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
Published online Aug 26, 2015. doi: 10.4331/wjbc.v6.i3.162
Factor(s) | Regulation | Regulator | Tissue/cell | Ref. |
Fish and lower vertebrates: Development and regeneration | ||||
Ttk protein kinase (mps1) | Upregulated mps1: a target of miR-133 | Downregulation of miR-133 by Fgf | Regeneration of Zebrafish caudal fin (appendage) | [68] |
RhoA | Downregulation of RhoA mRNA | Upregulation of miR-133b expression | Regenerating adult zebrafish spinal cord, axon outgrowth | [69] |
RhoA | Downregulation of RhoA protein | Upregulation of miR-1 and miR-133 expression | Zebrafish muscle gene expression and regulation of sarcomeric actin organization | [166] |
Cell cycle factors mps1, cdc37 and PA2G4, and cell junction components cx43 and cldn5 | Upregulated mps1, cdc37, PA2G4, cx43, cldn5 | Downregulated miR-133(a1) stimulates cardiac cell regeneration | Regenerating zebrafish cardiac muscle | [167] |
miR-133b | MiR-133b found in developing somites, little in CNS tissues | Whole zebrafish embryos - normal development | [168] | |
SRF activates muscle specific genes and miRs; | MiR-1 targets HDAC4, promoting myogenesis | In contrast, miR-133a represses SRF, enhancing myoblast proliferation | X. 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; MEF2 | Subunits UNC-29, UCR-63, and MEF2 downregulated | miR-1 upregulated | C. elegans muscle at the neuromuscular junction | [34] |
Mammalian pluripotent cells | ||||
Muscle-specific microRNAs: miR-1 and miR-133a | MiR-1 and miR-133a have opposing functions during differentiation of progenitor cardiac muscles | Muscle-specific | Promotion 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 effects | Dll-1 translationally repressed | miR-1 upregulation, promotes cardiomycete differentiation | Mouse and human ES cell differentiation into muscle | [13] |
SRF-/- EBs reflecting the loss of hematopoietic lineages in the absence of SRF | Early endoderm markers, Afp and Hnf4α: strongly down regulated | Increased miR-1 and miR-133a relieve the block on mesodermal differentiation | Mouse endoderm | [13] |
Blood cell -specific genes, such as Cd53, CxCl4, and Thbs1, dramatically down regulated | Cd53, 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 EBs | Nodal stimulated expression of endoderm markers Afp and Hnf4α in control EBs. Dramatically lower levels in mES(miR-1)- and mES(miR-133a)- EBs | miR-1 or miR-133 can each function as potent repressors of endoderm gene expression | mES cells, that lack either miR-1 or miR-133(a) during differentiation into EBs | [13] |
IGF-1 | IGF-1 signalling and miR-133 co-regulate myoblast differentiation via a feedback loop | IGF-1 upregulates miR-133; | Myogenic differentiation of C2C12 myoblasts; Mouse during development from embryonic to mature skeletal muscle | [24] |
IGF-1R | miR-133 downregulates IGF-1R | |||
IGF-1 | IGF-1 signalling and miR-1 coregulate differentiation of myoblasts via a feedback loop | IGF-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 promoter | Reversine 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 Ezh2 | Reduced expression of myogenin, MyoD, Myf5 and Aurora A and B kinases | |||
FZD7 and FRS2 | miR-1 promotes cardiac differentiation; miR-1 targets FZD7 and FRS2 | Activitation of WNT and signalling cause MCPs differentiation into cardiomyocytes | Mouse and human ES cells | [169] |
miR-206/133b cluster | PAX7 gene expression unchanged; | miR-206/133b cistron knock-out mice cells | Muscle 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 alpha | Repression of Idl-3 protein expression | miR-206 up-regulated | Mouse skeletal muscle differentiation | [42] |
Repression of p180 subunit of DNA polymerase alpha | ||||
MEF2 transcription factor | MEF2 activates of miR-1-2 and 133a-1 transcription; binds muscle-specific enhancer | Bicistronic primary transcript of miR-1-2 and 133a-1 | Development of mammalian skeletal muscle | [9] |
MRFs, Myf5, MyoD, Myogenin and MRF4 | Myf5 essential for miR-1 and miR-206 expression during skeletal muscle myogenesis | Forced expression of MRFs in neural tube induces miR-1 and miR-206 expression | Chicken and mouse embryonic muscle | [171] |
PTB and neuronal homolog nPTB, exon splicing factors | Downregulation of PTB protein by miR-133 (and miR-206) | Concurrent upregulation of miR-133 and induction of splicing of several PTB-repressed exons | During myoblast differentiation, microRNAs control a developmental exon splicing program | [172] |
BDNF | BDNF downregulated | miR-206 upregulated | Differentiation of C2C12 myoblasts into myotubes | [48] |
Fstl1 and Utrn | Fstl1 and Utrn downregulated | miR-206 upregulated | Skeletal muscle differentiation | [40] |
Utrophin A (muscle) | Utrophin A down-regulated by both miRs | Upregulated miR-133b, miR-206 | C2C12 mouse myoblasts, mouse soleus muscle | [173] |
CNN3 gene | Negative correlation between miR-1 expression and CNN3 mRNA expression | Normal skeletal muscle | Tongcheng (Chinese) and Landrace (Danish) pigs | [174] |
FGFR1 and PP2AC, members of ERK1/2 signalling pathway | miR-133 (a and b) activities increase during myogenesis | miR-133 directly downregulates expression of FGFR1 and PP2AC | Mouse C2C12 myoblast cells | [31] |
ERK1/2 signalling pathway activity | ERK1/2 signalling activity suppresses miR-133 expression | Downregulation of expression of miR-133 | A reciprocal mechanism for regulating myogenesis | |
BAF chromatin remodelling complex (BAF60a, BAF60b and BAF60c) | Positive inclusion of BAF60c in the BAF chromatin remodeling complex | Expression of miR-133 and miR-1/206 | Progression of developing somites in chick embryos | [63] |
BAF chromatin remodelling complex | Negative regulation of BAF60a and BAF60b; exclusion from BAF chromatin remodelling complex | Expression of miR-133 | Progression of developing somites in chick embryos | [63] |
BAF chromatin remodelling complex | Exogenous upregulation of BAF60a and BAF60b | Delay in developing somites in chick embryos | [63] | |
Mitochondrial UCP2 and UCP3 | MyoD activates miR-133a expression which in turn directly downregulates UCP2 mRNA | Feedback network involving MyoD-miR-133a-UCP2 | Mouse skeletal and cardiac muscles; UCP2 imposes developmental repression | [56] |
Mitochondrial UCP2 and UCP3 | Exogenous overexpression of myogenin and MyoD transcription factors | Strong increase in UCP3 promoter, expression, weak effect at the UCP2 promoter | Mouse C2C12 myoblasts | [57] |
Proliferating myogenic skeletal muscle cells | ||||
MiR-206/133b cluster | MiR-206/133b cluster is not required for survival and regeneration of skeletal muscle | Muscle regeneration proceeds in Mdx mice in vivo | miR-206/133b cistron knock-out mice | [170] |
Enhanced translation of specific mitochondrial genome-encoded transcripts | miR-1 enters muscle mitochondria and binds mtRNA targets along with Ago factor | Increased expression of mtRNA targets | Proliferating myogenic skeletal muscle cells after muscle injury | [53] |
mTOR (serine/threonine kinase) | MyoD stability regulated by mTOR | Regulates miR-1 expression via MyoD availability | Regenerating mouse skeletal muscle and differentiating myoblast cells | [32] |
AMPK-CRTC2-CREB and Raptor-mTORC-4EBP1 pathways | mTORC regulates timing of satellite cell proliferation during myogenesis | Knockdown of mTORC reduces miR-1 expression | Myogenenic satellite SCs proliferating and differentiating into myogenic precursors following rat skeletal muscle injury | [58] |
HDAC4 regulates Pax7-dependent muscle regeneration | Pax7 stimulates SCs differentiation toward the muscle lineage, and limits adipogenic differentiation | HDAC4 upregulated in SCs differentiating into muscle cells | Myogenenic satellite SCs | [175] |
pcRNA encoded by the H strand of the rat mitochondrial genome | Introduction of mt pcRNAs into injured muscle restoring mitochondrial mRNA levels; Intramuscular ATP levels were elevated after pcRNA treatment of injured muscle | Enhanced organellar translation and respiration; similarly reactive oxygen species were reduced; Resulted in accelerated rate of wound resolution | Injured 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-133 | Reprogrammed human fibroblasts show sarcomere-like structures and calcium transients; Some cells have spontaneous contractility | Forced over-expression of GATA binding protein 4, Hand2, T-box5, myocardin, and microRNAs miR-1 and miR-133 | Human embryonic and adult fibroblasts activated to express cardiac markers | [15] |
SRF, MyoD and Mef2 transcription factors | miR-1-1 and miR-1-2 | miR-1 genes upregulated; | Cardiac muscle precursor cells | [30] |
During cardiogenesis miR-1 genes titrate critical cardiac regulatory proteins, control ratio of differentiation to proliferation | Elevated miR-1 targets downregulation of Hand2 | |||
