Vous êtes sur la page 1sur 12

Review

Crosstalk between TGF-b signaling and


the microRNA machinery
Henriett Butz1, Karoly Racz1, Laszlo Hunyady2 and Attila Patocs3,4
1

2nd Department of Medicine, Faculty of Medicine, Semmelweis University, Budapest, Hungary


Department of Physiology, Semmelweis University, Budapest, Hungary
3
Molecular Medicine Research Group, Hungarian Academy of Sciences, Budapest, Hungary
4
Department of Laboratory Medicine, Semmelweis University, Budapest, Hungary
2

The activin/transforming growth factor-b (TGF-b) pathway plays an important role in tumorigenesis either by
its tumor suppressor or tumor promoting effect. Loss of
members of the TGF-b signaling by somatic mutations
or epigenetic events, such as DNA methylation or regulation by microRNA (miRNA), may affect the signaling
process. Most members of the TGF-b pathway are
known to be targeted by one or more miRNAs. In addition, the biogenesis of miRNAs is also regulated by TGFb both directly and through SMADs. Based on these
interactions, it appears that autoregulatory feedback
loops between TGF-b and miRNAs influence the fate
of tumor cells. Our aim is to review the crosstalk between TGF-b signaling and the miRNA machinery to
highlight potential novel therapeutic targets.
Members and functions of the TGF-b signaling pathway
The activin/TGF-b family consists of evolutionarily conserved polypeptides, which play prominent roles in the
regulation of embryonic development, reproduction and
tumor formation [1]. TGF-b signaling is often considered
a pathogenic factor in tumorigenesis due to its tumor
suppressor or tumor promoting effects (see below). Altered
TGF-b signaling has been frequently shown in different
tumor types, including breast, prostate, endometrial, colorectal, thyroid, parathyroid and pancreas neoplasms [2].
The TGF-b family comprises more than 35 members,
including TGF-bs, bone morphogenetic proteins (BMPs),
growth differentiation factors (GDFs), activins, inhibins,
Mullerian inhibiting substance (MIS), Nodal and leftys [1].
They are secreted in an inactive form, and TGF-bs become
active after cleavage of the N-terminal pro-region, referred
to as the latency-associated peptide (LAP). Until this
cleavage occurs, the LAP-associated TGF-bs (L-TGF-bs)
cannot interact with their receptors, and hence they are
biologically inactive [1]. The active TGF-b molecule is a
dimer composed of two TGF-b molecules linked by a disulfide bridge between the ninth cysteine of each monomer [1].
TGF-b receptors are serine/threonine kinase receptors
and are divided into three groups: type I, type II and type
III. In mammals there are seven different members of type
I (ALK1ALK7) and five members of type II receptors
(ACVR2, ACVR2B, AMHR2, BMPR2, TGF-bR2). Upon
ligand binding, an active ligand-type I/type II receptor
Corresponding author: Patocs, A. (patatt@bel2.sote.hu).
Keywords: TGF-b signaling pathway; miRNA; endocrine neoplasm.

382

complex is formed, type II receptors activate type I receptors by phosphorylation, and type I receptors subsequently
phosphorylate downstream SMAD proteins which transmit the signal to the nucleus (Figure 1). Type III receptors
(betaglycan and endoglin) have a higher molecular weight
than type I and type II receptors. Betaglycan is a membrane-anchored proteoglycan that can bind TGF-bs and
facilitates interaction of TGF-b-type II receptor with TGFb [1]. It can also promote binding of inhibin to type II
receptor, thereby antagonizing activin signaling. Endoglin
is a membrane glycoprotein with a large extracellular
domain containing an integrin recognition motif and a
short cytoplasmic tail with serine and threonine residues,
which can be phosphorylated by the TGF-b receptors.
Endoglin phosphorylation seems to play a regulatory role
for ALK1-dependent endothelial cell growth and adhesion,
which is confirmed by the findings that endoglin was found
to be overexpressed in several tumor types [3,4].
Tumor suppressor and tumor promoting effects of the
TGF-b pathway
TGF-b may inhibit cell proliferation at multiple levels.
Well-known tumor suppressors, such as p15Ink4b and
p21Waf1 are induced by TGF-b signaling, whereas oncogenic factors such as c-Myc, a transcription factor that promotes cell proliferation, and Id proteins, nuclear factors
that inhibit cell differentiation, are repressed via TGF-b
signaling [2]. TGF-b can also activate apoptosis [5]. This
function is mediated by its downstream targets, such as
death-associated protein kinase (DAPK), growth arrest
and DNA-damage inducible b (GADD45b), and Bcl2-like
11 (BCL2L11 or BIM). TGF-b signaling also inhibits tumor
growth by repressing hepatocyte growth factor (HGF),
macrophage-stimulating protein (MSP) and TGF-a [2].
In addition to its tumor suppressor effects, TGF-b signaling has tumor promoting downstream targets. For
example, through induction of deleted in esophageal cancer 1 (DEC1), platelet-derived growth factor beta polypeptide (PDGF-B), protein snail homolog 1 (SNAIL) and high
mobility group AT-hook 2 (HMGA2), TGF-b signaling may
mediate antiapoptotic effects, growth stimulation and epithelialmesenchymal transition (EMT). EMT is a biological process through which a polarized cell that normally
interacts with a basement membrane (epithelial phenotype) switches to a mesenchymal phenotype that is characterized by invasiveness and increased cell mobility [6].

0165-6147/$ see front matter 2012 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.tips.2012.04.003 Trends in Pharmacological Sciences, July 2012, Vol. 33, No. 7

Review

Trends in Pharmacological Sciences July 2012, Vol. 33, No. 7

TGF-/activin

TGFBR II

Smad
2/3

TGFBR I

SARA

Smad
2/3
miRNAs

Smad4

miRNA processing

Smurf1/2
Smad4
Smad
2/3

Smad3

sm

Nucleus

p68

Cytopla

Smad4
Smad
P
2/3

p72

pre-miRNA

DGCR8
Drosha
p68

p72

DGCR8
Drosha

Smad4

m7G

AAAA

pri-miRNA

Smad4
Smad TF
2/3

SBE

Smad4 TF
P Smad2/3

TGF-specific genes

SBE

TGF-specific miRNA genes

miRNA genes
TRENDS in Pharmacological Sciences

Figure 1. Outline of the crosstalk between members of the canonical TGF-b signaling and the miRNA machinery. The TGF-b/activin binds to its receptor. Through ligand binding
the type II receptors phosphorylate (hence activate) type I receptors. This complex then intracellularly activates SMAD molecules (R-SMADs) by phosphorylation. Binding of RSMADs to the type I receptor is mediated by the protein named SARA (SMAD anchor for receptor activation). SMAD2 and SMAD3 are TGF-b-specific, whereas SMAD1, SMAD5
and SMAD8 are BMP-, AMH- and GDF-specific. After activation by TGF-b, SMAD2/3 interacts with SMAD4 and this complex translocates to the nucleus. Here SMAD complex can
interact with other cofactors and transcription factors, and binds to specific DNA sequences, referred to as SBE (SMAD-binding element), in promoters of TGF-b target genes.
Among SMAD family members, SMAD6 and SMAD7 have inhibitory roles (I-Smad). SMAD6 preferentially inhibits phosphorylation of SMAD1/5/8 via BMP type I receptor,
whereas SMAD7 in a complex with Smurf1/2 (E3 ubiquitin ligases) translocates from nucleus and associates with TGF-b/activin type I receptor causing its degradation. SMAD7
can inhibit both the TGF-b and BMP signaling pathways [1,2,10]. TGF-b target genes include genes encoding both proteins and miRNAs (left lower part of the figure). Red and
green arrows indicate the connections between TGF-b signaling and biogenesis of miRNAs. In the nucleus, SMAD3 can interact with Drosha, a member of miRNA
microprocessing complex, and can enhance processing of both the T/B miRNAs (see details in the text) and miRNAs transcribed from non-T/B miRNA genes (right lower part of
the figure). After the pre-miRNA mature miRNA process, mature miRNA can target several members of TGF-b signaling (highlighted as red combs).

