You are on page 1of 9

Biomedicine & Pharmacotherapy 123 (2020) 109778

Contents lists available at ScienceDirect

Biomedicine & Pharmacotherapy


journal homepage: www.elsevier.com/locate/biopha

Krüppel-like factors in breast cancer: Function, regulation and clinical T


relevance
Jianping Zhanga,b,1, Guangliang Lic,1, Lifeng Fengb, Haiqi Lua,b,*, Xian Wanga,b,*
a
Department of Medical Oncology, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
b
Laboratory of Cancer Biology, Key Lab of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
c
Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China

A R T I C LE I N FO A B S T R A C T

Keywords: Breast cancer has accounted for the leading cause of cancer-related mortality among women worldwide.
Krüppel-like factors (KLFs) Although the progress in its diagnosis and treatment has come at a remarkable pace during the past several
Breast cancer decades, there are still a wide array of problems regarding its progression, metastasis and treatment resistance
Function that have not yet been fully clarified. Recently, an increasing number of studies have revealed that some
Regulation
members of Krüppel-like factors(KLFs) are significantly associated with cell proliferation, apoptosis, metastasis,
Clinical relevance
cancer stem cell regulation and prognostic and predictive value for patients in breast cancer, indicating their
promising prognostic and predictive potential for breast cancer survival and outcome. In this review, we will
summarize our current knowledge of the functions, regulations and clinical relevance of KLFs in breast cancer.

1. Introduction closely implicated with cancer-related regulatory processes and sig-


naling transduction pathways involved in cell proliferation differ-
As for the most common type of cancer and the most frequent cause entiation, metabolism, apoptosis, invasion and migration, as well as the
of cancer-related mortality among women, breast cancer is the main establishment and maintenance of cell pluripotency and the initiation,
reason for leading to over 500,000 deaths per year worldwide [1–3]. promotion and progression in breast cancer [14]. Intriguingly, it is
Breast cancer is a kind of complicated and heterogeneous diseases, worthy of in-depth exploration that KLFs can function as oncogenes
mainly divided into hormone-receptor positive [4], human epidermal and/or tumor suppressors in distinct cancer-specific contexts,implying
growth factor receptor-2 overexpressing (Her2+) [5] and triple nega- their potential roles in predicting prognosis in different cancer patients.
tive breast cancer(TNBC) [6] according to different molecular sub- Up to now, the function of KLFs have been systematic summarized in
typing and histological features [7]. Over the last several decades, many other diseases such as female reproductive system pathologies
breakthroughs have been made in the field of breast cancer treatment and leukocyte diseases [15,16]. However, there is no comprehensive
including surgery [8], radiotherapy [9], chemotherapy [6], endocrine generalization of the functions of Krüppel-like factors in breast cancer.
therapy [4], targeted therapy [10], immunotherapy [11] and so on. As a result, in this review, we will focus on the latest advances on the
However, the pathogenesis of breast cancer initiation and progression regulation and function of KLFs and highlight their molecular me-
and its drug resistance mechanisms in different subtypes still have not chanisms, biological roles and clinical relevance in breast cancer to
yet been fully elucidated. Remarkably, a large amount of transcriptional provide a novel therapeutic strategies to breast cancer prevention and
factors including KLFs have been demonstrated to be associated with treatment (Fig. 1).
the process of cancer initiation, promotion and progression [12,13].
Named for a mammalian homologue of the Drosophila melanogaster 2. Overview of Krupple-like factors
gene Krüppel identified in 1993 firstly, KLFs are a family of DNA-
binding transcriptional regulators which play essential roles in several KLFs, get its name from the first discovery of Krüppel protein in
crucial cellular processes such as proliferation, differentiation, apop- Drosophila melanogaster, a member of the “gap” class of segmentation
tosis, migration and cancer cell stem regulation [8,9]. To date, gene products that regulates body segmentation in the thorax and
mounting reports have demonstrated that several KLF factors are anterior abdomen of the Drosophila embryo [17]. Up to now, 17


Corresponding authors at: Department of Medical Oncology, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China.
E-mail addresses: haiqilu@zju.edu.cn (H. Lu), wangx118@zju.edu.cn (X. Wang).
1
These authors contributed equally to this work.

https://doi.org/10.1016/j.biopha.2019.109778
Received 21 September 2019; Received in revised form 27 November 2019; Accepted 29 November 2019
0753-3322/ © 2019 Published by Elsevier Masson SAS. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/BY-NC-ND/4.0/).
J. Zhang, et al. Biomedicine & Pharmacotherapy 123 (2020) 109778

Fig. 1. The structure of KLF.


KLF family is composed of the conversed C-
terminal regions and highly divergent N-term-
inal regions. C-terminus consists of three
tandem Cys2His2 zinc-finger motifs which act
as a DNA-binding domain recognizing GC rich
target sequences.N-terminus contains tran-
scriptional regulatory domains which can bind
co-factors and contribute to distinct functions
of KLFs.

members of KLFs have been identified in human tissue and got depth
lucubrated. It is well acknowledged that all KLFs members contain
three C2H2-type zinc fingers which is similar to the DNA binding do-
main structure of transcription factor Sp1, therefore, KLFs is a subgroup
of Sp1/KLF family [13]. In conservation and phylogenetic analysis,
KLFs proteins are conserved among mammals from human to rat [15].
However, the tissue expression of the KLFs are varies, most KLFs are
ubiquitously expressed(e.g. KLFs 6, 10, and 11), while others are found
to be developmentally or temporally expressed in tissue- and cell-type
specific manner (KLF1 is expressed predominantly in bone marrow,
KLF2 is highly expressed in adipose tissue, and KLFs 4, 5 are very
abundant in the gastrointestinal tract) [17]. Based on the similarity in
sequences of their DNA binding domains, KLFs can be phylogenetically
analyzed as Fig. 2 which defines evolutionary distances of individual
family members. In the structure, besides above-mentioned DNA
binding domains, transcriptional regulation domain and nuclear loca-
lization signal (NLS) domain are the remaining two parts, although NLS
is only located in several KLFs (KLF 1, 4, 8, 11), which occur im-
mediately adjacent to or within the Zinc-finger motifs, with the function
of nuclear localization of KLF [18]. Specifically, DNA binding domain is
Fig. 2. Phylogenetic tree of human KLFs.
composed of three tandem Cys2His2 zinc-finger motifs in C-terminal
According to the structural similarity approximation of DNA-binding domain,
region with the function of recognizing similar target sequences (GC-
the KLFs family can be divided into three major parts. Subgroup I consists of
rich sequences with a preference for the 5′-CACCC-3′core motif). Due to
KLF3,5,6,7,8,12 which are highly related with each other, Subgroup II consists
of KLF 1,2,4,15,17, the remaining KLFs are belong to Subgroup III. The tree was this special structure, evolutionally similar KLFs may have same tar-
generated using Genetic Computer Group (GCG) sequence analysis software geting promoters and antagonize their mutual transcriptional activity
mostly because of physical competition. By contrast, the transcriptional

Fig. 3. The role of KLFs in breast cancer


metastasis.
KLFs take part in the process of metastasis in-
cluding EMT, ECM change, invasion and an-
giopoiesis. The specific molecular mechanisms
are shown in Fig. 3. Blue square represents the
upstream of KLFs while the green rhombus
represents downstream of KLFs.Blue-white box
and Red-white box represent negatively and
positively regulate the process of metastases,
respectively. KLF4 negatively regulates angio-
poiesis via inhibiting VEGF expression, KLF8
promotes but KLF6 and KLF9 inhibit EMC
change, KLF8 stimulates invasion by FAK and
EPSTI1 activation, KLF10 downregulates inva-
sion by inhibiting EGFR signal pathway. KLF5
accumulates EMT by promoting Slug tran-
scription,However, BAP1 can deubiquitinate
KLF5 and promote EMT. KLF4 promotes E-
cadherin to restrain EMT development,while
KLF8 inhibits E-cadherin to facilitate EMT.
KLF6-SV boosts EMT by increaing TWIST1 but
KLF17 suppresses EMT through ID-1 inactiva-
tion and this supression can be rescured by DJ-
1 and P53.

