Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Nat Rev Genet ; 2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38424237

ABSTRACT

RNA therapeutics (RNATx) aim to treat diseases, including cancer, by targeting or employing RNA molecules for therapeutic purposes. Amongst the most promising targets are long non-coding RNAs (lncRNAs), which regulate oncogenic molecular networks in a cell type-restricted manner. lncRNAs are distinct from protein-coding genes in important ways that increase their therapeutic potential yet also present hurdles to conventional clinical development. Advances in genome editing, oligonucleotide chemistry, multi-omics and RNA engineering are paving the way for efficient and cost-effective lncRNA-focused drug discovery pipelines. In this Review, we present the emerging field of lncRNA therapeutics for oncology, with emphasis on the unique strengths and challenges of lncRNAs within the broader RNATx framework. We outline the necessary steps for lncRNA therapeutics to deliver effective, durable, tolerable and personalized treatments for cancer.

2.
Biochim Biophys Acta ; 1863(7 Pt B): 1953-60, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26620798

ABSTRACT

Cardiac development, function and pathological remodelling in response to stress depend on the dynamic control of tissue specific gene expression by distant acting transcriptional enhancers. Recently, super-enhancers (SEs), also known as stretch or large enhancer clusters, are emerging as sentinel regulators within the gene regulatory networks that underpin cellular functions. It is becoming increasingly evident that long noncoding RNAs (lncRNAs) associated with these sequences play fundamental roles for enhancer activity and the regulation of the gene programs hardwired by them. Here, we review this emerging landscape, focusing on the roles of SEs and their derived lncRNAs in cardiovascular development and disease. We propose that exploration of this genomic landscape could provide novel therapeutic targets and approaches for the amelioration of cardiovascular disease. Ultimately we envisage a future of ncRNA therapeutics targeting the SE landscape to alleviate cardiovascular disease. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Integration of Developmental and Environmental Cues in the Heart edited by Marcus Schaub and Hughes Abriel.


Subject(s)
Cardiovascular Diseases/genetics , Cardiovascular System , Enhancer Elements, Genetic , RNA, Long Noncoding/genetics , Animals , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/physiopathology , Cardiovascular System/embryology , Cardiovascular System/metabolism , Cardiovascular System/physiopathology , Gene Expression Regulation, Developmental , Genetic Predisposition to Disease , Genomics/methods , Humans , Morphogenesis , Phenotype , RNA, Long Noncoding/metabolism , Risk Factors
3.
Am J Physiol Endocrinol Metab ; 310(5): E303-12, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26670484

ABSTRACT

The POU4F2/Brn-3b transcription factor has been identified as a potentially novel regulator of key metabolic processes. Loss of this protein in Brn-3b knockout (KO) mice causes profound hyperglycemia and insulin resistance (IR), normally associated with type 2 diabetes (T2D), whereas Brn-3b is reduced in tissues taken from obese mice fed on high-fat diets (HFD), which also develop hyperglycemia and IR. Furthermore, studies in C2C12 myocytes show that Brn-3b mRNA and proteins are induced by glucose but inhibited by insulin, suggesting that this protein is itself highly regulated in responsive cells. Analysis of differential gene expression in skeletal muscle from Brn-3b KO mice showed changes in genes that are implicated in T2D such as increased glycogen synthase kinase-3ß and reduced GLUT4 glucose transporter. The GLUT4 gene promoter contains multiple Brn-3b binding sites and is directly transactivated by this transcription factor in cotransfection assays, whereas chromatin immunoprecipitation assays confirm that Brn-3b binds to this promoter in vivo. In addition, correlation between GLUT4 and Brn-3b in KO tissues or in C2C12 cells strongly supports a close association between Brn-3b levels and GLUT4 expression. Since Brn-3b is regulated by metabolites and insulin, this may provide a mechanism for controlling key genes that are required for normal metabolic processes in insulin-responsive tissues and its loss may contribute to abnormal glucose uptake.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Homeodomain Proteins/genetics , Hyperglycemia/genetics , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , RNA, Messenger/metabolism , Transcription Factor Brn-3B/genetics , Animals , Body Weight/genetics , Chromatin Immunoprecipitation , Gene Expression Profiling , Gene Expression Regulation , Glucaric Acid/pharmacology , Glucose Intolerance/genetics , Glucose Tolerance Test , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Homeodomain Proteins/drug effects , Homeodomain Proteins/metabolism , Immunoblotting , Insulin/pharmacology , Mice , Mice, Knockout , Muscle Fibers, Skeletal/drug effects , Mutation , RNA, Messenger/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factor Brn-3B/drug effects , Transcription Factor Brn-3B/metabolism
4.
Eur Heart J ; 36(6): 353-68a, 2015 Feb 07.
Article in English | MEDLINE | ID: mdl-24786300