Histone deacetylase inhibitor, trichostatin A forces differentiation, yet reduced miR-1 and miR-133a | miR-1 and miR-133a reduce cardiac specific Nkx2.5 protein and Cdk9 | miR-1 and miR-133a increase during spontaneous differentiation of cardiac myoblasts | Mouse cardiac stem cells (ES cells) | [10] |
Specific inhibition of HDAC4 modulates CSCs to facilitate myocardial repair | Positively proliferative myocytes increased in MI hearts receiving HDAC4 downregulated CSCs | CSCs with downregulated HDAC4 expression improved ventricular function, attenuated ventricular remodeling, promoted regeneration and neovascularization in MI hearts | Mouse CSCs transplanted into MI mouse hearts | [177] |
Snai1 | Overexpression of miR-133a (miR-133), Gata4, Mef2c, and Tbx5 (GMT) or GMT plus Mesp1 and MyocD improved cardiac cell reprogramming from mouse or human fibroblasts | miR-133a directly represses Snai1 expression, which silences fibroblast signatures; a key molecular process during cardiac reprogramming | Mouse/human fibroblasts more efficiently reprogrammed into cardiomycete-like cells | [16] |
β1AR signal transduction cascade | Adenylate cyclase VI and the catalytic subunit of the cAMP-dependent PKA are components of β1AR transduction cascade | miR-133 directly targets β1AR, Adenylate cyclase VI and PKA | TetON-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 GPx | miR-133 produced a reduction of ROS and MDA levels, and an increase in SOD activity and GPx levels | Overexpression of miR-133, a recognized anti-apoptotic miRNA | In vitro rat cardiomyocytes | [18] |
Caspase-9 | miR-133 directly suppresses caspase-9 expression resulting in downregulation of downstream apoptotic pathways | Overexpression of miR-133 | In vitro rat cardiomyocytes | [18] |
Spred1 | miR-1 directly targets Spred1 | miR-1 is upregulated in hCMPCs during angiogenic differentiation | hCMPCs | [178] |
miRNA-1 and miRNA-133a | miRNA-1 and miRNA-133a have antagonistic roles in the regulation of cardiac differentiation | Forced overexpression of miR-1 alone enhanced cardiac differentiation, in contrast overexpression of miR-133a reduced cardiac differentiation, compared to control cells | Pluripotent 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 Hand2 | Cardiac myocytes show defective heart development, altered cardiac morphogenesis, channel activity, and cell cycling | miR-1-2-/- gene knockout | Cardiac myocytes with knockout of both miR-1-2 genes | [180] |
mt-COX1 mRNA | 3’-UTR of mt-COX1 mRNA bound by miR-181c and Ago1 factor | Overexpression of miR-181c significantly decreased mt-COX1 protein, but not mt-COX1 mRNA level | Overexpression of miR-181c increased mitochondrial respiration and reactive oxygen species in neonatal rat ventricular myocytes | [54] |
mt-COX1 mRNA | In vivo elevation of miR-181c in rat heart, reduces levels of mt-COX1 protein | Results in reduced capacity for strenuous exercise and evidence of heart failure | Rat cardiac muscle | [55] |
Carvedilol, a β-adrenergic blocker | Induces upregulation of miR-133 | Cytoprotective effects against cardiomyocyte apoptosis | Rat cardiac tissue, in vivo | [18] |
GLUT4, and SRF | Both miRs downregulate SRF and KLF15 | Both miR-133a and miR-133b target KLF15 | Mouse cardiac myocytes | [181] |
GLUT4 expression | Both basal and insulin-stimulated glucose uptake are increased | KLF15 | Mouse muscle cell lines | [182] |
MEF2 transcription factor | MEF2 directly activates transcription of miR-1-2 and 133a-1 binding muscle-specific enhancer between the genes | Bicistronic primary transcript of miR-1-2 and 133a-1 | Development of mammalian cardiac muscle | [9] |
Myocardium tissue | Enriched in miR-1, miR-133b, miR-133a | Heart structures of rat, Beagle dog and cynomolgus monkey | [183] | |
Gelsolin | One common miR-133a isomiR targets gelsolin gene more efficiently than standard isomer; New second rat miR-1 gene | Many isomiRs were detected by deep sequencing at higher frequency than the canonical sequence in miRBase | miRNA/isomiR expression profiles in the left ventricular wall of rat heart | [184] |
CTGF | CTGF downregulated by both miRs | Exogenous upregulation of miR-133b (and miR-30c) | Cultured cardiomyocytes and ventricular fibroblasts | [185] |
MT1-MMP | miR-133a upregulated | miR-133a targets MT1-MMP | Human left ventricular fibroblasts | [186] |
Injured and regenerating cardiac muscle | ||||
SERCA2a | Akt/FoxO3A-dependent pathway | Downregulation of miR-1 expression in failing heart muscle | Failing mouse heart muscle | [187] |
Activated SERC2a reduces phosphorylation of FoxO3a, allowing entry to nucleus and activation of miR-1 expression | ||||
IGF-1 | IGF-1 signalling and miR-1 co-regulate differentiation of myoblasts via a feedback loop | IGF-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 Bmf | Only miR-133a expression enhanced under in vitro oxidative stress | miR-133a targets proapoptotic genes Bim and Bmf | Rat adult CPCs | [188] |
miR-1 favors differentiation of CPCs, whereas | ||||
Bim and Bmf | CPCs overexpressing miR-133a improved cardiac function by reducing Bim and Bmf | CPCs overexpressing miR-133a improved cardiac function, increasing vascularization and cardiomyocyte proliferation, reduced fibrosis and hypertrophy | CPCs overexpressing miR-133a in rat myocardial infarction model | [188] |
MT1-MMP activity increased in both. Ischemia and reperfusion regions | Interstitial miR-133a decreased with ischemia in vitro and in vivo; reperfusion returned to steady-state | Phosphorylated Smad2 increased within the ischemia-reperfusion region | Ischemia-reperfusion Yorkshire pigs (90 min ischemia/120 min reperfusion) | [186] |
Cardiovascular disease | ||||
CNN2 | Strong upregulation of CNN2 expression | miR-133b downregulated; miR-133b directly targets CNN2 | Pre-inflammatory events in diseased cardiac tissues | [65] |
Circulating platelet derived microparticles | Elevated miR-133 | Patients with stable and unstable coronary artery disease | [189] | |
Acute MI causes upregulation of circulating serum miRs | miR-1, -133a, -133b, and -499-5p were about 15- to 140-fold elevated over control | Acute STEMI patients and experimental mouse MI model | [190] | |
Circulating miRNAs in serum of cardiovascular disease patients | Released miR-1 and miR-133a are localized in exosomes, and are released by Ca(2+) stimulation | Levels of miR-1, miR-133a, reduced in infarcted mouse myocardium model heart | miR release indicates myocardial damage | [191] |
LVM after valve replacement in aortic stenosis | microRNA-133a is a significant positive predictor of LVM normalisation | miR-133 is a key element of the reverse remodelling process | Patients following valve replacement | [192] |
Circulating levels of miR-133a | Elevated miR-133a (11-fold) | Troponin-positive acute coronary syndrome patients | [193] | |
Circulating levels of miR-133a | Elevated miR-133a | Improved potential regression of Left Ventricular Hypertrophy after valve replacement | Patients with aortic stenosis surgery | [194] |
Apelin treatment reduces elevated circulating miRs | Elevated miR-133a, miR-208 and miR-1 reduced | High-fat diet elevated miRs and increased left ventricular diastolic and systolic diameters, and wall thickness | Obesity-associated cardiac dysfunction in mouse model | [195] |
NAC treatment | Expressed miR-1, miR-499, miR-133a, and miR-133b were strongly depressed in the diabetic cardiomyocytes | NAC restored expression of miR-499, miR-1, miR-133a, and miR-133b significantly in the myocardium | Diabetic rat hearts | [196] |
Myocardial junctin elevated | miR-1 targets junctin | NAC reduces junction levels | Development of diabetic cardiomyopathy in rat hearts | [196] |
CAD associated ischemic heart failure | miR-133 expression decreased with increased severity of heart failure | Patients with CAD | [197] | |
Runx2 | miR-133a targets Runx2 | Transition of VSMCs to osteoblast-like cells | [198] | |
Increased alkaline phosphatase activity, osteocalcin secretion and Runx2 expression | miR-133a was decreased during osteogenic differentiation | Transition of VSMCs to osteoblast-like cells | [198] | |
Circulating miR-133a and 208a levels | Cardiac muscle-enriched microRNAs (miR-133a, miR-208a) elevated | Patients with coronary artery disease | [199] | |
Hypertrophic cardiac muscle | ||||
Cx43 increased | miR-1 targets Cx43 | Downregulation of miR-1 mediates induction of pathologic cardiac hypertrophy | Hypertrophic rat cardiomyocytes in vitro and in vivo | [200] |
Cx43 downregulated | miR-1 targets Cx43 | Cx43 protein downregulated in miR-1 Tg mice compared to WT mice | Cardiac-specific miR-1 transgenic (Tg) mouse model | [201] |
Twf1 upregulated | miR-1 targets Twf1 | Strong downregulation of miR-1 in pathologic hypertrophic cardiac cells compared to normal, induces Twf1 expression | In vivo in hypertrophic mouse left ventricle; and in vitro in phenylephrine-induced hypertrophic cardiomyocytes | [202] |
