Skip to main content

Advertisement

Log in

Dgcr8 knockout approaches to understand microRNA functions in vitro and in vivo

  • Review
  • Published:
Cellular and Molecular Life Sciences Aims and scope Submit manuscript

A Correction to this article was published on 29 April 2019

This article has been updated

Abstract

Biologic function of the majority of microRNAs (miRNAs) is still unknown. Uncovering the function of miRNAs is hurdled by redundancy among different miRNAs. The deletion of Dgcr8 leads to the deficiency in producing all canonical miRNAs, therefore, overcoming the redundancy issue. Dgcr8 knockout strategy has been instrumental in understanding the function of miRNAs in a variety of cells in vitro and in vivo. In this review, we will first give a brief introduction about miRNAs, miRNA biogenesis pathway and the role of Dgcr8 in miRNA biogenesis. We will then summarize studies performed with Dgcr8 knockout cell models with a focus on embryonic stem cells. After that, we will summarize results from various in vivo Dgcr8 knockout models. Given significant phenotypic differences in various tissues between Dgcr8 and Dicer knockout, we will also briefly review current progresses on understanding miRNA-independent functions of miRNA biogenesis factors. Finally, we will discuss the potential use of a new strategy to stably express miRNAs in Dgcr8 knockout cells. In future, Dgcr8 knockout approaches coupled with innovations in miRNA rescue strategy may provide further insights into miRNA functions in vitro and in vivo.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2

Similar content being viewed by others

Change history

  • 29 April 2019

    The section: “miRNA‑independent functions of DICER” was missed between the section “miRNA‑independent functions of DROSHA and DGCR8” and the section “The Dgcr8 knockout strategy to study miRNA functions” in the original publications.

Abbreviations

APA:

Alternative polyadenylation

AGO:

Argonaute

cKO:

Conditional knockout

cNCCs:

Cardiac neural crest cells

COX-2:

Cyclooxygenase 2

cTECs:

Cortical thymic epithelial cells

DGCR:

DiGeorge syndrome chromosomal (or critical) region

DISME:

DGCR8-independent stable miRNA expression strategy

EMT:

Epithelial–mesenchymal transition

EpiLC:

Epiblast-like cells

EpiSCs:

Epiblast stem cells

ESC:

Embryonic stem cell

ESCC:

ESC-specific cell cycle regulating

GPCs:

Glia progenitor cells

iKO:

Inducible knockout

iPSCs:

Induced pluripotent stem cells

LINE-1:

Long interspersed element 1

lncRNA:

Long noncoding RNA

MC:

Microprocessor complex

miRNA:

microRNA

mTECs:

Medullary thymic epithelial cells

NK:

Natural killer

NSCs:

Neural stem cells

PACT:

Protein activator of PKR

PTA:

Persistent truncus arteriosus

RISC:

RNA-induced silencing complex

RPE:

Retinal pigmented epithelium

rRNA:

Ribosomal RNA

SCs:

Schwann cells

Shh:

Sonic hedgehog

shRNA:

Short hairpin RNA

snoRNA:

Small nucleolar RNA

TDP43:

TAR DNA-binding protein 43

TECs:

Thymic epithelial cells

TRBP:

HIV trans-activating response RNA-binding protein

Th cells:

Helper T cells

tRNA:

Transfer RNA

VSD:

Ventricular septal defect

References

  1. Lee RC, Feinbaum RL, Ambros V (1993) The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 75(5):843–854

    Article  CAS  PubMed  Google Scholar 

  2. Chakraborty C et al (2017) Therapeutic miRNA and siRNA: moving from bench to clinic as next generation medicine. Mol Ther Nucleic Acids 8:132–143

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Christopher AF et al (2016) MicroRNA therapeutics: discovering novel targets and developing specific therapy. Perspect Clin Res 7(2):68–74

    Article  PubMed  PubMed Central  Google Scholar 

  4. Ha M, Kim VN (2014) Regulation of microRNA biogenesis. Nat Rev Mol Cell Biol 15(8):509–524

    Article  CAS  PubMed  Google Scholar 

  5. Lin S, Gregory RI (2015) MicroRNA biogenesis pathways in cancer. Nat Rev Cancer 15(6):321–333

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Lee Y et al (2003) The nuclear RNase III Drosha initiates microRNA processing. Nature 425(6956):415–419