ABSTRACT

AIM: Heart disease is recognized as a consequence of dysregulation of cardiac gene regulatory networks. Previously, unappreciated components of such networks are the long non-coding RNAs (lncRNAs). Their roles in the heart remain to be elucidated. Thus, this study aimed to systematically characterize the cardiac long non-coding transcriptome post-myocardial infarction and to elucidate their potential roles in cardiac homoeostasis. METHODS AND RESULTS: We annotated the mouse transcriptome after myocardial infarction via RNA sequencing and ab initio transcript reconstruction, and integrated genome-wide approaches to associate specific lncRNAs with developmental processes and physiological parameters. Expression of specific lncRNAs strongly correlated with defined parameters of cardiac dimensions and function. Using chromatin maps to infer lncRNA function, we identified many with potential roles in cardiogenesis and pathological remodelling. The vast majority was associated with active cardiac-specific enhancers. Importantly, oligonucleotide-mediated knockdown implicated novel lncRNAs in controlling expression of key regulatory proteins involved in cardiogenesis. Finally, we identified hundreds of human orthologues and demonstrate that particular candidates were differentially modulated in human heart disease. CONCLUSION: These findings reveal hundreds of novel heart-specific lncRNAs with unique regulatory and functional characteristics relevant to maladaptive remodelling, cardiac function and possibly cardiac regeneration. This new class of molecules represents potential therapeutic targets for cardiac disease. Furthermore, their exquisite correlation with cardiac physiology renders them attractive candidate biomarkers to be used in the clinic.


Subject(s)
Myocardial Infarction/genetics , RNA, Long Noncoding/genetics , Transcriptome/genetics , Analysis of Variance , Animals , Biomarkers/metabolism , Cell Differentiation/physiology , Cells, Cultured , Chromatin/genetics , Embryonic Stem Cells/cytology , Gene Expression Profiling/methods , Humans , Male , Mice, Inbred C57BL , RNA, Long Noncoding/metabolism , Transfection , Vascular Remodeling/genetics
5.
J Mol Cell Cardiol ; 89(Pt A): 17-26, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26408097

ABSTRACT

Recent advances in sequencing and genomic technologies have resulted in the discovery of thousands of previously unannotated long noncoding RNAs (lncRNAs). However, their function in the cardiovascular system remains elusive. Here we review and discuss considerations for cardiovascular lncRNA discovery, annotation and functional characterization. Although we primarily focus on the heart, the proposed pipeline should foster functional and mechanistic exploration of these transcripts in various cardiovascular pathologies. Moreover, these insights could ultimately lead to novel therapeutic approaches targeting lncRNAs for the amelioration of cardiovascular diseases including heart failure.


Subject(s)
Cardiovascular System/metabolism , RNA, Long Noncoding/genetics , Animals , Humans , Molecular Sequence Annotation , RNA, Long Noncoding/metabolism
6.
J Mol Cell Cardiol ; 89(Pt A): 98-112, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26423156

