDL)-cholesterol was significantly increased in anti-miR-33-treated mice but only when they were fed a chow diet. However, HDL isolated from anti-miR-33-treated mice showed an increase cholesterol efflux capacity compared with HDL isolated from nontargeting oligonucleotide-treated mice. Analysis of atherosclerosis revealed a significant reduction of plaque size and macrophage content in mice receiving anti-miR-33. In contrast, no differences in collagen content and necrotic areas were observed among the 3 groups. CONCLUSIONS: Long-term anti-miR-33 therapy significantly reduces the progression of atherosclerosis and improves HDL functionality. The antiatherogenic effect is independent of plasma HDL-cholesterol levels.


Subject(s)
Atherosclerosis/genetics , Atherosclerosis/therapy , Genetic Therapy/methods , MicroRNAs/genetics , Receptors, LDL/genetics , Animal Feed , Animals , Atherosclerosis/pathology , Cholesterol, HDL/blood , Disease Progression , Gene Silencing , Injections, Subcutaneous , Mice , Mice, Knockout , Oligonucleotides/genetics , Oligonucleotides/pharmacology
9.
Hum Mol Genet ; 22(15): 3077-92, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23585551

ABSTRACT

Alzheimer's disease (AD) is a multifactorial and fatal neurodegenerative disorder for which the mechanisms leading to profound neuronal loss are incompletely recognized. MicroRNAs (miRNAs) are recently discovered small regulatory RNA molecules that repress gene expression and are increasingly acknowledged as prime regulators involved in human brain pathologies. Here we identified two homologous miRNAs, miR-132 and miR-212, downregulated in temporal cortical areas and CA1 hippocampal neurons of human AD brains. Sequence-specific inhibition of miR-132 and miR-212 induces apoptosis in cultured primary neurons, whereas their overexpression is neuroprotective against oxidative stress. Using primary neurons and PC12 cells, we demonstrate that miR-132/212 controls cell survival by direct regulation of PTEN, FOXO3a and P300, which are all key elements of AKT signaling pathway. Silencing of these three target genes by RNAi abrogates apoptosis caused by the miR-132/212 inhibition. We further demonstrate that mRNA and protein levels of PTEN, FOXO3a, P300 and most of the direct pro-apoptotic transcriptional targets of FOXO3a are significantly elevated in human AD brains. These results indicate that the miR-132/miR-212/PTEN/FOXO3a signaling pathway contributes to AD neurodegeneration.


Subject(s)
Alzheimer Disease/genetics , Apoptosis/genetics , Forkhead Transcription Factors/genetics , Gene Expression Regulation , MicroRNAs/genetics , Neurons/metabolism , Alzheimer Disease/metabolism , Animals , Apoptosis/drug effects , Brain/metabolism , Brain/pathology , Down-Regulation , Forkhead Box Protein O3 , Forkhead Transcription Factors/metabolism , Humans , Hydrogen Peroxide/pharmacology , Mice , MicroRNAs/metabolism , Models, Biological , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , RNA Interference , Rats , Signal Transduction , p300-CBP Transcription Factors/metabolism
10.
Circ Res ; 112(12): 1602-12, 2013 Jun 07.
Article in English | MEDLINE | ID: mdl-23519696