During this process, basement membrane degradation


occurs, cells lose E-cadherin and produce vimentin, a
mesenchymal cell-specific intermediate filament. EMT
has key roles in embryogenesis and wound healing, and
has also been described as a crucial mechanism for the
acquisition of malignant phenotypes of epithelial cells [7].
Various factors are reported to mediate TGF-b-induced
EMT, including SNAIL1, SNAIL2 (Slug), ZEB1, ZEB2
(SIP1). These are transcriptional repressors that bind to
E-box motifs of the DNA and repress transcription of
various genes, including E-cadherin [2].
It is generally accepted that TGF-b signaling exerts
tumor suppressor effects during the early phase of tumorigenesis and, in certain situations, it switches from a
tumor suppressor to a tumor promoter [2]. For the tumor

promoting effect, additional secondary molecular mechanisms, including mutation of the TP53 gene encoding tumor
suppressor p53 [8] or Ras activation [9], are also involved.
Ras enhances induction of SNAIL by TGF-b; however, this
process is also regulated by glycogen synthase kinase-3b
(GSK-3b). GSK-3b is apparently one of the major mediators
of environment-dependent TGF-b-induced responses, as a
downstream collector of multiple signaling pathways, including mitogen-activated protein kinase (MAPK), PI3K/
Akt and Wnt [2].
In addition to the effects of genes encoding members
of the TGF-b signaling, regulation of TGF-b expression
by epigenetic mechanisms via DNA methylation or miRNAs has also been demonstrated in different tumors
[10,11].
383

Review

Trends in Pharmacological Sciences July 2012, Vol. 33, No. 7

Canonical miRNA biogenesis

RNA Pol II

Non-canonical steps

RNA Pol II

Individual miRNA gene

RNA Pol II

miRtron

Exon1

miRNA

miRNA

Exon2

miRNA genes in cluster

miRNA1

miRNA2

miRNAn

Transcription

Transcription
Exon1

Exon

m7 G

AAAA
pri-miRNA

m7G

AAAA

pri-miRNA

DGCR8
Drosha

Nucleus

Splicosome

Processing

Splicing

Microprocessor
complex

m7 G

Exon1

Exon2

AAAA

Host gene mRNA

Cytoplasm

pre-miRNA

Ran GTP
Exportin 5

AGO2
Cleavage

pre-miRNA
pre-miRNA
ac-pre-miRNA

TRBP
Dicer

Cleavage

miRISC

Incorporation
to miRISC

Passenger strand degradation


miRNA*

miRNA:miRNA* duplex

Mature miRNA

Targeting mRNA

AAAA

Ribosome

AGO2
3 UTR

Translational repression,
mRNA degradation by cleavage or deadenylation
TRENDS in Pharmacological Sciences

Figure 2. Biogenesis and function of miRNAs. Genes encoding miRNAs can be located in the genome individually or in clusters (upper part of the figure) of noncoding
sequences, or in introns of protein-coding genes (called miRtrons) [74]. miRNA clusters are transcribed together. Both individual and clustered miRNA genes are
transcribed by RNA polymerase II. They have a 7-methyl guanosine cap and are polyadenylated similar to mRNA molecules [117119]. The primary transcript (primary
miRNA, pri-miRNA) is processed by an RNase III (Drosha) containing complex [120]. Pri-miRNAs are processed by an RNase III (Drosha) containing complex. Drosha cleaves
both strands into a 6070 nucleotide precursor-miRNA (pre-miRNA), which has hairpin secondary structure. The microprocessor complex contains an RNA-binding
protein (DGCR8 or Pasha) and other components including DEAD box helicases p68 and p72 [120]. The pre-miRNA molecule is transported to the cytoplasm by Exportin-5
[121] and processed by another RNase III enzyme (Dicer) complexed with transactivation-responsive RNA binding protein (TRBP). Dicer cleaves pre-miRNA into 21 nt
miRNA:miRNA* duplexes [122]. One strand of this RNA duplex (guide strand or matured miRNA) is incorporated into miRNA-induced silencing complex (miRISC), whereas
the other strand (passenger strand or miRNA*) is usually degraded [123]. However, recent data suggest that miRNA* could also be loaded into miRISC, which has functional

384

Review
Biogenesis and function of miRNAs
miRNAs are approximately 1925 nucleotides long (with
an average of 22 nucleotides), noncoding RNA molecules
that post-transcriptionally regulate gene expression via
RNA interference by binding either to the 30 UTR or
50 UTR or the coding sequence of protein-encoding
mRNAs [1216] (Figure 2). Approximately 3050% of
all protein-coding genes might be controlled by miRNAs
[17,18]. One miRNA has the potential to affect the expression of several proteins, and one protein is influenced
by several miRNAs. As the expression pattern of the
miRome is highly tissue-specific, miRNAs provide finetuning of protein expression level that renders them celland context-specific regulators of the adaptation process
[19]. As miRNAs may influence many different mRNAs,
they can participate in the regulation of several physiological and pathological cellular processes. Their roles
have already been considered in development, cell proliferation, differentiation, apoptosis and tumorigenesis
[20,21].
Experimentally validated interactions between TGF-b
signaling and miRNAs suggest that miRNAs influence the
TGF-b pathway at multiple levels. In addition, TGF-b
signaling itself enhances the maturation of miRNAs
[22], resulting in a bidirectional functional link.
Regulation of the TGF-b signaling by miRNAs via direct
interaction with downstream members of canonical
signaling pathways
Most, if not all, members of the canonical TGF-b signaling
pathway may be influenced by miRNAs. Using in silico
miRNAmRNA target predictions, several possible interactions can be obtained. However, these predictions always
need to be confirmed experimentally. Owing to the lack of
high-throughput screening (HTS) methods for monitoring
miRNAmRNA interactions, few such interactions have
been demonstrated to date (Table 1). Starting from TGF-b
receptors, it has been shown that TGF-b type 1 receptor
(TGF-bR1) and SMAD2 were upregulated in most primary
anaplastic thyroid carcinoma-derived cells, whereas miRNAs (miR-30 and/or miR-200 families) potentially targeting these molecules were downregulated [23]. Inhibition of
the TGF-bR1 in these cells induced EMT and a concomitant increase of the miR-200 family, suggesting their role
in TGF-b-mediated EMT.
In acute promyelocytic leukemia (APL) cells, miR-146a
was downregulated by all-trans-retinoid acid treatment
during APL differentiation. This miRNA may possibly
influence cell proliferation in this cell line via SMAD4 [24].
In addition to tumorigenesis, TGF-b signaling also plays
an important role in the development of several organs.

Trends in Pharmacological Sciences July 2012, Vol. 33, No. 7

Regulation of the TGF-bR1 by let-7 may modulate and


control TGF-b signaling activity to the necessary level at
each developmental stage [25]. The miR-23b cluster (including miR-23b, miR-27b and miR-24-1) was found to
repress bile duct gene expression in fetal hepatocytes
through downregulation of SMADs (SMAD3, SMAD4
and SMAD5), but low levels of the miR-23b cluster was
required in cholangiocytes to allow TGF-b signaling
necessary for bile duct formation [26].
TGF-b has also been implicated in regulation of fibrogenesis. In the heart, downregulation of miR-133 and miR590 via TGF-b1 and TGF-bR2 contribute to the enhancement of TGF-b signaling [27], whereas in the liver and
lung, miR-21 targeting the negative regulator SMAD7 can
also enhance TGF-b signaling [28,29]. Structural remodeling in vascular smooth muscle cell (VSMC) can lead to
atherosclerosis or abdominal aortic aneurysm. In this
process, miR-26a was found to be a potential pathogenic
factor by altering TGF-b signaling through direct targeting
of SMAD1 [30]. miR-141 and miR-200a directly inhibit
TGF-b2 in rat proximal tubular epithelial cells (NRK52E),
and their downregulation may be responsible for the development and progression of TGF-b-dependent EMT and
fibrosis [31].
SMAD3 has also been found to be a potential miRNA
target in stem cells and in the pituitary. Interestingly, five
of seven miRNAs that negatively correlated with tumor
size in pituitary adenomas have been potentially predicted
to target SMAD3, and among them miR-140 was already
validated experimentally [11,32].
Regulation of TGF-b signaling by miRNAs that interact
directly with TGF-b target genes
TGF-b target genes can also be regulated by miRNAs
(Figure 3a). The miR-106b/25 cluster (miR-106b, miR-93
and miR-25) was found to be upregulated and correlated
with the loss of tumor suppressor activity of TGF-b signaling. These miRNAs directly target the cell cycle inhibitor
p21Waf1/Cip1 and the pro-apoptotic protein BCL2L11 (BIM)
in gastric cancer by interfering with TGF-b-induced cell
cycle arrest and TGF-b-mediated apoptosis [5]. The miR106b/25 cluster accumulates prostate and pancreatic cancers, neuroendocrine tumors, neuroblastoma and multiple
myeloma. In B cells, BIM expression is also affected by a
well-characterized oncogenic miRNA cluster, miR-17/92
[33]. This cluster impairs TGF-b effects not only by targeting individual TGF-b responsive genes as p21Waf1/Cip1 and
BIM but also by targeting canonical TGF-b signaling
molecules (TGF-bR2, SMAD2, SMAD4) [34,35].
As discussed above, miRNAs regulate EMT by targeting
ZEB1, ZEB2 (see also below), SNAIL1, SNAIL2 and

consequences in certain cases [124,125]. This mechanism is called canonical miRNA processing. There are two noncanonical steps in the miRNA maturization process.
Some pre-miRNAs are transcribed from very short introns (called miRtrons) as a result of splicing and debranching [126]. In the cytoplasm some of these pre-miRNAs are
cleaved by AGO2, an argonaute protein into AGO2-cleaved precursor miRNA (ac-pre-miRNA). The single-stranded matured miRNA directly associates with argonaute
proteins (in mammals AGO14), which are core components of miRISC. Here miR interacts with 30 UTR of its target mRNA by base-pairing and represses expression of the
targets. In this process the seed region of miRNA is essential, although there is evidence that the central loop region of miRNA is also involved in the target determination
[127]. The seed region is defined as the consecutive stretch of approximately seven nucleotides starting from either the first or the second nucleotide at the 50 end of the
miRNA molecule. The mechanistic details of miRNA-mediated translational repression are not fully understood. Of the numerous factors that influence pairing, each
predicted miRNAmRNA target pair needs to be experimentally verified because a simple, high-throughput method for biological validation of miRNA targets does not
exist. Although several studies revealed spatial or temporal avoidance of miRNA coexpression with target genes, these may support the negative correlation between
miRNA and its potential target mRNA expression strengthening target pairing [8,22]. However, the negative correlation between mRNA and miRNA expression is not
exclusive because experimentally valid targets can be found even without changes in mRNA expression [32]. The repression of the target is realized by three major
processes: mRNA cleavage by AGO2, mRNA degradation by deadenylation and inhibition of different steps of the translation process [128132].