RhoA, Cdc42, Nelf-A/WHSC2 | Increased levels of RhoA, Cdc42, Nelf-A/WHSC2 | Reduction miR-133a | Hypertrophic cardiac muscle | [6] |
Calcineurin, agonist of cardiac hypertrophy | Increased Calcineurin activity; | Reduced miR-133a; | Hypertrophic cardiac muscle; | [203] |
Cyclosporin A inhibits calcineurin | Prevents miR-133 down-regulation | Cardiac hypertrophy reduced | ||
NFATc4 | NFAFc4 targetted by miR-133a | miR-133a | Cardiomyocyte hypertrophic repression | [204] |
Interdependent Calcineurin-NFAT and MEK1-ERK1/2 signalling pathways in cardiomyocytes | MEK1-ERK1/2 signalling augments NFAT and NFAF gene expression; Activated calcineurin activates NFAT, inducing cardiac hypertrophy | MEK1 is part of mitogen-activated protein kinase (MAPK) cascade; MEK1 activates ERK directly | Hypertrophic growth response of mouse cardiomyocytes | [205] |
Innervating skeletal muscle | ||||
Innervated skeletal muscle | MyoD, Myf5, Mrt4, nAChRα | Myogenin expression | Mouse skeletal muscle | [50,51] |
Each is strongly repressed | ||||
Denervated muscle (unstimulated) | Myogenin expression up-regulated MyoD, Myf5, Mrt4, nAChRα | Mouse skeletal muscle | [51] | |
All strongly stimulated | ||||
Electrically stimulated - Denervated muscle | Myogenin, MyoD, Myf5, Mrt4, partly stimulated; nAChRα inhibited | Mouse skeletal muscle | [51] | |
HDAC4 | miR-1 promotes myogenesis by targetting HDAC4 | miR-133 enhances myoblast proliferation by targetting SRF | Skeletal 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 HDAC4 | HDAC4 inhibits activation of muscle transcription factor MEF2; results in progressive muscle dysfunction | MEF-2 activity strongly inhibited in denervated mouse skeletal muscle and in ALS muscle | [49] |
Innervation and formation of airway smooth muscle | Sonic hedgehog (Shh) /miR-206/ BDNF | Shh signalling blocks miR-206 expression, which in turn increases BDNF protein | Shh coordinates innervation and formation of airway smooth muscle | [206] |
nAChR subunits (UNC-29 and UNC-63); retrograde signalling | Subunits UNC-29, UCR-63 and MEF2 downregulated | miR-1 upregulated | C. elegans muscle at the neuromuscular junction | [34] |
MEF2 | Hnrpu, Lsamp, MGC108776, MEF2, Npy, and Ppfibp2 downregulated | miR-206 upregulated | Rat skeletal muscle/re-innervating muscle | [43] |
HDAC4 | HDAC4 (miR-206 target, prospective miR-133b target) downregulated | miR-206/-133b upregulated (and miR-1/-133a downregulated) | Mouse fast twitch skeletal muscle/re-innervating muscle | [12] |
Regenerating injured muscle | ||||
Hnrpu and Npy downregulated | miR-1 upregulated | miR-1, -133a, downregulated 1 mo after denervation, then increased 2 × at 4 mo after re-innervation | Rat skeletal soleus muscle after sciatic nerve injury and subsequent re-innervation | [43] |
Ptprd downregulated | miR-133a upregulated | |||
Hnrpu, Lsamp, MGC108776, MEF2, Npy, and Ppfibp2 downregulated | 3 × increase in miR-206 1 mo later, after reinnervation; elevated at least 4 mo | Predominant type II fiber at 4 mo, after nerve re-innervation | Rat skeletal soleus muscle after sciatic nerve injury and subsequent re-innervation | [43] |
PP2A B56a | PP2A B56a downregulated | 133a upregulated | Canine heart failure model: myocytes | [207] |
CaMKII-dependent hyperphosphorylation of RyR2 | VF myocytes had increased reactive oxygen species and increased RyR oxidation | miR-1 upregulated | Canine post-myocardial infarction model | [208] |
Collagen upregulated | TGF-b1 and TGFbRII: upregulated | miR-133a or miR-590: downregulated | Canine model of acute nicotine exposure. Atrial fibrosis in vivo; cultured canine atrial fibroblasts in vitro | [209] |
miR-208 upregulated | miR-1 and miR-133a downregulated | Human MI compared to healthy adult hearts | [210] | |
Myogenic proteins, MyoD1, myogenin and Pax7 | Induced expression of MyoD1, myogenin and Pax7 several days after miR injection | Exogenous injection of miR-1, -133 and -206 promotes myotube differentiation | Regenerating injured mouse skeletal muscle | [211] |
Cyclin D1/ Sp1 | Cyclin D1/ Sp1 downregulated | miR-1/133 upregulated | Regenerating rat skeletal muscle | [212] |
PRP, source of pro-inflammatory cytokines | Stong 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-cristallin | Stimulated 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 TWEAK | TWEAK upregulated | miR-1-1, miR-1-2, miR-133a, miR-133b and miR-206 downregulated | Degenerating/wasting mouse skeletal muscle | [59] |
HMOX1 mediated by codependent inhibition of c/EBPδ binding to myoD promoter | HMOX1 inhibits differentiation of myoblasts and modulates miRNA processing | Downregulation 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-206 | Downregulation of MyoD, myogenin and myosin, and disturbed formation of myotubes. Upregulation of SDF-1 and miR-146a | |||
Dystrophic muscular disease | ||||
Circulating serum microRNAs | miR-1, miR-133a, and miR-206 highly abundant in Mdx serum | miR-1, miR-133a, and miR-206 downregulated or modestly upregulated in muscle | Muscle tissue from patients with Duchenne muscular dystrophy (Mdx) | [213] |
Laminin α2 chain deficiency | miR-1, miR-133a, and miR-206 are deregulated in laminin α2 chain-deficient muscle | Laminin α2 chain-deficient mouse | Congenital muscular dystrophy type 1A tissue | [214] |
Dystrophic process advances from prominent inflammation with necrosis and regeneration to prominent fibrosis | Deficiency in calpain leads initially to accelerated myofiber formation followed by depletion of satellite cells | Pax7-positive SCs highest in the fibrotic patient group; correlated with down-regulation of miR-1, miR-133a, and miR-206 | Muscle 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 heart | Normal skeletal muscle and heart development | Mdx 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 hybridization | Only miR-206 showed an over-expression in majority of DM1 patients | No 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 regeneration | HDAC inhibitors can activate FAPs towards muscle regeneration | Inhibition of HDAC induces MyoD and BAF60C expression, which causes up-regulation of miR-1-2, miR-133, and miR-206 expression | Early stage disease dystrophic mouse muscles, regeneration of myofibres | [62] |
TDP-43 | TDP-43 interacts with miR-1/-206 isomers, but not miR-133 isomers | Depleted miR-1/-206 allow targets IGF-1 and HDAC4 to accumulate in ALS muscle | Mouse ALS model injured motor neurons and muscle | [33] |
Inflammation response in muscle | ||||
Inflammatory myopathies | Increased expression of TNFα | Associated with decreased expression of miR-1, miR-133a, and miR-133b | Inflammatory myopathies including dermatomyositis, polymyositis, and inclusion body myositis | [64] |
hBSMCs sensitized with IL-13 | Increased muscle RhoA | Reduction of muscle miR-133a | Sensitized human bronchial smooth muscle cells (hBSMCs) | [218] |
Factor(s) | Regulation | Regulator | Tissue/cell | Ref. |
Nerve tissues | ||||
Pitx3 | Pitx3 downregulated | miR-133b | Mammalian midbrain DNs | [73] |
Exosome-mediated transfer of miR-133b from MSC to brain astrocytes | miR-133b transfer from multipotent mesenchymal stromal cells to neural cells | miR-133b upregulated | Mouse MSCs to neural cells | [47] |
Ctgf and RhoA | Ctgf and RhoA downregulated | miR-133b upregulated | Multipotent MSCs/Rat brain parenchymal cells | [72] |
miR-133b null mice: Striatum dopamine levels unchanged, Pitx3 expression unaffected; motor coordination unaltered | miR-133b has no significant role on mDA neuron development and maintenance in vivo | Normal numbers of mDA neurons during development and aging of miR-133b null mice | Mouse mDA neuron development in -/-miR-133b mutant mice | [45] |
Acute or chronic morphine administration, or morphine withdrawal | miR-133b levels not affected | Rat VTA/ nucleus accumbens shell | [219] | |
GPM6A, a neuronal glycoprotein | microRNA-133b upregulation | Reduction in gmp6a at mRNA and protein level. Cell filopodium density was reduced | Hippocampus and prefrontal cortex of neonatal male rats stressed when in utero | [220] |
Tac1 gene (neurotransmitter substance P) | Tac1 downregulated | miR-206 upregulated | MSCs-derived neural cells | [221] |
Ketamine (antidepressive) administration | BDNF, a direct target gene of miR-206, was upregulated | miR-206 was downregulated by ketamine | Rat hippocampus tissue | [222] |
Adipogenic tissues | ||||
IGF-1 and IGF-1R | IGF-1 signalling and miR-133b co-regulate ADSC differentiation via a feedback loop | miR-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 | ||||
Pdrm16 | miR-133a directly targets Prdm16. | Downregulation of miR-133 resultsin differentiation of pre-adipocyte precursors into BAT | Mouse adipocyte differentiation to BAT | [74] |