ABSTRACT

RATIONALE: The bile acid receptor farnesoid X receptor (FXR) regulates many aspects of lipid metabolism by variouscomplex and incompletely understood molecular mechanisms. We set out to investigate the molecular mechanisms for FXR-dependent regulation of lipid and lipoprotein metabolism. OBJECTIVE: To identify FXR-regulated microRNAs that were subsequently involved in regulating lipid metabolism. METHODS AND RESULTS: ATP binding cassette transporter A1 (ABCA1) is a major determinant of plasma high-density lipoprotein (HDL)-cholesterol levels. Here, we show that activation of the nuclear receptor FXR in vivo increases hepatic levels of miR-144, which in turn lowers hepatic ABCA1 and plasma HDL levels. We identified 2 complementary sequences to miR-144 in the 3' untranslated region of ABCA1 mRNA that are necessary for miR-144-dependent regulation. Overexpression of miR-144 in vitro decreased both cellular ABCA1 protein and cholesterol efflux to lipid-poor apolipoprotein A-I protein, whereas overexpression in vivo reduced hepatic ABCA1 protein and plasma HDL-cholesterol. Conversely, silencing miR-144 in mice increased hepatic ABCA1 protein and HDL-cholesterol. In addition, we used tissue-specific FXR-deficient mice to show that induction of miR-144 and FXR-dependent hypolipidemia requires hepatic, but not intestinal, FXR. Finally, we identified functional FXR response elements upstream of the miR-144 locus, consistent with direct FXR regulation. CONCLUSIONS: We have identified a novel pathway involving FXR, miR-144, and ABCA1 that together regulate plasma HDL-cholesterol.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Cholesterol, HDL/blood , Hepatocytes/drug effects , Isoxazoles/pharmacology , MicroRNAs/metabolism , Quinolines/pharmacology , Receptors, Cytoplasmic and Nuclear/agonists , 3' Untranslated Regions , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/genetics , Animals , Apolipoprotein A-I/metabolism , Cell Line, Tumor , Gene Expression Regulation , HEK293 Cells , Hepatocytes/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , RNA Interference , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Response Elements , Time Factors , Transfection
11.
J Biol Chem ; 288(10): 7105-16, 2013 Mar 08.
Article in English | MEDLINE | ID: mdl-23364798

ABSTRACT

Perisynaptic astrocytes express important glutamate transporters, especially excitatory amino acid transporter 2 (EAAT2, rodent analog GLT1) to regulate extracellular glutamate levels and modulate synaptic activation. In this study, we investigated an exciting new pathway, the exosome-mediated transfer of microRNA (in particular, miR-124a), in neuron-to-astrocyte signaling. Exosomes isolated from neuron-conditioned medium contain abundant microRNAs and small RNAs. These exosomes can be directly internalized into astrocytes and increase astrocyte miR-124a and GLT1 protein levels. Direct miR-124a transfection also significantly and selectively increases protein (but not mRNA) expression levels of GLT1 in cultured astrocytes. Consistent with our in vitro findings, intrastriatal injection of specific antisense against miR-124a into adult mice dramatically reduces GLT1 protein expression and glutamate uptake levels in striatum without reducing GLT1 mRNA levels. MiR-124a-mediated regulation of GLT1 expression appears to be indirect and is not mediated by its suppression of the putative GLT1 inhibitory ligand ephrinA3. Moreover, miR-124a is selectively reduced in the spinal cord tissue of end-stage SOD1 G93A mice, the mouse model of ALS. Subsequent exogenous delivery of miR-124a in vivo through stereotaxic injection significantly prevents further pathological loss of GLT1 proteins, as determined by GLT1 immunoreactivity in SOD1 G93A mice. Together, our study characterized a new neuron-to-astrocyte communication pathway and identified miRNAs that modulate GLT1 protein expression in astrocytes in vitro and in vivo.


Subject(s)
Astrocytes/metabolism , Excitatory Amino Acid Transporter 2/genetics , Exosomes/metabolism , MicroRNAs/genetics , Neurons/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Animals , Cells, Cultured , Corpus Striatum/cytology , Corpus Striatum/metabolism , Endocytosis , Excitatory Amino Acid Transporter 2/metabolism , Exosomes/ultrastructure , Gene Expression Regulation , Gene Knockdown Techniques , Glutamates/metabolism , HEK293 Cells , Humans , Immunoblotting , Mice , Mice, Knockout , Mice, Transgenic , Microscopy, Electron , Protein Biosynthesis , Signal Transduction/genetics , Spinal Cord/metabolism , Spinal Cord/pathology , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
12.
Nat Cell Biol ; 14(12): 1330-5, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23143398

ABSTRACT

Brown adipose tissue (BAT) uses the chemical energy of lipids and glucose to produce heat, a function that can be induced by cold exposure or diet. A key regulator of BAT is the gene encoding PR domain containing 16 (Prdm16), whose expression can drive differentiation of myogenic and white fat precursors to brown adipocytes. Here we show that after cold exposure, the muscle-enriched miRNA-133 is markedly downregulated in BAT and subcutaneous white adipose tissue (SAT) as a result of decreased expression of its transcriptional regulator Mef2. miR-133 directly targets and negatively regulates PRDM16, and inhibition of miR-133 or Mef2 promotes differentiation of precursors from BAT and SAT to mature brown adipocytes, thereby leading to increased mitochondrial activity. Forced expression of miR-133 in brown adipogenic conditions prevents the differentiation to brown adipocytes in both BAT and SAT precursors. Our results point to Mef2 and miR-133 as central upstream regulators of Prdm16 and hence of brown adipogenesis in response to cold exposure in BAT and SAT.