385

Review

Trends in Pharmacological Sciences July 2012, Vol. 33, No. 7

Table 1. Regulation of members of canonical TGF-b signaling by miRNAs


Name of miRNA
miR-133
miR-744
miR-200a
miR-141
miR-210
miR-21

let-7
miR-200 family,
miR-141
miR-106b,
miR-93
miR-17-5p,
miR-20
miR-204
miR-20a
miR-21
miR-21
miR-590
miR-155
miR-26
miR-26a
miR-199a
miR-26a
miR-141,
miR-200a,c,
miR-30d,e
miR-155
miR-140
miR-23b cluster
(miR-23b, -27b, -24)
miR-146a
miR-146b-5p
miR-18
miR-224
miR-130a
miR-124
miR-155

miRNA target
TGF-b1
TGF-b1
TGF-b2
TGF-b2
AcvR1B (ALK4)
TGF-b components
(TGF-bR2, TGF-bR3;
DAXX, BMPR2)
TGF-bR1
TGF-bR1

Tissue/cell where this miRNAmRNA regulation was demonstrated


Atrial fibroblast cells
Proximal tubular epithelial cells (HK-2)
NRK52E kidney proximal tubular epithelial cells
NRK52E kidney proximal tubular epithelial cells
ST2 bone marrow-derived stromal cells
U251and U87glioblastoma cells

Species
Dog
Human
Rat
Rat
Human
Human

Refs
[27]
[133]
[31]
[31]
[60]
[58]

Human
Human, rat

[25]
[23,134]

TGF-bR2

Adult and 912 week human embryonic liver tissues


Anaplastic thyroid carcinoma tissues and ATC-derived cells,
proximal tubular epithelial cells (NRK52E)
SH-SY5Y neuroblastoma cells, embryonic fibroblasts (MEF)

Human, mouse

[135,136]

TGF-bR2

HCT116 and DLD1 colon carcinoma cell line

Human

[81]

TGF-bR2
TGF-bR2
TGF-bR2

hfRPE human fetal retinal pigment epithelial cells


Lung cancer tissues
Myometrial smooth muscle cells, leiomyoma smooth muscle cells,
transformed LSMC and SKLM-S1 leiomyosarcoma cell line
hADSC human adipose tissues derived stem cell
Atrial fibroblast
Burkitts lymphoma cell line (Mutu I)
HeLa S3
hADSC human adipose tissues derived stem cell
Pluripotent C3H10T1/2 stem cells
Vascular smooth muscle cells

Human
Human
Human

[36]
[137]
[57]

Human
Human
Human
Human
Human
Human
Human

[56]
[27]
[73]
[138]
[139]
[140]
[30]

Anaplastic thyroid carcinoma tissues and ATC-derived cells

Human

[23]

SMAD2
SMAD3
SMAD3, -4, -5

THP1 monocyte cell line


Pluripotent C3H10T1/2 stem cells
HBC-3 fetal mouse liver stem cell

Human
Human
Mouse

[70]
[32]
[26]

SMAD4
SMAD4
SMAD4
SMAD4
SMAD4
SMAD5
SMAD5

NB4 cell (acute promyelocytic leukemia cell) and dermal fibroblast


Papillary carcinoma cell lines (TPC-1 and BCPAP)
HCT116 and DLD1 colon carcinoma cell line
Preantral granulosa cells (GCs)
HEK-293 kidney, A549 lung, 32Del3 myeloid precursor cell line
HeLa S3
Diffuse large B cell lymphoma and Burkitts lymphoma
cell line (Mutu I)
Lungs of mice with bleomycin-induced fibrosis and
lungs of patients with idiopathic pulmonary fibrosis
HCV-infected human liver tissues

Human
Human
Human
Mouse
Human
Human
Human

[25,141]
[142]
[63]
[138]
[143]
[144]
[71,72]

Human, mouse

[29]

Human

[28]

TGF-bR2
TGF-bR2
SMAD1
SMAD1
SMAD1
SMAD1
SMAD1
(SMAD2/3/4 reporter)
SMAD2

miR-21

SMAD7

miR-21

SMAD7

HMGA2. TGF-bR2 and SNAIL2 are direct targets of miR204. The expression of this miRNA was significantly lower
in the NCI60 tumor cell line panel than in normal tissues
[36]. In addition, SNAIL2 is an essential mediator of EMT,
which underlines the pathogenic role of miR-204 in promoting tumor dissemination [37]. The HMGAs are low
molecular weight, nonhistone chromosomal proteins that
interact with the minor groove of many AT-rich promoters
and enhancers, and mutations of the HMGA gene associates with many common diseases, including benign and
malignant tumors [38,39]. HMGA2 was found to cooperate
with the TGF-b pathway in regulating the expression of
SNAIL1 and SNAIL2 [40,41]. There is a physical interaction between HMGA2 and SMAD molecules, leading to an
increased binding of SMADs to the SNAIL1 promoter [41].
Because SNAIL1 is a master downstream effector of
386

HMGA2 during induction of EMT, miRNAs may also


influence EMT by regulating HMGA2. Indeed, the frequently downregulated miRNA let-7 targets HMGA2
resulting in its overexpression, which has already been
demonstrated in numerous tumors, including ovarian carcinoma, retinoblastoma, uterine leiomyosarcoma, neuroendocrine tumors and pituitary adenomas [4246]. In
these tumors, expression of HMGA proteins is associated
with malignant phenotypes and poor prognosis.
TGF-b alters miRNA expression directly and by
regulating the maturation process of miRNAs
As described above, miRNAs directly regulate the expression of members of the canonical TGF-b signaling and the
expression of TGF-b target genes. However, TGF-b itself
can alter the expression of numerous miRNAs through

Review

Trends in Pharmacological Sciences July 2012, Vol. 33, No. 7

(a)

(b)

TGF-
signaling

c-MYC

Drosha

Tumor suppression
c-MYC

p21
p15

Apoptosis

GADD45b
DAPK
PDCD4
BIM

EMT
DEC1
PDGFB
HMGA2
SNAIL
GAM

miR-106b/25
miR-17/92

miR-223
miR-429

miR-183

miR106b/25
miR-17/92

miR-17/92

miR-204

let-7

TGFBRII TGF-
SMAD4 signaling

ZEB1, ZEB2, p21, BIM


TGF- downstream targets
TRENDS in Pharmacological Sciences

Figure 3. Schematic representation of the role of the TGF-b pathway in tumorigenesis. (a) Main downstream effectors of the TGF-b signaling and its fine-tuning by miRNAs
(detailed in the text). Green arrows indicate downregulation and red arrows show upregulation. (b) Schematic regulatory feedback loop involving TGF-b signaling (C-MYC,
ZEB1, ZEB2, p21, BIM), Drosha, miR-17/92 cluster and GAM.

binding of p68, a component of the Drosha microprocessor complex. These issues are discussed in the following
sections.
Changes in miRNA expression levels after TGF-b
treatment
Upon TGF-b treatment, changes in the expression of numerous miRNAs have been detected in different cells
(Table 2).
The miR-200 family (miR-200a, miR-200b, miR-200c,
miR-141 and miR-429) and miR-205 were markedly downregulated in response to TGF-b treatment in kidney
(MDCK) and rat proximal tubular epithelial cells
(NRK52E) [31]. These miRNAs regulate the expression
of the E-cadherin transcriptional repressors, ZEB1 and
ZEB2, which are implicated in EMT and tumor metastasis
[47,48]. In murine mammary epithelial cells (NMuMG),
expression of the miR-200 family was lost after induction of
EMT by TGF-b stimulation [47]. Enforced expression of the
miR-200 family alone was sufficient to prevent TGF-binduced EMT, and its inhibition was sufficient to induce
EMT in a process requiring upregulation of ZEB1 and/or
ZEB2 [49]. However, miR-200a and miR-141 directly target TGF-b and TGF-bR1 in mesenchymal anaplastic thyroid carcinoma-derived and NRK52E cells [23,31]. ZEB1
directly suppresses transcription of miR-141 and miR200c, and triggers a miRNA-mediated feed-forward loop,
which stabilizes EMT and promotes invasion of cancer cells
[50]. This network, with the miR-200 family in the center,
contributes to the regulation of EMT in an environmentally-dependent manner [48].
TGF-b signaling can also regulate the expression of a
subset of miRNAs via transcription regulation by SMAD3.
miRNA let-7d was repressed in tissue samples of patients
with idiopathic pulmonary fibrosis (IPF) [51], and miR-24
was also repressed in C2C12 cells leading to reduced
expression of myogenic differentiation markers [52].