Pdrm16 | miR-133 directly targets Prdm16 | Downregulation of miR-133 resulted in differentiation of pre-adipocyte precursors into BAT | Mouse primary brown adipocyte (and myogenic) progenitor cells - differentiate into BAT or SAT | [75] |
Pdrm16 | miR-133 targets Prdm16 controlling brown adipose determination in skeletal muscle satellite cells | miR-133 downregulates Prdm16 | Adult mouse skeletal muscle stem cells (satellite cells) differentiate into BAT | [76] |
HDAC4 downregulation directs SCs towards adipocyte differentiation | Brown adipose master regulator Prdm16 is upregulated, while its inhibitor miR-133 is also downregulated | HDAC4 downregulated in SCs differentiating into adipocyte progenitor cells | Myogenenic satellite SCs | [175] |
GLUT4 expression | Both basal and insulin-stimulated glucose uptake are increased | KLF15 | Mouse 3T3-L1 preadipocytes differentiating into adipocytes | [182] |
Intrinsic insulin resistance | Elevated miR-133b | Undefined role | Adipose tissue of women with PCOS | [223] |
Upregulation of LIM homeobox 8 and Zic family member 1 and downregulation of Homeobox C8 and Homeobox C9 | Undefined relation of upregulated miR-206, miR-133b | Undefined relation with parallel upregulation of brite/beige markers, TBX1 and TMEM26 | Human BAT from the supraclavicular region | [224] |
Obesity development | Downregulation of miR-133b, miR-1 | Undefined role | Adipose tissue from obese male C57BLJ6 mice | [225] |
LXRα regulation of lipogenic genes | miR-1/miR-206 represses LXRα expression at both mRNA and protein levels | miR-1/miR-206-induces a decrease in lipogenic gene levels and lipid droplet accumulation | Mouse hepatocytes | [226] |
Osteogenic tissues | ||||
Development of bone on organic or inorganic substrates | miR-133 differentially expressed in osteoblasts grown on different substrates | Osteoblast | [227] | |
Runx2 | miR-133 directly down-regulates Runx2 | miR-133 up-regulated | Osteogenic differentiation from C2C12 mesenchymal cells | [228] |
HDAC4 | HDAC4 downregulates Runx2 | miR-1 targets HDAC4, increasing Runx2 activity | Chondrocyte proliferation in cartilage growth plate | [77] |
Aggrecan | miR-1 promotes late-stage differentiation of growing cartilage cells | miR-1 targets Aggrecan gene expression | Chicken chondrocytes and human HCS-2/8 cells | [78] |
Alveolar cells | ||||
VAMP2/ lung surfactant secretion | miR-206 targets VAMP-2 | miR-206 overexpression decreased lung surfactant secretion | Lung alveolar type II cells | [229] |
Hormonal regulation | ||||
L-thyroxine | miR-206/miR-133b downregulated | L-thyroxine treatment | L-thyroxine treated hypothroidic skeletal muscle from thyroidectomized patients | [230] |
miR-206/miR-133b upregulated | - | Hypothroidic human skeletal muscle | ||
Thyroid hormone/TEAD1 | Thyroid hormone inhibits the slow muscle phenotype by upregulation of miR-133a1 which downregulates TEAD1 | miR-133a1 is enriched in fast-twitch muscle and regulates slow-to-fast muscle fiber type conversion | Mouse muscle | [231] |
Thyroid hormone/miR-133a1 TEAD1 | myosin heavy chain I expression downregulated | TH indirectly downregulates myosin heavy chain I via miR-133a/TEAD1 | Mouse muscle | [232] |
L-thyroxine | pre-miR-206 and pre-mir-133b downregulated | L-thyroxine | L-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 cardiomycetes | Downregulation KLF15, which downregulates GLUT4 | Forced overexpression of miR-133a and miR-133b | Rat cardiac myocytes | [181] |
Cardiac myocyte glucose metabolism | Upregulation KLF15, which upregulates GLUT4 | Silencing endogenous miR-133 | Rat cardiac myocytes | [181] |
Metabolic control of glucose uptake by GLUT4 transporter | Downregulates KLF15, which results in downregulation of GLUT4 levels | Chronic heart failure has depressed miR-133a and -133b levels | Rat cardiac myocytes during chronic heart failure and cardiac hyperthrophy | [181] |
Atrial natriuretic factor expression upregulation | Enhanced at LVH and dramatically increased at CHF stage | Both miR-133a and miR-133b downregulated at CHF stage | LVH and CHF in salt-sensitive Dahl rats | [181] |
Estrogen | Estrogen replacement strongly decreased IGF-1 protein level in muscles at 1 wk | Ovariectomized rat skeletal muscle | [233] | |
Multiple targets | miR-133a upregulated in BTBR mice | Pancreatic 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 levels | miRNAs miR-1, miR-133a and miR-206 specifically expressed both in brown pre- and mature adipocytes | miRNAs miR-1, miR-133a and miR-206 were absent from white adipocytes | Mouse brown adipocytes | [235] |
Foxl2 | miR-133b targets Foxl2; | Foxl2 regulates StAR and CYP19A1 transcriptionally | Estradiol 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 astrocytes | miR-133b transfer from multipotent mesenchymal stromal cells to neural cells | miR-133b upregulated | Mouse multipotent MSCs to neural cells | [47] |
Cell to cell transfer of exosome-enriched extracellular particles | mir-133b promotes neural plasticity and recovery of function after stroke induced damage | miR-133b upregulated | Rat multipotent MSCs via transfer of exosome-enriched extracellular particles | [72] |
Transplanted stem cells | ||||
MSCs expressing miR-1 | Upregulated miR-1 | Increased rate of recovery, enhanced survival of transplanted MSCs and cardiomyogenic differentiation | Experimental ligation of the mouse left coronary artery to model myocardial infarction | [237] |
Knockdown of Hes-1, member of Notch pathway | Upregulated miR-1 promotes the differentiation of MSCs into cardiac lineage | Role in survival of transplanted MSCs and cardiomyogenic differentiation | Mouse MSCs | [238] |
Notch signalling and cardiomyocyte markers, Nkx2.5, GATA-4, cTnT, and Cx43 | MSCs expressing exogenous miR-1 | Mouse MSCs | [238] | |
Tissue inflammation | ||||
Selective release of miRs during inflammation into serum | miR-133 selectively released | Review | [239] | |
Inflammation and cancer | MicroRNA, free radical, cytokine and p53 pathways | Review | [240] | |
Immunological switch which shapes tissue responses | TWEAK/Fn14 pathway | Review | [241] | |
Tumor biology | HMOX1 | Review | [242] | |
GM-CSF | Direct supression of GM-CSF expression by miR-133 | Elevated expression of miR-133a/-133b during oxidative stress | Mouse alveolar epithelial cells during oxidative stress | [82] |
PI3K/Akt and IGF-1 pathways | Activation of PI3K/Akt and IGF-1 pathway activities | Downregulation of miR-133a (and other miRs) by AOM/DSS induced chronic inflammation | Mouse model: AOM/DSS-induced colitis-associated gastro-intestinal cancer | [83] |
CTGF, SMA, and COL1A1 | Increased expression of CTGF, SMA and COL1A1, which are miR-133b targets | Strong downregulation of miR-133b (and other miRs) | TGF-β treated rabbit corneal fibroblasts; Recovering mouse cornea after laser ablation, | [70] |
IL-10 and TGF-β | Exogenous IL-10 and TGF-β induces miR-133b expression | Upregulation of miR-133b | Human tolerogenic dendritic cells during maturation | [79] |
IL-17-producing T-cells | Upregulation of Il17a/f gene expression | miR-133b/-206 cistron transcription occurs along with nearby Il17a/f gene expression | Immunocompetent mouse Th17 cells | [80] |
NLRP3 inflammasome which processes IL-1β by caspase-1 cleavage | miR-133a-1 suppresses activation of inflammasomes via suppression of expression of mitochondrial UCP2 | miR-133a-1 overexpression in cells increases caspase-1 p10 and IL-1β p17 cleavage, | Differentiated mouse THP1 cells | [81] |
Concanavalin A-induced fulminant hepatitis | miR-133a is the most strongly differentially upregulated miR | Mouse liver following ConA injection | [243] | |
Infection/immune response to influenza virus (H1N2) | miR-206 expression | Experimental 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 PASMCs | miR-206 targets HIF-1α directly. Hypoxia-induced down-regulation of miR-206 promotes PH in PASMCs | Hypoxia-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 activity | Astrocyte-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 regulator | Negative regulator | Regulated target miR | Tissue/cell | Ref. |
Myogenin, MyoD | Upregulates miR-1-1 and miR-133a-2 | Primary human myoblasts; C2C12 cells | [11] | |
Upregulates miR-1-2 and miR-133a-1 | ||||
SRF, MyoD and MEF2 | Upregulates miR-1-2 | Muscle somites | [30] | |
MEF2 | Upregulates miR-1 and miR-133a | Skeletal muscle | [9] | |
KSRP (part of Drosha and Dicer complexes) | miR-206 binds 3’-UTR of KSRP and inhibits its expression | KSRP upregulates miR-1 expression | Skeletal muscle | [35,37] |
RNA-binding protein LIN28 | LIN28 upregulates miR-1 expression; LIN28 promotes pre-miR-1 uridylation by ZCCHC11 (TUT4) | Cardiac muscle of patients with muscular dystrophy | [36] | |
MBNL1 | MBNL1 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-1 | Cardiac muscle of patients with muscular dystrophy | [36] | |