    Article  CAS  PubMed  Google Scholar 

  7. Denli AM et al (2004) Processing of primary microRNAs by the Microprocessor complex. Nature 432(7014):231–235

    Article  CAS  PubMed  Google Scholar 

  8. Gregory RI et al (2004) The Microprocessor complex mediates the genesis of microRNAs. Nature 432(7014):235–240

    Article  CAS  PubMed  Google Scholar 

  9. Han J et al (2004) The Drosha-DGCR8 complex in primary microRNA processing. Genes Dev 18(24):3016–3027

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Landthaler M, Yalcin A, Tuschl T (2004) The human DiGeorge syndrome critical region gene 8 and its D. melanogaster homolog are required for miRNA biogenesis. Curr Biol 14(23):2162–2167

    Article  CAS  PubMed  Google Scholar 

  11. Bohnsack MT, Czaplinski K, Gorlich D (2004) Exportin 5 is a RanGTP-dependent dsRNA-binding protein that mediates nuclear export of pre-miRNAs. RNA 10(2):185–191

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Lund E et al (2004) Nuclear export of microRNA precursors. Science 303(5654):95–98

    Article  CAS  PubMed  Google Scholar 

  13. Yi R et al (2003) Exportin-5 mediates the nuclear export of pre-microRNAs and short hairpin RNAs. Genes Dev 17(24):3011–3016

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Zhang H et al (2004) Single processing center models for human Dicer and bacterial RNase III. Cell 118(1):57–68

    Article  CAS  PubMed  Google Scholar 

  15. Park JE et al (2011) Dicer recognizes the 5′ end of RNA for efficient and accurate processing. Nature 475(7355):201–205

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Fukunaga R et al (2012) Dicer partner proteins tune the length of mature miRNAs in flies and mammals. Cell 151(3):533–546

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Mourelatos Z et al (2002) miRNPs: a novel class of ribonucleoproteins containing numerous microRNAs. Genes Dev 16(6):720–728

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Kawamata T, Tomari Y (2010) Making RISC. Trends Biochem Sci 35(7):368–376

    Article  CAS  PubMed  Google Scholar 

  19. Bartel DP (2018) Metazoan MicroRNAs. Cell 173(1):20–51

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Huntzinger E, Izaurralde E (2011) Gene silencing by microRNAs: contributions of translational repression and mRNA decay. Nat Rev Genet 12(2):99–110

    Article  CAS  PubMed  Google Scholar 

  21. Yekta S, Shih IH, Bartel DP (2004) MicroRNA-directed cleavage of HOXB8 mRNA. Science 304(5670):594–596

    Article  CAS  PubMed  Google Scholar 

  22. Shin C et al (2010) Expanding the microRNA targeting code: functional sites with centered pairing. Mol Cell 38(6):789–802

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Karginov FV et al (2010) Diverse endonucleolytic cleavage sites in the mammalian transcriptome depend upon microRNAs, Drosha, and additional nucleases. Mol Cell 38(6):781–788

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Chiang HR et al (2010) Mammalian microRNAs: experimental evaluation of novel and previously annotated genes. Genes Dev 24(10):992–1009

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Kozomara A, Griffiths-Jones S (2014) miRBase: annotating high confidence microRNAs using deep sequencing data. Nucleic Acids Res 42(Database issue):D68–D73

    Article  CAS  PubMed  Google Scholar 

  26. Lewis BP, Burge CB, Bartel DP (2005) Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell 120(1):15–20

    Article  CAS  PubMed  Google Scholar 

  27. Friedman RC et al (2009) Most mammalian mRNAs are conserved targets of microRNAs. Genome Res 19(1):92–105

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Roush S, Slack FJ (2008) The let-7 family of microRNAs. Trends Cell Biol 18(10):505–516

    Article  CAS  PubMed  Google Scholar 

  29. Fischer S et al (2015) Unveiling the principle of microRNA-mediated redundancy in cellular pathway regulation. RNA Biol 12(3):238–247

    Article  PubMed  PubMed Central  Google Scholar 

  30. Olive V, Minella AC, He L (2015) Outside the coding genome, mammalian microRNAs confer structural and functional complexity. Sci Signal 8(368):re2

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Herbert KM et al (2016) A heterotrimer model of the complete Microprocessor complex revealed by single-molecule subunit counting. RNA 22(2):175–183

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Nguyen TA et al (2015) Functional anatomy of the human microprocessor. Cell 161(6):1374–1387

    Article  CAS  PubMed  Google Scholar 

  33. Nguyen TA et al (2018) Microprocessor depends on hemin to recognize the apical loop of primary microRNA. Nucleic Acids Res 46(11):5726–5736

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Partin AC et al (2017) Heme enables proper positioning of Drosha and DGCR8 on primary microRNAs. Nat Commun 8(1):1737