ABSTRACT

Long noncoding RNAs (lncRNAs) are emerging as important regulators of developmental pathways. However, their roles in human cardiac precursor cell (CPC) remain unexplored. To characterize the long noncoding transcriptome during human CPC cardiac differentiation, we profiled the lncRNA transcriptome in CPCs isolated from the human fetal heart and identified 570 lncRNAs that were modulated during cardiac differentiation. Many of these were associated with active cardiac enhancer and super enhancers (SE) with their expression being correlated with proximal cardiac genes. One of the most upregulated lncRNAs was a SE-associated lncRNA that was named CARMEN, (CAR)diac (M)esoderm (E)nhancer-associated (N)oncoding RNA. CARMEN exhibits RNA-dependent enhancing activity and is upstream of the cardiac mesoderm-specifying gene regulatory network. Interestingly, CARMEN interacts with SUZ12 and EZH2, two components of the polycomb repressive complex 2 (PRC2). We demonstrate that CARMEN knockdown inhibits cardiac specification and differentiation in cardiac precursor cells independently of MIR-143 and -145 expression, two microRNAs located proximal to the enhancer sequences. Importantly, CARMEN expression was activated during pathological remodeling in the mouse and human hearts, and was necessary for maintaining cardiac identity in differentiated cardiomyocytes. This study demonstrates therefore that CARMEN is a crucial regulator of cardiac cell differentiation and homeostasis.


Subject(s)
Body Patterning/genetics , Cell Differentiation/genetics , Heart/embryology , Homeostasis/genetics , RNA, Long Noncoding/metabolism , Animals , Cell Lineage/genetics , Enhancer Elements, Genetic/genetics , Enhancer of Zeste Homolog 2 Protein , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Humans , Mice , Myocardium/pathology , Polycomb Repressive Complex 2/metabolism , RNA, Long Noncoding/genetics , Stem Cells/cytology , Transcriptome/genetics
7.
J Mol Cell Cardiol ; 76: 55-70, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25149110

ABSTRACT

The key information processing units within gene regulatory networks are enhancers. Enhancer activity is associated with the production of tissue-specific noncoding RNAs, yet the existence of such transcripts during cardiac development has not been established. Using an integrated genomic approach, we demonstrate that fetal cardiac enhancers generate long noncoding RNAs (lncRNAs) during cardiac differentiation and morphogenesis. Enhancer expression correlates with the emergence of active enhancer chromatin states, the initiation of RNA polymerase II at enhancer loci and expression of target genes. Orthologous human sequences are also transcribed in fetal human hearts and cardiac progenitor cells. Through a systematic bioinformatic analysis, we identified and characterized, for the first time, a catalog of lncRNAs that are expressed during embryonic stem cell differentiation into cardiomyocytes and associated with active cardiac enhancer sequences. RNA-sequencing demonstrates that many of these transcripts are polyadenylated, multi-exonic long noncoding RNAs. Moreover, knockdown of two enhancer-associated lncRNAs resulted in the specific downregulation of their predicted target genes. Interestingly, the reactivation of the fetal gene program, a hallmark of the stress response in the adult heart, is accompanied by increased expression of fetal cardiac enhancer transcripts. Altogether, these findings demonstrate that the activity of cardiac enhancers and expression of their target genes are associated with the production of enhancer-derived lncRNAs.


Subject(s)
Enhancer Elements, Genetic , Heart/embryology , RNA, Long Noncoding/physiology , Animals , Cells, Cultured , Embryonic Stem Cells/physiology , Gene Expression , Gene Expression Regulation, Developmental , Heart Diseases/genetics , Heart Diseases/metabolism , Humans , Mice , Muscle Proteins/metabolism , Primary Cell Culture
8.
Biochim Biophys Acta ; 1833(4): 923-33, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22951218

ABSTRACT

Cardiovascular diseases and in particular heart failure are major causes of morbidity and mortality in the Western world. Recently, the notion of promoting cardiac regeneration as a means to replace lost cardiomyocytes in the damaged heart has engendered considerable research interest. These studies envisage the utilization of both endogenous and exogenous cellular populations, which undergo highly specialized cell fate transitions to promote cardiomyocyte replenishment. Such transitions are under the control of regenerative gene regulatory networks, which are enacted by the integrated execution of specific transcriptional programs. In this context, it is emerging that the non-coding portion of the genome is dynamically transcribed generating thousands of regulatory small and long non-coding RNAs, which are central orchestrators of these networks. In this review, we discuss more particularly the biological roles of two classes of regulatory non-coding RNAs, i.e. microRNAs and long non-coding RNAs, with a particular emphasis on their known and putative roles in cardiac homeostasis and regeneration. Indeed, manipulating non-coding RNA-mediated regulatory networks could provide keys to unlock the dormant potential of the mammalian heart to regenerate. This should ultimately improve the effectiveness of current regenerative strategies and discover new avenues for repair. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Cardiac Pathways of Differentiation, Metabolism and Contraction.