CX43 and CACNA1C calcium channel | CX43 and CACNA1C both increased in both DM1-/DM2-affected hearts, contributing to the cardiac dysfunctions | CX43 and CACNA1C are direct targets of miR-1 repression | Cardiac muscle of patients with muscular dystrophy; | [36] |
CACNA1C and CX43 encode the main calcium- and gap-junction channels in heart | ||||
Utrophin A | miR-206 and KSRP are negative regulators of utrophin A | Overexpression of miR-206 promotes the upregulation of utrophin A, via the downregulation of KSRP | Normal 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 | ||||
Myostatin | Downregulates miR-1, miR-133a, miR-133b, miR-206 | Mouse (35 d) pectoralis skeletal muscle | [29] | |
SRF | Downregulates miRs-133a | Skeletal muscle | [1,3] | |
Prmt5 and Prmt4 | Upregulates myomiR expression during differentiation | Mouse skeletal muscle | [247] | |
Smooth muscle | ||||
Sp-1 transcription factor | pERK1/2 | Upregulates miR-133(a) | VSMCs | [248] |
Brg1 | Upregulates miR-133 (ChIP complex with SRF) | Smooth muscle | [249] | |
Cardiac muscle | ||||
GATA4, Nkx2.5, Myocardin, SRF | Upregulates miR-1 and miR-133a | Differentiating cardiac muscle | [5] | |
SRF plus Myocardin | Upregulates miR-1-1 and miR-1-2 | Cardiomycetes | [30] | |
Calcineurin | Downregulates miR-133a | Hypertrophic cardiac muscle | [203] | |
miR-206/ miR-133b | ||||
Skeletal muscle | ||||
Mrf5 | Upregulates miR-1, miR-206 | Skeletal muscle | [171] | |
Myogenin, MyoD | Upregulates miR-206 | Primary human myoblasts; C2C12 cells | [11] | |
MyoD | Upregulates linc MD1 (encodes miR-133b) | Differentiating myoblasts | [11, 38] | |
Binds to (E-box) enhancer of miR-206, miR-133b | skeletal muscle (mouse) | [12,40] | ||
Upregulates miR-206/miR-133b | Differentiated human foetal skeletal muscle cells | [250] | ||
FGF2 allows upregulation of Sp1/Cyclin D1 | Downregulates p38-mediated miR-1/133 expression | Regenerating rat skeletal muscle | [212] | |
Myostatin | Downregulates miR-133a, mir-133b, miR-1, and miR-206 | Mouse (35 d) pectoralis skeletal muscle | [29] | |
TWEAK downregulates myoD and MEF2c | Downregulates miR-1-1 and miR-133 | Degenerating/wasting skeletal muscle | [59] | |
HMOX1 downregulates MyoD and myogenin | Downregulates all myomiRs | Inflamed skeletal muscle | [60] | |
L-Thyroxine treatment | Downregulation of pri-miR-206 and pri-miR-133b | Human skeletal muscle | [230] | |
No effect on miR-1/miR-133a pairs | ||||
Smooth muscle | ||||
p-ERK | Activated extracellular signal-regulated kinase p-ERK inversely correlated with VSMC growth | Downregulates miR-133 expression | VSMCs | [248] |
Other tissues | ||||
Myogenin | Binds miR-206 enhancer (ChIP) | Fibroblast cell line: | [40] | |
IGF-I signalling | Upregulates miR-133b | Mouse Adipose derived stem cells | [71] | |
L-Thyroxine deficiency | Upregulated Col5a3 | Strong upregulation of miR-133a and -133b | Hypothyroid mouse liver | [232] |
Downregulated Slc17a8, Gp2, Phlda1, Klk1d3, Klk1 and Dmbt1 | Strong upregulation of miRs -1, -206 | |||
Upregulated Vldlr and Akr1c19, and downregulated Upp2, Gdp2, Mup1, Nrp1, and Serpini2 | ||||
L-Thyroxine treatment | Pre-miR-206 and Pre-miR-133b down-regulated | Upregulation of Gdp2 andMup1 | Hypothyroid mouse liver in vivo, and in vitro mouse hepatocyte AML12 cells | [232] |
PA2G4, mps1, cdc37, cx43, cldn5; cx43 is a miR-133 target | Upregulation of cell cycle factors mps1, cdc37, and PA2G4, and cell junction components cx43 and cldn5 | Suppression of miR-133a1 stimulates cardiac cell proliferation | Regeneration of damaged Zebrafish cardiac muscle, associated with reduced miR-133a1 | [167] |
Fgf | Upregulated Fgf | Downregulates miR-133 | Zebrafish regenerating fin blastema | [67] |
SHP (nuclear receptor) | Downregulation of miR-206 in nuclear receptor SHP(-/-) mice | SHP(-/-) mice strain, mouse liver | [251] | |
AP1 transcription factor complex | AP1 induced miR-206 promoter transactivity and expression; this is repressed by YY1 | ChIP analysis shows physical association of AP1 (c-Jun) and YY1 with miR-206 promoter | SHP(-/-) nuclear receptor mice strain, mouse liver | [251] |
NR3B3 | YY1 promoter transactivated by ERRgamma; this inhibited by SHP (NROB2) | Nuclear receptor ERRgamma (NR3B3) binding site on the YY1 promoter | Mouse 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-206 | Mouse liver | [251] |
Il17a/f locus | miR-133b and miR-206 expression | Coregulated with IL-17 production | αβ and γδ T cells | [80] |
MiR-1 | MiR-206 | MiR-133a | MiR-133b | Cancer type | Ref. |
+ | + | 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] |
Downregulated miR-1 | ||||
miR-1 downregulation influences multiple cancer-related pathway processes, and promotes cell proliferation and motility | Epigenetic promoter hypermethylation reduces miR-1/-133a expression in (a subset of) human prostate tumors | Reduced 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 PTMA | miR-1 downregulation associated with upregulation of multiple cancer-related pathway processes | Reduced 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 | ||||
HSPB1 | HSPB1 restores oncogenic pathways in prostate cancer cells | Downregulates miR-1 expression | Progressive prostate cancer PCa cells | [252] |
XPO6 and TWF1 (PTK9) | Inverse expression between miR-1, XPO6 and TWF1 proteins in prostate cancer cell lines | Downregulated miR-1 expression | Prostate cancer cell cultures | [253] |
CCND2, CXCR4, and SDF-1α | Inverse expression between miR-1 and CXCR4 and SDF-1α protein levels in thyroid carcinomas | Strongly downregulated miR-1 expression in thyroid adenomas and carcinomas | Thyroid adenomas and carcinomas | [254] |
MET | MET upregulated | Reduced miR-1 | Colon cancer | [101] |
Reduced miR-1, -133b | Colon cancer | [87] | ||
MET, Pim-1 (Ser/Thr kinase), FoxP1 and HDAC4 | miR-1 downregulated, | MET, Pim-1, FoxP1 and HDAC4 are often upregulated in lung cancer | NSCLC tissue and A549 cell line | [100] |
miR-1 targets MET, Pim-1, and may regulate FoxP1 and HDAC4 | ||||
Fibronectin1 | Fibronectin1 upregulated | miR-1 downregulated | Laryngeal SCC Hep2 cells | [255] |
Met, Twf1 and Ets1 and Bag4 | Met, Twf1 and Ets1 and Bag4 activities upregulated | miR-1 downregulated | Mouse cutaneous squamous cell carcinomas | [256] |
Mediator complex subunit 1 (Med1) and 31 (Med31) | Med1 and Med31 activation result in increased Met activity | Reduced miR-1; miR-1 targets Med 1 and Med 31 | Osteosarcoma | [257] |
NOTCH3 upregulates Asef expression, activating the Asef promoter, enhancing cell migration | NOTCH3 upregulated | Reduced miR-1; miR-1 targets NOTCH3 | Colorectal tumor cells | [258] |
Overexpression of PIK3CA correlates with low miR-1 expression in NSCLC tissues | 71% of NSCLC samples had high PIK3CA expression | 69% of NSCLC samples had low miR-1 expression | Predictors of lymph node metastasis in NSCLC tissues | [259] |
SLUG expression downregulated by miR-1 | Transcriptional repressor of E-cadherin, or an inducer of epithelial-to-mesenchymal transition | Overexpression of miR-1 induces morphological change from a mesenchymal to an epithelial character | NSCLC A549 cell line | [260] |
SLUG expression high in chordoma tissue | miR-1 inhibited cell proliferation both time- and dose-dependently in chordoma | Transfection of MiR-1 inhibited Slug expression | mR-1 transfected chordoma cells | [261] |
Slug overexpressed in advanced chordoma tissues and chordoma cells | ||||
MET expression high in chordoma tissue | miR-1 downregulated 97% of chordoma samples | MiR-1 directly targets MET | Decreasing miR-1 expression levels correlated with severity of clinical prognosis | [262] |
SRSF9/SRp30c | Exogenous upregulation of miR-1 expression | Novel apoptosis pathway involving SRSF9/SRp30c mediates tumor suppression | Bladder cancer (TCC) cells | [263] |
ANXA2 is essential for glioblastoma growth and invasion | ANXA2 is highly abundant protein in glioblastoma-derived extracellular vesicles | miR-1 directly targets ANXA2; | Human Glioblastoma cells; miR-1 orchestrates glioblastoma extracellular vesicle function | [264] |
Reduced miR-1 in glioblastoma | ||||
EDN1 | miR-1 downregulated in gastric cancer | miR-1 causes ET-1 silencing in gastric cancer cell lines | Gastric cancer tissue compared with adjacent normal tissue | [265] |
EDN1 | Elevated expression of EDN1 and reduced miR-1 level | miR-1 directly targets EDN1 | Human liver cancer tissues | [266, 267] |
Overexpressed EDN1 | Enhanced in vitro cell proliferation and cell migration. Upregulation of several cell cycle/proliferation- and migration-specific genes | Upregulated UPR pathway mediators, spliced XBP1, ATF6, IRE1, and PERK at both RNA and protein levels | 293T cells | [267] |