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Faller M et al (2007) Heme is involved in microRNA processing. Nat Struct Mol Biol 14(1):23–29

    Article  CAS  PubMed  Google Scholar 

  36. Di Carlo V et al (2013) TDP-43 regulates the microprocessor complex activity during in vitro neuronal differentiation. Mol Neurobiol 48(3):952–963

    Article  CAS  PubMed  Google Scholar 

  37. Kawahara Y, Mieda-Sato A (2012) TDP-43 promotes microRNA biogenesis as a component of the Drosha and Dicer complexes. Proc Natl Acad Sci USA 109(9):3347–3352

    Article  PubMed  PubMed Central  Google Scholar 

  38. Mori M et al (2014) Hippo signaling regulates microprocessor and links cell-density-dependent miRNA biogenesis to cancer. Cell 156(5):893–906

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Hong S et al (2013) Signaling by p38 MAPK stimulates nuclear localization of the microprocessor component p68 for processing of selected primary microRNAs. Sci Signal 6(266):ra16

    Article  CAS  PubMed  Google Scholar 

  40. Motino O et al (2015) Regulation of MicroRNA 183 by cyclooxygenase 2 in liver is DEAD-box helicase p68 (DDX5) dependent: role in insulin signaling. Mol Cell Biol 35(14):2554–2567

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Murai K et al (2016) The TLX-miR-219 cascade regulates neural stem cell proliferation in neurodevelopment and schizophrenia iPSC model. Nat Commun 7:10965

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Stark KL et al (2008) Altered brain microRNA biogenesis contributes to phenotypic deficits in a 22q11-deletion mouse model. Nat Genet 40(6):751–760

    Article  CAS  PubMed  Google Scholar 

  43. Hornstein E, Shomron N (2006) Canalization of development by microRNAs. Nat Genet 38(Suppl):S20–S24

    Article  CAS  PubMed  Google Scholar 

  44. Fenelon K et al (2011) Deficiency of Dgcr8, a gene disrupted by the 22q11.2 microdeletion, results in altered short-term plasticity in the prefrontal cortex. Proc Natl Acad Sci USA 108(11):4447–4452

    Article  PubMed  PubMed Central  Google Scholar 

  45. Merico D et al (2014) MicroRNA dysregulation, gene networks, and risk for schizophrenia in 22q11.2 deletion syndrome. Front Neurol 5:238

    Article  PubMed  PubMed Central  Google Scholar 

  46. Amin H et al (2017) Developmental excitatory-to-inhibitory GABA-polarity switch is disrupted in 22q11.2 deletion syndrome: a potential target for clinical therapeutics. Sci Rep 7(1):15752

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Han J et al (2009) Posttranscriptional crossregulation between Drosha and DGCR8. Cell 136(1):75–84

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Triboulet R et al (2009) Post-transcriptional control of DGCR8 expression by the microprocessor. RNA 15(6):1005–1011

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Wang Y et al (2007) DGCR8 is essential for microRNA biogenesis and silencing of embryonic stem cell self-renewal. Nat Genet 39(3):380–385

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Shiohama A et al (2003) Molecular cloning and expression analysis of a novel gene DGCR8 located in the DiGeorge syndrome chromosomal region. Biochem Biophys Res Commun 304(1):184–190

    Article  CAS  PubMed  Google Scholar 

  51. Babiarz JE et al (2008) Mouse ES cells express endogenous shRNAs, siRNAs, and other microprocessor-independent, dicer-dependent small RNAs. Genes Dev 22(20):2773–2785

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Aguado LC et al (2017) RNase III nucleases from diverse kingdoms serve as antiviral effectors. Nature 547(7661):114–117

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Shapiro JS et al (2014) Drosha as an interferon-independent antiviral factor. Proc Natl Acad Sci USA 111(19):7108–7113

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Maillard PV et al (2013) Antiviral RNA interference in mammalian cells. Science 342(6155):235–238

    Article  CAS  PubMed  Google Scholar 

  55. Li Y et al (2013) RNA interference functions as an antiviral immunity mechanism in mammals. Science 342(6155):231–234

    Article  CAS  PubMed  Google Scholar 

  56. Li Y et al (2016) Induction and suppression of antiviral RNA interference by influenza A virus in mammalian cells. Nat Microbiol 2:16250

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Qiu Y et al (2017) Human virus-derived small RNAs can confer antiviral immunity in mammals. Immunity 46(6):992–1004

    Article  CAS  PubMed  Google Scholar 

  58. Calses PC et al (2017) DGCR8 mediates repair of UV-induced DNA damage independently of RNA processing. Cell Rep 19(1):162–174