Subject(s)
Adipocytes/cytology , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , DNA-Binding Proteins/metabolism , MicroRNAs/metabolism , Transcription Factors/metabolism , Adipocytes/metabolism , Animals , Blotting, Northern , Blotting, Western , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Cells, Cultured , Computational Biology , DNA-Binding Proteins/genetics , Humans , Lipolysis/genetics , Lipolysis/physiology , Male , Mice , MicroRNAs/genetics , Real-Time Polymerase Chain Reaction , Transcription Factors/genetics
13.
Nature ; 478(7369): 404-7, 2011 Oct 19.
Article in English | MEDLINE | ID: mdl-22012398

ABSTRACT

Cardiovascular disease remains the leading cause of mortality in westernized countries, despite optimum medical therapy to reduce the levels of low-density lipoprotein (LDL)-associated cholesterol. The pursuit of novel therapies to target the residual risk has focused on raising the levels of high-density lipoprotein (HDL)-associated cholesterol in order to exploit its atheroprotective effects. MicroRNAs (miRNAs) have emerged as important post-transcriptional regulators of lipid metabolism and are thus a new class of target for therapeutic intervention. MicroRNA-33a and microRNA-33b (miR-33a/b) are intronic miRNAs whose encoding regions are embedded in the sterol-response-element-binding protein genes SREBF2 and SREBF1 (refs 3-5), respectively. These miRNAs repress expression of the cholesterol transporter ABCA1, which is a key regulator of HDL biogenesis. Recent studies in mice suggest that antagonizing miR-33a may be an effective strategy for raising plasma HDL levels and providing protection against atherosclerosis; however, extrapolating these findings to humans is complicated by the fact that mice lack miR-33b, which is present only in the SREBF1 gene of medium and large mammals. Here we show in African green monkeys that systemic delivery of an anti-miRNA oligonucleotide that targets both miR-33a and miR-33b increased hepatic expression of ABCA1 and induced a sustained increase in plasma HDL levels over 12 weeks. Notably, miR-33 antagonism in this non-human primate model also increased the expression of miR-33 target genes involved in fatty acid oxidation (CROT, CPT1A, HADHB and PRKAA1) and reduced the expression of genes involved in fatty acid synthesis (SREBF1, FASN, ACLY and ACACA), resulting in a marked suppression of the plasma levels of very-low-density lipoprotein (VLDL)-associated triglycerides, a finding that has not previously been observed in mice. These data establish, in a model that is highly relevant to humans, that pharmacological inhibition of miR-33a and miR-33b is a promising therapeutic strategy to raise plasma HDL and lower VLDL triglyceride levels for the treatment of dyslipidaemias that increase cardiovascular disease risk.


Subject(s)
Chlorocebus aethiops , Gene Expression Regulation/drug effects , Lipoproteins, HDL/blood , Lipoproteins, VLDL/blood , Liver/drug effects , MicroRNAs/antagonists & inhibitors , Oligoribonucleotides, Antisense/pharmacology , Triglycerides/blood , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/metabolism , Animals , Cells, Cultured , Chlorocebus aethiops/blood , Chlorocebus aethiops/genetics , Chlorocebus aethiops/metabolism , Cholesterol, LDL/blood , Gene Silencing , HEK293 Cells , Humans , Liver/metabolism , Male , MicroRNAs/metabolism , Time Factors
14.
J Clin Invest ; 121(7): 2921-31, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21646721