An onco-miRNA, miR-21 was found to be upregulated by


TGF-b treatment in breast cancer and proliferating tubular epithelial cells (TECs) [5355], and its direct interaction with TGF-bR2 was also demonstrated [56,57].
Downregulation of miR-21 in glioblastoma cells caused
growth repression, increased apoptosis and cell cycle arrest
[58]. miR-21 was also among those miRNAs (miR-21, miR32, miR-137, miR-346, miR-136, miR-192, miR-210, miR211), which were suggested to participate in the regulation
of EMT [59].
In addition to its role in tumorigenesis, upregulation of
miR-21 was also detected in myofibroblasts obtained both
from lungs of mice having bleomycin-induced fibrosis and
patients with IPF [29].
Another miR, miR-210, was also found to be overexpressed after BMP4 administration and was considered to
act as a positive regulator of osteoblastic differentiation by
inhibiting TGF-b signaling through inhibition of ACVR1B
(ALK4) [60].
As mentioned above, TGF-b1 is a key mediator of fibrotic diseases. The pathomechanism of this process includes
SMAD7, a direct target of mir-21. Upregulation of miR-21
in primary pulmonary fibroblasts inhibits SMAD7, and
inhibition of this inhibitory SMAD7 results in enhanced
TGF-b signaling [28,29]. SMAD3 also regulates miR-192
by binding to its promoter and hence participating in the
regulation of renal fibrosis [61].
Members of the miR-17/92 cluster implicated in tumorigenesis (miR-17, miR-18a, miR-19a, miR-20a, miR-19b-1,
miR-92a-1) were upregulated by TGF-b administration in
several cell lines (Table 2) [62]. However, miR-17/92 directly targets TGF-bR2 and SMAD4, and thus it participates in the regulation of an autoregulatory feedback loop
similar to that reported between miR-200a and TGF-b/
TGF-bR1 [63]. In this loop, a zinc finger protein 512B
which is also known as GM632 or GAM (GAM/ZFp/
ZNF512B), a vertebrate-specific zinc finger factor, has a
387

Review

Trends in Pharmacological Sciences July 2012, Vol. 33, No. 7

Table 2. miRNAs regulated by TGF-b administration


Name of miRNA
let-7d
miR-142-3p
miR-145
miR-145
miR-146a
miR-155

Direction of miRNA
expression changes
Decreased
Elevated
Decreased
Elevated
Elevated
Elevated

miR-155
miR-17/92 cluster

Decreased
Elevated

miR-18
miR-181b
miR-192

Elevated
Elevated
Elevated

miR-200a/b
miR-200a/b/c
miR-205;
miR-200 family
miR-206
miR-21

Decreased
Decreased
Decreased

miR-216/217
miR-216a
miR-224
miR-23a/27a/24 cluster
miR-24
miR-24

miR-24
miR-27b
miR-29

Elevated
Elevated
Elevated
Elevated
Decreased
Elevated for short time,
decreased for long time
treatment
Elevated
Decreased
Decreased

miR-34
miR-451

Decreased
Elevated

Decreased
Elevated

Tissue/cell

Species

Refs

Idiopathic fibrosis pulmonary tissue


Limb primary mesenchymal cells
Mesenchymal stem cells
Coronary artery smooth muscle cell
Langerhans cells
Normal mouse mammary gland epithelial
cells (NMUMG)
Lung fibroblasts
HEK-293 kidney, HepG2 liver, MCF7 breast
cancer cell line
HeLa (cervix epithelial adenocarcinoma)
Hepatocellular carcinoma, breast cancer
Human kidney tubular epithelial cells
Mouse mesangial cells (MMCs)
Rat proximal tubular epithelial cells (NRK52E)
Gastric cancer cell line
Proximal tubular epithelial cells (NRK52E)
Mesenchymal cells

Human
Chicken
Murine
Human
Human
Mouse

[51]
[145]
[146]
[147]
[148]
[68]

Human
Human

[69]
[62]

Human
Human
Human
Mouse
Rat
Human
Rat
Human

[149]
[150,151]
[61,152,153]

C2C12 myoblasts
Breast cancer,
human proliferating tubular epithelial cells (TECs),
rat proximal tubular epithelial cells (NRK52E)
Glomerular mesangial cells
Mouse mesangial cell (MMCs)
Ovarian granulosa cells
Huh-7, HepG2, Hep3B liver cells
C2C12 myoblasts
HaCaT keratinocytes

Mouse
Human,
rat

[155]
[5355,134,156]

Human
Mouse
Mouse
Human
Human
Human

[157]
[152]
[158]
[55]
[52]
[159]

HeLa (cervix epithelial adenocarcinoma)


Cardiomyocytes
Primary murine hepatic stellate cells,
immortalized murine hepatic stellate cells,
C2C12 myoblasts
HeLa (cervix epithelial adenocarcinoma)
Glioblastoma stem (CD133+) cells

Human
Mouse
Mouse

[149]
[160]
[161,155]

Human
Human

[149]
[65]

key role. miR-17/92, together with let-7, reduces GAM


expression by directly targeting its 30 UTR, whereas
GAM downregulates miR-17/92 via three mechanisms:
(i) impairs transcriptional activation of the miR-17/92
cluster via C-MYC; (ii) decreases the transcriptional activity of SMADs; and (iii) by interacting directly with Drosha
is involved in pri-miRNA (primary transcript of miRNA)
miR-17/92 processing (Figure 3b). GAM increases apoptosis, reduces cell proliferation and modulates levels of E2F1
and Ras. C-MYC itself is a TGF-b target gene that activates the promoter of this miRNA cluster. In addition, the
miR-17/92 cluster also targets individual TGF-b responsive genes, such as p21Waf1/Cip1 and BIM [35,64].
Expression of miR-451 was shown to be induced by
SMAD3 and SMAD4 through SMAD target sites in their
promoter regions in glioblastoma cells, resulting in cell
growth inhibition [65].
The miR-24 cluster (miR-23a, miR-27a and miR-24) was
also induced in response to TGF-b in human hepatocellular
carcinoma cells (Huh-7) in a SMAD-dependent manner.
This cluster can function as an antiapoptotic and proliferation-promoting factor in liver cancer cells, because its
388

[154]
[31]
[49]

expression is highly upregulated in hepatocellular carcinoma tissues compared with normal liver [66].
TGF-b induces miR-216a and miR-217, which were
shown to activate Akt. Via this mechanism, TGF-b signaling participates in fibrosis, hypertrophy and survival in
glomerular mesangial cells [67]. Both miR-216a and miR217 target phosphatase and tensin homolog (PTEN), an
inhibitor of Akt activation. Because PTEN protein acts as a
tumor suppressor, this interaction may have a delicate role
in carcinogenesis beyond the development of diabetic kidney disease.
Upon TGF-b treatment of normal murine mammary
gland (NMuMG) epithelial cells, miR-155 was shown to
be among the most significantly elevated miRNAs. This
induction was SMAD4-dependent [68]. Inhibition of miR155 was sufficient to suppress the TGF-b-induced EMT,
making this miRNA a potential target in breast cancer
treatment. Interestingly, miR-155 was downregulated by
TGF-b in normal human lung fibroblasts, but its ectopic
overexpression increased cell migration [69]. In mouse
models of lung fibrosis the expression level of miR-155
was correlated with the degree of fibrosis [69]. TGF-b

Review
treatment may cause opposite effects on the expression
of miR-155 in different cells, underlining again that
both TGF-b and miRNA machineries work in a cell- and
environment-dependent manner, and emphasizing that
the exact mechanisms and the direct targets of miR-155
have to be identified in each cell type. miR-155 directly
targets TGF-b-specific SMAD2 in THP-1 monocyte cell
lines [70] and BMP-specific SMAD1 and SMAD5 [71
73], suggesting the presence of another possible feedback
regulatory loop.
SMADS influence miRNA processing
Davis et al. presented another mechanism that contributes
to the modulation of miR expression [74,75]. TGF-b treatment resulted in upregulation of pre-miRNAs and matured
miRNAs, but not that of pri-miRNAs. These miRs are
regulated post-transcriptionally by a genome-independent
mechanism through association of receptor-specific SMAD
(R-SMAD), SMAD1 and SMAD5, but not SMAD4 proteins,
with p68, an RNA helicase component of the Drosha microprocessor complex [75]. This subset of miRNAs is called
TGF-b/BMP-regulated miRNAs (T/B miRNAs) [75]. T/B
miRNAs contain in their primary transcripts a conserved
sequence, identified as R-SBE, which is similar to SMADbinding element (SBE) [76]. SMAD proteins through their
amino terminus MH1 domain directly associate with RSBE. Davis et al. demonstrated that mutations in the RSBE region abolished TGF-b/BMP-mediated induction of
pre-miRNA synthesis and impaired pri-miRNA binding to
Drosha and DGCR8 in vivo [76]. In the human genome, 44
T/B miRNAs have been identified [76]. The nucleocytoplasmic shuttling of SMADs (controlled by phosphorylation of
serine residues by the TGF-bR1) is crucial for SMADmediated miRNA maturation. MAPK and GSK-3b can also
alter the subcellular localization of SMADs through phosphorylation [77,78]. Therefore, it was suggested that
SMAD regulation of miRNA processing could be modulated
independently of TGF-b and BMPs by signals that alter the
nuclear localization of SMADs (ERKMAPK and the Wnt
pathways) [76].
Similar to SMAD proteins, the RNA helicases p68 have
been shown to interact with several other transcription
factors, including MyoD, Runx2, androgen receptor, estrogen receptor and p53. Association of p53 and p68 facilitated
Drosha processing of a subset of miRNAs, which were
different from T/B miRNAs [79].
Pharmacological interventions affecting TGF-b
signaling
The TGF-b signaling pathway is a promising target in
cancer therapy. Indeed, several compounds affecting this
signaling pathway are under preclinical development or
even in clinical trial phase, as summarized in several
recent reviews [8082].
From a theoretical point of view, there are three major
possibilities in targeting the TGF-b pathway. Ligand traps
include TGF-b antibodies and soluble TGF-b receptors. In
mice, anti-TGF-b antibodies suppressed metastasis formation [83], whereas in the rat, they arrested progressive
nephropathy [84]. TGF-b antibodies increase the immune
response in animal experiments [85]. Anti-TGF-b1, -b2

Trends in Pharmacological Sciences July 2012, Vol. 33, No. 7

and pan-TGF-b antibodies are under preclinical/clinical


trials, and have been tested for scleroderma, prevention of
scarring, metastatic melanoma and renal cell carcinoma
[80,84,86,87]. Soluble receptors, TGF-bR2 (sTbRII) and
TGF-bR3 (sTbRIII) can also inhibit TGF-b signaling. In
hepatoma cells transfected with sTbRII, decreased tumor
formation was observed in an in vivo animal model [88]. In
a transgenic mouse mammary tumor model, increased
apoptosis in primary tumors, reduced tumor cell motility,
reduced intravasation and a decreased number of lung
metastases were detected after systemic treatment with
sTbRII [89]. sTbRIII was also tested and proved to suppress cell growth and metastasis of human breast cancer
and colon carcinoma cells [90].
Silencing of TGF-b signaling by antisense oligonucleotides is another possibility for therapy. DNA oligonucleotides can inhibit the synthesis of TGF-b1 and -b2 by
specific binding to their mRNAs. It has been shown that
administration of TGF-b antisense oligonucleotides can
reactivate tumor-specific immune responses [91,92].
TGF-b antisense nucleotides have been tested in preclinical and clinical studies for the treatment of glioma, pancreatic, colorectal, prostate and non-small cell lung cancer
[86,9399].
A third option of TGF-b-targeted therapy is the use of
intracellularly-acting TGF-bR1 kinase inhibitors. Numerous compounds are under development, with promising
results; they are effective in blocking TGF-b-induced EMT
in mammary epithelial cells, pancreatic carcinoma cells
and in inhibiting glioma tumor growth, as well as suppressing renal fibrosis in obstructive nephropathy [100103].
Because miRNAs influence cellular processes at several
points, they are both promising therapeutic agents and
targets [104]. Potential drugs that inhibit overexpressed
(oncogenic) miRNAs or those that substitute underexpressed (e.g., tumor suppressor) miRNAs would be useful
in cancer therapy. At present, miRNA-based therapeutic
approaches are in experimental, preclinical or in early
clinical phases. The principal problem is the delivery of
miRNAs to the targeted cell [105]. Treatment with miRNAs appears to be difficult even when a local delivery
approach may be suitable to ensure the cell-specific effect.
Systemic delivery is more dangerous; cytotoxic effects
related to unconjugated miRNAs or vectors used for delivery, as well as immunogenic reactions, present major
difficulties. For this purpose, adenoviral vectors have been
commonly used. Their application as therapeutic agents
has been reported as local or targeted treatments (e.g.,
intratumoral delivery in hepatocellular carcinoma [106]
and transnasal administration for lung cancer treatment
[107]). Systemic administration of miR-10b inhibited the
metastasis formation in a mouse mammary tumor model.
However, cytotoxic effects related to unconjugated miRNAs or vectors used for delivery, as well as immunogenic
reactions, present difficulties and cause serious problems
during systemic delivery [108,109]. For miRNA silencing,
several strategies, including anti-miRNA oligonucleotides
(AMOs), miRNA sponges and miRNA masking, have already been tested. AMOs are synthetic antisense oligonucleotides that bind their target miRNAs and thereby
competitively inhibit the miRNAmRNA interaction
389

Review
[110]. In clinical trials, AMOs have been tested against
hypoxia-induced factor 1 (HIF-1a), miR-122 and protein
kinase N3 (PKN3) in solid tumors [111] and lymphomas
[112]. Another potential area in which miRNA therapy
may be considered is treatment of viral infections. The first
experimental drug was an antagomir, which inhibits miR122, essential for the accumulation of hepatitis C virus
(HCV) in hepatic cells. Subcutaneous administration dramatically reduced HCV load in the liver and blood [113].
Effective treatments may require knockdown of multiple,
rather than individual, miRNAs. For this specific aim,
miRNA sponges have been developed. These compounds
are oligonucleotide-based constructs with multiple binding
sites against an miRNA cluster inserted in vectors containing a strong promoter [114]. Also, target protection or
miRNA masking represents an interesting novel strategy
for knockdown of the effect of miRNAs. For this purpose,
single-stranded, chemically-modified oligoribonucleotides,
perfectly complementary to the miRNA binding site of the
30 UTR of target mRNA, have been used in vivo in a zebrafish model [115].
Double-stranded RNAs, which mimic the effect of endogenous miRNAs, have been developed for miRNA substitution [105]. Application of miRNAs in combination with
other therapeutic agents may also contribute to a more
successful treatment. Adjuvant miRNA therapy may enhance the effect of other systemic therapies through influencing radiosensitivity or increasing sensitivity to DNAdamaging drugs. It has been reported that inhibition of the
overexpressed miR-128a (which target TGF-b-R1) leads to
resensitization for the growth inhibitory effects of TGF-b in
letrozole-resistant breast cancer [116].
Concluding remarks
The TGF-b signaling pathway is a complex network that
controls many physiological and pathophysiological processes. Its regulation and interaction with miRNAs in a
cell- and context-specific manner provides a fine-tuning,
dynamic and adaptive control of protein expression. In the
process of tumorigenesis, alterations of the TGF-b pathway either by genetic or epigenetic events result in a switch
from a tumor suppressor to a tumor promoting effect.
Recent knowledge on targeting members of this signaling
cascade by miRNAs or other agents may lead to the development of novel approaches in the therapy of cancer and
other diseases.
Disclosure statement
The authors have nothing to disclose.
Acknowledgments
The authors acknowledge the financial support from the Hungarian
MOP-4.2.2.B-10/B-10/
Ministry of National Resources (ETT40/09) and TA
1-2010-0013. A.P. is a recipient of the Janos Bolyai Research Fellowship.

References
1 Chang, H. et al. (2002) Genetic analysis of the mammalian
transforming growth factor-beta superfamily. Endocr. Rev. 23,
787823
2 Ikushima, H. and Miyazono, K. (2010) Cellular context-dependent
colors of transforming growth factor-beta signaling. Cancer Sci. 101,
306312
390