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Tang KF et al (2008) Decreased Dicer expression elicits DNA damage and up-regulation of MICA and MICB. J Cell Biol 182(2):233–239

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Wei W et al (2012) A role for small RNAs in DNA double-strand break repair. Cell 149(1):101–112

    Article  CAS  PubMed  Google Scholar 

  61. Francia S et al (2012) Site-specific DICER and DROSHA RNA products control the DNA-damage response. Nature 488(7410):231–235

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Wang Q, Goldstein M (2016) Small RNAs recruit chromatin-modifying enzymes MMSET and Tip60 to reconfigure damaged DNA upon double-strand break and facilitate repair. Cancer Res 76(7):1904–1915

    Article  CAS  PubMed  Google Scholar 

  63. Chen X et al (2017) Dicer regulates non-homologous end joining and is associated with chemosensitivity in colon cancer patients. Carcinogenesis 38(9):873–882

    Article  CAS  PubMed  Google Scholar 

  64. Pong SK, Gullerova M (2018) Noncanonical functions of microRNA pathway enzymes—drosha, DGCR8, dicer and ago proteins. FEBS Lett 592(17):2973–2986

    Article  CAS  PubMed  Google Scholar 

  65. Yang JS, Lai EC (2011) Alternative miRNA biogenesis pathways and the interpretation of core miRNA pathway mutants. Mol Cell 43(6):892–903

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Burger K, Gullerova M (2015) Swiss army knives: non-canonical functions of nuclear Drosha and Dicer. Nat Rev Mol Cell Biol 16(7):417–430

    Article  CAS  PubMed  Google Scholar 

  67. Macias S et al (2012) DGCR8 HITS-CLIP reveals novel functions for the microprocessor. Nat Struct Mol Biol 19(8):760–766

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Heras SR et al (2013) The Microprocessor controls the activity of mammalian retrotransposons. Nat Struct Mol Biol 20(10):1173–1181

    Article  CAS  PubMed  Google Scholar 

  69. Kim B, Jeong K, Kim VN (2017) Genome-wide mapping of DROSHA cleavage sites on primary MicroRNAs and noncanonical substrates. Mol Cell 66(2):258–269

    Article  CAS  PubMed  Google Scholar 

  70. Knuckles P et al (2012) Drosha regulates neurogenesis by controlling neurogenin 2 expression independent of microRNAs. Nat Neurosci 15(7):962–969

    Article  CAS  PubMed  Google Scholar 

  71. Hoffmann N et al (2018) DGCR8 promotes neural progenitor expansion and represses neurogenesis in the mouse embryonic neocortex. Front Neurosci 12:281

    Article  PubMed  PubMed Central  Google Scholar 

  72. Rolando C et al (2016) Multipotency of adult hippocampal NSCs in vivo is restricted by drosha/NFIB. Cell Stem Cell 19(5):653–662

    Article  CAS  PubMed  Google Scholar 

  73. Marinaro F et al (2017) MicroRNA-independent functions of DGCR8 are essential for neocortical development and TBR1 expression. EMBO Rep 18(4):603–618

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Johanson TM et al (2015) Drosha controls dendritic cell development by cleaving messenger RNAs encoding inhibitors of myelopoiesis. Nat Immunol 16(11):1134–1141

    Article  CAS  PubMed  Google Scholar 

  75. Wagschal A et al (2012) Microprocessor, Setx, Xrn2, and Rrp6 co-operate to induce premature termination of transcription by RNAPII. Cell 150(6):1147–1157

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Gromak N et al (2013) Drosha regulates gene expression independently of RNA cleavage function. Cell Rep 5(6):1499–1510

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Dhir A et al (2015) Microprocessor mediates transcriptional termination of long noncoding RNA transcripts hosting microRNAs. Nat Struct Mol Biol 22(4):319–327

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Lee D, Nam JW, Shin C (2017) DROSHA targets its own transcript to modulate alternative splicing. RNA 23(7):1035–1047

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Havens MA, Reich AA, Hastings ML (2014) Drosha promotes splicing of a pre-microRNA-like alternative exon. PLoS Genet 10(5):e1004312

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Wu H et al (2000) Human RNase III is a 160-kDa protein involved in preribosomal RNA processing. J Biol Chem 275(47):36957–36965

    Article  CAS  PubMed  Google Scholar 

  81. Macias S et al (2015) DGCR8 acts as an adaptor for the exosome complex to degrade double-stranded structured RNAs. Mol Cell 60(6):873–885

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Bernstein E et al (2001) Role for a bidentate ribonuclease in the initiation step of RNA interference. Nature 409(6818):363–366

    Article  CAS  PubMed  Google Scholar 

  83. Doi N et al (2003) Short-interfering-RNA-mediated gene silencing in mammalian cells requires Dicer and eIF2C translation initiation factors. Curr Biol 13(1):41–46

    Article  CAS  PubMed  Google Scholar 

  84. Okamura K, Lai EC (2008) Endogenous small interfering RNAs in animals. Nat Rev Mol Cell Biol 9(9):673–678

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Yang N, Kazazian HH Jr (2006) L1 retrotransposition is suppressed by endogenously encoded small interfering RNAs in human cultured cells. Nat Struct Mol Biol 13(9):763–771

    Article  CAS  PubMed  Google Scholar 

  86. Kaneko H et al (2011) DICER1 deficit induces Alu RNA toxicity in age-related macular degeneration. Nature 471(7338):325–330

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Hu Q et al (2012) DICER- and AGO3-dependent generation of retinoic acid-induced DR2 Alu RNAs regulates human stem cell proliferation. Nat Struct Mol Biol 19(11):1168–1175

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Rybak-Wolf A et al (2014) A variety of dicer substrates in human and C. elegans. Cell 159(5):1153–1167

    Article  CAS  PubMed  Google Scholar 

  89. Krol J et al (2007) Ribonuclease dicer cleaves triplet repeat hairpins into shorter repeats that silence specific targets. Mol Cell 25(4):575–586

    Article  CAS  PubMed  Google Scholar 

  90. Much C et al (2016) Endogenous mouse dicer is an exclusively cytoplasmic protein. PLoS Genet 12(6):e1006095

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. White E et al (2014) Human nuclear Dicer restricts the deleterious accumulation of endogenous double-stranded RNA. Nat Struct Mol Biol 21(6):552–559

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Neve J et al (2016) Subcellular RNA profiling links splicing and nuclear DICER1 to alternative cleavage and polyadenylation. Genome Res 26(1):24–35

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Wang Y et al (2008) Embryonic stem cell-specific microRNAs regulate the G1-S transition and promote rapid proliferation. Nat Genet 40(12):1478–1483

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Wang Y et al (2013) miR-294/miR-302 promotes proliferation, suppresses G1-S restriction point, and inhibits ESC differentiation through separable mechanisms. Cell Rep 4(1):99–109

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Gu KL et al (2016) Pluripotency-associated miR-290/302 family of microRNAs promote the dismantling of naive pluripotency. Cell Res 26(3):350–366

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Guo WT et al (2015) Suppression of epithelial-mesenchymal transition and apoptotic pathways by miR-294/302 family synergistically blocks let-7-induced silencing of self-renewal in embryonic stem cells. Cell Death Differ 22(7):1158–1169

    Article  CAS  PubMed  Google Scholar 

  97. Cao Y et al (2015) miR-290/371-Mbd2-Myc circuit regulates glycolytic metabolism to promote pluripotency. EMBO J 34(5):609–623

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Wu DR et al (2018) Opposing roles of miR-294 and MBNL1/2 in shaping the gene regulatory network of embryonic stem cells. EMBO Rep 19(6):e45667

    Article  CAS  Google Scholar 

  99. Wray J, Kalkan T, Smith AG (2010) The ground state of pluripotency. Biochem Soc Trans 38(4):1027–1032

    Article  CAS  PubMed  Google Scholar 

  100. Yan Y et al (2017) Significant differences of function and expression of microRNAs between ground state and serum-cultured pluripotent stem cells. J Genet Genomics 44(4):179–189

    Article  PubMed  Google Scholar 

  101. Melton C, Judson RL, Blelloch R (2010) Opposing microRNA families regulate self-renewal in mouse embryonic stem cells. Nature 463(7281):621–626

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Kondoh H et al (2007) A high glycolytic flux supports the proliferative potential of murine embryonic stem cells. Antioxid Redox Signal 9(3):293–299

    Article  CAS  PubMed  Google Scholar 

  103. Prigione A et al (2010) The senescence-related mitochondrial/oxidative stress pathway is repressed in human induced pluripotent stem cells. Stem Cells 28(4):721–733

    Article  CAS  PubMed  Google Scholar 

  104. Varum S et al (2011) Energy metabolism in human pluripotent stem cells and their differentiated counterparts. PLoS One 6(6):e20914

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Gambardella G et al (2017) The impact of microRNAs on transcriptional heterogeneity and gene co-expression across single embryonic stem cells. Nat Commun 8:14126