Subject(s)
Heart Failure/genetics , MicroRNAs/genetics , Myocardium/metabolism , Myocytes, Cardiac/metabolism , RNA, Long Noncoding/genetics , Recovery of Function/genetics , Regeneration , Animals , Cell Differentiation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Expression Regulation , Gene Regulatory Networks , Heart Failure/metabolism , Heart Failure/pathology , Heart Failure/therapy , Homeostasis , Humans , MicroRNAs/metabolism , Myocardium/pathology , Myocytes, Cardiac/pathology , RNA, Long Noncoding/metabolism , Signal Transduction
9.
Heliyon ; 9(1): e13087, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36747920

ABSTRACT

Cardiovascular disorders such as heart failure are leading causes of mortality. Patient stratification via identification of novel biomarkers could improve management of cardiovascular diseases of complex etiologies. Long-noncoding RNAs (lncRNAs) are highly tissue-specific in nature and have emerged as important biomarkers in human diseases. In this study, we aimed to identify cardiac-enriched lncRNAs as potential biomarkers for cardiovascular conditions. Deep RNA sequencing and quantitative PCR identified differentially expressed lncRNAs between failing and non-failing hearts. An independent dataset was used to evaluate the enrichment of lncRNAs in normal hearts. We identified a panel of 2906 lncRNAs, named FIMICS, that were either cardiac-enriched or differentially expressed between failing and non-failing hearts. Expression of lncRNAs in blood samples differentiated patients with myocarditis and acute myocardial infarction. We hereby present the FIMICS panel, a readily available tool to provide insights into cardiovascular pathologies and which could be helpful for diagnosis, monitoring and prognosis purposes.

10.
JACC Basic Transl Sci ; 8(6): 658-674, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37426530

ABSTRACT

After myocardial infarction (MI), fibroblasts progress from proliferative to myofibroblast states, resulting in fibrosis. Platelet-derived growth factors (PDGFs) are reported to induce fibroblast proliferation, myofibroblast differentiation, and fibrosis. However, we have previously shown that PDGFs improve heart function post-MI without increasing fibrosis. We treated human cardiac fibroblasts with PDGF isoforms then performed RNA sequencing to show that PDGFs reduced cardiac fibroblasts myofibroblast differentiation and downregulated cell cycle pathways. Using mouse/pig MI models, we reveal that PDGF-AB infusion increases cell-cell interactions, reduces myofibroblast differentiation, does not affect proliferation, and accelerates scar formation. RNA sequencing of pig hearts after MI showed that PDGF-AB reduces inflammatory cytokines and alters both transcript variants and long noncoding RNA expression in cell cycle pathways. We propose that PDGF-AB could be used therapeutically to manipulate post-MI scar maturation with subsequent beneficial effects on cardiac function.

11.
Nat Commun ; 14(1): 7024, 2023 11 02.
Article in English | MEDLINE | ID: mdl-37919291

ABSTRACT

After myocardial infarction in the adult heart the remaining, non-infarcted tissue adapts to compensate the loss of functional tissue. This adaptation requires changes in gene expression networks, which are mostly controlled by transcription regulating proteins. Long non-coding transcripts (lncRNAs) are taking part in fine-tuning such gene programs. We describe and characterize the cardiomyocyte specific lncRNA Sweetheart RNA (Swhtr), an approximately 10 kb long transcript divergently expressed from the cardiac core transcription factor coding gene Nkx2-5. We show that Swhtr is dispensable for normal heart development and function but becomes essential for the tissue adaptation process after myocardial infarction in murine males. Re-expressing Swhtr from an exogenous locus rescues the Swhtr null phenotype. Genes that depend on Swhtr after cardiac stress are significantly occupied and therefore most likely regulated by NKX2-5. The Swhtr transcript interacts with NKX2-5 and disperses upon hypoxic stress in cardiomyocytes, indicating an auxiliary role of Swhtr for NKX2-5 function in tissue adaptation after myocardial injury.