Trends in Pharmacological Sciences July 2012, Vol. 33, No. 7

3 Nassiri, F. et al. (2011) Endoglin (CD105): a review of its role in


angiogenesis and tumor diagnosis, progression and therapy.
Anticancer Res. 31, 22832290
4 Perez-Gomez, E. et al. (2010) The role of the TGF-b coreceptor
endoglin in cancer. Sci. World J. 10, 23672384
5 Petrocca, F. et al. (2008) E2F1-regulated microRNAs impair TGFbdependent cell-cycle arrest and apoptosis in gastric cancer. Cancer
Cell 13, 272286
6 Kalluri, R. and Weinberg, R.A. (2009) The basics of epithelial
mesenchymal transition. J. Clin. Invest. 119, 14201428
7 Brabletz, T. et al. (2005) Invasion and metastasis in colorectal cancer:
epithelialmesenchymal
transition,
mesenchymalepithelial
transition, stem cells and beta-catenin. Cells Tissues Organs 179,
5665
8 Adorno, M. et al. (2009) A mutant-p53/Smad complex opposes p63 to
empower TGFb-induced metastasis. Cell 137, 8798
9 Oft, M. et al. (1996) TGF-beta1 and Ha-Ras collaborate in modulating
the phenotypic plasticity and invasiveness of epithelial tumor cells.
Genes Dev. 10, 24622477
10 Bruna, A. et al. (2007) High TGFb-Smad activity confers poor
prognosis in glioma patients and promotes cell proliferation
depending on the methylation of the PDGF-B gene. Cancer Cell 11,
147160
11 Butz, H. et al. (2011) MicroRNA profile indicates downregulation of
the TGFb pathway in sporadic non-functioning pituitary adenomas.
Pituitary 14, 112124
12 Lagos-Quintana, M. et al. (2001) Identification of novel genes coding
for small expressed RNAs. Science 294, 853858
13 Place, R.F. et al. (2008) MicroRNA-373 induces expression of genes
with complementary promoter sequences. Proc. Natl. Acad. Sci.
U.S.A. 105, 16081613
14 Orom, U.A. et al. (2008) MicroRNA-10a binds the 50 UTR of ribosomal
protein mRNAs and enhances their translation. Mol. Cell 30, 460471
15 Zhang, J. et al. (2010) MiR-145, a new regulator of the DNA
fragmentation factor-45 (DFF45)-mediated apoptotic network. Mol.
Cancer 9, 211
16 Huang, S. et al. (2010) MicroRNA-181a modulates gene expression of
zinc finger family members by directly targeting their coding regions.
Nucleic Acids Res. 38, 72117218
17 Chen, K. and Rajewsky, N. (2006) Natural selection on human miRNA
binding sites inferred from SNP data. Nat. Genet. 38, 14521456
18 Lewis, B.P. et al. (2005) Conserved seed pairing, often flanked by
adenosines, indicates that thousands of human genes are microRNA
targets. Cell 120, 1520
19 Mattick, J.S. and Makunin, I.V. (2005) Small regulatory RNAs in
mammals. Hum. Mol. Genet. 14, R121R132
20 Sayed, D. and Abdellatif, M. (2011) MicroRNAs in development and
disease. Physiol. Rev. 91, 827887
21 Lima, R.T. et al. (2011) MicroRNA regulation of core apoptosis
pathways in cancer. Eur. J. Cancer 47, 163174
22 Davis-Dusenbery, B.N. and Hata, A. (2010) Mechanisms of control of
microRNA biogenesis. J. Biochem. 148, 381392
23 Braun, J. et al. (2010) Downregulation of microRNAs directs the EMT
and invasive potential of anaplastic thyroid carcinomas. Oncogene 29,
42374244
24 Zhong, H. et al. (2010) Targeting Smad4 links microRNA-146a to the
TGF-beta pathway during retinoid acid induction in acute
promyelocytic leukemia cell line. Int. J. Hematol. 92, 129135
25 Tzur, G. et al. (2009) Comprehensive gene and microRNA expression
profiling reveals a role for microRNAs in human liver development.
PLoS ONE 4, e7511
26 Rogler, C.E. et al. (2009) MicroRNA-23b cluster microRNAs regulate
transforming growth factor-beta/bone morphogenetic protein
signaling and liver stem cell differentiation by targeting Smads.
Hepatology 50, 575584
27 Shan, H. et al. (2009) Downregulation of miR-133 and miR-590
contributes to nicotine-induced atrial remodelling in canines.
Cardiovasc. Res. 83, 465472
28 Marquez, R.T. et al. (2010) Correlation between microRNA expression
levels and clinical parameters associated with chronic hepatitis C
viral infection in humans. Lab. Invest. 90, 17271736
29 Liu, G. et al. (2010) miR-21 mediates fibrogenic activation of
pulmonary fibroblasts and lung fibrosis. J. Exp. Med. 207, 15891597

Review
30 Leeper, N.J. et al. (2010) MicroRNA-26a is a novel regulator of
vascular smooth muscle cell function. J. Cell Physiol. 226, 10351043
31 Wang, B. et al. (2011) miR-200a prevents renal fibrogenesis through
repression of TGF-b2 expression. Diabetes 60, 280287
32 Pais, H. et al. (2010) Analyzing mRNA expression identifies Smad3 as
a microRNA-140 target regulated only at protein level. RNA 16,
489494
33 Fontana, L. et al. (2008) Antagomir-17-5p abolishes the growth of
therapy-resistant neuroblastoma through p21 and BIM. PLoS ONE 3,
e2236
34 Mestdagh, P. et al. (2010) The miR-17-92 microRNA cluster regulates
multiple components of the TGF-b pathway in neuroblastoma. Mol.
Cell 40, 762773
35 Petrocca, F. et al. (2008) Emerging role of miR-106b-25/miR-17-92
clusters in the control of transforming growth factor beta signaling.
Cancer Res. 68, 81918194
36 Wang, F.E. et al. (2010) MicroRNA-204/211 alters epithelial
physiology. FASEB J. 24, 15521571
37 Casas, E. et al. (2011) Snail2 is an essential mediator of Twist1induced epithelial mesenchymal transition and metastasis. Cancer
Res. 71, 245254
38 Reeves, R. and Nissen, M.S. (1990) The A-T-DNA-binding domain of
mammalian high mobility group I chromosomal proteins. A novel
peptide motif for recognizing DNA structure. J. Biol. Chem. 265,
85738582
39 Sgarra, R. et al. (2004) Nuclear phosphoproteins HMGA and their
relationship with chromatin structure and cancer. FEBS Lett. 574,
18
40 Thuault, S. et al. (2006) Transforming growth factor-beta employs
HMGA2 to elicit epithelialmesenchymal transition. J. Cell Biol. 174,
175183
41 Thuault, S. et al. (2008) HMGA2 and Smads co-regulate SNAIL1
expression during induction of epithelial-to-mesenchymal transition.
J. Biol. Chem. 283, 3343733446
42 Mahajan, A. et al. (2010) HMGA2: a biomarker significantly
overexpressed in high-grade ovarian serous carcinoma. Mod.
Pathol. 23, 673681
43 Mu, G. et al. (2010) Correlation of overexpression of HMGA1 and
HMGA2 with poor tumor differentiation, invasion, and proliferation
associated with let-7 down-regulation in retinoblastomas. Hum.
Pathol. 41, 493502
44 Shi, G. et al. (2009) Let-7 repression leads to HMGA2 overexpression
in uterine leiomyosarcoma. J. Cell. Mol. Med. 13, 38983905
45 Rahman, M.M. et al. (2009) Frequent overexpression of HMGA1 and 2
in gastroenteropancreatic neuroendocrine tumours and its
relationship to let-7 downregulation. Br. J. Cancer 100, 501510
46 Qian, Z.R. et al. (2009) Overexpression of HMGA2 relates to reduction
of the let-7 and its relationship to clinicopathological features in
pituitary adenomas. Mod. Pathol. 22, 431441
47 Korpal, M. et al. (2008) The miR-200 family inhibits epithelial
mesenchymal transition and cancer cell migration by direct
targeting of E-cadherin transcriptional repressors ZEB1 and ZEB2.
J. Biol. Chem. 283, 1491014914
48 Park, S.M. et al. (2008) The miR-200 family determines the epithelial
phenotype of cancer cells by targeting the E-cadherin repressors ZEB1
and ZEB2. Genes Dev. 22, 894907
49 Gregory, P.A. et al. (2008) The miR-200 family and miR-205 regulate
epithelial to mesenchymal transition by targeting ZEB1 and SIP1.
Nat. Cell Biol. 10, 593601
50 Burk, U. et al. (2008) A reciprocal repression between ZEB1 and
members of the miR-200 family promotes EMT and invasion in cancer
cells. EMBO Rep. 9, 582589
51 Pandit, K.V. et al. (2010) Inhibition and role of let-7d in idiopathic
pulmonary fibrosis. Am. J. Respir. Crit. Care Med. 182, 220229
52 Sun, Q. et al. (2008) Transforming growth factor-beta-regulated miR24 promotes skeletal muscle differentiation. Nucleic Acids Res. 36,
26902699
53 Qian, B. et al. (2009) High miR-21 expression in breast cancer
associated with poor disease-free survival in early stage disease
and high TGF-beta1. Breast Cancer Res. Treat. 117, 131140
54 Godwin, J.G. et al. (2010) Identification of a microRNA signature of
renal ischemia reperfusion injury. Proc. Natl. Acad. Sci. U.S.A. 107,
1433914344