2
J. Zhang, et al. Biomedicine & Pharmacotherapy 123 (2020) 109778

regulation domain located in N-terminal is highly variable and contain 3.2. KLFs in apoptosis
domains including acidic transactivation domains, Sin-3 interacting
repressor domains and C-Terminal Binding Protein (CtBP)-Binding re- As proliferative and apoptotic signaling pathways are often inter-
pressor domains that interact with specific co-activators and co-re- connected, many of the factors (p53, E2F, MYC, MAPKs etc) and
pressors, resulting in functional diversity and specificity. Interestingly, pathways important for KLFs in cancer cell proliferation are also re-
It has been reported that there is overlap between Sin-3 interacting levant for apoptosis. Importantly, KLFs can regulate several classical
domain and CtBP-Binding domain, indicating that the function of Sin- apoptotic factors such as BAX and survival factors including MCL1,
3and CtBP may exist completion relationship. By regulating gene BCL-2, JNK and so on [14]. It has been reported that KLF5 inhibits
transcription, the function of KLFs are involved in many physiologic apoptosis through downregulating tumor necrosis factor TNF-α [35]
and pathologic processes, such as cell differentiation, proliferation, cell with increasing the anti-apoptotic factors like B-cell lymphoma-XL(BCL-
growth, and apoptosis, during normal development or under different XL) [36]. Furthermore, KLF10 promotes breast cancer cell apoptosis via
disease conditions [13]. downregulating the inhibition of BAX (BI-1) expression as well as in-
creasing Ca2+ release from endoplasmic reticulum to cytoplasm in
estrogen-responding adenocarcinoma cells [37,38]. KLF11 enhances
3. The function of KLFs in breast cancer the apoptosis of breast cancer cells by inhibiting the expression of Bcl-2
while upregulating Bax protein level. During the process of apoptosis in
3.1. KLFs in cell proliferation breast cancer cells, KLFs are also modulated by some kinds of ncRNAs
such as microRNAs. The low expression of KLF11 could be caused by
Cell proliferation is one of the important physiological functions of miR-30d through targeting 3’UTR [39]. Moreover, miR-205 can directly
living cells and an important life feature of organisms which is the basis target 3’-UTR of KLF12 and induce the apoptosis. However, the anti-
for growth, development, reproduction, and inheritance of organisms apoptotic mechanism of KLF12 remained largely unknown [40,41].
[19–21]. Infinite ability to proliferate is a critical hallmark of tumor Collectively, KLF5 inhibit whereas KLF10, KLF11 and KLF12 accelerate
cells [22]. During the proliferation, genetic alterations found in cancers the apoptosis of breast cancer cells.
can alter specific regulatory pathways and provide a selective growth
advantage by activation of transforming oncogenes or repression of 3.3. KLFs in tumor metastasis
tumor suppressors [23]. However, whether the role of KLFs acting as
oncogenes or tumor suppressors has not reach a consensus and need Metastasis is the spread of cancer to another part of the body distant
deeper exploration in the proliferation of breast cancer. Remarkably, from the original primary cancer which occurs through the transfer of
KLF4 is demonstrated as a well-recognized tumor suppressor in breast malignant or cancerous cells via lymphatic or blood circulation. During
cancer by its well-.documented cell cycle inhibition. Mechanically, the process of metastasis, Epithelial–mesenchymal transition (EMT),
KLF4 acts as a transcriptional factor which binds to the promoter of extracellular matrix change, invasion and angiogenesis all play critical
CDKN1A (p21) and CDKN1B (p27) to promote their transcription and roles and have long been the research focus [42–44]. Remarkably, an
cell cycle G1/S arrest [24,25]. However, other recent findings reported increasing amount of reports indicates that KLFs may take part in this
that KLF4 may act as oncogene in breast cancer [23,26]. K. Wade Foster process.
etc revealed that KLF4 is high expressed in breast cancer tissues and
Loss-of-function mutations frequently affect p53 and p16/CDKN2 to 3.3.1. Epithelial-mesenchymal transition
inhibit the proliferation [23]. Another research indicated that trans- As one of the most well-known characters of malignant transfor-
fection of the KLF4 gene inhibited the proliferation of breast cancer mation, the epithelial–mesenchymal transition (EMT) is a process by
cells MDA-MB-231, suggesting that KLF4 is important for the initiation which epithelial cells lose their cell polarity and cell-cell adhesion, si-
and progression of breast cancer [26]. Although these evidences are multaneously gain migratory and invasive properties to become me-
limited and controversial, it may provide us a novel perspective on the senchymal cells [45,46]. The most remarkable character is loss of E-
role of KLF4 dependent on different cell and tissue situation. Intrigu- cadherin which maintains cell–cell contacts and adherent junctions
ingly, KLF4α, the splice of KLF4, stimulates breast cancer cell cycle and [47]. E-cadherin is downregulated by transcription factors like Snail,
proliferation by acting as a KLF4 antagonist through abrogating its Slug, RhoB, TWIST1 and some growth factors like TGF-β, EGF, FGF and
nuclear localization to activate the transcription of target genes such as IGF-1 [47–50]. Recently, a large amount of researches indicated that
CDKN1A and CDKN1B [27]. Similarly, CDKN1A transcription can be KLFs are highly associated to EMT in breast cancer [28,51]. For ex-
activated by KLF6 while be attenuated by miR-4262 [4,24]. Different ample, KLF4 inhibits EMT by promoting E-cadherin expression in
from KLF4, KLF5 is highly overexpressed and functions as an oncogene transcriptional activation, which is necessary for maintenance the epi-
in breast cancer cells [28–30]. Mechanically, KLF5 can participate in a thelial phenotype [52]. Strinkingly, KLF5 induces EMT by increasing
diverse set of biological processes such as chromatin licensing and the expression of slug [29]. Moreover, KLF6-SV, the splice of KLF6,
modulated a vast array of cell cycle related genes such as cyclin A2 increases EMT by promoting TWIST1 transcription. Thus KLF6-SV ac-
(CCNA2) [31], cyclinD1(CCND1) [32], DNA replication factor 1 (CDT1) celerates breast cancer metastasis in mouse models and indicate a poor
and E2F transcription factor 3(E2F3) [31], which controls cell cycle- survival of breast cancer patients [53–55]. Contrary to KLF4, KLF8
dependent gene expression like fibroblast growth factor binding pro- promotes EMT via directly binding to the GT box in the E-cadherin
tein1(FGF-BP1) [30]. Besides, KLF5 binds to the promoter of micro- promoter but represses E-cadherin gene expression [56,57]. Taken to-
somal prostaglandin E2 synthase1(mPGES1)to facilitate its transcrip- gether, these seemingly controversial but plausible explanations about
tion so as to enhance the activity to suppress CDK inhibitors including the consequences of how KLFs regulate E-cadherin might indicate that
p21 and p27 [33]. Furthermore, another research indicated that KLF5 further studies should be undertaken to understand subtle differences of
recruits p300 to the promoter of lncRNA RP1 to accelerate its expres- their binding in the promoter of E-cadherin the exquisite and specific
sion at transcriptional level. The high expressional lncRNA RP1 binds to regulation of their downstream target genes and their completely re-
translational initiation complex p-4E-BP1/eIF4E to downregulate the levant biological mechanisms. Compared with the promotive effect of
expression of tumor suppressor p27 [34]. Nevertheless, how other KLFs above-mentioned KLFs in EMT, KLF17 is a negative regulator of EMT by
members regulate cell cycle have not yet been reported. Therefore, suppressing the transcription of the inhibitor of DNA binding-1 (ID-1)
further investigations will be urgent to shed light upon the other KLFs which is an important member controlling cancer metastasis [58,59].
and how they orchestrate their crosstalk relationship on regulating cell Besides, the expression of KLF17 is also regulated by p53 and DJ-1 [59].
cycle genes. Mutant-p53 interacts with KLF17 and antagonizes KLF17 mediated