Subject(s)
Heart Injuries , Myocardial Infarction , RNA, Long Noncoding , Male , Mice , Animals , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Myocytes, Cardiac/metabolism , Cardiomegaly/genetics , Cardiomegaly/metabolism , Myocardial Infarction/metabolism
12.
Breast Cancer Res ; 13(1): R5, 2011 Jan 17.
Article in English | MEDLINE | ID: mdl-21241485

ABSTRACT

INTRODUCTION: In cancer cells, elevated transcription factor-related Brn-3a regulator isolated from brain cDNA (Brn-3b) transcription factor enhances proliferation in vitro and increases tumour growth in vivo whilst conferring drug resistance and migratory potential, whereas reducing Brn-3b slows growth both in vitro and in vivo. Brn-3b regulates distinct groups of key target genes that control cell growth and behaviour. Brn-3b is elevated in >65% of breast cancer biopsies, but mechanisms controlling its expression in these cells are not known. METHODS: Bioinformatics analysis was used to identify the regulatory promoter region and map transcription start site as well as transcription factor binding sites. Polymerase chain reaction (PCR) cloning was used to generate promoter constructs for reporter assays. Chromatin immunoprecipitation and site-directed mutagenesis were used to confirm the transcription start site and autoregulation. MCF-7 and Cos-7 breast cancer cells were used. Cells grown in culture were transfected with Brn-3b promoter and treated with growth factors or estradiol to test for effects on promoter activity. Quantitative reverse transcriptase PCR assays and immunoblotting were used to confirm changes in gene and protein expression. RESULTS: We cloned the Brn-3b promoter, mapped the transcription start site and showed stimulation by estradiol and growth factors, nerve growth factor and epidermal growth factor, which are implicated in breast cancer initiation and/or progression. The effects of growth factors are mediated through the mitogen-activated protein kinase pathway, whereas hormone effects act via oestrogen receptor α (ERα). Brn-3b also autoregulates its expression and cooperates with ERα to further enhance levels. CONCLUSIONS: Key regulators of growth in cancer cells, for example, oestrogens and growth factors, can stimulate Brn-3b expression, and autoregulation also contributes to increasing Brn-3b in breast cancers. Since increasing Brn-3b profoundly enhances growth in these cells, understanding how Brn-3b is increased in breast cancers will help to identify strategies for reducing its expression and thus its effects on target genes, thereby reversing its effects in breast cancer cells.


Subject(s)
Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/metabolism , Transcription Factor Brn-3B/genetics , Binding Sites/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , Cloning, Molecular , Epidermal Growth Factor/pharmacology , Female , Gene Expression Regulation, Neoplastic , Gene Order , Homeostasis , Humans , Mutation , Nerve Growth Factor/pharmacology , Promoter Regions, Genetic/drug effects , Transcription Factor Brn-3B/metabolism , Transcription Initiation Site
13.
Article in English | MEDLINE | ID: mdl-31932317

ABSTRACT

The exquisite transcriptional control of developmental gene programs is critical for hardwiring the complex expression patterns that govern cell-fate determination and differentiation during heart development. During the past several years, studies have illuminated our understanding of a complex noncoding transcriptional landscape, primarily associated with long noncoding RNAs (lncRNAs), that is implicated in these developmental processes and has begun to reveal key functions of these transcripts. In this review, we discuss the expanding roles for lncRNAs in the earliest points of cardiac development and through differentiation and maturation of multiple cell types within the adult heart. We go on to outline the diverse mechanisms by which cardiovascular lncRNAs orchestrate these transcriptional programs, explore the challenges linked to the study of lncRNAs in developmental phenotypes, and summarize the implications for these molecules in human cardiovascular disorders.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Heart/growth & development , RNA, Long Noncoding/metabolism , Animals , Embryo, Mammalian , Humans , RNA, Long Noncoding/genetics
14.
BMC Mol Biol ; 9: 50, 2008 May 19.
Article in English | MEDLINE | ID: mdl-18489770