ABSTRACT

Plasma HDL levels have a protective role in atherosclerosis, yet clinical therapies to raise HDL levels have remained elusive. Recent advances in the understanding of lipid metabolism have revealed that miR-33, an intronic microRNA located within the SREBF2 gene, suppresses expression of the cholesterol transporter ABC transporter A1 (ABCA1) and lowers HDL levels. Conversely, mechanisms that inhibit miR-33 increase ABCA1 and circulating HDL levels, suggesting that antagonism of miR-33 may be atheroprotective. As the regression of atherosclerosis is clinically desirable, we assessed the impact of miR-33 inhibition in mice deficient for the LDL receptor (Ldlr-/- mice), with established atherosclerotic plaques. Mice treated with anti-miR33 for 4 weeks showed an increase in circulating HDL levels and enhanced reverse cholesterol transport to the plasma, liver, and feces. Consistent with this, anti-miR33-treated mice showed reductions in plaque size and lipid content, increased markers of plaque stability, and decreased inflammatory gene expression. Notably, in addition to raising ABCA1 levels in the liver, anti-miR33 oligonucleotides directly targeted the plaque macrophages, in which they enhanced ABCA1 expression and cholesterol removal. These studies establish that raising HDL levels by anti-miR33 oligonucleotide treatment promotes reverse cholesterol transport and atherosclerosis regression and suggest that it may be a promising strategy to treat atherosclerotic vascular disease.


Subject(s)
Atherosclerosis/metabolism , Cholesterol, HDL/metabolism , MicroRNAs/antagonists & inhibitors , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Atherosclerosis/genetics , Atherosclerosis/pathology , Gene Expression , Humans , Lipid Metabolism , Liver/cytology , Liver/physiology , Macrophages/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/metabolism , Random Allocation , Receptors, LDL/genetics , Receptors, LDL/metabolism
15.
Cancer Res ; 71(10): 3563-72, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21471404

ABSTRACT

MicroRNA (miRNA) expression profiling studies revealed a number of miRNAs dysregulated in the malignant brain tumor glioblastoma. Molecular functions of these miRNAs in gliomagenesis are mainly unknown. We show that inhibition of miR-10b, a miRNA not expressed in human brain and strongly upregulated in both low-grade and high-grade gliomas, reduces glioma cell growth by cell-cycle arrest and apoptosis. These cellular responses are mediated by augmented expression of the direct targets of miR-10b, including BCL2L11/Bim, TFAP2C/AP-2γ, CDKN1A/p21, and CDKN2A/p16, which normally protect cells from uncontrolled growth. Analysis of The Cancer Genome Atlas expression data set reveals a strong positive correlation between numerous genes sustaining cellular growth and miR-10b levels in human glioblastomas, while proapoptotic genes anticorrelate with the expression of miR-10b. Furthermore, survival of glioblastoma patients expressing high levels of miR-10 family members is significantly reduced in comparison to patients with low miR-10 levels, indicating that miR-10 may contribute to glioma growth in vivo. Finally, inhibition of miR-10b in a mouse model of human glioma results in significant reduction of tumor growth. Altogether, our experiments validate an important role of miR-10b in gliomagenesis, reveal a novel mechanism of miR-10b-mediated regulation, and suggest the possibility of its future use as a therapeutic target in gliomas.


Subject(s)
Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Glioma/metabolism , MicroRNAs/biosynthesis , Animals , Brain Neoplasms/metabolism , Cell Cycle , Cell Line, Tumor , Disease Models, Animal , Gene Expression Profiling , Gene Silencing , HeLa Cells , Humans , Mice , Mice, Nude
16.
Biochem Biophys Res Commun ; 406(4): 518-23, 2011 Mar 25.
Article in English | MEDLINE | ID: mdl-21329664