3
J. Zhang, et al. Biomedicine & Pharmacotherapy 123 (2020) 109778

EMT gene transcription such as CD44, PAI-1 and Cyclin-D1 [60]. DJ-1 3.4. KLFs in signaling transduction
binds to the promoter of KLF17 to inhibit its transcription [59]. Col-
lectively, an increasing number of KLFs have been recognized to play 3.4.1. Hormone receptors signaling pathway
pivotal roles in the process of EMT, raising some pending problems that Hormone receptor signaling pathways play a fundamental role in
whether different KLFs are fine tuned to carry out their functions de- the development of breast cancer. The roles of KLFs in estrogen re-
pendent on cancer cell-specific microenvironment. Thus, future in- ceptors-related signaling pathways are complicated. For example, oes-
vestigations will be urgent to explore their crosstalk and mutual reg- trogen upregulates the expression of KLF4 in protein level. Von Hippel-
ulation with each other. Lindau (pVHL), downregulated by estrogen, facilitates accumulation of
KLF4 in a proteolytic regulation way. While KLF4 knockdown inhibits
oestrogen-induced growth of tumor cells [25], which indicates KLF4
3.3.2. Extracellular matrix (ECM) change acting a positive regulatory role in the proliferation induced by es-
Extracellular matrix (ECM) change is a crucial part of metastasis. trogen. However, another research indicated that KLF4 suppresses es-
Matrix metallo proteinases (MMPs) can degrade various protein com- trogen-dependent breast cancer growth by interacting with the DNA-
ponents in ECM and destroy the histological barrier of tumor cell in- binding region of ERa, competitively antagonist binding of ERa to es-
vasion, thus playing a key role in tumor invasion and metastasis trogen response elements at the presence of estrogen, resulting in a
[61,62]. For instance, KLF6 can inhibit breast cancer metastasis by reduction in ERa target gene transcription. In another word, KLF4 in-
reinforcing Tissue factor pathway inhibitor-2 (TFPI-2) transcription, hibit only the transcriptional activity not expression of ERα [75]. In a
which is a matrix-associated Kunitz inhibitor to restrain the activation word, these seemingly controversial but plausible explanations about
of zymogen matrix metalloproteinases [63]. Different from KLF6, KLF8 the consequences of function of KLF4 in estrogen signal pathway might
is a positive regulator of MMPs, which could enhance the activity of indicate that further studies should be undertaken to understand subtle
MMP2 but not influence its expression. Besides, as an active upstream differences of their functions From my perspective, negative feedback
regulator of MMP2, MMP14 is a direct target of KLF8. As a result, KLF8 may be responsible for this controversy, estrogen induce KLF4 accu-
enhances the transcription of MMP14, which increases the activity of mulation while excessive KLF4 inhibit ERa to estrogen response ele-
MMP2. In another part, KLF8 accelerates to form β-catenin/TCF1 ments of target genes at the presence of estrogen. As for whether KLF4
transactivation complex which facilitates the transcription of MMP14 turnover function is dependent on stage-specific microenvironment.
[64]. The aforementioned evidences directly demonstrated that KLF8 Additionally, KLF5 inhibits the proliferation of ER(+) cells by inter-
can promote ECM change in breast cancer [65,66]. Besides, KLF9 boosts acting with ERα to form a complex with the result of target genes in-
metastasis by impairing matrix metalloproteinase 9 (MMP9) in breast activation [76,77]. However, KLF6 inhibits estrogen receptor-mediated
cancer tissues [67–69]. KLF9 recedes MMP9 by antagonizing NF-κB cell growth in ER (+) breast cancer via disrupting the interaction of
p50/p65 binding to the promoter of MMP9. In addition to MMP9, KLF9 ERα and c-Src, ultimately leading to the inactivation of c-Src to MAPK
also down-regulated several other NF-κB targets such as TNF-α, VEGFA signaling pathway [78,79]. Moreover, KLF17 attenuates ERα-mediated
and uPA in breast cancer cells [68]. signaling by impeding ERα function. As a feedback, ER signaling
pathway inhibits the transcription of KLF17 [80]. Besides, the down-
regulation of KLF17 expression in breast cancer is probably due to
3.3.3. Invasion UHRF1-dependent promoter hypermethylation [81]. Above all, further
KLFs are also implicated in the invasion of breast cancer cells. KLF4 investigations will be urgent to shed light upon the roles of KLFs in
positively regulates invasion via activating Notch signal pathway and estrogen signaling pathway and how they orchestrate their crosstalk
promotes migration of breast cancer [70]. Intriguingly, on the one relationship with other KLFs members. Besides, Proliferation induced
hand, KLF8 promotes lung metastasis of breast cancer in nude mice by by progesterone in breast cancer cells is also associated with KLF5.
augmenting epithelial stromal interaction 1(EPSTI1) expression [71] KLF5, upregulated by progesterone, facilitates cell proliferation in
which interacting Valois-containing protein(VCP)to activate NF-κB progesterone-treated breast cancer cells [31,82]. However, other KLFs
signaling pathway. On the other hand, KLF8 facilitates the transcription have not been identified in this signal pathway, thus, it’s necessary for
of CXCR4, which towards to CXCL12 and activates focal adhesion ki- further clarification of other KLFs functions in progesterone-receptor
nase (FAK), fascinatingly, as a positive feedback, FAK further enhances signal pathway, which will break a new path for exploring the me-
the expression of KLF8 [56]. Nevertheless, KLF10 inhibits breast cancer chanisms involved in KLFs regulate proliferation.
cell invasion by descending the transcription of EGFR via binding with
HDAC1 to form a complex, which binds to Sp1 sites on the EGFR pro-
3.4.2. KLFs and other signaling pathways
moter and inhibits its transcription by suppressing histone acetylation
Besides hormone signaling pathway, KLFs also participate in several
[66,72]. Moreover, KLF11 is supposed to be a suppressor in breast
other signaling pathways like retinoic acid (RA) signaling pathway,
cancer invasion whereas the specific mechanism is still obsure [39].
Wnt/β-catenin signaling pathway, Notch signaling pathway as well as
During the invasion, angiogenesis is recognized as an essential part of
EGFR signaling pathway.
metastasis which delivers nutrition to cancer cells and transfers tumor
cells to a distant site [73]. Vascular endothelial growth factor (VEGF) is
an important angiogenic factor that promotes angiogenesis in a series of 3.4.2.1. Retinoic acid (RA) signaling pathway. KLF2 is downregulated in
pathological conditions, including cancer, inflammation and ischemic breast cancer and acts as a tumor suppressor which is positively
disorders. Breast cancer cells such as MDA-MB-231 are more aggressive correlated with breast cancer patient survival [83–85]. It inhibits
than normal breast epithelial cell MCF-10A, this transfer ability might breast cancer cell growth by shifting retinoic acid (RA) signaling
result from the high expression of VEGF. For instance, KLF4 inhibits pathway from the pro-carcinogenic Fatty acid-binding protein 5
angiopoiesis via inhibiting VEGF transcription. KLF4 recruits histone (FABP5)/peroxisome proliferators-activated receptor /δ(PPARβ/δ)
deacetylases (HDACs) -2 and -3 at the VEGF promoter which is over- pathway to the growth-suppressing cellular retinoic acid-binding
lapping with SAF-1- binding elements, whereas SAF-1 enhances but proteinⅡ(CRABPⅡ) / retinoic acid receptor (RAR) pathway [84]. In
KLF-4 attenuates VEGF expression in normal MCF-10A cells, which can another word, KLF2 facilitates the expression of CRABPⅡwith RAR but
explain the reason why MDA-MB-231cells are more aggressive than suppresses FABP5 with PPARβ/δ [86–88]. Besides, KLF2 can also be
MCF-10A cells [74]. epigenetic silenced by the Polycomb group protein Enhancer of Zeste
Homolog 2 (EZH2), which may explain the low-expression of KLF2 in
tumor samples from breast cancer patients [85].