ABSTRACT

BACKGROUND: Myocyte stress 1 (MS1) is a striated muscle actin binding protein required for the muscle specific activity of the evolutionary ancient myocardin related transcription factor (MRTF)/serum response factor (SRF) transcriptional pathway. To date, little is known about the molecular mechanisms that govern skeletal muscle specific expression of MS1. Such mechanisms are likely to play a major role in modulating SRF activity and therefore muscle determination, differentiation and regeneration. In this study we employed a comparative in silico analysis coupled with an experimental promoter characterisation to delineate these mechanisms. RESULTS: Analysis of MS1 expression in differentiating C2C12 muscle cells demonstrated a temporal differentiation dependent up-regulation in ms1 mRNA. An in silico comparative sequence analysis identified two conserved putative myogenic regulatory domains within the proximal 1.5 kbp of 5' upstream sequence. Co-transfecting C2C12 myoblasts with ms1 promoter/luciferase reporters and myogenic regulatory factor (MRF) over-expression plasmids revealed specific sensitivity of the ms1 promoter to MyoD. Subsequent mutagenesis and EMSA analysis demonstrated specific targeting of MyoD at two distinct E-Boxes (E1 and E2) within identified evolutionary conserved regions (ECRs, alpha and beta). Chromatin immunoprecipitation (ChIP) analysis indicates that co-ordinated binding of MyoD at E-Boxes located within ECRs alpha and beta correlates with the temporal induction in ms1 mRNA. CONCLUSION: These findings suggest that the tissue specific and differentiation dependent up-regulation in ms1 mRNA is mediated by temporal binding of MyoD at distinct evolutionary conserved E-Boxes within the ms1 5' upstream sequence. We believe, through its activation of ms1, this is the first study to demonstrate a direct link between MyoD activity and SRF transcriptional signalling, with clear implications for the understanding of muscle determination, differentiation and regeneration.


Subject(s)
Computational Biology , Muscle Development , Muscle Proteins/genetics , MyoD Protein/metabolism , Promoter Regions, Genetic , Animals , Binding Sites , Gene Regulatory Networks , Humans , Mice , Mutagenesis, Site-Directed , NIH 3T3 Cells
15.
J Thorac Dis ; 9(Suppl 1): S5-S8, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28446963

ABSTRACT

Mammalian genomes are pervasively transcribed generating thousands of long noncoding RNAs (lncRNAs) with emergent regulatory roles. Many of these lncRNAs exhibit highly specialised expression patterns during development and typically flank and regulate key developmental factors. In this review, we discuss and summarise the latest advances in our understanding of the roles of lncRNAs during mesendoderm (ME) specification, a key step during gastrulation and the formation of the primitive streak (PS).

17.
Sci Transl Med ; 9(395)2017 06 21.
Article in English | MEDLINE | ID: mdl-28637928

ABSTRACT

Long noncoding RNAs (lncRNAs) are emerging as powerful regulators of cardiac development and disease. However, our understanding of the importance of these molecules in cardiac fibrosis is limited. Using an integrated genomic screen, we identified Wisper (Wisp2 super-enhancer-associated RNA) as a cardiac fibroblast-enriched lncRNA that regulates cardiac fibrosis after injury. Wisper expression was correlated with cardiac fibrosis both in a murine model of myocardial infarction (MI) and in heart tissue from human patients suffering from aortic stenosis. Loss-of-function approaches in vitro using modified antisense oligonucleotides (ASOs) demonstrated that Wisper is a specific regulator of cardiac fibroblast proliferation, migration, and survival. Accordingly, ASO-mediated silencing of Wisper in vivo attenuated MI-induced fibrosis and cardiac dysfunction. Functionally, Wisper regulates cardiac fibroblast gene expression programs critical for cell identity, extracellular matrix deposition, proliferation, and survival. In addition, its association with TIA1-related protein allows it to control the expression of a profibrotic form of lysyl hydroxylase 2, implicated in collagen cross-linking and stabilization of the matrix. Together, our findings identify Wisper as a cardiac fibroblast-enriched super-enhancer-associated lncRNA that represents an attractive therapeutic target to reduce the pathological development of cardiac fibrosis in response to MI and prevent adverse remodeling in the damaged heart.