ABSTRACT

Numerous microRNAs (miRNAs) are reported as differentially expressed in cancer, however the consequence of miRNA deregulation in cancer is unknown for many miRNAs. We report that two miRNAs located on chromosome 17p13, miR-132 and miR-212, are over-expressed in pancreatic adenocarcinoma (PDAC) tissues. Both miRNAs are predicted to target the retinoblastoma tumor suppressor, Rb1. Validation of this interaction was confirmed by luciferase reporter assay and western blot in a pancreatic cancer cell line transfected with pre-miR-212 and pre-miR-132 oligos. Cell proliferation was enhanced in Panc-1 cells transfected with pre-miR-132/-212 oligos. Conversely, antisense oligos to miR-132/-212 reduced cell proliferation and caused a G(2)/M cell cycle arrest. The mRNA of a number of E2F transcriptional targets were increased in cells over expressing miR-132/-212. Exposing Panc-1 cells to the ß2 adrenergic receptor agonist, terbutaline, increased the miR-132 and miR-212 expression by 2- to 4-fold. We report that over-expression of miR-132 and miR-212 result in reduced pRb protein in pancreatic cancer cells and that the increase in cell proliferation from over-expression of these miRNAs is likely due to increased expression of several E2F target genes. The ß2 adrenergic pathway may play an important role in this novel mechanism.


Subject(s)
Adenocarcinoma/metabolism , MicroRNAs/biosynthesis , Pancreatic Neoplasms/metabolism , Retinoblastoma Protein/antagonists & inhibitors , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adrenergic beta-2 Receptor Antagonists/pharmacology , Cell Line, Tumor , Humans , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Receptors, Adrenergic, beta-2/metabolism , Retinoblastoma Protein/metabolism
17.
PLoS One ; 5(6): e11264, 2010 Jun 22.
Article in English | MEDLINE | ID: mdl-20582318

ABSTRACT

Nocturnin is a circadian clock-regulated deadenylase thought to control mRNA expression post-transcriptionally through poly(A) tail removal. The expression of Nocturnin is robustly rhythmic in liver at both the mRNA and protein levels, and mice lacking Nocturnin are resistant to diet-induced obesity and hepatic steatosis. Here we report that Nocturnin expression is regulated by microRNA-122 (miR-122), a liver specific miRNA. We found that the 3'-untranslated region (3'-UTR) of Nocturnin mRNA harbors one putative recognition site for miR-122, and this site is conserved among mammals. Using a luciferase reporter construct with wild-type or mutant Nocturnin 3'-UTR sequence, we demonstrated that overexpression of miR-122 can down-regulate luciferase activity levels and that this effect is dependent on the presence of the putative miR-122 recognition site. Additionally, the use of an antisense oligonucleotide to knock down miR-122 in vivo resulted in significant up-regulation of both Nocturnin mRNA and protein expression in mouse liver during the night, resulting in Nocturnin rhythms with increased amplitude. Together, these data demonstrate that the normal rhythmic profile of Nocturnin expression in liver is shaped in part by miR-122. Previous studies have implicated Nocturnin and miR-122 as important post-transcriptional regulators of both lipid metabolism and circadian clock controlled gene expression in the liver. Therefore, the demonstration that miR-122 plays a role in regulating Nocturnin expression suggests that this may be an important intersection between hepatic metabolic and circadian control.


Subject(s)
Circadian Rhythm , Gene Expression Profiling , Liver/enzymology , MicroRNAs/physiology , Nuclear Proteins/genetics , Transcription Factors/genetics , 3' Untranslated Regions , Animals , Base Sequence , Blotting, Northern , Cell Line , Humans , Male , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Polymerase Chain Reaction , RNA, Messenger/genetics
18.
Pancreas ; 38(7): e190-9, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19730150

ABSTRACT

OBJECTIVES: The contribution of overexpressed microRNA-21 and -221 (miR-21 and miR-221) to the malignant phenotype was determined by inhibiting these miRNAs using antisense oligonucleotides. METHODS: The effects of antisense to miR-21 and miR-221 on cell proliferation, cell cycle arrest, induction of apoptosis, combinatorial effects with gemcitabine, and effects on target protein levels were studied. RESULTS: Low nanomolar concentrations of both antisense oligonucleotides reduced proliferation of pancreatic cancer cell lines. Reduced proliferation was less pronounced in the normal ductal epithelial cell line human pancreatic Nestin-expressing cell or in pancreatic cancer cell lines exposed to an irrelevant control oligonucleotide. Inhibition of miR-21 and miR-221 increased the amount of apoptosis in HS766T cells by 3- to 6-fold compared with the control oligonucleotide. HS766T cells exposed to miR-21 antisense resulted in cell cycle arrest (G1 phase). Protein levels of tumor suppressor targets of the miRNAs were increased by antisense to miR-21 (PTEN and RECK) and miR-221 (p27). Antisense to miR-21 and miR-221 sensitized the effects of gemcitabine, and the antisense-gemcitabine combinations were synergistic at high fraction affected. CONCLUSIONS: We demonstrate that antisense to miR-21 and miR-221 results in significant cell killing under various conditions and that antisense oligonucleotides targeted to miRNA represents a potential new therapy for pancreatic cancer.