4
J. Zhang, et al. Biomedicine & Pharmacotherapy 123 (2020) 109778

3.4.2.2. Wnt/β-catenin pathway. KLF5 promotes cancer cell kinase pathway is implicated in FBXO32-mediated ubiquitination of
proliferation via stimulating Wnt/β-catenin pathway in triple KLF4 [99]. KLF5 can be modified by ubiquitin E3 ligases such as
negative breast cancer. Mechanically, KLF5 binds to the promoter of WWOX, FBXW7, WWP1 [100–102], facilitating KLF5 degradation in a
CTNNB1(β-catenin) and reinforce the CTNNB1 transcription. proteasome-mediated degradation. On the contrary, some deubiquiti-
Interestingly, this promotive function of KLF5 can be upregulated by nases like ATXN3L, BAP1, USP3 exert their activitation to deubiquiti-
lncRNA PVT1. Mechanistically, lncRNA PVT1 promotes the interaction nate and stabilize KLF5 so as to promote proliferation in breast cancer
between KLF5 and deubiquitinase BAP1, preventing KLF5 from cells [103,104]. Mechanismly,take USP3 for example, C terminus of
ubiquitination degradation [89]. As a result, accumulated KLF5 USP3 interacted with N terminus of KLF5 to downregulate the ubiqui-
continuously activate Wnt/β-catenin pathway and boost breast cancer tinate level of KLF5 and promote proliferation of breast cancer [105].
proliferation.
4.2. Methylation
3.4.2.3. EGFR and Notch signaling pathway. KLF8 enhances EGFR
signaling pathway to drive breast cancer proliferation by repressing Protein methylation generally refers to the methylation of arginine
miR-141 transcription, which targets the 3′-untranslational region of [106] or lysine in a protein sequence [107]. Arginine can be methylated
EGFR, leading EGFR translation activation [90]. Additionally, KLF8 once (called monomethylarginine) or twice (arginine methyltransferase
activates Notch signaling pathway by increasing the transcription of (PRMTs) simultaneously transfer two methyl groups to the same ni-
miR-146a, which targets the 3’-UTR of Notch signaling inhibitor trogen at the end of the arginine polypeptide a symmetric methylargi-
(NUMB),as a result, the obstacle of Notch signaling pathway is nine at the atom, or a methyl group at each nitrogen end to become a
downregulated [65]. Thus, KLF8 takes part in the activation of EGFR symmetric dimethylarginine [106]. Zhou Z etc found that KLF4 could
signaling pathway and Notch signaling pathway. Nevertheless, whether be methylated by PRMT5, whose methylation on amino acids R374,
KLF8 simultaneously activate this two pathways or which pathway is R376, and R377 counteracts VHL/VBC-mediated KLF4 ubiquitination,
the dominating target is under consideration. averting degradation of KLF4 and promotes breast cancer proliferation
Collectively, these data imply that KLFs play a vital role in gov- [108].
erning proliferation-related signal pathway. However, how they or-
chestrate their crosstalk relationship with other signal pathway remain 5. KLFs in prognosis, treatment and drug resistance
to be further investigated.
5.1. Prognosis
3.5. KLFs in cancer stem cell regulation
As KLFs play key roles in the development of breast cancer, the
Cancer stem cells are rare immortal cells within a tumor that can expression level of KLFs can be a potent prognostic factor for patients
both self-renew by dividing and give rise to many cell types that con- with breast cancer. KLF4 is associated with lower pCR in locally ad-
stitute the tumor. Such cells have been found in various types of human vanced breast cancer patients undergoing neoadjuvant chemotherapy,
tumors [91] and might be attractive targets for cancer treatment [92]. suggesting that KLF4 may serve as a predictor for pCR in patients with
Breast cancer stem cells (BCSCs) are a rare population of cells with breast cancer after neoadjuvant chemotherapy [109]. However, in
critical roles in the initiation, growth, metastasis, and relapse of breast triple-negative breast cancer patients, KLF4 higher-expression group
cancer. BCSCs can enter the circulation and form a secondary tumor. had more favorable overall survival (OS) and disease-free survival
KLF4 is a general recognized stem-cell related genes which is high ex- (DFS) rates than KLF4 lower-expression group [110]. As a result, KLF4
pression in cancer stem cells and increase the stem cell marker (the functions as a biomarker depending on the type of breast cancer and
ratio of CD44/CD24) no matter in triple-negative or ERBB2 positive whether received treatment. Positive expression of KLF5 is significantly
breast cancer with a promotion in self-renewal and brain metastasis associated with increased risk of recurrence and worse clinical outcome
[70,93–95]. However, most of other members of KLFs family have not in breast cancer patients [82]. In addition, KLF5 is positively correlated
yet been fully elucidated, suggesting that further studies are extremely with HER2 and Ki67 expression but negatively correlated with the age
urgent to explore other members in cancer stem cell regulation. of the patients at diagnosis [111]. KLF6-SV is a marker of increased
metastatic potential indicating poor survival which is higher in patients
4. KLFs in post-translational regulation with metastasis than in patients without lymph node metastasis [53].
Nevertheless, KLF10 is negatively associated with outcomes. Low ex-
Post-translational modulation (PTM), which occurs after the com- pression of KLF10 predicts malignancy, lymph node metastasis and
pletion of protein translation, regulates protein structures and functions poor survival [112]. To a certain extent, KLF10 could give a hint about
by covalent addition of functional groups, peptides or other complex the status of ER and the response to endocrine therapy [113] (Table 1).
molecules reversibly or irreversibly, leading to extend and diversify
protein properties and enhance the ability of modulating protein sta- 5.2. Treatment and drug resistance
bility, localization and activity themselves or their interacting proteins
[96]. As KLFs control multiple processes and are indispensable for proper
function of the breast cancer. KLFs were proposed to be potential
4.1. Ubiquitination therapeutic targets. Zoledronic acid, used for the treatment breast
cancer bone metastases, can upregulate the expression of KLF2 and
Ubiquitin (Ub), consisting of 76 amino acid residues, is an evolu- KLF6 and induce apoptosis in MCF7 cells [83,114]. Besides, KLF5 acts
tionarily conserved protein dedicated to tagging target proteins for as an oncogene in the triple-negative breast cancer, and a large amount
degradation. Ubiquitination is a well-recognized PTM process, which of drugs exert their functions by depending on restraining the expres-
covalently attaches Ub to the modified proteins and regulates their sion of KLF5. For example, Mifepristone, a progesterone receptor (PR)
stability, functions and localizations involved in multiple cell functions and glucocorticoid receptor (GR) antagonist which is widely approved
and diseases, especially in cancer development [97,98]. KLF4 has re- for abortion can inhibit triple-negative breast cancer by downregulating
cently been demonstrated as a novel substrate of ubiquitin ligase SCF- KLF5 translation through mi-R153 binding to the 3’-UTR in transcripts
FBXO32, which physically interacts with the N-terminus (1–60 aa) of of KLF5. Mithramycin A, a DNA and RNA polymerase inhibitor, can also
KLF4 via its C-terminus (228–355 aa) and directly targets KLF4 for inhibit KLF5 in protein level in triple-negative breast cancer, but the
ubiquitination and degradation. Besides, p38 mitogen-activated protein mechanism is still not certain [115–117]. Velcade, a proteasome

5
J. Zhang, et al. Biomedicine & Pharmacotherapy 123 (2020) 109778

Table 1
KLFs in the breast cancer.
Name Process Regulation Biological functions Ref

KLF2 Proliferation down Shift RA signaling fromFABP5/PPARβ/δ to CRABP2/RAR [84]


KLF4 Cycle cycle down promote the cell cycle inhibitor p21, p27 [75]
metastasis down Inhibit VEGF transcription [74]
KLF4α Proliferation up recruit KLF4 to form complex and antagonize the role of KLF4 [27]
KLF5 proliferation up upregulate the cell cycle genes cyclin A2, cyclinD1, downregulate CDK inhibitors like p21 and p27 [28,29]
apoptosis down downregulate TNF-a and increase the anti-apoptotic factors like MCL1 and BCL-XL [39]
metastasis down upregulate E-cadherin [13]
KLF6 proliferation down promote CDKN1A transcription [78]
metastasis down upregulate E-cadherin [56]
KLF6-SV metastasis up Promote the transcription of TWIST1 [53]
KLF8 proliferation up Upregulate cyclinD1 [65]
metastasis up repress E-cadherin gene expression, [60,67–70]
promote MMP9 and MMP14 expression
KLF9 metastasis down Inhibit MMP9 expression [71,72]
KLF10 proliferation down The mechanism is not mentioned [26]
apoptosis up modulation of BI-1 (inhibition of BAX) expression and Ca2+ homeostasis [41]
KLF11 apoptosis up inhibit Bcl-2 expression, whilst upregulate Bax protein level [43]
KLF12 apoptosis down The mechanism is not mentioned [44,45]
KLF17 proliferation down attenuates estrogen receptor alpha-mediated signaling [28]
metastasis down Inhibit ID-1 expression [62]

inhibitor, accelerates breast cancer apoptosis by upregulating tumor 6. Conclusions and perspectives
suppressor KLF10 via inhibiting it’s proteolytic degradation [38]. Re-
markably, Melatonin, 2-Hydroxycinnamaldehyde and Paclitaxel can As DNA-binding transcriptional regulators, KLFs control a series of
stimulate KLF17, a suppressor of EMT and metastasis in breast cancer, essential cellular processes such as proliferation, differentiation, apop-
to play its repressive role in the metastasis of breast cancer [118–120]. tosis and migration in breast cancer. The levels of KLF factors are al-
What’s more, with the extension of the treatment cycle and disease tered during the development of breast cancer. Thus, KLFs might be
progression, drug resistance, a daunting challenge to the successful potential markers to indicate the malignancy of breast cancer.
treatment for breast cancer is gradually emerging. KLFs are related with Importantly, targeting KLFs is proposed as a novel and feasible ap-
this process involved in endocrine therapy resistance, Anti-Her2 treat- proach in breast cancer treatment and prevention. Significant pro-
ment resistance and chemotherapy resistance. For instance, in tamox- gresses have been achieved in understanding the mechanisms by which
ifen resistance breast cancer patients, KLF4 is downregulated and how KLFs function. However, there are still an amount of questions to
overexpression of KLF4 can contribute to TAM sensitivity via inhibiting be solved. For example, whether some KLFs have other functions be-
phosphorylation modification of ERK and p38 signaling, suggesting that sides transcriptional role is still unclear. Some KLFs play promotive
enhancing KLF4 expression may be a potential therapeutic strategy for roles in the development of breast cancer, but whether these factors
breast cancer treatment, especially for the TAM-resistant patients. Be- have synergistic or contrary effect remains to be further explored. So
sides, in Her-2 positive breast cancer, inhibiting the expression of KLF4 far, it is obscure that how the organism balances the relationship be-
and KLF5 can rescue Lapatinib resistance by downregulating anti- tween oncogenetic KLFs and tumor suppressive KLFs, maybe depending
apoptotic factors myeloid cell leukemia 1 (MCL1) and B-cell lymphoma- on identical cellular context and tissue-restricted function. During the
extra-large (BCL-XL) [36]. Lacking of hormone receptor and Human process of tumorigenesis when a KLF expression is altered, other KLFs
epidermal growth factor receptor 2, chemotherapy is the mainstay of will make compensatory changes, compensating for its function to keep
treatment for triple-negative breast cancer (TNBC). Dexamethasone the body in a steady state, and when the external stimulus exceeded the
induces the drug resistance of docetaxel and cisplatin partially through scope of compensation, some other KLFs will have positive feedback,
up-regulating KLF5 in protein level in TNBC, thus targeting KLF5 may making the body develop towards cancer. Besides, the organism ba-
provide a promising therapy to overcome chemotherapy resistance lanced the relationship between oncogenes and tumor suppressors is
[121] Collectively, we summarize of note small molecular compounds dependent on the expression levels of different KLFs, like teeter-board,
capable of modifying expression and activities of KLF family in Table2. which leans to the heavier. Moreover, tumor microenvironment, on-
cogenic mutations, genomic instability and distinct cancer-specific
contexts are also orchestrated to break this balance. In addition, the
problem that drugs targeting some oncogenes may attenuate other

Table 2
Agents That Target KLF Proteins in breast cancer.
Compound Affected KLF family Mode of action Ref
members

Zoledronic acid KLF2, KLF6 Zoledronic acid upregulate the tumor suppressors KLF2 and KLF6 and induce apoptosis [83]
Mifepristone KLF5 Mifepristone suppresses the expression of KLF5 through inducing the expression of miR-153 and induce apoptosis [115]
Mithramycin A KLF5 MithramycinA suppress KLF5 transcription and induce apoptosis [116]
Homoharringtonine KLF10 Homoharringtonine upregulate KLF10 and increase BAX2 and Bim, meanwhile decrease Bcl-2 and BCL-XL, active [38]
Velcade caspase3.
Melatonin KLF17 Melatonin reduce DJ-1 and ID-1 expression and increase the transcription of KLF17, what,s more, Melatonin [118]
induce GSK3-beta nuclear localization and suppress Snail and rise E-cadherin expression to inhibit EMT
2-Hydroxycinnamaldehyde KLF17 Same to Melatonin [119]
Paclitaxel KLF17 Paclitaxel reduce DJ-1 and ID-1 expression and increase the transcription of KLF17 [120]

6
J. Zhang, et al. Biomedicine & Pharmacotherapy 123 (2020) 109778

tumor suppressors due to the homology of KLFs is also worthy of under Y. Wang, R. Liu, J. Feng, C. Chen, KLF5 promotes breast cancer proliferation,
consideration. Collectively, better understanding of KLFs in breast migration and invasion in part by upregulating the transcription of TNFAIP2,
Oncogene 35 (2016) 2040–2051.
cancer and clarifying their intrinsic molecular mechanisms will provide [29] R. Liu, P. Shi, Z. Zhou, H. Zhang, W. Li, H. Zhang, C. Chen, Krupple-like factor 5 is
ideas for the development of novel cancer therapeutic intervention in essential for mammary gland development and tumorigenesis, J. Pathol. 246
breast cancer. (2018) 497–507.
[30] H.Q. Zheng, Z. Zhou, J. Huang, L. Chaudhury, J.T. Dong, C. Chen, Kruppel-like
factor 5 promotes breast cell proliferation partially through upregulating the
Declaration of Competing Interest transcription of fibroblast growth factor binding protein 1, Oncogene 28 (2009)
3702–3713.
[31] R. Liu, Z. Zhou, D. Zhao, C. Chen, The induction of KLF5 transcription factor by
We had no conflict interest to claim. progesterone contributes to progesterone-induced breast cancer cell proliferation
and dedifferentiation, Mol. Endocrinol. 25 (2011) 1137–1144.
Acknowledgements [32] W. Zhou, F. Song, Q. Wu, R. Liu, L. Wang, C. Liu, Y. Peng, S. Mao, Feng J and Chen
C. miR-217 inhibits triple-negative breast cancer cell growth, migration, and in-
vasion through targeting KLF5, PLoS One 12 (2017) e0176395.
This work was supported by Zheng Shu Medical Elite Scholarship [33] H. Xia, C. Wang, W. Chen, H. Zhang, L. Chaudhury, Z. Zhou, R. Liu, C. Chen,
Fund. The National Natural Science Foundation of China (81702976). Kruppel-like factor 5 transcription factor promotes microsomal prostaglandin E2
Zhejiang TCM science and technology program (2014Zb069). Zhejiang synthase 1 gene transcription in breast cancer, J. Biol. Chem. 288 (2013)
26731–26740.
Provincial Program for the Cultivation of High-Level Innovative Health [34] X. Jia, L. Shi, X. Wang, L. Luo, L. Ling, J. Yin, Y. Song, Z. Zhang, N. Qiu, H. Liu,
Talents. M. Deng, Z. He, H. Li, G. Zheng, KLF5 regulated lncRNA RP1 promotes the growth
and metastasis of breast cancer via repressing p27kip1 translation, Cell Death Dis.
10 (2019) 373.
References [35] J. Shi, C. Liu, A. Zhang, N. Cui, B. Wang, B. Chen, Z. Ma, [Roles of KLF5 in in-
hibition TNFalpha-induced SK-BR-3 breast cancer cell apoptosis], Zhonghua Yi
[1] C. Woolston, Breast cancer, Nature 527 (2015) S101. Xue Za Zhi 94 (2014) 2004–2007.
[2] N. Harbeck, M. Gnant, Breast cancer, Lancet 389 (2017) 1134–1150. [36] M.K. Farrugia, S.B. Sharma, C.C. Lin, S.L. McLaughlin, D.B. Vanderbilt,
[3] A.G. Waks, E.P. Winer, Breast cancer treatment: a review, JAMA 321 (2019) A.G. Ammer, M.A. Salkeni, P. Stoilov, Y.M. Agazie, C.J. Creighton, J.M. Ruppert,
288–300. Regulation of anti-apoptotic signaling by Kruppel-like factors 4 and 5 mediates
[4] N.C. Turner, P. Neven, S. Loibl, F. Andre, Advances in the treatment of advanced lapatinib resistance in breast cancer, Cell Death Dis. 6 (2015) e1699.
oestrogen-receptor-positive breast cancer, Lancet 389 (2017) 2403–2414. [37] C.F. Hsu, C.L. Sui, W.C. Wu, J.J. Wang, D.H. Yang, Y.C. Chen, W.C. Yu, H.S. Chang,
[5] J.M. Specht, N.E. Davidson, Optimal duration of trastuzumab for early HER2- Klf10 induces cell apoptosis through modulation of BI-1 expression and Ca2+
positive breast cancer, Lancet 389 (2017) 1167–1168. homeostasis in estrogen-responding adenocarcinoma cells, Int. J. Biochem. Cell
[6] C. Denkert, C. Liedtke, A. Tutt, G. von Minckwitz, Molecular alterations in triple- Biol. 43 (2011) 666–673.
negative breast cancer-the road to new treatment strategies, Lancet 389 (2017) [38] W. Jin, G. Di, J. Li, Y. Chen, W. Li, J. Wu, T. Cheng, M. Yao, Z. Shao, TIEG1 induces
2430–2442. apoptosis through mitochondrial apoptotic pathway and promotes apoptosis in-
[7] Comprehensive molecular portraits of human breast tumours, Nature 490 (2012) duced by homoharringtonine and velcade, FEBS Lett. 581 (2007) 3826–3832.
61–70. [39] M. Han, Y. Wang, G. Guo, L. Li, D. Dou, X. Ge, P. Lv, F. Wang, Y. Gu, microRNA-
[8] C.B. Matsen, L.A. Neumayer, Breast cancer: a review for the general surgeon, 30d mediated breast cancer invasion, migration, and EMT by targeting KLF11 and
JAMA Surg. 148 (2013) 971–979. activating STAT3 pathway, J. Cell. Biochem. 119 (2018) 8138–8145.
[9] J.S. Vaidya, M. Bulsara, F. Wenz, J.S. Tobias, D. Joseph, M. Baum, Targeted [40] B. Guan, Q. Li, L. Shen, Q. Rao, Y. Wang, Y. Zhu, X.J. Zhou, X.H. Li, MicroRNA-205
radiotherapy for early breast cancer, Lancet 391 (2018) 26–27. directly targets Kruppel-like factor 12 and is involved in invasion and apoptosis in
[10] L. The, Breast cancer targeted therapy: successes and challenges, Lancet 389 basal-like breast carcinoma, Int. J. Oncol. 49 (2016) 720–734.
(2017) 2350. [41] B. Guan, Q. Li, X.H. Li, X.J. Zhou, [MicroRNA-205 targeted Kruppel-like factor 12
[11] L.A. Emens, Breast cancer immunotherapy: facts and hopes, Clin. Cancer Res. 24 and regulated MDA-MB-468 cells apoptosis in basal-like breast carcinoma],
(2018) 511–520. Zhonghua Yi Xue Za Zhi 96 (2016) 2070–2075.
[12] B.B. McConnell, V.W. Yang, Mammalian Kruppel-like factors in health and dis- [42] A.C. Chiang, J. Massague, Molecular basis of metastasis, N. Engl. J. Med. 359
eases, Physiol. Rev. 90 (2010) 1337–1381. (2008) 2814–2823.
[13] C. Bureau, N. Hanoun, J. Torrisani, J.P. Vinel, L. Buscail, P. Cordelier, Expression [43] I.J. Fidler, The pathogenesis of cancer metastasis: the’ seed and soil’ hypothesis
and function of Kruppel Like-Factors (KLF) in carcinogenesis, Curr. Genomics 10 revisited, Nat. Rev. Cancer 3 (2003) 453–458.
(2009) 353–360. [44] G.P. Gupta, J. Massague, Cancer metastasis: building a framework, Cell 127
[14] M.P. Tetreault, Y. Yang, J.P. Katz, Kruppel-like factors in cancer, Nat. Rev. Cancer (2006) 679–695.
13 (2013) 701–713. [45] A. Singh, J. Settleman, EMT, cancer stem cells and drug resistance: an emerging
[15] Z. Cao, X. Sun, B. Icli, A.K. Wara, M.W. Feinberg, Role of Kruppel-like factors in axis of evil in the war on cancer, Oncogene 29 (2010) 4741–4751.
leukocyte development, function, and disease, Blood 116 (2010) 4404–4414. [46] J.P. Thiery, Epithelial-mesenchymal transitions in tumour progression, Nat. Rev.
[16] R.C. Simmen, M.E. Heard, A.M. Simmen, M.T. Montales, M. Marji, S. Scanlon, Cancer 2 (2002) 442–454.
J.M. Pabona, The Kruppel-like factors in female reproductive system pathologies, [47] A. Cano, M.A. Perez-Moreno, I. Rodrigo, A. Locascio, M.J. Blanco, M.G. del Barrio,
J. Mol. Endocrinol. 54 (2015) R89–R101. F. Portillo, M.A. Nieto, The transcription factor snail controls epithelial-me-
[17] J. Kaczynski, T. Cook, R. Urrutia, Sp1- and Kruppel-like transcription factors, senchymal transitions by repressing E-cadherin expression, Nat. Cell Biol. 2 (2000)
Genome Biol. 4 (2003) 206. 76–83.
[18] S.K. Swamynathan, Kruppel-like factors: three fingers in control, Hum. Genomics 4 [48] M.A. Nieto, M.G. Sargent, D.G. Wilkinson, J. Cooke, Control of cell behavior
(2010) 263–270. during vertebrate development by Slug, a zinc finger gene, Science 264 (1994)
[19] J.M. Wells, F.M. Watt, Diverse mechanisms for endogenous regeneration and re- 835–839.
pair in mammalian organs, Nature 557 (2018) 322–328. [49] J. Feng, D. Song, S. Jiang, X. Yang, T. Ding, H. Zhang, J. Luo, J. Liao, Q. Yin,
[20] A. Schulze, A.L. Harris, How cancer metabolism is tuned for proliferation and Quercetin restrains TGF-beta1-induced epithelial-mesenchymal transition by in-
vulnerable to disruption, Nature 491 (2012) 364–373. hibiting Twist1 and regulating E-cadherin expression, Biochem. Biophys. Res.
[21] J. Massague, G1 cell-cycle control and cancer, Nature 432 (2004) 298–306. Commun. 498 (2018) 132–138.
[22] G.I. Evan, K.H. Vousden, Proliferation, cell cycle and apoptosis in cancer, Nature [50] E. Batlle, E. Sancho, C. Franci, D. Dominguez, M. Monfar, J. Baulida, A. Garcia De
411 (2001) 342–348. Herreros, The transcription factor snail is a repressor of E-cadherin gene expres-
[23] K.W. Foster, A.R. Frost, P. McKie-Bell, C.Y. Lin, J.A. Engler, W.E. Grizzle, sion in epithelial tumour cells, Nat. Cell Biol. 2 (2000) 84–89.
J.M. Ruppert, Increase of GKLF messenger RNA and protein expression during [51] J.L. Yori, E. Johnson, G. Zhou, M.K. Jain, R.A. Keri, Kruppel-like factor 4 inhibits
progression of breast cancer, Cancer Res. 60 (2000) 6488–6495. epithelial-to-mesenchymal transition through regulation of E-cadherin gene ex-
[24] S.H. Kim, S.V. Singh, Role of kruppel-like factor 4-p21(CIP1) Axis in breast Cancer pression, J. Biol. Chem. 285 (2010) 16854–16863.
Stem-like cell inhibition by benzyl isothiocyanate, Cancer Prev. Res. (Phila.) 12 [52] H.K. Lee, D.S. Lee, J.C. Park, Nuclear factor I-C regulates E-cadherin via control of
(2019) 125–134. KLF4 in breast cancer, BMC Cancer 15 (2015) 113.
[25] D. Hu, Z. Zhou, N.E. Davidson, Y. Huang, Y. Wan, Novel insight into KLF4 pro- [53] R. Hatami, A.M. Sieuwerts, S. Izadmehr, Z. Yao, R.F. Qiao, L. Papa, M.P. Look,
teolytic regulation in estrogen receptor signaling and breast carcinogenesis, J. M. Smid, J. Ohlssen, A.C. Levine, D. Germain, D. Burstein, A. Kirschenbaum,
Biol. Chem. 287 (2012) 13584–13597. A. DiFeo, J.A. Foekens, G. Narla, KLF6-SV1 drives breast cancer metastasis and is
[26] X. Song, Y.M. Xing, W. Wu, G.H. Cheng, F. Xiao, G. Jin, Y. Liu, X. Zhao, Expression associated with poor survival, Sci. Transl. Med. 5 (2013) 169ra112.
of Kruppel-like factor 4 in breast cancer tissues and its effects on the proliferation [54] An oncogenic splice variant drives EMT and metastasis in breast cancer, Cancer
of breast cancer MDA-MB-231 cells, Exp. Ther. Med. 13 (2017) 2463–2467. Discov. 3 (2013) Of16.
[27] J. Ferralli, R. Chiquet-Ehrismann, M. Degen, KLF4alpha stimulates breast cancer [55] O.C. Olson, J.A. Joyce, A splicing twist on metastasis, Sci. Transl. Med. 5 (2013)
cell proliferation by acting as a KLF4 antagonist, Oncotarget 7 (2016) 169fs162.
45608–45621. [56] X. Wang, H. Lu, A.M. Urvalek, T. Li, L. Yu, J. Lamar, C.M. DiPersio, P.J. Feustel,
[28] L. Jia, Z. Zhou, H. Liang, J. Wu, P. Shi, F. Li, Z. Wang, C. Wang, W. Chen, H. Zhang, J. Zhao, KLF8 promotes human breast cancer cell invasion and metastasis by

7
J. Zhang, et al. Biomedicine & Pharmacotherapy 123 (2020) 109778

transcriptional activation of MMP9, Oncogene 30 (2011) 1901–1911. 741–750.