Subject(s)
Cardiomyopathies/genetics , RNA, Long Noncoding/genetics , Cardiomyopathies/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis/genetics , Fibrosis/pathology , Humans , RNA, Long Noncoding/physiology , Ventricular Remodeling
18.
Nat Commun ; 8(1): 1806, 2017 11 27.
Article in English | MEDLINE | ID: mdl-29180618

ABSTRACT

Enhancers and long noncoding RNAs (lncRNAs) are key determinants of lineage specification during development. Here, we evaluate remodeling of the enhancer landscape and modulation of the lncRNA transcriptome during mesendoderm specification. We sort mesendodermal progenitors from differentiating embryonic stem cells (ESCs) according to Eomes expression, and find that enhancer usage is coordinated with mesendoderm-specific expression of key lineage-determining transcription factors. Many of these enhancers are associated with the expression of lncRNAs. Examination of ESC-specific enhancers interacting in three-dimensional space with mesendoderm-specifying transcription factor loci identifies MesEndoderm Transcriptional Enhancer Organizing Region (Meteor). Genetic and epigenetic manipulation of the Meteor enhancer reveal its indispensable role during mesendoderm specification and subsequent cardiogenic differentiation via transcription-independent and -dependent mechanisms. Interestingly, Meteor-deleted ESCs are epigenetically redirected towards neuroectodermal lineages. Loci, topologically associating a transcribed enhancer and its cognate protein coding gene, appear to represent therefore a class of genomic elements controlling developmental competence in pluripotency.


Subject(s)
Ectoderm/physiology , Embryonic Stem Cells/physiology , Enhancer Elements, Genetic/physiology , Mesoderm/physiology , RNA, Long Noncoding/physiology , Animals , Cell Differentiation/genetics , Cell Line , Cell Lineage/genetics , Ectoderm/cytology , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental/physiology , Humans , Induced Pluripotent Stem Cells , Mesoderm/cytology , Mice , Neural Plate/cytology , Neural Plate/physiology
19.
Cell Stem Cell ; 18(5): 561-2, 2016 05 05.
Article in English | MEDLINE | ID: mdl-27152437

ABSTRACT

Long noncoding RNAs (lncRNAs) comprise a class of regulatory molecules that may control diverse stem cell properties. Now in Cell Stem Cell, Luo et al. (2016) show that a specific group of lncRNAs, those transcribed divergently from protein coding genes, activate key developmental genes to control embryonic stem cell fate.


Subject(s)
Cell Differentiation , RNA, Long Noncoding/genetics , Embryonic Stem Cells
20.
Cardiovasc Res ; 110(1): 73-84, 2016 May 01.
Article in English | MEDLINE | ID: mdl-26857418

ABSTRACT

AIMS: The adult mammalian heart has poor regenerative capacity. In contrast, the zebrafish heart retains a robust capacity for regeneration into adulthood. These distinct responses are consequences of a differential utilization of evolutionary-conserved gene regulatory networks in the damaged heart. To systematically identify miRNA-dependent networks controlling cardiac repair following injury, we performed comparative gene and miRNA profiling of the cardiac transcriptome in adult mice and zebrafish. METHODS AND RESULTS: Using an integrated approach, we show that 45 miRNA-dependent networks, involved in critical biological pathways, are differentially modulated in the injured zebrafish vs. mouse hearts. We study, more particularly, the miR-26a-dependent response. Therefore, miR-26a is down-regulated in the fish heart after injury, whereas its expression remains constant in the mouse heart. Targets of miR-26a involve activators of the cell cycle and Ezh2, a component of the polycomb repressive complex 2 (PRC2). Importantly, PRC2 exerts repressive functions on negative regulators of the cell cycle. In cultured neonatal cardiomyocytes, inhibition of miR-26a stimulates, therefore, cardiomyocyte proliferation. Accordingly, miR-26a knockdown prolongs the proliferative window of cardiomyocytes in the post-natal mouse heart. CONCLUSIONS: This novel strategy identifies a series of miRNAs and associated pathways, in particular miR-26a, which represent attractive therapeutic targets for inducing repair in the injured heart.


Subject(s)
Cell Proliferation/genetics , Gene Regulatory Networks/genetics , MicroRNAs/metabolism , Wound Healing/genetics , Animals , Cell Cycle , Gene Expression Profiling/methods , Mice, Inbred C57BL , MicroRNAs/genetics , Myocytes, Cardiac/physiology , Regeneration , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL