Review
Copyright ©The Author(s) 2015.
World J Biol Chem. Aug 26, 2015; 6(3): 162-208
Published online Aug 26, 2015. doi: 10.4331/wjbc.v6.i3.162
Table 1 Roles and targets of the myomiRs, miR-1, -206, -133a, -133b
Factor(s)RegulationRegulatorTissue/cellRef.
Fish and lower vertebrates: Development and regeneration
Ttk protein kinase (mps1)Upregulated mps1: a target of miR-133Downregulation of miR-133 by FgfRegeneration of Zebrafish caudal fin (appendage)[68]
RhoADownregulation of RhoA mRNAUpregulation of miR-133b expressionRegenerating adult zebrafish spinal cord, axon outgrowth[69]
RhoADownregulation of RhoA proteinUpregulation of miR-1 and miR-133 expressionZebrafish muscle gene expression and regulation of sarcomeric actin organization[166]
Cell cycle factors mps1, cdc37 and PA2G4, and cell junction components cx43 and cldn5Upregulated mps1, cdc37, PA2G4, cx43, cldn5Downregulated miR-133(a1) stimulates cardiac cell regenerationRegenerating zebrafish cardiac muscle[167]
miR-133bMiR-133b found in developing somites, little in CNS tissuesWhole zebrafish embryos - normal development[168]
SRF activates muscle specific genes and miRs;MiR-1 targets HDAC4, promoting myogenesisIn contrast, miR-133a represses SRF, enhancing myoblast proliferationX. laevis embryos: skeletal muscle proliferation and differentiation in cultured myoblasts in vitro and in embryos in vivo[7]
HDAC4 represses muscle gene expression
nAChR subunits UNC-29, UCR-63; MEF2Subunits UNC-29, UCR-63, and MEF2 downregulatedmiR-1 upregulatedC. elegans muscle at the neuromuscular junction[34]
Mammalian pluripotent cells
Muscle-specific microRNAs: miR-1 and miR-133aMiR-1 and miR-133a have opposing functions during differentiation of progenitor cardiac musclesMuscle-specificPromotion of mesoderm formation from mouse ES cells[13]
microRNAs, miR-1 and miR-133(a) upregulated
Notch signalling, promotes neural differentiation and inhibits muscle differentiation; opposes miR-1 effectsDll-1 translationally repressedmiR-1 upregulation, promotes cardiomycete differentiationMouse and human ES cell differentiation into muscle[13]
SRF-/- EBs reflecting the loss of hematopoietic lineages in the absence of SRFEarly endoderm markers, Afp and Hnf4α: strongly down regulatedIncreased miR-1 and miR-133a relieve the block on mesodermal differentiationMouse endoderm[13]
Blood cell -specific genes, such as Cd53, CxCl4, and Thbs1, dramatically down regulatedCd53, CxCl4, and Thbs1 expression was reinitiated by reintroduction of miR-1 or miR-133
mES(miR-1)- and mES(miR-133a)- EBs compared to in control EBsNodal stimulated expression of endoderm markers Afp and Hnf4α in control EBs. Dramatically lower levels in mES(miR-1)- and mES(miR-133a)- EBsmiR-1 or miR-133 can each function as potent repressors of endoderm gene expressionmES cells, that lack either miR-1 or miR-133(a) during differentiation into EBs[13]
IGF-1IGF-1 signalling and miR-133 co-regulate myoblast differentiation via a feedback loopIGF-1 upregulates miR-133;Myogenic differentiation of C2C12 myoblasts; Mouse during development from embryonic to mature skeletal muscle[24]
IGF-1RmiR-133 downregulates IGF-1R
IGF-1IGF-1 signalling and miR-1 coregulate differentiation of myoblasts via a feedback loopIGF-1 signalling downregulates miR-1 by repression of FoxO3a;Differentiating C2C12 myoblasts[25]
miR-1 down-regulates IGF-1
Reversine [2-(4-morpholinoanilino)-N6-cyclohexyladenine]Decrease in active histone modifications; including trimethylation of histone H3K4/ H3K36, phosphorylation of H3S10;miR-133a expression strongly inhibited by reversine; reduced acetylation of H3K14 at miR-133a promoterReversine dedifferentiates murine C2C12 myoblasts back into multipotent progenitor cells, via extensive epigenetic modification of histones resulting in chromatin remodelling, and altered gene expression[20-23]
Stimulates expression of polycomb genes Phc1 and Ezh2Reduced expression of myogenin, MyoD, Myf5 and Aurora A and B kinases
FZD7 and FRS2miR-1 promotes cardiac differentiation; miR-1 targets FZD7 and FRS2Activitation of WNT and signalling cause MCPs differentiation into cardiomyocytesMouse and human ES cells[169]
miR-206/133b clusterPAX7 gene expression unchanged;miR-206/133b cistron knock-out mice cellsMuscle satellite cell differentiation in vitro[170]
miR-206/133b cluster is not required for development, and survival of skeletal muscle cells
Differentiating skeletal muscle
DNA polymerase alphaRepression of Idl-3 protein expressionmiR-206 up-regulatedMouse skeletal muscle differentiation[42]
Repression of p180 subunit of DNA polymerase alpha
MEF2 transcription factorMEF2 activates of miR-1-2 and 133a-1 transcription; binds muscle-specific enhancerBicistronic primary transcript of miR-1-2 and 133a-1Development of mammalian skeletal muscle[9]
MRFs, Myf5, MyoD, Myogenin and MRF4Myf5 essential for miR-1 and miR-206 expression during skeletal muscle myogenesisForced expression of MRFs in neural tube induces miR-1 and miR-206 expressionChicken and mouse embryonic muscle[171]
PTB and neuronal homolog nPTB, exon splicing factorsDownregulation of PTB protein by miR-133 (and miR-206)Concurrent upregulation of miR-133 and induction of splicing of several PTB-repressed exonsDuring myoblast differentiation, microRNAs control a developmental exon splicing program[172]
BDNFBDNF downregulatedmiR-206 upregulatedDifferentiation of C2C12 myoblasts into myotubes[48]
Fstl1 and UtrnFstl1 and Utrn downregulatedmiR-206 upregulatedSkeletal muscle differentiation[40]
Utrophin A (muscle)Utrophin A down-regulated by both miRsUpregulated miR-133b, miR-206C2C12 mouse myoblasts, mouse soleus muscle[173]
CNN3 geneNegative correlation between miR-1 expression and CNN3 mRNA expressionNormal skeletal muscleTongcheng (Chinese) and Landrace (Danish) pigs[174]
FGFR1 and PP2AC, members of ERK1/2 signalling pathwaymiR-133 (a and b) activities increase during myogenesismiR-133 directly downregulates expression of FGFR1 and PP2ACMouse C2C12 myoblast cells[31]
ERK1/2 signalling pathway activityERK1/2 signalling activity suppresses miR-133 expressionDownregulation of expression of miR-133A reciprocal mechanism for regulating myogenesis
BAF chromatin remodelling complex (BAF60a, BAF60b and BAF60c)Positive inclusion of BAF60c in the BAF chromatin remodeling complexExpression of miR-133 and miR-1/206Progression of developing somites in chick embryos[63]
BAF chromatin remodelling complexNegative regulation of BAF60a and BAF60b; exclusion from BAF chromatin remodelling complexExpression of miR-133Progression of developing somites in chick embryos[63]
BAF chromatin remodelling complexExogenous upregulation of BAF60a and BAF60bDelay in developing somites in chick embryos[63]
Mitochondrial UCP2 and UCP3MyoD activates miR-133a expression which in turn directly downregulates UCP2 mRNAFeedback network involving MyoD-miR-133a-UCP2Mouse skeletal and cardiac muscles; UCP2 imposes developmental repression[56]
Mitochondrial UCP2 and UCP3Exogenous overexpression of myogenin and MyoD transcription factorsStrong increase in UCP3 promoter, expression, weak effect at the UCP2 promoterMouse C2C12 myoblasts[57]
Proliferating myogenic skeletal muscle cells
MiR-206/133b clusterMiR-206/133b cluster is not required for survival and regeneration of skeletal muscleMuscle regeneration proceeds in Mdx mice in vivomiR-206/133b cistron knock-out mice[170]
Enhanced translation of specific mitochondrial genome-encoded transcriptsmiR-1 enters muscle mitochondria and binds mtRNA targets along with Ago factorIncreased expression of mtRNA targetsProliferating myogenic skeletal muscle cells after muscle injury[53]
mTOR (serine/threonine kinase)MyoD stability regulated by mTORRegulates miR-1 expression via MyoD availabilityRegenerating mouse skeletal muscle and differentiating myoblast cells[32]
AMPK-CRTC2-CREB and Raptor-mTORC-4EBP1 pathwaysmTORC regulates timing of satellite cell proliferation during myogenesisKnockdown of mTORC reduces miR-1 expressionMyogenenic satellite SCs proliferating and differentiating into myogenic precursors following rat skeletal muscle injury[58]
HDAC4 regulates Pax7-dependent muscle regenerationPax7 stimulates SCs differentiation toward the muscle lineage, and limits adipogenic differentiationHDAC4 upregulated in SCs differentiating into muscle cellsMyogenenic satellite SCs[175]
pcRNA encoded by the H strand of the rat mitochondrial genomeIntroduction of mt pcRNAs into injured muscle restoring mitochondrial mRNA levels; Intramuscular ATP levels were elevated after pcRNA treatment of injured muscleEnhanced organellar translation and respiration; similarly reactive oxygen species were reduced; Resulted in accelerated rate of wound resolutionInjured rat skeletal muscle is associated with general downregulation of mitochondrial function; reduced ATP, and increased ROS[176]