AKT inhibitor diminished the unfolded protein response and eliminated EDN1-induced cell migration | EDN1 effects act via activation of the AKT pathway | Results to enhance the UPR and subsequently activate the expression of downstream genes | 293T cells | [267] |
Edn1 | Induced steatosis, fibrosis, glycogen accumulation, bile duct dilation, hyperplasia, and HCC | Liver-specific edn1 expression | Transgenic Zebrafish liver | [267] |
API5 | API5 expression upregulated thus inhibiting apoptosis | miR-1 expression downregulated | Human liver cancer tissues | [268] |
Apoptosis activated, API5 reduced | Overexpression of miR-1 | HepG2 liver cancer cells | ||
Phosphorylation of ERK and AKT; LASP1 | Overexpression of miR-1 inhibits phosphorylation of ERK and AKT and reverses EMT process via inhibition of MAPK and PI3K/AKT pathways | MAPK and PI3K/AKT pathways | Transgenic miR-1 expressing CRC cell lines | [269] |
LASP1 expression upregulated | Upregulated LASP1 stimulates EMT resulting in cell proliferation and migration | miR-1 downregulated | Colorectal tumor tissue | [269] |
PIK3CA | Increased expression of PIK3CA | Downregulated miR-1 expression in lung cancer | NSCLC tissue with poor patient prognosis | [259] |
PIK3CA indirectly regulating pAKT and survivin proteins | Overexpressed miR-1 downregulated PIK3CA causing reduced pAKT and survivin proteins | Exogenously 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 adhesion | Ectopic expression of miR-1 and miR-145 downregulates VEGFA and AXL, respectively | Highly downregulated expression of miR-1, miR-133, miR-143 and miR-145 in gall bladder cancer | Gall bladder tumor samples and GBC NOZ cell line | [271] |
lncRNA UCA1 | Lnc RNA UCA1 upregulated in bladder cancer (TCC); | Downregulated miR-1 expression in bladder cancer (TCC); miR-1 targets lnc RNA UCA1 for downregulation | Human bladder cancer (TCC) tissue | [156] |
Inverse relationship between miR-1 and lnc UCA1 | ||||
Downregulated miR-133a | ||||
Moesin | Moesin upregulated | Reduced miR-133a | HNSCC | [143] |
ARPC5 | ARPC5 upregulated | Reduced miR-133a | HNSCC | [272] |
ARPC5 and GSTP1 | ARPC5 and GSTP1 upregulated | Reduced miR-133a (and miR-206) | Lung carcinoma | [115] |
IGF-1R, TGFBR1, and EGFR are downregulated | Restoration of ectopic-expression of miR-133a in NSCLC suppresses metastatic capacity | miR-133a inhibits cell invasiveness and cell growth via suppression of IGF-1R, TGFBR1 and EGFR | NSCLCs | [131] |
Low expression of miR-133a is characteristic of pancreas tissue | Reduced miR-133a | PDAC | [103] | |
CDC42 | CDC42 upregulated causing downstream activation of PAKs | miR-133 downregulated | Gastric cancer tissues | [273] |
GSTP1 | Upregulated GSTP1 | Downregulation of miR-133a in cancer | Bladder 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 specimens | GSTP1 upregulated | Reduced miR-133a | Bladder cancer (TCC) tissue | [144] |
Actin-binding protein, FSCN1 | Upregulated FSCN1; | Downregulation of miR-133a; | Bladder cancer (TCC) tissue | [274] |
Enforced downregulation of FSCN1 inhibits cell proliferation, migration and invasion | Forced UP exp of miR-133a inhibits cell proliferation, migration and invasion | |||
EGFR/AKT signalling pathway | Upregulated EGFR; | Downregulated miR-133a; | Human MCF-7 and MDA-MB-231 breast cancer cell lines | [275] |
Activated pAkt-1 | Enforced expression of miR-133a inhibits EGRF translation; causes inhibition of Akt protein phosphorylation and its nuclear translocation | |||
Bcl-xL and Mcl-1 expression | Upregulated Bcl-xL and Mcl-1 | Downregulated miR-133a correlated with tumor progression and poor patient prognosis; | Primary human osteosarcoma tissues; | [276] |
Osteosarcoma cell lines | ||||
E3 ubiquitin protein ligase | Downregulation of p21 and p53 proteins | Downregulated miR-133a | Primary CRC tissues | [277] |
Enhanced sensitivity to doxorubicin and oxaliplatin | Enhancing apoptosis and inhibited cell proliferation | Ectopic upregulation of miR-133a | CRC cell lines | [277] |
LASP1 upregulated | miR-133a expression downregulated | miR-133a targets LASP1 | CRC tissues and cell lines | [278] |
FTL protein upregulated | miR-133a expression downregulated | miR-133a targets downregulation of FTL protein | Patient breast cancer tissue | [279] |
Increased sensitivity to chemotherapeutic drugs doxorubicin and cisplatin | Exogenous upregulation of miR-133a expression | Downregulation of FTL protein | Human MCF-7 breast cancer cells | [279] |
Poor survival during breast cancer; upregulated FSCN1 | Loss of miR-133a expression | FSCN1 is a direct target gene of miR-133a | Breast cancer tissue | [280] |
FSCN1 downregulated | Restoration of miR-133a expression | Inhibited breast cancer cell growth and invasion | Breast cancer cell line | [280] |
lncRNA Malat1/Srf/miR-133 regulatory loop | Malat1 transcript has a functional miR-133 target site, miR-133 acts as a competing endogenous RNA, regulating Malat1 levels | In vitro depletion of Malat1 in C2C12 cells reduces Srf activity, Srf is an enhancer of miR-133 expression; feed-back regulation loop involving miR-133 | Mouse myoblast C2C12 cells | [164] |
lncRNA MALAT1 | MALAT1 is overexpressed in 46% of ESCC tissues, primarily in high-stage tumors, high expression correlates with lymph node metastasis | In vitro depletion of MALAT1 suppresses tumor cell proliferation, cell migration and invasion; G2/M phase arrest was induced and the ratio of apoptotic cells increased | Human ESCC | [162] |
WIF1/lncRNA MALAT1 | WIF1 (strong tumor suppressor) is systematically downregulated in glioblastoma | WIF1 down regulation correlates with strong upregulation of MALAT1. In vitro depletion of MALAT1 suppresses tumor cell proliferation | Glioblastoma | [163] |
Downregulated miR-133b | ||||
Fascin-1 mRNA | FSCN1 upregulated | Reduced miR-133b | High-grade GIST tissue | [281] |
BCL-2 family (MCL-1 and BCL2L2) | MCL-1 and BCL2L2 upregulated | Reduced miR-133b | Lung cancer | [85] |
FAIM antiapoptotic protein and GSTP1 | miR-133b directly targets FAIM and GSTP1 | Downregulated miR-133b | miR-133b expression significantly downregulated in 75% of prostate cancer tumor specimens | [282] |
Gli1 | Gli1 upregulated | Gli1 inversely correlated with downregulated expression of miR-133b | Gastric cancer | [283] |
Bcl-w and Akt1 | Bcl-w and Akt1 proteins overexpressed significantly | miR-133b significantly downregulated | Bladder cancer tissues | [284] |
miR-133b downregulated in tumors compared to surrounding tissue | Gastric and esophageal adenocarcinomas | [285] | ||
Endometrial sarcoma, leiomyosarcoma, and mixed epithelial-mesenchymal tumors | [286] | |||
Downregulated miR-206 | ||||
Notch3/ miR-206 | Downregulated Notch3, blocking of the anti-apoptotic activity of Notch3 | Forced expression of miR-206 strongly induced apoptotic cell death via; also inhibited cell migration and focus formation | HeLa cells | [287] |
Met | Upregulated Met | miR-206 downregulated | Human rhabdomyosarcoma | [288] |
HGFR | Upregulated HGFR | miR-206 downregulated | Human breast cancer cells | [289] |
KLF4 | Upregulated KLF4 | miR-206 downregulated | RK3E breast epithelium cells | [108] |
KLF4; RAS-ERK signalling | Upregulated KLF4 promotes RAS-ERK signalling | miR-206 downregulated | TNBC cells | [290] |
Endogenous KLF4 binds the promoter regions stimulates expression of miR-206 | ||||
RASA1 and SPRED1 | miR-206 inhibits translation of the RAS pathway suppressors RASA1 and SPRED1 | Suppression of RASA1 or SPRED1 increased levels of GTP-bound, wild-type RAS and activated ERK 1/2 | ||
VEGF | VEGF upregulated in Laryngeal SCC tissues | MiR-206 strongly downregulated in LSCC tissues | Laryngeal SCC cancer tissue and cells | [113] |
VEGF | VEGF upregulated in ccRCC tissues | MiR-206 strongly downregulated in ccRCC tissues | ccRCC tissues assayed by Deep Sequencing | [291] |
Cdc42, MMP-2 and MMP-9 | Upregulated Cdc42, MMP-2 and MMP-9 | miR-206 downregulated | Human breast cancer tissues | [292] |
ERα | miR-206 directly targets ERα 3'-untranslated region | MiR-206 inhibited by ERα agonists, indicating a mutually (double) inhibitory feedback loop; | Estrogen stimulated breast cancer cell lines | [293] |
miR-206 downregulated | [109] | |||
Upregulated ERα | MCF-7 breast cancer cells | [294] | ||
ERα | Upregulated ERα | miR-206 downregulated | EEC tissue | [110] |
K-Ras | K-Ras is direct target of miR-206; | Low miR-206 potentiates metastases, and shorter overall survival | OSCC tissue samples and cell lines | [295] |
MiR-206 expression significantly downregulated and k-Ras upregulated on OSCC tissues | ||||
MiR-206 | Enforced upregulated of miR-206 attenuated cell proliferation, increased apoptosis and inhibited cell migration and invasion | MiR-206 strongly downregulated in lung cancer tissues | Lung 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 phenotype | EGFR signalling represses estrogenic responses in MCF-7 cells by enhancing miR-206 activity | miR-206 downregulates steroid receptor co-activators SRC-1 and SRC-3 and GATA-3 transcription factor, directly | MCF-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 stage | miR-206 expression strongly downregulated | Correlates with tumor progression | Human gastric cancer tissue | [297] |
CCND2 | miR-206 expression strongly downregulated | Correlates with upregulation of CCND2 and cancer progression | Human breast cancer | [298] |
Human gastric cancer | [299] | |||
MET | miR-206 expression strongly downregulated | Upregulation of MET | Papillary thyroid carcinoma | [300] |
Prognostic signature of metastatic colorectal cancer | miR-206 expression strongly downregulated | Prognostic signature of metastases: miRs 21, 135a, 335, 206 and let-7a | Metastatic 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-3 | miR-206 is a potent tumor supressor | p57 and Bax upregulated, and cleaved caspase-3 protein upregulated | Reduced apoptosis, and cell migration in HepG2 cells overexpressing miR-206 | |
STC2, HDAC4, KLF4, IGF1R, FRS2, SFRP1, BCL2, BDNF and K-ras | Exogenous upregulation of miR-206 expression; | STC2, HDAC4, KLF4, IGF1R, FRS2, SFRP1, BCL2, BDNF, and K-ras downregulated strongly in SCG-7901 cells overexpressing miR-206 | Gastric carcinoma SCG-7901 cells | [303] |
miR-206 is a potent tumor supressor | Reduced apoptosis, and cell migration in SCG-7901 cells overexpressing miR-206 | |||
Cyclin C, CCND1 and CDK4 | Cyclin C, CCND1 and CDK4 upregulated in melanoma tissue; | hsa-miR-206 downregulated in melanoma tissue | Human 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 protein | Silencing of coronin expression reduced tumor cell migration and altered the cellular actin skeleton and cell morphology, but did not effect cell proliferation | Downregulated 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 APOBEC3 | DND1 blocks miRNA interaction with 3'-UTR of specific mRNAs, restores protein expression; APOBEC3G binds DND1 counteracts repression and restores miRNA activity | APOBEC3G blocks DND1 to restore miR-206 inhibition of CX43 translation | Mouse cells | [306] |
Advanced clinical stage, T classification, metastasis and poor histological differentiation | Significant association with decreased miR-206 expression | Paired human osteosarcoma and normal adjacent tissues | [307] | |
Ellagic acid inhibits E2-induced mammary tumorigenesis | Reverses the downregulation of miR-206 | ACI model rat mammary tissue | [308] | |
Actin-like 6A (BAF53a), a subunit of the SWI/SNF chromatin remodeling complex | Elevated BAF53a | Downregulation of miR-206 | Primary rhabdomyosarcoma tumors | [309] |
Actin-like 6A (BAF53a) | BAF53a transcript is significantly higher in primary rhabdomyosarcomas than in normal muscle | Restoration 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 Lef1 | Exogenous upregulation of miR-206 expression | Inhibition of Wnt, Tbx3 and Lef1 activities | Estrogen receptor alpha (ER-α)-positive human breast cancer; developing mammary buds | [310] |
ANXA2 and KRAS | Stimulation of KRAS activity then induces NFKB1 expression; | Downregulated miR-206 in PDAC | PDAC tissues and cell lines | [311] |
Induces NFKB1 | Increased KRAS results in stimulation of cytokines CXCR2, CXCL1, CCL2, as well as CSF2 (GM-CSF) and VEGFC | Increased cell cycle progression, cell proliferation, migration and invasion | ||
Downregulated miR-1 and miR-133a | ||||
PNP | PNP upregulated | Reduced miR-1, -133a | Prostate cancer | [97] |
TAGLN2 | TAGLN2 upregulated | Reduced miR-1, -133a | RCC | [136] |
TAGLN2 and PNP | TAGLN2 and PNP upregulated | Reduced miR-1, -133a | MSSCC | [137] |
PTMA and PNP | PTMA and PNP upregulated | Reduced miR-1, -133a | Bladder cancer (TCC) | [312] |
LASP1 | LASP1 upregulated | Reduced 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 expression | Inverse correlation with TAGLN2 levels | CpG islands upstream of miR-1-133a hypermethylated | Colorectal 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 cancer | miR-1 and mir-133b are significantly downregulated in recurrent prostate cancer tissue specimens | Recurrent prostate cancer tissue | [96] | |
Downregulated miR-1 and miR-206 | ||||
NRF2 upregulated | Downregulated miR-1 and miR-206 expression | Upregulated expression of NRF2 induces increased expression HDAC4 | Primary lung adenocarcinoma; DU145 human prostate cancer cell line | [99] |
Loss of NRF2 | Decreased 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 expression | A549 human NSCLC cell line | [99] |
c-Met | c-Met upregulated | miR-1 and -206 downregulated | Human rhabdomyosarcoma | [89] |
ARPC5 and GSTP1 | ARPC5 and GSTP1 upregulated | Reduced miR-133a (and miR-206) | Lung SCC cell lines | [115] |
Downregulated miR-133a and miR-133b | ||||
PKM2 | PKM2 upregulated | Downregulated miR-133a, -133b | TSCC | [111] |
FSCN1 | FSCN1 upregulated | Downregulated miR-133a, -133b, (miR-145) | ESCC | [132] |
miR-133a, miR-133b downregulated | ESCC | [94] | ||
KRT7 | KRT7 upregulated | Downregulated (miR-133a and miR-133b) | Bladder cancer (TCC) and in vitro in BC KK47 cells | [315] |
Downregulated miR-1, miR-206 and miR-133 | ||||
myomiRs | Patient to patient variation in the up or down regulation of miR expression in both tumor and matched normal tissues | In 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 RCC | Each of miR-206, miR-1, miR-133b strongly downregulated | Restored expression strongly inhibited cancer cell proliferation, | RCC | [316] |
Shorter overall survival and disease-free survival | Correlated with increased downregulated of miR-133b and/or miR-206 | Both miR-133b and miR-206 significantly downregulated | Osteosarcoma tissues | [317] |
Cell invasion and metastasis | miR-1, miR-133a, miR-133b downregulated | miR-133a, miR-133b involved in invasion and metastasis | ESCC | [94] |
Factor(s) | Regulation | Regulator | Tissue/cell | Ref. |
Upregulated miR-133b | ||||
Activated p-ERK, pAKT1 cause in vitro proliferation of cervical cancer cell lines, and promote in vivo tumorigenesis and metastasis | Downregulation of MST2, CDC42, RHOA | Upregulated miR-133b | Human cervical carcinoma tissue compared to surrounding normal cervical tissue | [121] |
Decreased patient survival | Upregulated miR-133b | Progression bladder cancer | [122] | |
Androgen receptor | miR-133b directly represses CDC2L5, PTPRK, RB1CC1, CPNE3 | miR-133b directly upregulated by AR | Hormone-sensitive human prostate cancer (LNCaP) cells stimulated by androgen | [123] |
Activativated neuroendocrine neoplasia proliferation | Mutation in von Hippel-Lindau tumor suppressor, E3 ubiquitin protein ligase gene (VHL) | Upregulated miR-133b expression in VHL- deficient pheochromocytoma | Human pheochromocytoma (PCCs) and paraganglioma (PGLs) neuroendocrine neoplasias | [318] |
Upregulated miR-206 | ||||
Cell reprogramming factor KLF4 | KLF4 downregulated in colon cancer tissue, associated with increased miR-206 | miR-206 strongly upregulated in colon cancer tissues | Human colon cancer tissue | [116] |
Upregulated miR-1 and miR-133 | ||||
Decreased survival of R172 IDH2-mutated subset of CN-AML patients, increases resistance to chemotherapy | Distinctive gene and microRNA expression profiles accurately predicted R172 IDH2 mutations | Upregulated expression of miR-1 and miR-133 | De novo CN-AML patient bone marrow and blood samples | [151] |
EVI1 increases aggressive cancer growth | EVI1 expression upregulated in established patient samples | Upregulated expression of miR-1-2 and miR-133-a-1 | EVI1 expressing AML subset of patients | [120] |
ChIP assays show EVI1 binds to miR-1-2 gene promoter directly | ||||
CCND2 | miR-1 and miR-133a were specifically overexpressed in the cases with t(14;16) translocation, correlates with down-regulated CCND2 expression | Upregulated miR-1 and miR-133-a | Multiple myeloma | [152] |
Secreted myomiRs | ||||
miRs selectively released into serum (within exosome microparticles) | miR-1, miR-133a, and miR-133b selectively released | Human breast cancer | [319] | |
Circulating microRNA | Tumor-derived exosomes | Human non-small-cell lung cancer | [320] |
Up regulated cell factor | Down regulated cell factor | Cancer type | Ref. | |
Downregulated myomiRs | ||||
Downregulated miR-1 | ||||
Mediator complex subunit 1 (Med1) and 31 (Med31) upregulated | miR-1 downregulated | Osteosarcoma | [257] | |
Slug expression upregulated; enhanced cell migratory and invasive activities | miR-1 downregulated | Chordoma | [261] | |
Slug expression upregulated; stimulation of EMT process | miR-1 reduced increasingly with cancer progression | Prostate adenocarcinoma | [321] | |
Downregulated miR-133a | ||||
ARPC5 upregulated; | Downregulated miR-133a (> miR-206) | Lung SCC | [115] | |
HNSCC | [272] | |||
CAV1 upregulated; | miR-133a downregulated | HNSCC | [322] | |
Moesin upregulated; | miR-133a downregulated | HNSCC | [143] | |
FSCN1 upregulated; | miR-133a/ miR-133b downregulated | ESCC | [132] | |
Bladder cancer (TCC) | [274] | |||
GSTP1 upregulated; | miR-133a downregulated | HNSCC | [323] | |
Bladder cancer (TCC) | [144] | |||
Lung SCC | [115] | |||
LASP1 upregulated; | miR-133a downregulated in 83% of colorectal tumors | Colorectal 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 downregulated | Bladder cancer (TCC) | [313] | |
PKM2 upregulated; | miR-133a downregulated | TSCC | [111] | |
Moesin upregulated; | miR-133a downregulated | HNSCC | [143] | |
EGFR upregulated; | miR-133 downregulated | Hormone-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 downregulated | ESCC | [326] | |
Reduced miR-133a expression correlated significantly with advanced clinical stages, poor histological differentiation and lymph node metastasis | Marked downregulation of miR-133a in primary EOC tumors and OVCAR-3 cell line | Epithelial ovarian cancer (EOC), and in OVCAR-3 cell line | [327] | |
Downregulated miR-133b | ||||
FSCN1 upregulated; | miR-133a/-133b downregulated | ESCC | [132] | |
FSCN1 mRNA upregulated; | miR-133b downregulated | Progressive GIST | [281] | |
BCL2L2 upregulated; | miR-133b downregulated | Lung cancer | [85] | |
MCL1 upregulated; | miR-133b downregulated | Lung cancer | [85] | |
MET upregulated; | miR-133b downregulated | Colorectal cancer | [87] | |
MET protein upregulated; | miR-133b downregulated | high grade osteosarcoma tumor samples and cell lines | [328] | |
EGFR upregulated; | miR-133b downregulated | NSCLC | [105] | |
Multiple cell factors elevated; | miR-133b downregulated | Prostate cancer | [282] | |
FGFR1 downregulated; | miR-133b downregulated | Gastric cancer | [329] | |
Gli1 protein downregulated by miR133b, Gli1 target genes, OPN and Zeb2, are indirectly regulated | miR-133b downregulated | Gastric cancer | [283] | |
TAp63 supresses metastasis; downregulation target of miR-133b | miR-133b is a transcription target of TAp63, downregulated | Colon cancer cells | [330] | |
Chemokine (C-X-C motif) receptor 4 protein downregulated by miR133b; upregulated in advanced cancer | miR-133b downregulated | CRC | [331] | |
TBP-like 1 mRNA and protein are upregulated in CRC | miR-133b downregulated in CRC | CRC | [332] | |
Strong additional down regulation of miR-133b aids liver metastatic niche for CRC cells | miR-133b downregulated 3 × (significant) in liver metastasis compared to primary CRC | miR-133b downregulated in primary CRC compared to surrounding tissue | Metastatic 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 D1 | miR-133a and -133b downregulated | Gastric cancer | [334] | |
miR-133b target MMP-9 is upregulated | miR-133b downregulated | RCC | [335] | |
Downregulated miR-206 | ||||
ERα | ERα downregulates miR-206 | ERα-positive breast cancer; | [294] | |
miR-206 downregulated | Double feedback loop | [109] | ||
miR-206 downregulated | [293,336] | |||
ERα | miR-206 downregulated | EEC tissue | [110] | |
SRC-1, SRC-3 and GATA-3 proteins contribute to estrogenic signalling | miR-206 downregulated | ERα-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 translation | Breast cancer cells and normal cells | [108] | |
FGBP1 | miR-206 gene double knockdown | miR-206-/- mouse skeletal muscle. | [12] | |
VEGF upregulated | miR-206 downregulated | Laryngeal SCC cells | [113] | |
VEGF upregulated | miR-206 downregulated | CRC tumors compared to matched normal tissue; (1DS assay) | [337] | |
miR-206 correlates with negative ER status, negative PR status, and negative HER-2 status | Downregulated miR-206 | Breast cancer tumor tissue | [338] | |
miR-206 was downregulated in clinical TNBC tumor samples, one of its targets, actin-binding protein coronin was upregulated | Downregulated miR-206 associates with increased metastasis potential in breast cancers | High metastatic capacity TNBC tumors | [305] | |
Downregulated miR-1 and miR-133a | ||||
PNP upregulated | miR-1/miR-133a downregulated | MSSCC | [137] | |
Prostate cancer | [97] | |||
Bladder cancer (TCC) | [312] | |||
TAGLN2 upregulated; | miR-1/miR-133a downregulated | MSSCC; | [137] | |
RCC | [138] | |||
HNSCC | [339] | |||
Bladder cancer (TCC) | [93] | |||
PTMA upregulated | miR-1 and miR-133a downregulated | Bladder cancer (TCC) | [312] | |
Downregulated miR-1 and miR-206 | ||||
MET levels correlated inversely with miR-1/206 expression | miR-1/206 downregulated | Up-regulation of MET in rhabdomyosarcoma | [89,288] | |
HGFR upregulated | miR-1/206 downregulated | Breast cancer cells | [289] | |
G6PD; PGD; TKT; GPD2 upregulated | miR-1/206 downregulated | Primary lung adenocarcinoma | [99] | |
Upregulated myomiRs | ||||
Upregulated miR-133b | ||||
miR-133b | miR-133b strongly upregulated | MST1, CDC42, RHOA, and DUSP1 downregulated | Cervical carcinoma | [121] |
miR-133b | miR-133b is directly upregulated by AR | miR-133b represses CDC2L5, PTPRK, RB1CC1, and CPNE3 | PCa prostate cancer cell line | [123] |
Upregulated miR-206 | ||||
miR-206 | Strongly upregulated miR-206 | KLF4 downregulated | Human colon cancer tissue | [116] |
Upregulated miR-1 and miR-133a | ||||
miR-1-2 and miR-133-a-1 | Upregulated miR-1-2 and miR-133-a-1 | EVI1 (transcriptional activator of miR-1 and miR-133b) | AML | [120,151] |
miR-1 and miR-133-a | Upregulated miR-1 and miR-133-a | Downregulated CCND2 | Multiple myeloma | [152] |
Up-regulation of exogenous myomiR expression in cell lines | ||||
Reduced cell proliferation | Estrogen receptor alpha | Overexpression of miR-206 has an inhibitory effect on cell proliferation | ERα-positive breast cancer cells over expressing mir-206 | [289] |
miR-133b | GSTP1 downregulated | Transgenic miR-133b overexpression | HeLa cervical cancer cells | [282] |
miR-133b | FAIM downregulated | Transgenic miR-133b overexpression | HeLa cervical cancer cells | [282] |
Apoptosis increased | TNFα-induced cell death is activated | Transgenic miR-133b overexpression | HeLa cervical cancer cells | [282] |
Increased cell proliferation and migration | Downregulation of MST2 | Transgenic miR-133b overexpression | CaSki cervical cancer cells | [121] |
Downregulation of CDC42 | ||||
Downregulation of RHOA | ||||
Increased cell proliferation and migration | Indirect upregulation of p-AKT1 activity | Transgenic miR-133b overexpression | CaSki cervical cancer cells | [121] |
Indirect upregulation of p-ERK activity | ||||
RB1CC1 downregulated | Exogenous upregulation of miR-133b; | miR-133bm promotes cell apoptosis, but suppressed cell proliferation and cell-cycle progression in aggressive PC-3 cells | PC3 prostate cancer cell line | [106] |
miR-133b directly targets RB1CC1 in LNCaP cells | In contrast in low-aggression LNCaP cells, miR-133b stimulate cell proliferation and cell-cycle progression, but inhibit apoptosis | Hormone sensitive prostate cancer LNCaP cell line | ||
Cell proliferation decreased and apoptosis increased | Met, Twf1 and Ets1 and Bag4 activities downregulated | miR-1 expression is lower in mouse cSCCs compared to normal skin | Mouse cutaneous squamous cell carcinomas (cSCCs); A5 and B9 cSCCcell lines | [256] |
Transgenic miR-1 overexpression | ||||
Ets1 proto-oncogene | Repression of Ets1 expression inhibited HepG2 cell invasion and migration | Transgenic miR-1 overexpression | HCC HepG2 cells | [340] |
lncRNA UCA1 | Knockdown of lnc UCA1 expression phenocopied the effects of upregulation of hsa-miR-1 | hsa-miR-1 decreased the expression of lnc UCA1 in bladder cancer cells in an Ago2-slicer-dependent manner | Human bladder cancer (TCC) cells | [156] |
NOTCH3 signalling | miR-206 had a direct inhibition of NOTCH3 signalling and indirect interaction with other signalling pathways via CDH2 and MMP-9 | miR-206 upregulation blocks the cell cycle, inhibits cancer cell proliferation and migration and activates cell apoptosis | SW480 (plus its metastatic strain) and SW620 colon cancer cell lines | [341] |
FSCN1 | miR-133b targets FSCN1 in GC cells; the direct knockdown of FSCN1 can also inhibit GC cell growth and invasion | Up regulation of miR-133b in GC cells inhibits cell proliferation, cell migration and invasion | miR-133b is significantly downregulated in GC tissues compared with adjacent normal tissues, as well as in GC cell lines | [342] |
FSCN1 | miR-133a targets FSCN1 in CRC cells; | Up regulation of miR-133a expression and downregulation of FSCN1 protein expression both suppress colorectal cancer cell invasion | miR-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 |
- Citation: Mitchelson KR, Qin WY. Roles of the canonical myomiRs miR-1, -133 and -206 in cell development and disease. World J Biol Chem 2015; 6(3): 162-208
- URL: https://www.wjgnet.com/1949-8454/full/v6/i3/162.htm
- DOI: https://dx.doi.org/10.4331/wjbc.v6.i3.162