MicroRNA control of muscle development and disease

https://doi.org/10.1016/j.ceb.2009.01.029Get rights and content

Cardiac and skeletal muscle development are controlled by evolutionarily conserved networks of transcription factors that coordinate the expression of genes involved in muscle growth, morphogenesis, differentiation, and contractility. In addition to regulating the expression of protein-coding genes, recent studies have revealed that myogenic transcription factors control the expression of a collection of microRNAs, which act through multiple mechanisms to modulate muscle development and function. In some cases, microRNAs fine-tune the expression of target mRNAs, whereas in other cases they function as ‘on–off’ switches. MicroRNA control of gene expression appears to be especially important during cardiovascular and skeletal muscle diseases, in which microRNAs participate in stress-dependent remodeling of striated muscle tissues. We review findings that point to the importance of microRNA-mediated control of gene expression during muscle development and disease, and consider the potential of microRNAs as therapeutic targets.

Introduction

The development of cardiac and skeletal muscle is orchestrated by evolutionarily conserved networks of transcription factors that regulate the expression of genes involved in muscle growth, differentiation, and contractility. The most ancient of these myogenic transcription factors is the MADS (MCM1, Agamous, Deficiens, Serum response factor) box transcription factor myocyte enhancer factor-2 (MEF2), which directly activates the majority of muscle genes through combinatorial interactions with other transcription factors [1]. In cardiac muscle, MEF2 and another MADS-box transcription factor, serum response factor (SRF), cooperatively activate cardiac gene expression by association with GATA, T-box, and Nkx2.5 transcription factors, as well as the myocardin family of transcriptional co-activators, whereas in skeletal muscle MEF2 activates the myogenic differentiation program in conjunction with the basic-helix-loop-helix (bHLH) transcription factors, MyoD and myogenin [2, 3]. Recent studies have revealed that, in addition to activating genes involved in muscle differentiation and muscle contraction, these myogenic transcription factors activate the expression of a set of conserved microRNAs (miRNAs) that function to ‘fine-tune’ the output of these transcriptional networks, resulting in precise cellular responses to developmental, physiologic, and pathologic signals. The integration of miRNAs into the core muscle transcriptional program expands the precision and complexity of gene regulation in muscle cells because individual miRNAs are capable of regulating hundreds of mRNAs, and individual mRNAs can be targeted by many miRNAs. In this review, we discuss recent publications that have illuminated the roles of miRNAs in muscle development and disease.

Section snippets

miRNA biogenesis and function

miRNAs are small, evolutionarily conserved noncoding RNAs that are transcribed by RNA polymerase II as long pri-miRNAs encoding one or more miRNAs. Most miRNAs are transcribed as independent transcripts, but approximately a third are embedded within introns of protein-coding genes and processed following splicing of pre-messenger RNAs [4]. Pri-miRNAs are processed in the nucleus by the proteins Drosha and DGCR8, which produce an ∼70 nucleotide hairpin RNA, termed the pre-miRNA, which is

Muscles without miRNAs

An essential role for miRNAs in mouse development was shown by a loss-of-function mutation in the miRNA-generating enzyme, Dicer, which results in embryonic lethality by day 7.5 [11]. In order to circumvent the lethality associated with the deletion of Dicer and to study the roles of Dicer in specific tissues, several groups have generated conditional null alleles of Dicer. Deletion of Dicer in cardiomyocytes using a Cre recombinase ‘knocked-in’ to the endogenous Nkx2.5 locus, which directs

Muscle-specific miRNAs

Several individual miRNAs are specifically expressed in cardiac and skeletal muscle (Table 1) [16]. Of these, the most widely studied are members of miR-1/206 and miR-133a/133b families, which originate from bicistronic transcripts on three separate chromosomes (Figure 1) [17]. miR-1-1 and miR-1-2 are identical and differ from miR-206 by four nucleotides, and miR-133a-1 and miR-133a-2 are identical and differ from miR-133b by two nucleotides. Cardiac and skeletal muscle specific transcription

MyomiRs

Proper function of the heart depends on the expression of myosin heavy chain (MHC) proteins MYH6 (α-MHC) and MYH7 (β-MHC). Cardiac injury results in a decrease in the expression of the adult myosin gene, Myh6, and an increase in the expression of the embryonic myosin gene, Myh7. Recently, it was discovered that these myosin genes also encode a family of miRNAs, called MyomiRs, which have important functions in regulating myosin content and stress-dependent cardiac remodeling [38].

α-MHC, the

miRNAs with functions in muscle

In addition to muscle-specific miRNAs, there are also examples of ubiquitously expressed miRNAs that have been shown to have functions in muscle. In zebrafish, miR-214 was shown to act as a positive regulator of the slow muscle phenotype, by targeting suppressor of fused (Su(fu)), a negative regulator of hedgehog signaling [40]. miR-138 was reported to contribute to chamber-specific gene expression patterns in the zebrafish heart by targeting aldehyde dehydrogenase-1a2 (RALDH2), a regulator of

miRNAs in muscle disease

Several important studies have indicated that miRNA expression is dysregulated in cardiac and skeletal muscle disease and in some cases individual miRNAs have been shown to cause or alleviate disease. The first series of such studies focused on the profiling of miRNAs in hypertrophic murine and human hearts and revealed a common set of miRNAs that are elevated in hypertrophic hearts [43, 44, 45, 46]. One of these miRNAs, miR-195, was shown to be sufficient to induce heart failure and death due

Therapeutic manipulation of miRNAs with antagomiRs and miR mimics

Therapeutic manipulation of miRNAs represents a potentially powerful approach to treat cardiovascular and skeletal muscle diseases. In this regard, cholesterol-modified antisense oligonucleotides, referred to as antagomiRs, show remarkable efficacy in the inhibition of miRNAs following intravenous delivery in vivo [53]. Engelhardt and co-workers showed that delivery of an antagomiR to miR-21, a miRNA upregulated in failing hearts, preserved cardiac function and reduced cardiac fibrosis

Conclusions

Although many important findings have been made within the past few years, our current knowledge about the function of miRNAs in muscle development and disease is still quite limited given the multitude of possible interactions between transcription factors, miRNAs, and their target mRNAs. Future studies will need to focus on characterizing the in vivo functions of individual vertebrate miRNAs followed by the identification of their downstream target mRNAs. Identification of the miRNA

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted as:

  • • of special interest

  • •• of outstanding interest

Acknowledgements

We apologize to the many researchers whose work was not cited in this review owing to space limitations. We thank Jose Cabrera for graphics and Jennifer Brown for editorial assistance. Work in the laboratory of ENO is supported by grants from the NIH, the Donald W Reynolds Center for Clinical Cardiovascular Research, the Leducq Foundation, the Sandler Foundation for Asthma Research, and the Robert A Welch Foundation. AHW is supported by a training grant from the National Heart, Lung, and Blood

References (59)

  • H. Wang et al.

    NF-kappaB-YY1-miR-29 regulatory circuitry in skeletal myogenesis and rhabdomyosarcoma

    Cancer Cell

    (2008)
  • T. Thum et al.

    MicroRNA-21 contributes to myocardial disease by stimulating MAP kinase signalling in fibroblasts

    Nature

    (2008)
  • E.N. Olson

    Gene regulatory networks in the evolution and development of the heart

    Science

    (2006)
  • G.C. Pipes et al.

    The myocardin family of transcriptional coactivators: versatile regulators of cell growth, migration, and myogenesis

    Genes Dev

    (2006)
  • M.J. Potthoff et al.

    MEF2: a central regulator of diverse developmental programs

    Development

    (2007)
  • E. van Rooij et al.

    MicroRNAs: powerful new regulators of heart disease and provocative therapeutic targets

    J Clin Invest

    (2007)
  • D. Baek et al.

    The impact of microRNAs on protein output

    Nature

    (2008)
  • M. Selbach et al.

    Widespread changes in protein synthesis induced by microRNAs

    Nature

    (2008)
  • S. Vasudevan et al.

    Switching from repression to activation: microRNAs can up-regulate translation

    Science

    (2007)
  • J.C. Schwartz et al.

    Antisense transcripts are targets for activating small RNAs

    Nat Struct Mol Biol

    (2008)
  • E. Bernstein et al.

    Dicer is essential for mouse development

    Nat Genet

    (2003)
  • Y. Zhao et al.

    Dysregulation of cardiogenesis, cardiac conduction, and cell cycle in mice lacking miRNA-1-2

    Cell

    (2007)
  • J.F. Chen et al.

    Targeted deletion of Dicer in the heart leads to dilated cardiomyopathy and heart failure

    Proc Natl Acad Sci U S A

    (2008)
  • J.R. O’Rourke et al.

    Essential role for Dicer during skeletal muscle development

    Dev Biol

    (2007)
  • J.J. McCarthy

    MicroRNA-206: the skeletal muscle-specific myomiR

    Biochim Biophys Acta

    (2008)
  • J.F. Chen et al.

    The role of microRNA-1 and microRNA-133 in skeletal muscle proliferation and differentiation

    Nat Genet

    (2006)
  • Y. Zhao et al.

    Serum response factor regulates a muscle-specific microRNA that targets Hand2 during cardiogenesis

    Nature

    (2005)
  • Z. Niu et al.

    Serum response factor orchestrates nascent sarcomerogenesis and silences the biomineralization gene program in the heart

    Proc Natl Acad Sci U S A

    (2008)
  • M.I. Rosenberg et al.

    MyoD inhibits Fstl1 and Utrn expression by inducing transcription of miR-206

    J Cell Biol

    (2006)
  • Cited by (301)

    • Assessment of diagnostic potential of some circulating microRNAs in Amyotrophic Lateral Sclerosis Patients, an Egyptian study

      2021, Clinical Neurology and Neurosurgery
      Citation Excerpt :

      MiR-206 is one of the highly expressed miRNAs in the skeletal muscle and is involved in the regulation of various processes during the development of skeletal muscle. Previous studies showed that miR-206 plays roles in repair of muscle injury, muscle regeneration, neuromuscular connectivity, reinnervation and neuromuscular gene expression [23–27]. In addition, a previous study revealed that there is a comparable increase of miR-206 levels in muscle of ALS patients as that observed in blood [27].

    • Calcium | Calcium signaling: Calmodulin-dependent phosphatase

      2021, Encyclopedia of Biological Chemistry: Third Edition
    View all citing articles on Scopus
    View full text