Cardiac muscle precursor cells
GATA binding protein 4, Hand2, T-box5, myocardin, and microRNAs miR-1 and miR-133Reprogrammed human fibroblasts show sarcomere-like structures and calcium transients; Some cells have spontaneous contractilityForced over-expression of GATA binding protein 4, Hand2, T-box5, myocardin, and microRNAs miR-1 and miR-133Human embryonic and adult fibroblasts activated to express cardiac markers[15]
SRF, MyoD and Mef2 transcription factorsmiR-1-1 and miR-1-2miR-1 genes upregulated;Cardiac muscle precursor cells[30]
During cardiogenesis miR-1 genes titrate critical cardiac regulatory proteins, control ratio of differentiation to proliferationElevated miR-1 targets downregulation of Hand2
Histone deacetylase inhibitor, trichostatin A forces differentiation, yet reduced miR-1 and miR-133amiR-1 and miR-133a reduce cardiac specific Nkx2.5 protein and Cdk9miR-1 and miR-133a increase during spontaneous differentiation of cardiac myoblastsMouse cardiac stem cells (ES cells)[10]
Specific inhibition of HDAC4 modulates CSCs to facilitate myocardial repairPositively proliferative myocytes increased in MI hearts receiving HDAC4 downregulated CSCsCSCs with downregulated HDAC4 expression improved ventricular function, attenuated ventricular remodeling, promoted regeneration and neovascularization in MI heartsMouse CSCs transplanted into MI mouse hearts[177]
Snai1Overexpression of miR-133a (miR-133), Gata4, Mef2c, and Tbx5 (GMT) or GMT plus Mesp1 and MyocD improved cardiac cell reprogramming from mouse or human fibroblastsmiR-133a directly represses Snai1 expression, which silences fibroblast signatures; a key molecular process during cardiac reprogrammingMouse/human fibroblasts more efficiently reprogrammed into cardiomycete-like cells[16]
β1AR signal transduction cascadeAdenylate cyclase VI and the catalytic subunit of the cAMP-dependent PKA are components of β1AR transduction cascademiR-133 directly targets β1AR, Adenylate cyclase VI and PKATetON-miR-133 inducible transgenic mice, subjected to transaortic constriction, maintained cardiac performance with attenuated apoptosis and reduced fibrosis via elevated miR-133 expression[17]
ROS, MDA, SOD and GPxmiR-133 produced a reduction of ROS and MDA levels, and an increase in SOD activity and GPx levelsOverexpression of miR-133, a recognized anti-apoptotic miRNAIn vitro rat cardiomyocytes[18]
Caspase-9miR-133 directly suppresses caspase-9 expression resulting in downregulation of downstream apoptotic pathwaysOverexpression of miR-133In vitro rat cardiomyocytes[18]
Spred1miR-1 directly targets Spred1miR-1 is upregulated in hCMPCs during angiogenic differentiationhCMPCs[178]
miRNA-1 and miRNA-133amiRNA-1 and miRNA-133a have antagonistic roles in the regulation of cardiac differentiationForced overexpression of miR-1 alone enhanced cardiac differentiation, in contrast overexpression of miR-133a reduced cardiac differentiation, compared to control cellsPluripotent P19.CL6 stem cells[179]
Overexpression of both miRNAs promoted mesodermal commitment and decreased expression of neural differentiation markers
Cardiac muscle
Induction of GATA6, Irx4/5, and Hand2Cardiac myocytes show defective heart development, altered cardiac morphogenesis, channel activity, and cell cyclingmiR-1-2-/- gene knockoutCardiac myocytes with knockout of both miR-1-2 genes[180]
mt-COX1 mRNA3’-UTR of mt-COX1 mRNA bound by miR-181c and Ago1 factorOverexpression of miR-181c significantly decreased mt-COX1 protein, but not mt-COX1 mRNA levelOverexpression of miR-181c increased mitochondrial respiration and reactive oxygen species in neonatal rat ventricular myocytes[54]
mt-COX1 mRNAIn vivo elevation of miR-181c in rat heart, reduces levels of mt-COX1 proteinResults in reduced capacity for strenuous exercise and evidence of heart failureRat cardiac muscle[55]
Carvedilol, a β-adrenergic blockerInduces upregulation of miR-133Cytoprotective effects against cardiomyocyte apoptosisRat cardiac tissue, in vivo[18]
GLUT4, and SRFBoth miRs downregulate SRF and KLF15Both miR-133a and miR-133b target KLF15Mouse cardiac myocytes[181]
GLUT4 expressionBoth basal and insulin-stimulated glucose uptake are increasedKLF15Mouse muscle cell lines[182]
MEF2 transcription factorMEF2 directly activates transcription of miR-1-2 and 133a-1 binding muscle-specific enhancer between the genesBicistronic primary transcript of miR-1-2 and 133a-1Development of mammalian cardiac muscle[9]
Myocardium tissueEnriched in miR-1, miR-133b, miR-133aHeart structures of rat, Beagle dog and cynomolgus monkey[183]
GelsolinOne common miR-133a isomiR targets gelsolin gene more efficiently than standard isomer; New second rat miR-1 geneMany isomiRs were detected by deep sequencing at higher frequency than the canonical sequence in miRBasemiRNA/isomiR expression profiles in the left ventricular wall of rat heart[184]
CTGFCTGF downregulated by both miRsExogenous upregulation of miR-133b (and miR-30c)Cultured cardiomyocytes and ventricular fibroblasts[185]
MT1-MMPmiR-133a upregulatedmiR-133a targets MT1-MMPHuman left ventricular fibroblasts[186]
Injured and regenerating cardiac muscle
SERCA2aAkt/FoxO3A-dependent pathwayDownregulation of miR-1 expression in failing heart muscleFailing mouse heart muscle[187]
Activated SERC2a reduces phosphorylation of FoxO3a, allowing entry to nucleus and activation of miR-1 expression
IGF-1IGF-1 signalling and miR-1 co-regulate differentiation of myoblasts via a feedback loopIGF-1 signalling down-regulates miR-1 by repression of FoxO3a;Mouse heart muscle during cardiac failure states[25]
miR-1 down-regulates IGF-1
Bim and BmfOnly miR-133a expression enhanced under in vitro oxidative stressmiR-133a targets proapoptotic genes Bim and BmfRat adult CPCs[188]
miR-1 favors differentiation of CPCs, whereas
Bim and BmfCPCs overexpressing miR-133a improved cardiac function by reducing Bim and BmfCPCs overexpressing miR-133a improved cardiac function, increasing vascularization and cardiomyocyte proliferation, reduced fibrosis and hypertrophyCPCs overexpressing miR-133a in rat myocardial infarction model[188]
MT1-MMP activity increased in both. Ischemia and reperfusion regionsInterstitial miR-133a decreased with ischemia in vitro and in vivo; reperfusion returned to steady-statePhosphorylated Smad2 increased within the ischemia-reperfusion regionIschemia-reperfusion Yorkshire pigs (90 min ischemia/120 min reperfusion)[186]
Cardiovascular disease
CNN2Strong upregulation of CNN2 expressionmiR-133b downregulated; miR-133b directly targets CNN2Pre-inflammatory events in diseased cardiac tissues[65]
Circulating platelet derived microparticlesElevated miR-133Patients with stable and unstable coronary artery disease[189]
Acute MI causes upregulation of circulating serum miRsmiR-1, -133a, -133b, and -499-5p were about 15- to 140-fold elevated over controlAcute STEMI patients and experimental mouse MI model[190]
Circulating miRNAs in serum of cardiovascular disease patientsReleased miR-1 and miR-133a are localized in exosomes, and are released by Ca(2+) stimulationLevels of miR-1, miR-133a, reduced in infarcted mouse myocardium model heartmiR release indicates myocardial damage[191]
LVM after valve replacement in aortic stenosismicroRNA-133a is a significant positive predictor of LVM normalisationmiR-133 is a key element of the reverse remodelling processPatients following valve replacement[192]
Circulating levels of miR-133aElevated miR-133a (11-fold)Troponin-positive acute coronary syndrome patients[193]
Circulating levels of miR-133aElevated miR-133aImproved potential regression of Left Ventricular Hypertrophy after valve replacementPatients with aortic stenosis surgery[194]
Apelin treatment reduces elevated circulating miRsElevated miR-133a, miR-208 and miR-1 reducedHigh-fat diet elevated miRs and increased left ventricular diastolic and systolic diameters, and wall thicknessObesity-associated cardiac dysfunction in mouse model[195]
NAC treatmentExpressed miR-1, miR-499, miR-133a, and miR-133b were strongly depressed in the diabetic cardiomyocytesNAC restored expression of miR-499, miR-1, miR-133a, and miR-133b significantly in the myocardiumDiabetic rat hearts[196]
Myocardial junctin elevatedmiR-1 targets junctinNAC reduces junction levelsDevelopment of diabetic cardiomyopathy in rat hearts[196]
CAD associated ischemic heart failuremiR-133 expression decreased with increased severity of heart failurePatients with CAD[197]
Runx2miR-133a targets Runx2Transition of VSMCs to osteoblast-like cells[198]
Increased alkaline phosphatase activity, osteocalcin secretion and Runx2 expressionmiR-133a was decreased during osteogenic differentiationTransition of VSMCs to osteoblast-like cells[198]
Circulating miR-133a and 208a levelsCardiac muscle-enriched microRNAs (miR-133a, miR-208a) elevatedPatients with coronary artery disease[199]
Hypertrophic cardiac muscle
Cx43 increasedmiR-1 targets Cx43Downregulation of miR-1 mediates induction of pathologic cardiac hypertrophyHypertrophic rat cardiomyocytes in vitro and in vivo[200]
Cx43 downregulatedmiR-1 targets Cx43Cx43 protein downregulated in miR-1 Tg mice compared to WT miceCardiac-specific miR-1 transgenic (Tg) mouse model[201]
Twf1 upregulatedmiR-1 targets Twf1Strong downregulation of miR-1 in pathologic hypertrophic cardiac cells compared to normal, induces Twf1 expressionIn vivo in hypertrophic mouse left ventricle; and in vitro in phenylephrine-induced hypertrophic cardiomyocytes[202]
RhoA, Cdc42, Nelf-A/WHSC2Increased levels of RhoA, Cdc42, Nelf-A/WHSC2Reduction miR-133aHypertrophic cardiac muscle[6]
Calcineurin, agonist of cardiac hypertrophyIncreased Calcineurin activity;Reduced miR-133a;Hypertrophic cardiac muscle;[203]
Cyclosporin A inhibits calcineurinPrevents miR-133 down-regulationCardiac hypertrophy reduced
NFATc4NFAFc4 targetted by miR-133amiR-133aCardiomyocyte hypertrophic repression[204]
Interdependent Calcineurin-NFAT and MEK1-ERK1/2 signalling pathways in cardiomyocytesMEK1-ERK1/2 signalling augments NFAT and NFAF gene expression; Activated calcineurin activates NFAT, inducing cardiac hypertrophyMEK1 is part of mitogen-activated protein kinase (MAPK) cascade; MEK1 activates ERK directlyHypertrophic growth response of mouse cardiomyocytes[205]
Innervating skeletal muscle
Innervated skeletal muscleMyoD, Myf5, Mrt4, nAChRαMyogenin expressionMouse skeletal muscle[50,51]
Each is strongly repressed
Denervated muscle (unstimulated)Myogenin expression up-regulated MyoD, Myf5, Mrt4, nAChRαMouse skeletal muscle[51]
All strongly stimulated
Electrically stimulated - Denervated muscleMyogenin, MyoD, Myf5, Mrt4, partly stimulated; nAChRα inhibitedMouse skeletal muscle[51]
HDAC4miR-1 promotes myogenesis by targetting HDAC4miR-133 enhances myoblast proliferation by targetting SRFSkeletal muscle proliferation and differentiation in myoblast cultures[7]
SRF
Neural activity effect on muscle (HDAC4 - MEF2 Axis)Loss of neural input leads to concomitant nuclear accumulation of HDAC4HDAC4 inhibits activation of muscle transcription factor MEF2; results in progressive muscle dysfunctionMEF-2 activity strongly inhibited in denervated mouse skeletal muscle and in ALS muscle[49]
Innervation and formation of airway smooth muscleSonic hedgehog (Shh) /miR-206/ BDNFShh signalling blocks miR-206 expression, which in turn increases BDNF proteinShh coordinates innervation and formation of airway smooth muscle[206]
nAChR subunits (UNC-29 and UNC-63); retrograde signallingSubunits UNC-29, UCR-63 and MEF2 downregulatedmiR-1 upregulatedC. elegans muscle at the neuromuscular junction[34]
MEF2Hnrpu, Lsamp, MGC108776, MEF2, Npy, and Ppfibp2 downregulatedmiR-206 upregulatedRat skeletal muscle/re-innervating muscle[43]
HDAC4HDAC4 (miR-206 target, prospective miR-133b target) downregulatedmiR-206/-133b upregulated (and miR-1/-133a downregulated)Mouse fast twitch skeletal muscle/re-innervating muscle[12]
Regenerating injured muscle
Hnrpu and Npy downregulatedmiR-1 upregulatedmiR-1, -133a, downregulated 1 mo after denervation, then increased 2 × at 4 mo after re-innervationRat skeletal soleus muscle after sciatic nerve injury and subsequent re-innervation[43]
Ptprd downregulatedmiR-133a upregulated
Hnrpu, Lsamp, MGC108776, MEF2, Npy, and Ppfibp2 downregulated3 × increase in miR-206 1 mo later, after reinnervation; elevated at least 4 moPredominant type II fiber at 4 mo, after nerve re-innervationRat skeletal soleus muscle after sciatic nerve injury and subsequent re-innervation[43]
PP2A B56aPP2A B56a downregulated133a upregulatedCanine heart failure model: myocytes[207]
CaMKII-dependent hyperphosphorylation of RyR2VF myocytes had increased reactive oxygen species and increased RyR oxidationmiR-1 upregulatedCanine post-myocardial infarction model[208]
Collagen upregulatedTGF-b1 and TGFbRII: upregulatedmiR-133a or miR-590: downregulatedCanine model of acute nicotine exposure. Atrial fibrosis in vivo; cultured canine atrial fibroblasts in vitro[209]
miR-208 upregulatedmiR-1 and miR-133a downregulatedHuman MI compared to healthy adult hearts[210]
Myogenic proteins, MyoD1, myogenin and Pax7Induced expression of MyoD1, myogenin and Pax7 several days after miR injectionExogenous injection of miR-1, -133 and -206 promotes myotube differentiationRegenerating injured mouse skeletal muscle[211]
Cyclin D1/ Sp1Cyclin D1/ Sp1 downregulatedmiR-1/133 upregulatedRegenerating rat skeletal muscle[212]
PRP, source of pro-inflammatory cytokinesStong upregulation of the mRNA of pro-inflammatory cytokines IL-1β and TGF-β1; stimulation of both inflammatory and myogenic pathways; elevated heat shock proteins and increased phosphorylation of αB-cristallinStimulated tissue recovery via increased myogenic regulators MyoD1, Myf5, Pax7, and IGF-1Eb (muscle isoform) together with SRF; acts via increased expression of miR-133a with reduced levels of apoptotic factors (NF-κB-p65 and caspase 3)Regenerating flexor sublimis muscle of rats, 5 d after injury and treated with PRP[66]
Muscle degeneration
Pro-inflammatory cytokine TWEAKTWEAK upregulatedmiR-1-1, miR-1-2, miR-133a, miR-133b and miR-206 downregulatedDegenerating/wasting mouse skeletal muscle[59]
HMOX1 mediated by codependent inhibition of c/EBPδ binding to myoD promoterHMOX1 inhibits differentiation of myoblasts and modulates miRNA processingDownregulation of miR-1, miR-133a, miR-133b, and miR-206.Degenerating/wasting mouse skeletal muscle[60]
HMOX1 effects partially reversed by enforced expression of miR-133b and miR-206Downregulation of MyoD, myogenin and myosin, and disturbed formation of myotubes. Upregulation of SDF-1 and miR-146a
Dystrophic muscular disease
Circulating serum microRNAsmiR-1, miR-133a, and miR-206 highly abundant in Mdx serummiR-1, miR-133a, and miR-206 downregulated or modestly upregulated in muscleMuscle tissue from patients with Duchenne muscular dystrophy (Mdx)[213]
Laminin α2 chain deficiencymiR-1, miR-133a, and miR-206 are deregulated in laminin α2 chain-deficient muscleLaminin α2 chain-deficient mouseCongenital muscular dystrophy type 1A tissue[214]
Dystrophic process advances from prominent inflammation with necrosis and regeneration to prominent fibrosisDeficiency in calpain leads initially to accelerated myofiber formation followed by depletion of satellite cellsPax7-positive SCs highest in the fibrotic patient group; correlated with down-regulation of miR-1, miR-133a, and miR-206Muscle from Limb-girdle muscular dystrophy 2 type I patients[215]
Transgenic overexpression of miR-133a1 (in dystrophin point mutation Mdx mice)Extensive overexpression in skeletal muscle, lesser increase in heartNormal skeletal muscle and heart developmentMdx mice (model for human muscular dystrophy), extensor digitorum longus muscle[216]
miR-206 located in nuclear in both normal and DM1 tissues by in situ hybridizationOnly miR-206 showed an over-expression in majority of DM1 patientsNo change in expression of profiled miRs, miR-1, miR-133 (miR-133a/-133b), miR-181 (miR-181a/-181b/-181c)Skeletal muscle (vastus lateralis) of from patients with myotonic dystrophy type 1 (DM1)[217]
FAPs facilitate myofiber regenerationHDAC inhibitors can activate FAPs towards muscle regenerationInhibition of HDAC induces MyoD and BAF60C expression, which causes up-regulation of miR-1-2, miR-133, and miR-206 expressionEarly stage disease dystrophic mouse muscles, regeneration of myofibres[62]
TDP-43TDP-43 interacts with miR-1/-206 isomers, but not miR-133 isomersDepleted miR-1/-206 allow targets IGF-1 and HDAC4 to accumulate in ALS muscleMouse ALS model injured motor neurons and muscle[33]
Inflammation response in muscle
Inflammatory myopathiesIncreased expression of TNFαAssociated with decreased expression of miR-1, miR-133a, and miR-133bInflammatory myopathies including dermatomyositis, polymyositis, and inclusion body myositis[64]
hBSMCs sensitized with IL-13Increased muscle RhoAReduction of muscle miR-133aSensitized human bronchial smooth muscle cells (hBSMCs)[218]
Table 2 Roles and targets of the myomiRs, miR-1, -206, -133a, -133b in other precursor cells and tissues
Factor(s)RegulationRegulatorTissue/cellRef.
Nerve tissues
Pitx3Pitx3 downregulatedmiR-133bMammalian midbrain DNs[73]
Exosome-mediated transfer of miR-133b from MSC to brain astrocytesmiR-133b transfer from multipotent mesenchymal stromal cells to neural cellsmiR-133b upregulatedMouse MSCs to neural cells[47]
Ctgf and RhoACtgf and RhoA downregulatedmiR-133b upregulatedMultipotent MSCs/Rat brain parenchymal cells[72]
miR-133b null mice: Striatum dopamine levels unchanged, Pitx3 expression unaffected; motor coordination unalteredmiR-133b has no significant role on mDA neuron development and maintenance in vivoNormal numbers of mDA neurons during development and aging of miR-133b null miceMouse mDA neuron development in -/-miR-133b mutant mice[45]
Acute or chronic morphine administration, or morphine withdrawalmiR-133b levels not affectedRat VTA/ nucleus accumbens shell[219]
GPM6A, a neuronal glycoproteinmicroRNA-133b upregulationReduction in gmp6a at mRNA and protein level. Cell filopodium density was reducedHippocampus and prefrontal cortex of neonatal male rats stressed when in utero[220]
Tac1 gene (neurotransmitter substance P)Tac1 downregulatedmiR-206 upregulatedMSCs-derived neural cells[221]
Ketamine (antidepressive) administrationBDNF, a direct target gene of miR-206, was upregulatedmiR-206 was downregulated by ketamineRat hippocampus tissue[222]
Adipogenic tissues
IGF-1 and IGF-1RIGF-1 signalling and miR-133b co-regulate ADSC differentiation via a feedback loopmiR-133b downregulation of Pitx3;Adipose tissue-derived stem cell differentiation into neuron-like cells[71]
IGF-1 upregulates miR-133b;
miR-133b downregulates IGF-1R
Pdrm16miR-133a directly targets Prdm16.Downregulation of miR-133 resultsin differentiation of pre-adipocyte precursors into BATMouse adipocyte differentiation to BAT[74]
Pdrm16miR-133 directly targets Prdm16Downregulation of miR-133 resulted in differentiation of pre-adipocyte precursors into BATMouse primary brown adipocyte (and myogenic) progenitor cells - differentiate into BAT or SAT[75]
Pdrm16miR-133 targets Prdm16 controlling brown adipose determination in skeletal muscle satellite cellsmiR-133 downregulates Prdm16Adult mouse skeletal muscle stem cells (satellite cells) differentiate into BAT[76]
HDAC4 downregulation directs SCs towards adipocyte differentiationBrown adipose master regulator Prdm16 is upregulated, while its inhibitor miR-133 is also downregulatedHDAC4 downregulated in SCs differentiating into adipocyte progenitor cellsMyogenenic satellite SCs[175]
GLUT4 expressionBoth basal and insulin-stimulated glucose uptake are increasedKLF15Mouse 3T3-L1 preadipocytes differentiating into adipocytes[182]
Intrinsic insulin resistanceElevated miR-133bUndefined roleAdipose tissue of women with PCOS[223]
Upregulation of LIM homeobox 8 and Zic family member 1 and downregulation of Homeobox C8 and Homeobox C9Undefined relation of upregulated miR-206, miR-133bUndefined relation with parallel upregulation of brite/beige markers, TBX1 and TMEM26Human BAT from the supraclavicular region[224]
Obesity developmentDownregulation of miR-133b, miR-1Undefined roleAdipose tissue from obese male C57BLJ6 mice[225]
LXRα regulation of lipogenic genesmiR-1/miR-206 represses LXRα expression at both mRNA and protein levelsmiR-1/miR-206-induces a decrease in lipogenic gene levels and lipid droplet accumulationMouse hepatocytes[226]
Osteogenic tissues
Development of bone on organic or inorganic substratesmiR-133 differentially expressed in osteoblasts grown on different substratesOsteoblast[227]
Runx2miR-133 directly down-regulates Runx2miR-133 up-regulatedOsteogenic differentiation from C2C12 mesenchymal cells[228]
HDAC4HDAC4 downregulates Runx2miR-1 targets HDAC4, increasing Runx2 activityChondrocyte proliferation in cartilage growth plate[77]
AggrecanmiR-1 promotes late-stage differentiation of growing cartilage cellsmiR-1 targets Aggrecan gene expressionChicken chondrocytes and human HCS-2/8 cells[78]
Alveolar cells
VAMP2/ lung surfactant secretionmiR-206 targets VAMP-2miR-206 overexpression decreased lung surfactant secretionLung alveolar type II cells[229]
Hormonal regulation
L-thyroxinemiR-206/miR-133b downregulatedL-thyroxine treatmentL-thyroxine treated hypothroidic skeletal muscle from thyroidectomized patients[230]
miR-206/miR-133b upregulated-Hypothroidic human skeletal muscle
Thyroid hormone/TEAD1Thyroid hormone inhibits the slow muscle phenotype by upregulation of miR-133a1 which downregulates TEAD1miR-133a1 is enriched in fast-twitch muscle and regulates slow-to-fast muscle fiber type conversionMouse muscle[231]
Thyroid hormone/miR-133a1 TEAD1myosin heavy chain I expression downregulatedTH indirectly downregulates myosin heavy chain I via miR-133a/TEAD1Mouse muscle[232]
L-thyroxinepre-miR-206 and pre-mir-133b downregulatedL-thyroxineL-thyroxine treated hypothyroidic mouse liver;[232]
50-500x increase expression of miR-1/-133a and miR-206/-133b-Hypothyroidic mouse liver
Reduced insulin-mediated glucose uptake in cardiomycetesDownregulation KLF15, which downregulates GLUT4Forced overexpression of miR-133a and miR-133bRat cardiac myocytes[181]
Cardiac myocyte glucose metabolismUpregulation KLF15, which upregulates GLUT4Silencing endogenous miR-133Rat cardiac myocytes[181]
Metabolic control of glucose uptake by GLUT4 transporterDownregulates KLF15, which results in downregulation of GLUT4 levelsChronic heart failure has depressed miR-133a and -133b levelsRat cardiac myocytes during chronic heart failure and cardiac hyperthrophy[181]
Atrial natriuretic factor expression upregulationEnhanced at LVH and dramatically increased at CHF stageBoth miR-133a and miR-133b downregulated at CHF stageLVH and CHF in salt-sensitive Dahl rats[181]
EstrogenEstrogen replacement strongly decreased IGF-1 protein level in muscles at 1 wkOvariectomized rat skeletal muscle[233]
Multiple targetsmiR-133a upregulated in BTBR micePancreatic islets, adipose tissue, and liver from diabetes-resistant (B6) and diabetes-susceptible (BTBR) mice[234]
Augmentation of adipocyte differentiation by norepinephrine does not alter myomiR levelsmiRNAs miR-1, miR-133a and miR-206 specifically expressed both in brown pre- and mature adipocytesmiRNAs miR-1, miR-133a and miR-206 were absent from white adipocytesMouse brown adipocytes[235]
Foxl2miR-133b targets Foxl2;Foxl2 regulates StAR and CYP19A1 transcriptionallyEstradiol production in ovarian granulosa cells[236]
miR-133b inhibits Foxl2 binding to StAR and CYP19A1 promoter sequences
Exosome release and cell to cell transfer
Exosome-mediated transfer of miR-133b from MSCs to brain astrocytesmiR-133b transfer from multipotent mesenchymal stromal cells to neural cellsmiR-133b upregulatedMouse multipotent MSCs to neural cells[47]
Cell to cell transfer of exosome-enriched extracellular particlesmir-133b promotes neural plasticity and recovery of function after stroke induced damagemiR-133b upregulatedRat multipotent MSCs via transfer of exosome-enriched extracellular particles[72]
Transplanted stem cells
MSCs expressing miR-1Upregulated miR-1Increased rate of recovery, enhanced survival of transplanted MSCs and cardiomyogenic differentiationExperimental ligation of the mouse left coronary artery to model myocardial infarction[237]
Knockdown of Hes-1, member of Notch pathwayUpregulated miR-1 promotes the differentiation of MSCs into cardiac lineageRole in survival of transplanted MSCs and cardiomyogenic differentiationMouse MSCs[238]
Notch signalling and cardiomyocyte markers, Nkx2.5, GATA-4, cTnT, and Cx43MSCs expressing exogenous miR-1Mouse MSCs[238]
Tissue inflammation
Selective release of miRs during inflammation into serummiR-133 selectively releasedReview[239]
Inflammation and cancerMicroRNA, free radical, cytokine and p53 pathwaysReview[240]
Immunological switch which shapes tissue responsesTWEAK/Fn14 pathwayReview[241]
Tumor biologyHMOX1Review[242]
GM-CSFDirect supression of GM-CSF expression by miR-133Elevated expression of miR-133a/-133b during oxidative stressMouse alveolar epithelial cells during oxidative stress[82]
PI3K/Akt and IGF-1 pathwaysActivation of PI3K/Akt and IGF-1 pathway activitiesDownregulation of miR-133a (and other miRs) by AOM/DSS induced chronic inflammationMouse model: AOM/DSS-induced colitis-associated gastro-intestinal cancer[83]
CTGF, SMA, and COL1A1Increased expression of CTGF, SMA and COL1A1, which are miR-133b targetsStrong downregulation of miR-133b (and other miRs)TGF-β treated rabbit corneal fibroblasts; Recovering mouse cornea after laser ablation,[70]
IL-10 and TGF-βExogenous IL-10 and TGF-β induces miR-133b expressionUpregulation of miR-133bHuman tolerogenic dendritic cells during maturation[79]
IL-17-producing T-cellsUpregulation of Il17a/f gene expressionmiR-133b/-206 cistron transcription occurs along with nearby Il17a/f gene expressionImmunocompetent mouse Th17 cells[80]
NLRP3 inflammasome which processes IL-1β by caspase-1 cleavagemiR-133a-1 suppresses activation of inflammasomes via suppression of expression of mitochondrial UCP2miR-133a-1 overexpression in cells increases caspase-1 p10 and IL-1β p17 cleavage,Differentiated mouse THP1 cells[81]
Concanavalin A-induced fulminant hepatitismiR-133a is the most strongly differentially upregulated miRMouse liver following ConA injection[243]
Infection/immune response to influenza virus (H1N2)miR-206 expressionExperimental influenza infection in pig lung[244]
HIF-1α, and its regulator Four-and-a-half LIM (Lin-11, Isl-1 and Mec-3) domain 1 (Fhl-1)Downregulation of miR-206 and upregulated HIF-1α and Fhl-1 in hypoxic lung tissue and PASMCsmiR-206 targets HIF-1α directly. Hypoxia-induced down-regulation of miR-206 promotes PH in PASMCsHypoxia-induced PH in hypoxic rat model in cultured hypoxic PASMCs[245]
miR-206/NR4A2/NFKB1;NFKB1 stimulates inflammatory cytokines (IL6, IL1B, CCL5)Liposaccharides induce miR-206 expression which targets NR4A2 downregulation, which in turn allows upregulation of NFKB1 activityAstrocyte-associated inflammation during recovery from chronic central nervous system injury[246]
Indirectly: inflammatory cytokines (IL6, IL1B, CCL5)
Cellular factors influencing myomir expression/activity
miR-1/miR-133a
Skeletal muscle
Positive regulatorNegative regulatorRegulated target miRTissue/cellRef.
Myogenin, MyoDUpregulates miR-1-1 and miR-133a-2Primary human myoblasts; C2C12 cells[11]
Upregulates miR-1-2 and miR-133a-1
SRF, MyoD and MEF2Upregulates miR-1-2Muscle somites[30]
MEF2Upregulates miR-1 and miR-133aSkeletal muscle[9]
KSRP (part of Drosha and Dicer complexes)miR-206 binds 3’-UTR of KSRP and inhibits its expressionKSRP upregulates miR-1 expressionSkeletal muscle[35,37]
RNA-binding protein LIN28LIN28 upregulates miR-1 expression; LIN28 promotes pre-miR-1 uridylation by ZCCHC11 (TUT4)Cardiac muscle of patients with muscular dystrophy[36]
MBNL1MBNL1 downregulates miR-1 expression; MBNL1 binds to UGC motif in the loop of pre-miR-1 and competes for the binding of LIN28; MBNL1 blocks DICER processing of pre-miR-1Cardiac muscle of patients with muscular dystrophy[36]
CX43 and CACNA1C calcium channelCX43 and CACNA1C both increased in both DM1-/DM2-affected hearts, contributing to the cardiac dysfunctionsCX43 and CACNA1C are direct targets of miR-1 repressionCardiac muscle of patients with muscular dystrophy;[36]
CACNA1C and CX43 encode the main calcium- and gap-junction channels in heart
Utrophin AmiR-206 and KSRP are negative regulators of utrophin AOverexpression of miR-206 promotes the upregulation of utrophin A, via the downregulation of KSRPNormal and dystrophic muscle cells;[37]
miR-206 can switch between (1) direct repression of utrophin A expression, and (2) activation of its expression by decreasing KSRP, allowing close regulation
MyostatinDownregulates miR-1, miR-133a, miR-133b, miR-206Mouse (35 d) pectoralis skeletal muscle[29]
SRFDownregulates miRs-133aSkeletal muscle[1,3]
Prmt5 and Prmt4Upregulates myomiR expression during differentiationMouse skeletal muscle[247]
Smooth muscle
Sp-1 transcription factorpERK1/2Upregulates miR-133(a)VSMCs[248]
Brg1Upregulates miR-133 (ChIP complex with SRF)Smooth muscle[249]
Cardiac muscle
GATA4, Nkx2.5, Myocardin, SRFUpregulates miR-1 and miR-133aDifferentiating cardiac muscle[5]
SRF plus MyocardinUpregulates miR-1-1 and miR-1-2Cardiomycetes[30]
CalcineurinDownregulates miR-133aHypertrophic cardiac muscle[203]
miR-206/ miR-133b
Skeletal muscle
Mrf5Upregulates miR-1, miR-206Skeletal muscle[171]
Myogenin, MyoDUpregulates miR-206Primary human myoblasts; C2C12 cells[11]
MyoDUpregulates linc MD1 (encodes miR-133b)Differentiating myoblasts[11, 38]
Binds to (E-box) enhancer of miR-206, miR-133bskeletal muscle (mouse)[12,40]
Upregulates miR-206/miR-133bDifferentiated human foetal skeletal muscle cells[250]
FGF2 allows upregulation of Sp1/Cyclin D1Downregulates p38-mediated miR-1/133 expressionRegenerating rat skeletal muscle[212]
MyostatinDownregulates miR-133a, mir-133b, miR-1, and miR-206Mouse (35 d) pectoralis skeletal muscle[29]
TWEAK downregulates myoD and MEF2cDownregulates miR-1-1 and miR-133Degenerating/wasting skeletal muscle[59]
HMOX1 downregulates MyoD and myogeninDownregulates all myomiRsInflamed skeletal muscle[60]
L-Thyroxine treatmentDownregulation of pri-miR-206 and pri-miR-133bHuman skeletal muscle[230]
No effect on miR-1/miR-133a pairs
Smooth muscle
p-ERKActivated extracellular signal-regulated kinase p-ERK inversely correlated with VSMC growthDownregulates miR-133 expressionVSMCs[248]
Other tissues
MyogeninBinds miR-206 enhancer (ChIP)Fibroblast cell line:[40]
IGF-I signallingUpregulates miR-133bMouse Adipose derived stem cells[71]
L-Thyroxine deficiencyUpregulated Col5a3Strong upregulation of miR-133a and -133bHypothyroid mouse liver[232]
Downregulated Slc17a8, Gp2, Phlda1, Klk1d3, Klk1 and Dmbt1Strong upregulation of miRs -1, -206
Upregulated Vldlr and Akr1c19, and downregulated Upp2, Gdp2, Mup1, Nrp1, and Serpini2
L-Thyroxine treatmentPre-miR-206 and Pre-miR-133b down-regulatedUpregulation of Gdp2 andMup1Hypothyroid mouse liver in vivo, and in vitro mouse hepatocyte AML12 cells[232]
PA2G4, mps1, cdc37, cx43, cldn5; cx43 is a miR-133 targetUpregulation of cell cycle factors mps1, cdc37, and PA2G4, and cell junction components cx43 and cldn5Suppression of miR-133a1 stimulates cardiac cell proliferationRegeneration of damaged Zebrafish cardiac muscle, associated with reduced miR-133a1[167]
FgfUpregulated FgfDownregulates miR-133Zebrafish regenerating fin blastema[67]
SHP (nuclear receptor)Downregulation of miR-206 in nuclear receptor SHP(-/-) miceSHP(-/-) mice strain, mouse liver[251]
AP1 transcription factor complexAP1 induced miR-206 promoter transactivity and expression; this is repressed by YY1ChIP analysis shows physical association of AP1 (c-Jun) and YY1 with miR-206 promoterSHP(-/-) nuclear receptor mice strain, mouse liver[251]
NR3B3YY1 promoter transactivated by ERRgamma; this inhibited by SHP (NROB2)Nuclear receptor ERRgamma (NR3B3) binding site on the YY1 promoterMouse liver[251]
Novel cascade "dual inhibitory" mechanism governing miR-206 gene transcription by SHP(1) SHP inhibition of ERRgamma leads to decreased YY1 expression(2) Derepression of YY1 on AP1 activity, leads to activation of miR-206Mouse liver[251]
Il17a/f locusmiR-133b and miR-206 expressionCoregulated with IL-17 productionαβ and γδ T cells[80]
Table 3 The deregulated expression of the canonical myomiRs in different cancers
MiR-1MiR-206MiR-133aMiR-133bCancer typeRef.
++Progressive bladder cancer (TCC)[122]
--Bladder cancer (TCC)[93,156]
---Bladder cancer (TCC)[92,144]
----Bladder cancer[127]1
----Bladder cancer[128]1
---Muscle-invasive bladder cancer[92]
-Proliferating breast cancer[108]
-ERα-positive breast cancer[109]
--Breast cancer[95]
+Progressive cervical carcinoma[117,121]
--Chordoma[90]
+Colon cancer[116]
-Colon cancer[101]
-CRC[104,136]
-CRC[86,87]
+/-Liver metastasis compared to primary CRC[118]
--Colon cancer[95]
--Progressive GIST[281]
-HNSCC[143]
-HNSCC[114]
-EEC[110]
-ESCC[132]
---ESCC[94]
-Laryngeal SCC cells[113]
--MSSCC[137]
-TSCC[111,112]
-HCC[98]
++Liver cancer[95]
-Lung cancers: (NSCLC, adenocarcinomas, lung SCC, large cell carcinoma, and bronchoalveolar cell carcinoma)[100]
-High metastasis lung tumors[107]
-Lung SCC tissue; lung-SCC cell lines[115]
-NSCLC[85,105]
--Lung adenocarcinomas; NSCLC cells[99]
--Lung carcinomas[85,115,131]
--Lung cancer[95]
--Lymphoma[95]
+AML[120,151]
+Multiple myeloma[152]
+Progressive prostate cancer[123]
-(-)-Prostate cancer[102]
--Prostate cancer[95]
--Recurrent prostate cancer compared to non-recurrent cancer[96]
--Hormone-insensitive prostate cancer cells[102]
---Osteosarcoma[91]
--Ovarian cancer[95]
-PDAC[103]
-RCC[138]
--Rhabdomyosarcoma[88,89]
--Testicular cancer[95]
Table 4 Targets and pathways influenced by the downregulated myomiRs in different cancers
Downregulated miR-1
miR-1 downregulation influences multiple cancer-related pathway processes, and promotes cell proliferation and motilityEpigenetic promoter hypermethylation reduces miR-1/-133a expression in (a subset of) human prostate tumorsReduced miR-1, miR-133a (and miR-206)Human prostate tumors[102]
Actin filament network-associated genes: FN1, LASP1, XPO6, CLCN3 and G6PD; Cell cycle and DNA damage control genes: BRCA1, CHK1, MCM7; Histone acetylation: HDAC4; Oncogenes: NOTCH3 and PTMAmiR-1 downregulation associated with upregulation of multiple cancer-related pathway processesReduced miR-1;Human prostate cell lines, LNCaP, 22Rv1, PC-3 and RWPE-1[102]
Exogenous introduction of miR-1 or miR-206 caused similar inhibition of various cancer-related pathway genes
HSPB1HSPB1 restores oncogenic pathways in prostate cancer cellsDownregulates miR-1 expressionProgressive prostate cancer PCa cells[252]
XPO6 and TWF1 (PTK9)Inverse expression between miR-1, XPO6 and TWF1 proteins in prostate cancer cell linesDownregulated miR-1 expressionProstate cancer cell cultures[253]
CCND2, CXCR4, and SDF-1αInverse expression between miR-1 and CXCR4 and SDF-1α protein levels in thyroid carcinomasStrongly downregulated miR-1 expression in thyroid adenomas and carcinomasThyroid adenomas and carcinomas[254]
METMET upregulatedReduced miR-1Colon cancer[101]
Reduced miR-1, -133bColon cancer[87]
MET, Pim-1 (Ser/Thr kinase), FoxP1 and HDAC4miR-1 downregulated,MET, Pim-1, FoxP1 and HDAC4 are often upregulated in lung cancerNSCLC tissue and A549 cell line[100]
miR-1 targets MET, Pim-1, and may regulate FoxP1 and HDAC4
Fibronectin1Fibronectin1 upregulatedmiR-1 downregulatedLaryngeal SCC Hep2 cells[255]
Met, Twf1 and Ets1 and Bag4Met, Twf1 and Ets1 and Bag4 activities upregulatedmiR-1 downregulatedMouse cutaneous squamous cell carcinomas[256]
Mediator complex subunit 1 (Med1) and 31 (Med31)Med1 and Med31 activation result in increased Met activityReduced miR-1; miR-1 targets Med 1 and Med 31Osteosarcoma[257]
NOTCH3 upregulates Asef expression, activating the Asef promoter, enhancing cell migrationNOTCH3 upregulatedReduced miR-1; miR-1 targets NOTCH3Colorectal tumor cells[258]
Overexpression of PIK3CA correlates with low miR-1 expression in NSCLC tissues71% of NSCLC samples had high PIK3CA expression69% of NSCLC samples had low miR-1 expressionPredictors of lymph node metastasis in NSCLC tissues[259]
SLUG expression downregulated by miR-1Transcriptional repressor of E-cadherin, or an inducer of epithelial-to-mesenchymal transitionOverexpression of miR-1 induces morphological change from a mesenchymal to an epithelial characterNSCLC A549 cell line[260]
SLUG expression high in chordoma tissuemiR-1 inhibited cell proliferation both time- and dose-dependently in chordomaTransfection of MiR-1 inhibited Slug expressionmR-1 transfected chordoma cells[261]
Slug overexpressed in advanced chordoma tissues and chordoma cells
MET expression high in chordoma tissuemiR-1 downregulated 97% of chordoma samplesMiR-1 directly targets METDecreasing miR-1 expression levels correlated with severity of clinical prognosis[262]
SRSF9/SRp30cExogenous upregulation of miR-1 expressionNovel apoptosis pathway involving SRSF9/SRp30c mediates tumor suppressionBladder cancer (TCC) cells[263]
ANXA2 is essential for glioblastoma growth and invasionANXA2 is highly abundant protein in glioblastoma-derived extracellular vesiclesmiR-1 directly targets ANXA2;Human Glioblastoma cells; miR-1 orchestrates glioblastoma extracellular vesicle function[264]
Reduced miR-1 in glioblastoma
EDN1miR-1 downregulated in gastric cancermiR-1 causes ET-1 silencing in gastric cancer cell linesGastric cancer tissue compared with adjacent normal tissue[265]
EDN1Elevated expression of EDN1 and reduced miR-1 levelmiR-1 directly targets EDN1Human liver cancer tissues[266, 267]
Overexpressed EDN1Enhanced in vitro cell proliferation and cell migration. Upregulation of several cell cycle/proliferation- and migration-specific genesUpregulated UPR pathway mediators, spliced XBP1, ATF6, IRE1, and PERK at both RNA and protein levels293T cells[267]
AKT inhibitor diminished the unfolded protein response and eliminated EDN1-induced cell migrationEDN1 effects act via activation of the AKT pathwayResults to enhance the UPR and subsequently activate the expression of downstream genes293T cells[267]
Edn1Induced steatosis, fibrosis, glycogen accumulation, bile duct dilation, hyperplasia, and HCCLiver-specific edn1 expressionTransgenic Zebrafish liver[267]
API5API5 expression upregulated thus inhibiting apoptosismiR-1 expression downregulatedHuman liver cancer tissues[268]
Apoptosis activated, API5 reducedOverexpression of miR-1HepG2 liver cancer cells
Phosphorylation of ERK and AKT; LASP1Overexpression of miR-1 inhibits phosphorylation of ERK and AKT and reverses EMT process via inhibition of MAPK and PI3K/AKT pathwaysMAPK and PI3K/AKT pathwaysTransgenic miR-1 expressing CRC cell lines[269]
LASP1 expression upregulatedUpregulated LASP1 stimulates EMT resulting in cell proliferation and migrationmiR-1 downregulatedColorectal tumor tissue[269]
PIK3CAIncreased expression of PIK3CADownregulated miR-1 expression in lung cancerNSCLC tissue with poor patient prognosis[259]
PIK3CA indirectly regulating pAKT and survivin proteinsOverexpressed miR-1 downregulated PIK3CA causing reduced pAKT and survivin proteinsExogenously overexpressed miR-1 targets PIK3CA directly.NSCLC A549 cell line[270]
Signalling pathways such as TGF-β, ErbB3, WNT and VEGFA, and cell motility or adhesionEctopic expression of miR-1 and miR-145 downregulates VEGFA and AXL, respectivelyHighly downregulated expression of miR-1, miR-133, miR-143 and miR-145 in gall bladder cancerGall bladder tumor samples and GBC NOZ cell line[271]
lncRNA UCA1Lnc RNA UCA1 upregulated in bladder cancer (TCC);Downregulated miR-1 expression in bladder cancer (TCC); miR-1 targets lnc RNA UCA1 for downregulationHuman bladder cancer (TCC) tissue[156]
Inverse relationship between miR-1 and lnc UCA1
Downregulated miR-133a
MoesinMoesin upregulatedReduced miR-133aHNSCC[143]
ARPC5ARPC5 upregulatedReduced miR-133aHNSCC[272]
ARPC5 and GSTP1ARPC5 and GSTP1 upregulatedReduced miR-133a (and miR-206)Lung carcinoma[115]
IGF-1R, TGFBR1, and EGFR are downregulatedRestoration of ectopic-expression of miR-133a in NSCLC suppresses metastatic capacitymiR-133a inhibits cell invasiveness and cell growth via suppression of IGF-1R, TGFBR1 and EGFRNSCLCs[131]
Low expression of miR-133a is characteristic of pancreas tissueReduced miR-133aPDAC[103]
CDC42CDC42 upregulated causing downstream activation of PAKsmiR-133 downregulatedGastric cancer tissues[273]
GSTP1Upregulated GSTP1Downregulation of miR-133a in cancerBladder cancer (TCC) cell lines[144]
Enforced downregulation of GSTP1 inhibits cell proliferation and growth;Enforced upregulation of miR-133a and miR-133b induces cell apoptosis
GSTP1 in cancer specimensGSTP1 upregulatedReduced miR-133aBladder cancer (TCC) tissue[144]
Actin-binding protein, FSCN1Upregulated FSCN1;Downregulation of miR-133a;Bladder cancer (TCC) tissue[274]
Enforced downregulation of FSCN1 inhibits cell proliferation, migration and invasionForced UP exp of miR-133a inhibits cell proliferation, migration and invasion
EGFR/AKT signalling pathwayUpregulated EGFR;Downregulated miR-133a;Human MCF-7 and MDA-MB-231 breast cancer cell lines[275]
Activated pAkt-1Enforced expression of miR-133a inhibits EGRF translation; causes inhibition of Akt protein phosphorylation and its nuclear translocation
Bcl-xL and Mcl-1 expressionUpregulated Bcl-xL and Mcl-1Downregulated miR-133a correlated with tumor progression and poor patient prognosis;Primary human osteosarcoma tissues;[276]
Osteosarcoma cell lines
E3 ubiquitin protein ligaseDownregulation of p21 and p53 proteinsDownregulated miR-133aPrimary CRC tissues[277]
Enhanced sensitivity to doxorubicin and oxaliplatinEnhancing apoptosis and inhibited cell proliferationEctopic upregulation of miR-133aCRC cell lines[277]
LASP1 upregulatedmiR-133a expression downregulatedmiR-133a targets LASP1CRC tissues and cell lines[278]
FTL protein upregulatedmiR-133a expression downregulatedmiR-133a targets downregulation of FTL proteinPatient breast cancer tissue[279]
Increased sensitivity to chemotherapeutic drugs doxorubicin and cisplatinExogenous upregulation of miR-133a expressionDownregulation of FTL proteinHuman MCF-7 breast cancer cells[279]
Poor survival during breast cancer; upregulated FSCN1Loss of miR-133a expressionFSCN1 is a direct target gene of miR-133aBreast cancer tissue[280]
FSCN1 downregulatedRestoration of miR-133a expressionInhibited breast cancer cell growth and invasionBreast cancer cell line[280]
lncRNA Malat1/Srf/miR-133 regulatory loopMalat1 transcript has a functional miR-133 target site, miR-133 acts as a competing endogenous RNA, regulating Malat1 levelsIn vitro depletion of Malat1 in C2C12 cells reduces Srf activity, Srf is an enhancer of miR-133 expression; feed-back regulation loop involving miR-133Mouse myoblast C2C12 cells[164]
lncRNA MALAT1MALAT1 is overexpressed in 46% of ESCC tissues, primarily in high-stage tumors, high expression correlates with lymph node metastasisIn vitro depletion of MALAT1 suppresses tumor cell proliferation, cell migration and invasion; G2/M phase arrest was induced and the ratio of apoptotic cells increasedHuman ESCC[162]
WIF1/lncRNA MALAT1WIF1 (strong tumor suppressor) is systematically downregulated in glioblastomaWIF1 down regulation correlates with strong upregulation of MALAT1. In vitro depletion of MALAT1 suppresses tumor cell proliferationGlioblastoma[163]
Downregulated miR-133b
Fascin-1 mRNAFSCN1 upregulatedReduced miR-133bHigh-grade GIST tissue[281]
BCL-2 family (MCL-1 and BCL2L2)MCL-1 and BCL2L2 upregulatedReduced miR-133bLung cancer[85]
FAIM antiapoptotic protein and GSTP1miR-133b directly targets FAIM and GSTP1Downregulated miR-133bmiR-133b expression significantly downregulated in 75% of prostate cancer tumor specimens[282]
Gli1Gli1 upregulatedGli1 inversely correlated with downregulated expression of miR-133bGastric cancer[283]
Bcl-w and Akt1Bcl-w and Akt1 proteins overexpressed significantlymiR-133b significantly downregulatedBladder cancer tissues[284]
miR-133b downregulated in tumors compared to surrounding tissueGastric and esophageal adenocarcinomas[285]
Endometrial sarcoma, leiomyosarcoma, and mixed epithelial-mesenchymal tumors[286]
Downregulated miR-206
Notch3/ miR-206Downregulated Notch3, blocking of the anti-apoptotic activity of Notch3Forced expression of miR-206 strongly induced apoptotic cell death via; also inhibited cell migration and focus formationHeLa cells[287]
MetUpregulated MetmiR-206 downregulatedHuman rhabdomyosarcoma[288]
HGFRUpregulated HGFRmiR-206 downregulatedHuman breast cancer cells[289]
KLF4Upregulated KLF4miR-206 downregulatedRK3E breast epithelium cells[108]
KLF4; RAS-ERK signallingUpregulated KLF4 promotes RAS-ERK signallingmiR-206 downregulatedTNBC cells[290]
Endogenous KLF4 binds the promoter regions stimulates expression of miR-206
RASA1 and SPRED1miR-206 inhibits translation of the RAS pathway suppressors RASA1 and SPRED1Suppression of RASA1 or SPRED1 increased levels of GTP-bound, wild-type RAS and activated ERK 1/2
VEGFVEGF upregulated in Laryngeal SCC tissuesMiR-206 strongly downregulated in LSCC tissuesLaryngeal SCC cancer tissue and cells[113]
VEGFVEGF upregulated in ccRCC tissuesMiR-206 strongly downregulated in ccRCC tissuesccRCC tissues assayed by Deep Sequencing[291]
Cdc42, MMP-2 and MMP-9Upregulated Cdc42, MMP-2 and MMP-9miR-206 downregulatedHuman breast cancer tissues[292]
ERαmiR-206 directly targets ERα 3'-untranslated regionMiR-206 inhibited by ERα agonists, indicating a mutually (double) inhibitory feedback loop;Estrogen stimulated breast cancer cell lines[293]
miR-206 downregulated[109]
Upregulated ERαMCF-7 breast cancer cells[294]
ERαUpregulated ERαmiR-206 downregulatedEEC tissue[110]
K-RasK-Ras is direct target of miR-206;Low miR-206 potentiates metastases, and shorter overall survivalOSCC tissue samples and cell lines[295]
MiR-206 expression significantly downregulated and k-Ras upregulated on OSCC tissues
MiR-206Enforced upregulated of miR-206 attenuated cell proliferation, increased apoptosis and inhibited cell migration and invasionMiR-206 strongly downregulated in lung cancer tissuesLung cancer - tissues and cell lines[107]
EGFR/MAPK signalling switches MCF-7 breast cancer cells from ERα-positive, Luminal-A phenotype to ERα-negative, basal-like phenotypeEGFR signalling represses estrogenic responses in MCF-7 cells by enhancing miR-206 activitymiR-206 downregulates steroid receptor co-activators SRC-1 and SRC-3 and GATA-3 transcription factor, directlyMCF-7 breast cancer cells[296]
Elevated miR-206 reduces cell proliferation, enhances apoptosis, and reduces numerous estrogen-responsive genes
Greater lymph node metastasis, venous invasion, and at a more advanced stagemiR-206 expression strongly downregulatedCorrelates with tumor progressionHuman gastric cancer tissue[297]
CCND2miR-206 expression strongly downregulatedCorrelates with upregulation of CCND2 and cancer progressionHuman breast cancer[298]
Human gastric cancer[299]
METmiR-206 expression strongly downregulatedUpregulation of METPapillary thyroid carcinoma[300]
Prognostic signature of metastatic colorectal cancermiR-206 expression strongly downregulatedPrognostic signature of metastases: miRs 21, 135a, 335, 206 and let-7aMetastatic CRC[301]
Notch3, Hes1, Bcl-2 and MMP-9;Exogenous upregulation of miR-206 expression;Notch3, Hes1, Bcl-2 and MMP-9 downregulated at both mRNA and protein level;Human HHC Hep2 cells.[302]
p57, Bax and caspase-3miR-206 is a potent tumor supressorp57 and Bax upregulated, and cleaved caspase-3 protein upregulatedReduced apoptosis, and cell migration in HepG2 cells overexpressing miR-206
STC2, HDAC4, KLF4, IGF1R, FRS2, SFRP1, BCL2, BDNF and K-rasExogenous upregulation of miR-206 expression;STC2, HDAC4, KLF4, IGF1R, FRS2, SFRP1, BCL2, BDNF, and K-ras downregulated strongly in SCG-7901 cells overexpressing miR-206Gastric carcinoma SCG-7901 cells[303]
miR-206 is a potent tumor supressorReduced apoptosis, and cell migration in SCG-7901 cells overexpressing miR-206
Cyclin C, CCND1 and CDK4Cyclin C, CCND1 and CDK4 upregulated in melanoma tissue;hsa-miR-206 downregulated in melanoma tissueHuman melanoma cancer tissue, and cell lines[304]
Exogenous upregulation of miR-206 expression reduced growth and migration/invasion of several melanoma cell lines;Overexpression of miR-206 in melanoma cells strongly downregulated cyclin C, CCND1 and CDK4
G1 arrest in melanoma cells
Coronin, actin-binding proteinSilencing of coronin expression reduced tumor cell migration and altered the cellular actin skeleton and cell morphology, but did not effect cell proliferationDownregulated miR-206 allowed upregulation of coronin, a direct target;TNBC cell lines[305]
Upregulated miR-206 reduced TNBC cell migration and cell proliferation
RNA binding protein DEAD-END (DND1), DNA cytosine deaminase (AICDA), and APOBEC3DND1 blocks miRNA interaction with 3'-UTR of specific mRNAs, restores protein expression; APOBEC3G binds DND1 counteracts repression and restores miRNA activityAPOBEC3G blocks DND1 to restore miR-206 inhibition of CX43 translationMouse cells[306]
Advanced clinical stage, T classification, metastasis and poor histological differentiationSignificant association with decreased miR-206 expressionPaired human osteosarcoma and normal adjacent tissues[307]
Ellagic acid inhibits E2-induced mammary tumorigenesisReverses the downregulation of miR-206ACI model rat mammary tissue[308]
Actin-like 6A (BAF53a), a subunit of the SWI/SNF chromatin remodeling complexElevated BAF53aDownregulation of miR-206Primary rhabdomyosarcoma tumors[309]
Actin-like 6A (BAF53a)BAF53a transcript is significantly higher in primary rhabdomyosarcomas than in normal muscleRestoration of miR-206 expression downregulated BAF53a, which inhibits proliferation and anchorage independent growth;Primary rhabdomyosarcoma tumors[309]
BAF53a and is a direct target of miR-206
Wnt and transcription factors Tbx3 and Lef1Exogenous upregulation of miR-206 expressionInhibition of Wnt, Tbx3 and Lef1 activitiesEstrogen receptor alpha (ER-α)-positive human breast cancer; developing mammary buds[310]
ANXA2 and KRASStimulation of KRAS activity then induces NFKB1 expression;Downregulated miR-206 in PDACPDAC tissues and cell lines[311]
Induces NFKB1Increased KRAS results in stimulation of cytokines CXCR2, CXCL1, CCL2, as well as CSF2 (GM-CSF) and VEGFCIncreased cell cycle progression, cell proliferation, migration and invasion
Downregulated miR-1 and miR-133a
PNPPNP upregulatedReduced miR-1, -133aProstate cancer[97]
TAGLN2TAGLN2 upregulatedReduced miR-1, -133aRCC[136]
TAGLN2 and PNPTAGLN2 and PNP upregulatedReduced miR-1, -133aMSSCC[137]
PTMA and PNPPTMA and PNP upregulatedReduced miR-1, -133aBladder cancer (TCC)[312]
LASP1LASP1 upregulatedReduced miR-1, 133a, (and miR-218)Bladder cancer (TCC)[313]
Forced expression of each miR decreased LASP1 in cell lines
DNA methylation regulates miR-1-1 and miR-133a-2 cistron expressionInverse correlation with TAGLN2 levelsCpG islands upstream of miR-1-133a hypermethylatedColorectal carcinoma tissue and liver cancer tissue[314]
Downregulated miR-1 and miR-133b
miR-1 and mir-133b have sufficient power to distinguish recurrent specimens from non-recurrent prostate cancermiR-1 and mir-133b are significantly downregulated in recurrent prostate cancer tissue specimensRecurrent prostate cancer tissue[96]
Downregulated miR-1 and miR-206
NRF2 upregulatedDownregulated miR-1 and miR-206 expressionUpregulated expression of NRF2 induces increased expression HDAC4Primary lung adenocarcinoma; DU145 human prostate cancer cell line[99]
Loss of NRF2Decreased expression histone deacetylase (HDAC4)Results in increased expression of miR-1 and miR-206; which inhibits PPP expression; Reduced PPP acts as a regulatory feedback loop stimulates HDAC4 expressionA549 human NSCLC cell line[99]
c-Metc-Met upregulatedmiR-1 and -206 downregulatedHuman rhabdomyosarcoma[89]
ARPC5 and GSTP1ARPC5 and GSTP1 upregulatedReduced miR-133a (and miR-206)Lung SCC cell lines[115]
Downregulated miR-133a and miR-133b
PKM2PKM2 upregulatedDownregulated miR-133a, -133bTSCC[111]
FSCN1FSCN1 upregulatedDownregulated miR-133a, -133b, (miR-145)ESCC[132]
miR-133a, miR-133b downregulatedESCC[94]
KRT7KRT7 upregulatedDownregulated (miR-133a and miR-133b)Bladder cancer (TCC) and in vitro in BC KK47 cells[315]
Downregulated miR-1, miR-206 and miR-133
myomiRsPatient to patient variation in the up or down regulation of miR expression in both tumor and matched normal tissuesIn tumors strong down regulation of highly expressed miR-1/133a; (downregulation of weakly expressed miR-206/-133b)Bladder cancer assayed by deep sequencing[315]
Candidate tumor suppressor miRNAs in RCCEach of miR-206, miR-1, miR-133b strongly downregulatedRestored expression strongly inhibited cancer cell proliferation,RCC[316]
Shorter overall survival and disease-free survivalCorrelated with increased downregulated of miR-133b and/or miR-206Both miR-133b and miR-206 significantly downregulatedOsteosarcoma tissues[317]
Cell invasion and metastasismiR-1, miR-133a, miR-133b downregulatedmiR-133a, miR-133b involved in invasion and metastasisESCC[94]
Table 5 Targets and pathways influenced by the upregulated myomiRs in different cancers
Factor(s)RegulationRegulatorTissue/cellRef.
Upregulated miR-133b
Activated p-ERK, pAKT1 cause in vitro proliferation of cervical cancer cell lines, and promote in vivo tumorigenesis and metastasisDownregulation of MST2, CDC42, RHOAUpregulated miR-133bHuman cervical carcinoma tissue compared to surrounding normal cervical tissue[121]
Decreased patient survivalUpregulated miR-133bProgression bladder cancer[122]
Androgen receptormiR-133b directly represses CDC2L5, PTPRK, RB1CC1, CPNE3miR-133b directly upregulated by ARHormone-sensitive human prostate cancer (LNCaP) cells stimulated by androgen[123]
Activativated neuroendocrine neoplasia proliferationMutation in von Hippel-Lindau tumor suppressor, E3 ubiquitin protein ligase gene (VHL)Upregulated miR-133b expression in VHL- deficient pheochromocytomaHuman pheochromocytoma (PCCs) and paraganglioma (PGLs) neuroendocrine neoplasias[318]
Upregulated miR-206
Cell reprogramming factor KLF4KLF4 downregulated in colon cancer tissue, associated with increased miR-206miR-206 strongly upregulated in colon cancer tissuesHuman colon cancer tissue[116]
Upregulated miR-1 and miR-133
Decreased survival of R172 IDH2-mutated subset of CN-AML patients, increases resistance to chemotherapyDistinctive gene and microRNA expression profiles accurately predicted R172 IDH2 mutationsUpregulated expression of miR-1 and miR-133De novo CN-AML patient bone marrow and blood samples[151]
EVI1 increases aggressive cancer growthEVI1 expression upregulated in established patient samplesUpregulated expression of miR-1-2 and miR-133-a-1EVI1 expressing AML subset of patients[120]
ChIP assays show EVI1 binds to miR-1-2 gene promoter directly
CCND2miR-1 and miR-133a were specifically overexpressed in the cases with t(14;16) translocation, correlates with down-regulated CCND2 expressionUpregulated miR-1 and miR-133-aMultiple myeloma[152]
Secreted myomiRs
miRs selectively released into serum (within exosome microparticles)miR-1, miR-133a, and miR-133b selectively releasedHuman breast cancer[319]
Circulating microRNATumor-derived exosomesHuman non-small-cell lung cancer[320]
Table 6 Validated myomiR targets related to cancer progression
Up regulated cell factorDown regulated cell factorCancer typeRef.
Downregulated myomiRs
Downregulated miR-1
Mediator complex subunit 1 (Med1) and 31 (Med31) upregulatedmiR-1 downregulatedOsteosarcoma[257]
Slug expression upregulated; enhanced cell migratory and invasive activitiesmiR-1 downregulatedChordoma[261]
Slug expression upregulated; stimulation of EMT processmiR-1 reduced increasingly with cancer progressionProstate adenocarcinoma[321]
Downregulated miR-133a
ARPC5 upregulated;Downregulated miR-133a (> miR-206)Lung SCC[115]
HNSCC[272]
CAV1 upregulated;miR-133a downregulatedHNSCC[322]
Moesin upregulated;miR-133a downregulatedHNSCC[143]
FSCN1 upregulated;miR-133a/ miR-133b downregulatedESCC[132]
Bladder cancer (TCC)[274]
GSTP1 upregulated;miR-133a downregulatedHNSCC[323]
Bladder cancer (TCC)[144]
Lung SCC[115]
LASP1 upregulated;miR-133a downregulated in 83% of colorectal tumorsColorectal cancer[136]
CAV1 downregulated with miR-133a levels, and is lowest in metastatic cancers;Contrastingly, higher levels of miR-133a correlate with poor prognosis and increased metastasis
FSCN1 upregulated in non-metastatic tumors
LASP1 upregulated;miR-133a downregulatedBladder cancer (TCC)[313]
PKM2 upregulated;miR-133a downregulatedTSCC[111]
Moesin upregulated;miR-133a downregulatedHNSCC[143]
EGFR upregulated;miR-133 downregulatedHormone-sensitive prostate cancer cell lines[324]
Human TERT telomerase catalytic subunit upregulated;miR-133a downregulated[325]
TCF7 transcription factor upregulated;miR-133a downregulated[325]
FSCN1 and MMP14 upregulated;miR-133a downregulatedESCC[326]
Reduced miR-133a expression correlated significantly with advanced clinical stages, poor histological differentiation and lymph node metastasisMarked downregulation of miR-133a in primary EOC tumors and OVCAR-3 cell lineEpithelial ovarian cancer (EOC), and in OVCAR-3 cell line[327]
Downregulated miR-133b
FSCN1 upregulated;miR-133a/-133b downregulatedESCC[132]
FSCN1 mRNA upregulated;miR-133b downregulatedProgressive GIST[281]
BCL2L2 upregulated;miR-133b downregulatedLung cancer[85]
MCL1 upregulated;miR-133b downregulatedLung cancer[85]
MET upregulated;miR-133b downregulatedColorectal cancer[87]
MET protein upregulated;miR-133b downregulatedhigh grade osteosarcoma tumor samples and cell lines[328]
EGFR upregulated;miR-133b downregulatedNSCLC[105]
Multiple cell factors elevated;miR-133b downregulatedProstate cancer[282]
FGFR1 downregulated;miR-133b downregulatedGastric cancer[329]
Gli1 protein downregulated by miR133b, Gli1 target genes, OPN and Zeb2, are indirectly regulatedmiR-133b downregulatedGastric cancer[283]
TAp63 supresses metastasis; downregulation target of miR-133bmiR-133b is a transcription target of TAp63, downregulatedColon cancer cells[330]
Chemokine (C-X-C motif) receptor 4 protein downregulated by miR133b; upregulated in advanced cancermiR-133b downregulatedCRC[331]
TBP-like 1 mRNA and protein are upregulated in CRCmiR-133b downregulated in CRCCRC[332]
Strong additional down regulation of miR-133b aids liver metastatic niche for CRC cellsmiR-133b downregulated 3 × (significant) in liver metastasis compared to primary CRCmiR-133b downregulated in primary CRC compared to surrounding tissueMetastatic cancer arising from primary hCRC[333]
Interestingly, miR-133b is not downregulated significantly in lung metastasis compared to primary CRC
SP1 targeted directly by miR-133, causing reduced expression of MMP-9 and Cyclin D1miR-133a and -133b downregulatedGastric cancer[334]
miR-133b target MMP-9 is upregulatedmiR-133b downregulatedRCC[335]
Downregulated miR-206
ERαERα downregulates miR-206ERα-positive breast cancer;[294]
miR-206 downregulatedDouble feedback loop[109]
miR-206 downregulated[293,336]
ERαmiR-206 downregulatedEEC tissue[110]
SRC-1, SRC-3 and GATA-3 proteins contribute to estrogenic signallingmiR-206 downregulatedERα-positive breast cancer[296]
Signalling contributes to Luminal-A phenotype
KLF4 over expressed in proliferating cells and cancers.miR-206 levels are KLF4 dependent. KLF4 and miR-206 feedback pathway oppositely affect KLF4 protein translationBreast cancer cells and normal cells[108]
FGBP1miR-206 gene double knockdownmiR-206-/- mouse skeletal muscle.[12]
VEGF upregulatedmiR-206 downregulatedLaryngeal SCC cells[113]
VEGF upregulatedmiR-206 downregulatedCRC tumors compared to matched normal tissue; (1DS assay)[337]
miR-206 correlates with negative ER status, negative PR status, and negative HER-2 statusDownregulated miR-206Breast cancer tumor tissue[338]
miR-206 was downregulated in clinical TNBC tumor samples, one of its targets, actin-binding protein coronin was upregulatedDownregulated miR-206 associates with increased metastasis potential in breast cancersHigh metastatic capacity TNBC tumors[305]
Downregulated miR-1 and miR-133a
PNP upregulatedmiR-1/miR-133a downregulatedMSSCC[137]
Prostate cancer[97]
Bladder cancer (TCC)[312]
TAGLN2 upregulated;miR-1/miR-133a downregulatedMSSCC;[137]
RCC[138]
HNSCC[339]
Bladder cancer (TCC)[93]
PTMA upregulatedmiR-1 and miR-133a downregulatedBladder cancer (TCC)[312]
Downregulated miR-1 and miR-206
MET levels correlated inversely with miR-1/206 expressionmiR-1/206 downregulatedUp-regulation of MET in rhabdomyosarcoma[89,288]
HGFR upregulatedmiR-1/206 downregulatedBreast cancer cells[289]
G6PD; PGD; TKT; GPD2 upregulatedmiR-1/206 downregulatedPrimary lung adenocarcinoma[99]
Upregulated myomiRs
Upregulated miR-133b
miR-133bmiR-133b strongly upregulatedMST1, CDC42, RHOA, and DUSP1 downregulatedCervical carcinoma[121]
miR-133bmiR-133b is directly upregulated by ARmiR-133b represses CDC2L5, PTPRK, RB1CC1, and CPNE3PCa prostate cancer cell line[123]
Upregulated miR-206
miR-206Strongly upregulated miR-206KLF4 downregulatedHuman colon cancer tissue[116]
Upregulated miR-1 and miR-133a
miR-1-2 and miR-133-a-1Upregulated miR-1-2 and miR-133-a-1EVI1 (transcriptional activator of miR-1 and miR-133b)AML[120,151]
miR-1 and miR-133-aUpregulated miR-1 and miR-133-aDownregulated CCND2Multiple myeloma[152]
Up-regulation of exogenous myomiR expression in cell lines
Reduced cell proliferationEstrogen receptor alphaOverexpression of miR-206 has an inhibitory effect on cell proliferationERα-positive breast cancer cells over expressing mir-206[289]
miR-133bGSTP1 downregulatedTransgenic miR-133b overexpressionHeLa cervical cancer cells[282]
miR-133bFAIM downregulatedTransgenic miR-133b overexpressionHeLa cervical cancer cells[282]
Apoptosis increasedTNFα-induced cell death is activatedTransgenic miR-133b overexpressionHeLa cervical cancer cells[282]
Increased cell proliferation and migrationDownregulation of MST2Transgenic miR-133b overexpressionCaSki cervical cancer cells[121]
Downregulation of CDC42
Downregulation of RHOA
Increased cell proliferation and migrationIndirect upregulation of p-AKT1 activityTransgenic miR-133b overexpressionCaSki cervical cancer cells[121]
Indirect upregulation of p-ERK activity
RB1CC1 downregulatedExogenous upregulation of miR-133b;miR-133bm promotes cell apoptosis, but suppressed cell proliferation and cell-cycle progression in aggressive PC-3 cellsPC3 prostate cancer cell line[106]
miR-133b directly targets RB1CC1 in LNCaP cellsIn contrast in low-aggression LNCaP cells, miR-133b stimulate cell proliferation and cell-cycle progression, but inhibit apoptosisHormone sensitive prostate cancer LNCaP cell line
Cell proliferation decreased and apoptosis increasedMet, Twf1 and Ets1 and Bag4 activities downregulatedmiR-1 expression is lower in mouse cSCCs compared to normal skinMouse cutaneous squamous cell carcinomas (cSCCs); A5 and B9 cSCCcell lines[256]
Transgenic miR-1 overexpression
Ets1 proto-oncogeneRepression of Ets1 expression inhibited HepG2 cell invasion and migrationTransgenic miR-1 overexpressionHCC HepG2 cells[340]
lncRNA UCA1Knockdown of lnc UCA1 expression phenocopied the effects of upregulation of hsa-miR-1hsa-miR-1 decreased the expression of lnc UCA1 in bladder cancer cells in an Ago2-slicer-dependent mannerHuman bladder cancer (TCC) cells[156]
NOTCH3 signallingmiR-206 had a direct inhibition of NOTCH3 signalling and indirect interaction with other signalling pathways via CDH2 and MMP-9miR-206 upregulation blocks the cell cycle, inhibits cancer cell proliferation and migration and activates cell apoptosisSW480 (plus its metastatic strain) and SW620 colon cancer cell lines[341]
FSCN1miR-133b targets FSCN1 in GC cells; the direct knockdown of FSCN1 can also inhibit GC cell growth and invasionUp regulation of miR-133b in GC cells inhibits cell proliferation, cell migration and invasionmiR-133b is significantly downregulated in GC tissues compared with adjacent normal tissues, as well as in GC cell lines[342]
FSCN1miR-133a targets FSCN1 in CRC cells;Up regulation of miR-133a expression and downregulation of FSCN1 protein expression both suppress colorectal cancer cell invasionmiR-133a is significantly downregulated in some colorectal cancer cell lines, as well as in colorectal cancer tissues compared with the normal adjacent tissues[343]
Overexpression of FSCN1 can reverse the inhibitory effect of miR-133a upregulation, reactivating CRC cell invasion