Subject(s)
Apoptosis/drug effects , Cell Cycle/drug effects , Deoxycytidine/analogs & derivatives , MicroRNAs/genetics , Oligonucleotides, Antisense/genetics , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Antimetabolites, Antineoplastic/pharmacology , Apoptosis/genetics , Blotting, Western , Cell Cycle/genetics , Cell Line , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Deoxycytidine/pharmacology , G1 Phase/drug effects , G1 Phase/genetics , Humans , PTEN Phosphohydrolase/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Gemcitabine
19.
Genes Dev ; 23(11): 1313-26, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19487572

ABSTRACT

In liver, most metabolic pathways are under circadian control, and hundreds of protein-encoding genes are thus transcribed in a cyclic fashion. Here we show that rhythmic transcription extends to the locus specifying miR-122, a highly abundant, hepatocyte-specific microRNA. Genetic loss-of-function and gain-of-function experiments have identified the orphan nuclear receptor REV-ERBalpha as the major circadian regulator of mir-122 transcription. Although due to its long half-life mature miR-122 accumulates at nearly constant rates throughout the day, this miRNA is tightly associated with control mechanisms governing circadian gene expression. Thus, the knockdown of miR-122 expression via an antisense oligonucleotide (ASO) strategy resulted in the up- and down-regulation of hundreds of mRNAs, of which a disproportionately high fraction accumulates in a circadian fashion. miR-122 has previously been linked to the regulation of cholesterol and lipid metabolism. The transcripts associated with these pathways indeed show the strongest time point-specific changes upon miR-122 depletion. The identification of Pparbeta/delta and the peroxisome proliferator-activated receptor alpha (PPARalpha) coactivator Smarcd1/Baf60a as novel miR-122 targets suggests an involvement of the circadian metabolic regulators of the PPAR family in miR-122-mediated metabolic control.


Subject(s)
Circadian Rhythm/physiology , Gene Expression Regulation , Liver/metabolism , MicroRNAs/metabolism , Animals , Circadian Rhythm/genetics , DNA-Binding Proteins/metabolism , Gene Expression Profiling , Genome/genetics , Male , Mice , Mice, Inbred C57BL , Nuclear Receptor Subfamily 1, Group D, Member 1 , Oligonucleotide Array Sequence Analysis , Peroxisome Proliferator-Activated Receptors/metabolism , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Time Factors
20.
Nucleic Acids Res ; 37(1): 70-7, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19015151

ABSTRACT

Chemically modified antisense oligonucleotides (ASOs) are widely used as a tool to functionalize microRNAs (miRNAs). Reduction of miRNA level after ASO inhibition is commonly reported to show efficacy. Whether this is the most relevant endpoint for measuring miRNA inhibition has not been adequately addressed in the field although it has important implications for evaluating miRNA targeting studies. Using a novel approach to quantitate miRNA levels in the presence of excess ASO, we have discovered that the outcome of miRNA inhibition can vary depending on the chemical modification of the ASO. Although some miRNA inhibitors cause a decrease in mature miRNA levels, we have identified a novel 2'-fluoro/2'-methoxyethyl modified ASO motif with dramatically improved in vivo potency which does not. These studies show there are multiple mechanisms of miRNA inhibition by ASOs and that evaluation of secondary endpoints is crucial for interpreting miRNA inhibition studies.


Subject(s)
MicroRNAs/antagonists & inhibitors , Oligonucleotides, Antisense/pharmacology , Animals , Gene Expression Regulation , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , MicroRNAs/analysis , MicroRNAs/metabolism , Oligonucleotides, Antisense/chemistry , Peptide Nucleic Acids/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...