Trends in Pharmacological Sciences July 2012, Vol. 33, No. 7

55 Yu, Y. et al. (2010) Context-dependent bidirectional regulation of the


mutS homolog 2 by transforming growth factor b contributes to
chemoresistance in breast cancer cells. Mol. Cancer Res. 8, 16331642
56 Kim, Y.J. et al. (2009) MiR-21 regulates adipogenic differentiation
through the modulation of TGF-b signaling in mesenchymal stem
cells derived from human adipose tissue. Stem Cells 27, 30933102
57 Pan, Q. et al. (2010) microRNA 21: response to hormonal therapies
and regulatory function in leiomyoma, transformed leiomyoma and
leiomyosarcoma cells. Mol. Hum. Reprod. 16, 215227
58 Papagiannakopoulos, T. et al. (2008) MicroRNA-21 targets a network
of key tumor-suppressive pathways in glioblastoma cells. Cancer Res.
68, 81648172
59 Zavadil, J. et al. (2007) Transforming growth factor-beta and
microRNA:mRNA regulatory networks in epithelial plasticity. Cells
Tissues Organs 185, 157161
60 Mizuno, Y. et al. (2009) miR-210 promotes osteoblastic differentiation
through inhibition of AcvR1b. FEBS Lett. 583, 22632268
61 Chung, A.C. et al. (2010) miR-192 mediates TGF-beta/Smad3-driven
renal fibrosis. J. Am. Soc. Nephrol. 21, 13171325
62 Tili, E. et al. (2010) GAM/ZFp/ZNF512B is central to a gene sensor
circuitry involving cell-cycle regulators, TGFb effectors, Drosha and
microRNAs with opposite oncogenic potentials. Nucleic Acids Res. 38,
76737688
63 Dews, M. et al. (2010) The myc-miR-1792 axis blunts TGFb signaling
and production of multiple TGFb-dependent antiangiogenic factors.
Cancer Res. 70, 82338246
64 Ventura, A. and Jacks, T. (2009) MicroRNAs and cancer: short RNAs
go a long way. Cell 136, 586591
65 Gal, H. et al. (2008) MIR-451 and Imatinib mesylate inhibit tumor
growth of glioblastoma stem cells. Biochem. Biophys. Res. Commun.
376, 8690
66 Huang, S. et al. (2008) Upregulation of miR-23a approximately 27a
approximately 24 decreases transforming growth factor-b-induced
tumor-suppressive activities in human hepatocellular carcinoma
cells. Int. J. Cancer 123, 972978
67 Kato, M. et al. (2010) Post-transcriptional up-regulation of Tsc-22 by
Ybx1, a target of miR-216a, mediates TGF-b-induced collagen
expression in kidney cells. J. Biol. Chem. 285, 3400434015
68 Kong, W. et al. (2008) MicroRNA-155 is regulated by the transforming
growth factor b/Smad pathway and contributes to epithelial cell
plasticity by targeting RhoA. Mol. Cell. Biol. 28, 67736784
69 Pottier, N. et al. (2009) Identification of keratinocyte growth factor as
a target of microRNA-155 in lung fibroblasts: implication in
epithelialmesenchymal interactions. PLoS ONE 4, e6718
70 Louafi, F. et al. (2010) Microrna-155 (miR-155) targets SMAD2 and
modulates the response of macrophages to transforming growth
factor-b (TGF-b). J. Biol. Chem. 285, 4132841336
71 Yin, Q. et al. (2008) MicroRNA-155 is an Epstein-Barr virus-induced
gene that modulates Epstein-Barr virus-regulated gene expression
pathways. J. Virol. 82, 52955306
72 Rai, D. et al. (2010) Targeting of SMAD5 links microRNA-155 to the
TGF-b pathway and lymphomagenesis. Proc. Natl. Acad. Sci. U.S.A.
107, 31113116
73 Yin, Q. et al. (2010) MicroRNA miR-155 inhibits bone morphogenetic
protein (BMP) signaling and BMP-mediated Epstein-Barr virus
reactivation. J. Virol. 84, 63186327
74 Krol, J. et al. (2010) The widespread regulation of microRNA
biogenesis, function and decay. Nat. Rev. Genet. 11, 597610
75 Davis, B.N. et al. (2008) SMAD proteins control DROSHA-mediated
microRNA maturation. Nature 454, 5661
76 Davis, B.N. et al. (2010) Smad proteins bind a conserved RNA sequence
to promote microRNA maturation by Drosha. Mol. Cell 39, 373384
77 Fuentealba, L.C. et al. (2007) Integrating patterning signals: Wnt/
GSK3 regulates the duration of the BMP/Smad1 signal. Cell 131,
980993
78 Kretzschmar, M. et al. (1997) Opposing BMP and EGF signalling
pathways converge on the TGF-beta family mediator Smad1. Nature
389, 618622
79 Suzuki, H.I. et al. (2009) Modulation of microRNA processing by p53.
Nature 460, 529533
80 Nagaraj, N.S. and Datta, P.K. (2010) Targeting the transforming
growth factor-b signaling pathway in human cancer. Expert Opin.
Investig. Drugs 19, 7791
391

Review
81 Korpal, M. and Kang, Y. (2010) Targeting the transforming growth
factor-b signalling pathway in metastatic cancer. Eur. J. Cancer 46,
12321240
82 Pennison, M. and Pasche, B. (2007) Targeting transforming growth
factor-beta signaling. Curr. Opin. Oncol. 19, 579585
83 Hoefer, M. and Anderer, F.A. (1995) Anti-(transforming growth factor
b) antibodies with predefined specificity inhibit metastasis of highly
tumorigenic human xenotransplants in nu/nu mice. Cancer Immunol.
Immunother. 41, 302308
84 Benigni, A. et al. (2003) Add-on anti-TGF-b antibody to ACE inhibitor
arrests progressive diabetic nephropathy in the rat. J. Am. Soc.
Nephrol. 14, 18161824
85 Arteaga, C.L. et al. (1993) Anti-transforming growth factor (TGF)-b
antibodies inhibit breast cancer cell tumorigenicity and increase
mouse spleen natural killer cell activity. Implications for a possible
role of tumor cell/host TGF-b interactions in human breast cancer
progression. J. Clin. Invest. 92, 25692576
86 Lahn, M. et al. (2005) TGF-b inhibitors for the treatment of cancer.
Expert Opin. Investig. Drugs 14, 629643
87 Mead, A.L. et al. (2003) Evaluation of anti-TGF-b2 antibody as a new
postoperative anti-scarring agent in glaucoma surgery. Invest.
Ophthalmol. Vis. Sci. 44, 33943401
88 Zhao, W. et al. (2002) Suppression of in vivo tumorigenicity of rat
hepatoma cell line KDH-8 cells by soluble TGF-beta receptor type II.
Cancer Immunol. Immunother. 51, 381388
89 Muraoka, R.S. et al. (2002) Blockade of TGF-b inhibits mammary
tumor cell viability, migration, and metastases. J. Clin. Invest. 109,
15511559
90 Bandyopadhyay, A. et al. (2002) Extracellular domain of TGFb type
III receptor inhibits angiogenesis and tumor growth in human cancer
cells. Oncogene 21, 35413551
91 Fakhrai, H. et al. (1996) Eradication of established intracranial rat
gliomas by transforming growth factor b antisense gene therapy.
Proc. Natl. Acad. Sci. U.S.A. 93, 29092914
92 Wu, R.S. et al. (2001) Comparative analysis of IFN-g B7.1 and
antisense TGF-b gene transfer on the tumorigenicity of a poorly
immunogenic metastatic mammary carcinoma. Cancer Immunol.
Immunother. 50, 229240
93 Bogdahn, U. et al. (2004) Specific therapy for high-grade glioma by
convection-enhanced delivery of the TGF-b2 specific antisense
oligonucleotide AP 12009. J. Clin. Oncol. 22, 1514
94 Stauder, G. et al. (2004) TGF-b2 suppression by the antisense
oligonucleotide AP 12009 as treatment for pancreatic cancer:
preclinical efficacy data. J. Clin. Oncol. 22, 4106
95 Schlingensiepen, K.H. et al. (2004) The TGF-b1 antisense
oligonucleotide AP 11014 for the treatment of non-small cell lung,
colorectal and prostate cancer: preclinical studies. J. Clin. Oncol.
22, 3132
96 Schlingensiepen, K.H. et al. (2008) Antisense therapeutics for tumor
treatment: the TGF-b2 inhibitor AP 12009 in clinical development
against malignant tumors. Recent Results Cancer Res. 177, 137150
97 Saunier, E.F. and Akhurst, R.J. (2006) TGF b inhibition for cancer
therapy. Curr. Cancer Drug Targets 6, 565578
98 Hau, P. et al. (2009) Treatment of malignant gliomas with TGF-b2
antisense oligonucleotides. Expert Rev. Anticancer Ther. 9, 16631674
99 Nemunaitis, J. et al. (2009) Phase II trial of Belagenpumatucel-L, a
TGF-b2 antisense gene modified allogeneic tumor vaccine in
advanced non small cell lung cancer (NSCLC) patients. Cancer
Gene Ther. 16, 620624
100 Peng, S.B. et al. (2005) Kinetic characterization of novel pyrazole
TGF-b receptor I kinase inhibitors and their blockade of the
epithelialmesenchymal transition. Biochemistry 44, 22932304
101 Subramanian, G. et al. (2004) Targeting endogenous transforming
growth factor b receptor signaling in SMAD4-deficient human
pancreatic carcinoma cells inhibits their invasive phenotype1.
Cancer Res. 64, 52005211
102 Uhl, M. et al. (2004) SD-208, a novel transforming growth factor beta
receptor I kinase inhibitor, inhibits growth and invasiveness and
enhances immunogenicity of murine and human glioma cells in
vitro and in vivo. Cancer Res. 64, 79547961
103 Moon, J.A. et al. (2006) IN-1130, a novel transforming growth factor-b
type I receptor kinase (ALK5) inhibitor, suppresses renal fibrosis in
obstructive nephropathy. Kidney Int. 70, 12341243
392

Trends in Pharmacological Sciences July 2012, Vol. 33, No. 7

104 He, S. et al. (2009) Applications of RNA interference in cancer


therapeutics as a powerful tool for suppressing gene expression.
Mol. Biol. Rep. 36, 21532163
105 Sotillo, E. and Thomas-Tikhonenko, A. (2011) Shielding the
messenger
(RNA):
microRNA-based
anticancer
therapies.
Pharmacol. Ther. 131, 1832
106 Zhang, X. et al. (2009) Up-regulated microRNA-143 transcribed by
nuclear factor k B enhances hepatocarcinoma metastasis by
repressing fibronectin expression. Hepatology 50, 490499
107 Trang, P. et al. (2010) Regression of murine lung tumors by the let-7
microRNA. Oncogene 29, 15801587
108 Grimm, D. et al. (2006) Fatality in mice due to oversaturation of cellular
microRNA/short hairpin RNA pathways. Nature 441, 537541
109 Aagaard, L. and Rossi, J.J. (2007) RNAi therapeutics: principles,
prospects and challenges. Adv. Drug Deliv. Rev. 59, 7586
110 Lu, Y. et al. (2009) A single anti-microRNA antisense
oligodeoxyribonucleotide (AMO) targeting multiple microRNAs
offers an improved approach for microRNA interference. Nucleic
Acids Res. 37, e24
111 Greenberger, L.M. et al. (2008) A RNA antagonist of hypoxiainducible factor-1a, EZN-2968, inhibits tumor cell growth. Mol.
Cancer Ther. 7, 35983608
112 Davis, M.E. et al. (2010) Evidence of RNAi in humans from
systemically administered siRNA via targeted nanoparticles.
Nature 464, 10671070
113 Lanford, R.E. et al. (2009) Therapeutic silencing of microRNA-122 in
primates with chronic hepatitis C virus infection. Science 327, 198201
114 Ebert, M.S. et al. (2007) MicroRNA sponges: competitive inhibitors of
small RNAs in mammalian cells. Nat. Methods 4, 721726
115 Choi, W.Y. et al. (2007) Target protectors reveal dampening and
balancing of Nodal agonist and antagonist by miR-430. Science
318, 271274
116 Masri, S. et al. (2010) The role of microRNA-128a in regulating TGFb
signaling in letrozole-resistant breast cancer cells. Breast Cancer Res.
Treat. 124, 8999
117 Kim, V.N. et al. (2009) Biogenesis of small RNAs in animals. Nat. Rev.
Mol. Cell Biol. 10, 126139
118 Zhang, X. and Zeng, Y. (2010) Regulation of mammalian microRNA
expression. J. Cardiovasc. Transl. Res. 3, 197203
119 Cai, X. et al. (2004) Human microRNAs are processed from capped,
polyadenylated transcripts that can also function as mRNAs. RNA 10,
19571966
120 Gregory, R.I. et al. (2004) The microprocessor complex mediates the
genesis of microRNAs. Nature 432, 235240
121 Lund, E. et al. (2004) Nuclear export of microRNA precursors. Science
303, 9598
122 Chendrimada, T.P. et al. (2005) TRBP recruits the Dicer complex to
Ago2 for microRNA processing and gene silencing. Nature 436, 740744
123 Gregory, R.I. et al. (2005) Human RISC couples microRNA biogenesis
and posttranscriptional gene silencing. Cell 123, 631640
124 Ghildiyal, M. et al. (2010) Sorting of Drosophila small silencing RNAs
partitions microRNA* strands into the RNA interference pathway.
RNA 16, 4356
125 Khvorova, A. et al. (2003) Functional siRNA and miRNAs exhibit
strand bias. Cell 115, 209216
126 Ruby, J.G. et al. (2007) Intronic microRNA precursors that bypass
Drosha processing. Nature 448, 8386
127 Ye, W. et al. (2008) The effect of central loops in miRNA:MRE
duplexes on the efficiency of miRNA-mediated gene regulation.
PLoS ONE 3, e1719
128 Zamore, P.D. et al. (2000) RNAi: double-stranded RNA directs the
ATP-dependent cleavage of mRNA at 21 to 23 nucleotide intervals.
Cell 101, 2533
129 Bartel, D.P. (2004) MicroRNAs: genomics, biogenesis, mechanism,
and function. Cell 116, 281297
130 Pillai, R.S. et al. (2005) Inhibition of translational initiation by Let-7
MicroRNA in human cells. Science 309, 15731576
131 Kiriakidou, M. et al. (2007) An mRNA m7G cap binding-like motif
within human Ago2 represses translation. Cell 129, 11411151
132 Zinovyev, A. et al. (2010) Dynamical modeling of microRNA action on
the protein translation process. BMC Syst. Biol. 4, 13
133 Martin, J. et al. (2011) Post-transcriptional regulation of transforming
growth factor b-1 by microRNA-744. PLoS ONE 6, e25044

Review
134 Denby, L. et al. (2011) miR-21 and miR-214 are consistently
modulated during renal injury in rodent models. Am. J. Pathol.
179, 661672
135 Wang, H. et al. (2010) miR-106b aberrantly expressed in a double
transgenic mouse model for Alzheimers disease targets TGF-b type II
receptor. Brain Res. 1357, 166174
136 Li, Z. et al. (2011) Small RNA-mediated regulation of iPS cell
generation. EMBO J. 30, 823834
137 Volinia, S. et al. (2006) A microRNA expression signature of human
solid tumors defines cancer gene targets. Proc. Natl. Acad. Sci. U.S.A.
103, 22572261
138 Lewis, B.P. et al. (2003) Prediction of mammalian microRNA targets.
Cell 115, 787798
139 Luzi, E. et al. (2008) Osteogenic differentiation of human adipose
tissue-derived stem cells is modulated by the miR-26a targeting of the
SMAD1 transcription factor. J. Bone Miner. Res. 23, 287295
140 Lin, E.A. et al. (2009) miR-199a, a bone morphogenic protein 2responsive microRNA, regulates chondrogenesis via direct
targeting to Smad1. J. Biol. Chem. 284, 1132611335
141 Liu, Z. et al. (2012) MicroRNA-146a modulates TGF-b1-induced
phenotypic differentiation in human dermal fibroblasts by
targeting SMAD4. Arch. Dermatol. Res. 304, 195202
142 Geraldo, M.V. et al. (2012) MicroRNA miR-146b-5p regulates signal
transduction of TGF-b by repressing SMAD4 in thyroid cancer.
Oncogene 31, 19101922
143 Hager, M. et al. (2011) MicroRNA-130a-mediated downregulation of
Smad4 contributes to reduced sensitivity to TGF-b1 stimulation in
granulocytic precursors. Blood 118, 66496659
144 Lim, L.P. et al. (2005) Microarray analysis shows that some
microRNAs downregulate large numbers of target mRNAs. Nature
433, 769773
145 Kim, D. et al. (2011) MicroRNA-142-3p regulates TGF-b3-mediated
region-dependent chondrogenesis by regulating ADAM9. Biochem.
Biophys. Res. Commun. 414, 653659
146 Yang, B. et al. (2011) MicroRNA-145 regulates chondrogenic
differentiation of mesenchymal stem cells by targeting Sox9. PLoS
ONE 6, e21679
147 Long, X. and Miano, J.M. (2011) Transforming growth factor-b1 (TGFb1) utilizes distinct pathways for the transcriptional activation of
microRNA 143/145 in human coronary artery smooth muscle cells. J.
Biol. Chem. 286, 3011930129

Trends in Pharmacological Sciences July 2012, Vol. 33, No. 7

148 Jurkin, J. et al. (2010) miR-146a is differentially expressed by myeloid


dendritic cell subsets and desensitizes cells to TLR2-dependent
activation. J. Immunol. 184, 49554965
149 Dogar, A.M. et al. (2011) Suppression of latent transforming growth
factor (TGF)-b1 restores growth inhibitory TGF-b signaling through
microRNAs. J. Biol. Chem. 286, 1644716458
150 Wang, B. et al. (2010) TGFb-mediated upregulation of hepatic miR181b promotes hepatocarcinogenesis by targeting TIMP3. Oncogene
29, 17871797
151 Wang, Y. et al. (2011) Transforming growth factor-b regulates the
sphere-initiating stem cell-like feature in breast cancer through
miRNA-181 and ATM. Oncogene 30, 14701480
152 Kato, M. et al. (2007) MicroRNA-192 in diabetic kidney glomeruli and
its function in TGF-b-induced collagen expression via inhibition of Ebox repressors. Proc. Natl. Acad. Sci. U.S.A. 104, 34323437
153 Sun, L. et al. (2011) Low-dose paclitaxel ameliorates fibrosis in the
remnant kidney model by down-regulating miR-192. J. Pathol. 225,
364377
154 Ahn, S.M. et al. (2011) Smad3 regulates E-cadherin via miRNA-200
pathway. Oncogene http://dx.doi.org/10.1038/onc.2011.484
155 Winbanks, C.E. et al. (2011) TGF-b regulates miR-206 and miR-29 to
control myogenic differentiation through regulation of HDAC4. J.
Biol. Chem. 286, 1380513814
156 Zhong, X. et al. (2011) Smad3-mediated upregulation of miR-21
promotes renal fibrosis. J. Am. Soc. Nephrol. 22, 16681681
157 Kato, M. et al. (2009) TGF-b activates Akt kinase through a
microRNA-dependent amplifying circuit targeting PTEN. Nat. Cell
Biol. 11, 881889
158 Yao, G. et al. (2010) MicroRNA-224 is involved in transforming
growth factor-b-mediated mouse granulosa cell proliferation and
granulosa cell function by targeting Smad4. Mol. Endocrinol. 24,
540551
159 Papadimitriou, E. et al. (2011) Differential regulation of the two
RhoA-specific GEF isoforms Net1/Net1A by TGF-b and miR-24:
role in epithelial-to-mesenchymal transition. Oncogene
http://
dx.doi.org/10.1038/onc.2011.457
160 Wang, J. et al. (2012) Cardiomyocyte overexpression of miR-27b
induces cardiac hypertrophy and dysfunction in mice. Cell Res. 22,
516527
161 Roderburg, C. et al. (2010) Micro-RNA profiling reveals a role for miR29 in human and murine liver fibrosis. Hepatology 53, 209218

393

Vous aimerez peut-être aussi