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Kumar RM et al (2014) Deconstructing transcriptional heterogeneity in pluripotent stem cells. Nature 516(7529):56–61

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Murchison EP et al (2005) Characterization of Dicer-deficient murine embryonic stem cells. Proc Natl Acad Sci USA 102(34):12135–12140

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Kanellopoulou C et al (2005) Dicer-deficient mouse embryonic stem cells are defective in differentiation and centromeric silencing. Genes Dev 19(4):489–501

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Ma Y et al (2015) Functional screen reveals essential roles of miR-27a/24 in differentiation of embryonic stem cells. EMBO J 34(3):361–378

    Article  CAS  PubMed  Google Scholar 

  110. Landgraf P et al (2007) A mammalian microRNA expression atlas based on small RNA library sequencing. Cell 129(7):1401–1414

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Xu N et al (2009) MicroRNA-145 regulates OCT4, SOX2, and KLF4 and represses pluripotency in human embryonic stem cells. Cell 137(4):647–658

    Article  CAS  PubMed  Google Scholar 

  112. Tay Y et al (2008) MicroRNAs to Nanog, Oct4 and Sox2 coding regions modulate embryonic stem cell differentiation. Nature 455(7216):1124–1128

    Article  CAS  PubMed  Google Scholar 

  113. Schouten M et al (2015) MicroRNA-124 and -137 cooperativity controls caspase-3 activity through BCL2L13 in hippocampal neural stem cells. Sci Rep 5:12448

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Santos MC et al (2016) miR-124, -128, and -137 orchestrate neural differentiation by acting on overlapping gene sets containing a highly connected transcription factor network. Stem Cells 34(1):220–232

    Article  CAS  PubMed  Google Scholar 

  115. Pons-Espinal M et al (2017) Synergic functions of miRNAs determine neuronal fate of adult neural stem cells. Stem Cell Reports 8(4):1046–1061

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Weinberger L et al (2016) Dynamic stem cell states: naive to primed pluripotency in rodents and humans. Nat Rev Mol Cell Biol 17(3):155–169

    Article  CAS  PubMed  Google Scholar 

  117. Tesar PJ et al (2007) New cell lines from mouse epiblast share defining features with human embryonic stem cells. Nature 448(7150):196–199

    Article  CAS  PubMed  Google Scholar 

  118. Brons IG et al (2007) Derivation of pluripotent epiblast stem cells from mammalian embryos. Nature 448(7150):191–195

    Article  CAS  PubMed  Google Scholar 

  119. Kalkan T, Smith A (2014) Mapping the route from naive pluripotency to lineage specification. Philos Trans R Soc Lond B Biol Sci 369(1657):20130540

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Shenoy A, Danial M, Blelloch RH (2015) Let-7 and miR-125 cooperate to prime progenitors for astrogliogenesis. EMBO J 34(9):1180–1194

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Morita S et al (2007) One Argonaute family member, Eif2c2 (Ago2), is essential for development and appears not to be involved in DNA methylation. Genomics 89(6):687–696

    Article  CAS  PubMed  Google Scholar 

  122. Bernstein E et al (2003) Dicer is essential for mouse development. Nat Genet 35(3):215–217

    Article  CAS  PubMed  Google Scholar 

  123. Zimmermann C et al (2014) Germ cell-specific targeting of DICER or DGCR8 reveals a novel role for endo-siRNAs in the progression of mammalian spermatogenesis and male fertility. PLoS One 9(9):e107023

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Suh N et al (2010) MicroRNA function is globally suppressed in mouse oocytes and early embryos. Curr Biol 20(3):271–277

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Tang F et al (2007) Maternal microRNAs are essential for mouse zygotic development. Genes Dev 21(6):644–648

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Murchison EP et al (2007) Critical roles for Dicer in the female germline. Genes Dev 21(6):682–693

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Tam OH et al (2008) Pseudogene-derived small interfering RNAs regulate gene expression in mouse oocytes. Nature 453(7194):534–538

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Watanabe T et al (2008) Endogenous siRNAs from naturally formed dsRNAs regulate transcripts in mouse oocytes. Nature 453(7194):539–543

    Article  CAS  PubMed  Google Scholar 

  129. Stein P et al (2015) Essential Role for endogenous siRNAs during meiosis in mouse oocytes. PLoS Genet 11(2):e1005013

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Kim YS et al (2016) Deficiency in DGCR8-dependent canonical microRNAs causes infertility due to multiple abnormalities during uterine development in mice. Sci Rep 6:20242

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Hawkins SM et al (2012) Dysregulation of uterine signaling pathways in progesterone receptor-Cre knockout of dicer. Mol Endocrinol 26(9):1552–1566

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Steiner DF et al (2011) MicroRNA-29 regulates T-box transcription factors and interferon-gamma production in helper T cells. Immunity 35(2):169–181

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Ma F et al (2011) The microRNA miR-29 controls innate and adaptive immune responses to intracellular bacterial infection by targeting interferon-gamma. Nat Immunol 12(9):861–869

    Article  CAS  PubMed  Google Scholar 

  134. Bezman NA et al (2010) Distinct requirements of microRNAs in NK cell activation, survival, and function. J Immunol 185(7):3835–3846

    Article  CAS  PubMed  Google Scholar 

  135. Leong JW, Sullivan RP, Fehniger TA (2012) Natural killer cell regulation by microRNAs in health and disease. J Biomed Biotechnol 2012:632329

    PubMed  PubMed Central  Google Scholar 

  136. Khan IS et al (2014) Canonical microRNAs in thymic epithelial cells promote central tolerance. Eur J Immunol 44(5):1313–1319

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Zuklys S et al (2012) MicroRNAs control the maintenance of thymic epithelia and their competence for T lineage commitment and thymocyte selection. J Immunol 189(8):3894–3904

    Article  CAS  PubMed  Google Scholar 

  138. Papadopoulou AS et al (2011) The thymic epithelial microRNA network elevates the threshold for infection-associated thymic involution via miR-29a mediated suppression of the IFN-alpha receptor. Nat Immunol 13(2):181–187

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Brandl A et al (2016) The microprocessor component, DGCR8, is essential for early B-cell development in mice. Eur J Immunol 46(12):2710–2718

    Article  CAS  PubMed  Google Scholar 

  140. Koralov SB et al (2008) Dicer ablation affects antibody diversity and cell survival in the B lymphocyte lineage. Cell 132(5):860–874

    Article  CAS  PubMed  Google Scholar 

  141. Ventura A et al (2008) Targeted deletion reveals essential and overlapping functions of the miR-17 through 92 family of miRNA clusters. Cell 132(5):875–886

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Lin HP et al (2015) Microprocessor complex subunit DiGeorge syndrome critical region gene 8 (Dgcr8) is required for schwann cell myelination and myelin maintenance. J Biol Chem 290(40):24294–24307

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Le N et al (2005) Analysis of congenital hypomyelinating Egr2Lo/Lo nerves identifies Sox2 as an inhibitor of Schwann cell differentiation and myelination. Proc Natl Acad Sci USA 102(7):2596–2601

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Yun B et al (2010) MicroRNA-deficient Schwann cells display congenital hypomyelination. J Neurosci 30(22):7722–7728

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Viader A et al (2011) MicroRNAs modulate Schwann cell response to nerve injury by reinforcing transcriptional silencing of dedifferentiation-related genes. J Neurosci 31(48):17358–17369

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Goebbels S et al (2006) Genetic targeting of principal neurons in neocortex and hippocampus of NEX-Cre mice. Genesis 44(12):611–621

    Article  CAS  PubMed  Google Scholar 

  147. Hsu R et al (2012) Loss of microRNAs in pyramidal neurons leads to specific changes in inhibitory synaptic transmission in the prefrontal cortex. Mol Cell Neurosci 50(3–4):283–292

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Davis TH et al (2008) Conditional loss of Dicer disrupts cellular and tissue morphogenesis in the cortex and hippocampus. J Neurosci 28(17):4322–4330

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Cheloufi S et al (2010) A dicer-independent miRNA biogenesis pathway that requires Ago catalysis. Nature 465(7298):584–589

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Babiarz JE et al (2011) A role for noncanonical microRNAs in the mammalian brain revealed by phenotypic differences in Dgcr8 versus Dicer1 knockouts and small RNA sequencing. RNA 17(8):1489–1501

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Gu S et al (2012) The loop position of shRNAs and pre-miRNAs is critical for the accuracy of dicer processing in vivo. Cell 151(4):900–911

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Wang XW et al (2017) A DGCR8-independent stable MicroRNA expression strategy reveals important functions of miR-290 and miR-183-182 families in mouse embryonic stem cells. Stem Cell Rep 9(5):1618–1629

    Article  CAS  Google Scholar 

  153. Kim HJ et al (2014) MicroRNAs are required for the feature maintenance and differentiation of brown adipocytes. Diabetes 63(12):4045–4056

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Busskamp V et al (2014) miRNAs 182 and 183 are necessary to maintain adult cone photoreceptor outer segments and visual function. Neuron 83(3):586–600

    Article  CAS  PubMed  Google Scholar 

  155. Ohana R et al (2015) MicroRNAs are essential for differentiation of the retinal pigmented epithelium and maturation of adjacent photoreceptors. Development 142(14):2487–2498

    Article  CAS  PubMed  Google Scholar 

  156. Sundermeier TR et al (2017) MicroRNA-processing enzymes are essential for survival and function of mature retinal pigmented epithelial cells in mice. J Biol Chem 292(8):3366–3378

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Chen Z et al (2012) DiGeorge syndrome critical region 8 (DGCR8) protein-mediated microRNA biogenesis is essential for vascular smooth muscle cell development in mice. J Biol Chem 287(23):19018–19028

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Pan Y et al (2011) Conditional deletion of Dicer in vascular smooth muscle cells leads to the developmental delay and embryonic mortality. Biochem Biophys Res Commun 408(3):369–374

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Zou Y et al (2018) Deletion of DGCR8 in VSMCs of adult mice results in loss of vascular reactivity, reduced blood pressure and neointima formation. Sci Rep 8(1):1468

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Albinsson S et al (2011) Smooth muscle miRNAs are critical for post-natal regulation of blood pressure and vascular function. PLoS One 6(4):e18869

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Choi YJ et al (2017) Deficiency of DGCR8 increases bone formation through downregulation of miR-22 expression. Bone 103:287–294

    Article  CAS  PubMed  Google Scholar 

  162. Gaur T et al (2010) Dicer inactivation in osteoprogenitor cells compromises fetal survival and bone formation, while excision in differentiated osteoblasts increases bone mass in the adult mouse. Dev Biol 340(1):10–21

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Sugatani T et al (2014) Expression of DGCR8-dependent microRNAs is indispensable for osteoclastic development and bone-resorbing activity. J Cell Biochem 115(6):1043–1047

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Mizoguchi F et al (2010) Osteoclast-specific Dicer gene deficiency suppresses osteoclastic bone resorption. J Cell Biochem 109(5):866–875

    CAS  PubMed  Google Scholar 

  165. Yi R et al (2009) DGCR8-dependent microRNA biogenesis is essential for skin development. Proc Natl Acad Sci USA 106(2):498–502

    Article  PubMed  Google Scholar 

  166. Yi R et al (2006) Morphogenesis in skin is governed by discrete sets of differentially expressed microRNAs. Nat Genet 38(3):356–362

    Article  CAS  PubMed  Google Scholar 

  167. Rao PK et al (2009) Loss of cardiac microRNA-mediated regulation leads to dilated cardiomyopathy and heart failure. Circ Res 105(6):585–594

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Chen JF et al (2008) Targeted deletion of Dicer in the heart leads to dilated cardiomyopathy and heart failure. Proc Natl Acad Sci USA 105(6):2111–2116

    Article  PubMed  PubMed Central  Google Scholar 

  169. Chapnik E et al (2012) Dgcr8 controls neural crest cells survival in cardiovascular development. Dev Biol 362(1):50–56

    Article  CAS  PubMed  Google Scholar 

  170. Huang ZP et al (2010) Loss of microRNAs in neural crest leads to cardiovascular syndromes resembling human congenital heart defects. Arterioscler Thromb Vasc Biol 30(12):2575–2586

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Bartram MP et al (2016) Mice lacking microRNAs in Pax8-expressing cells develop hypothyroidism and end-stage renal failure. BMC Mol Biol 17:11

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Bartram MP et al (2015) Loss of Dgcr8-mediated microRNA expression in the kidney results in hydronephrosis and renal malformation. BMC Nephrol 16:55

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Bartram MP et al (2013) Conditional loss of kidney microRNAs results in congenital anomalies of the kidney and urinary tract (CAKUT). J Mol Med (Berl) 91(6):739–748

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The research in Wang laboratory is supported by the National Key Research and Development Program of China (2016YFA0100701 and 2018YFA0107601) and the National Natural Science Foundation of China (31471222, 31622033, 31821091 and 91640116). WTG is supported by the Fundamental Research Funds for the Central Universities (3332018008). Due to the breadth of this review, we apologize for the unavoidable exclusion of references to research done by many outstanding investigators working in relevant areas.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Yangming Wang.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Guo, WT., Wang, Y. Dgcr8 knockout approaches to understand microRNA functions in vitro and in vivo. Cell. Mol. Life Sci. 76, 1697–1711 (2019). https://doi.org/10.1007/s00018-019-03020-9

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00018-019-03020-9

Keywords

Navigation