[57] X. Wang, M. Zheng, G. Liu, W. Xia, P.J. McKeown-Longo, M.C. Hung, J. Zhao, [83] R. Ebert, S. Zeck, J. Meissner-Weigl, B. Klotz, T.D. Rachner, P. Benad, L. Klein-
Kruppel-like factor 8 induces epithelial to mesenchymal transition and epithelial Hitpass, M. Rudert, L.C. Hofbauer, F. Jakob, Kruppel-like factors KLF2 and 6 and
cell invasion, Cancer Res. 67 (2007) 7184–7193. Ki-67 are direct targets of zoledronic acid in MCF-7 cells, Bone 50 (2012) 723–732.
[58] K. Gumireddy, A. Li, P.A. Gimotty, A.J. Klein-Szanto, L.C. Showe, D. Katsaros, [84] W. Zhang, L. Levi, P. Banerjee, M. Jain, N. Noy, Kruppel-like factor 2 suppresses
G. Coukos, L. Zhang, Q. Huang, KLF17 is a negative regulator of epithelial-me- mammary carcinoma growth by regulating retinoic acid signaling, Oncotarget 6
senchymal transition and metastasis in breast cancer, Nat. Cell Biol. 11 (2009) (2015) 35830–35842.
1297–1304. [85] H. Taniguchi, F.V. Jacinto, A. Villanueva, A.F. Fernandez, H. Yamamoto,
[59] I.A. Ismail, H.S. Kang, H.J. Lee, J.K. Kim, S.H. Hong, DJ-1 upregulates breast F.J. Carmona, S. Puertas, V.E. Marquez, Y. Shinomura, K. Imai, M. Esteller,
cancer cell invasion by repressing KLF17 expression, Br. J. Cancer 110 (2014) Silencing of Kruppel-like factor 2 by the histone methyltransferase EZH2 in human
1298–1306. cancer, Oncogene 31 (2012) 1988–1994.
[60] A. Ali, A.S. Shah, A. Ahmad, Gain-of-function of mutant p53: mutant p53 enhances [86] P. Kannan-Thulasiraman, D.D. Seachrist, G.H. Mahabeleshwar, M.K. Jain, N. Noy,
cancer progression by inhibiting KLF17 expression in invasive breast carcinoma Fatty acid-binding protein 5 and PPARbeta/delta are critical mediators of epi-
cells, Cancer Lett. 354 (2014) 87–96. dermal growth factor receptor-induced carcinoma cell growth, J. Biol. Chem. 285
[61] C. Bonnans, J. Chou, Z. Werb, Remodelling the extracellular matrix in develop- (2010) 19106–19115.
ment and disease, Nat. Rev. Mol. Cell Biol. 15 (2014) 786–801. [87] T.T. Schug, D.C. Berry, N.S. Shaw, S.N. Travis, N. Noy, Opposing effects of retinoic
[62] M.W. Pickup, J.K. Mouw, V.M. Weaver, The extracellular matrix modulates the acid on cell growth result from alternate activation of two different nuclear re-
hallmarks of cancer, EMBO Rep. 15 (2014) 1243–1253. ceptors, Cell 129 (2007) 723–733.
[63] H. Guo, Y. Lin, H. Zhang, J. Liu, N. Zhang, Y. Li, D. Kong, Q. Tang, D. Ma, Tissue [88] L.J. Donato, N. Noy, Suppression of mammary carcinoma growth by retinoic acid:
factor pathway inhibitor-2 was repressed by CpG hypermethylation through in- proapoptotic genes are targets for retinoic acid receptor and cellular retinoic acid-
hibition of KLF6 binding in highly invasive breast cancer cells, BMC Mol. Biol. 8 binding protein II signaling, Cancer Res. 65 (2005) 8193–8199.
(2007) 110. [89] J. Tang, Y. Li, Y. Sang, B. Yu, D. Lv, W. Zhang, H. Feng, LncRNA PVT1 regulates
[64] H. Lu, L. Hu, L. Yu, X. Wang, A.M. Urvalek, T. Li, C. Shen, D. Mukherjee, triple-negative breast cancer through KLF5/beta-catenin signaling, Oncogene 37
S.K. Lahiri, M.S. Wason, J. Zhao, KLF8 and FAK cooperatively enrich the active (2018) 4723–4734.
MMP14 on the cell surface required for the metastatic progression of breast [90] T. Li, H. Lu, D. Mukherjee, S.K. Lahiri, C. Shen, L. Yu, J. Zhao, Identification of
cancer, Oncogene 33 (2014) 2909–2917. epidermal growth factor receptor and its inhibitory microRNA141 as novel targets
[65] X. Wang, H. Lu, T. Li, L. Yu, G. Liu, X. Peng, J. Zhao, Kruppel-like factor 8 pro- of Kruppel-like factor 8 in breast cancer, Oncotarget 6 (2015) 21428–21442.
motes tumorigenic mammary stem cell induction by targeting miR-146a, Am. J. [91] T. Reya, H. Clevers, Wnt signalling in stem cells and cancer, Nature 434 (2005)
Cancer Res. 3 (2013) 356–373. 843–850.
[66] D. Mukherjee, H. Lu, L. Yu, C. He, S.K. Lahiri, T. Li, J. Zhao, Kruppel-like factor 8 [92] J.E. Visvader, Cells of origin in cancer, Nature 469 (2011) 314–322.
activates the transcription of C-X-C cytokine receptor type 4 to promote breast [93] R. Mimoto, Y. Imawari, S. Hirooka, H. Takeyama, K. Yoshida, Impairment of
cancer cell invasion, transendothelial migration and metastasis, Oncotarget 7 DYRK2 augments stem-like traits by promoting KLF4 expression in breast cancer,
(2016) 23552–23568. Oncogene 36 (2017) 1862–1872.
[67] F. Pu, S. Tanaka, M. Kawashima, K. Kawaguchi, M. Sugimoto, S. Saji, M. Toi, [94] D.M. Cittelly, J. Finlay-Schultz, E.N. Howe, N.S. Spoelstra, S.D. Axlund,
R. Limame, K.O. de Beeck, S. Van Laere, L. Croes, A. De Wilde, L. Dirix, G. Van P. Hendricks, B.M. Jacobsen, C.A. Sartorius, J.K. Richer, Progestin suppression of
Camp, M. Peeters, O. De Wever, F. Lardon, P. Pauwels, Expression profiling of miR-29 potentiates dedifferentiation of breast cancer cells via KLF4, Oncogene 32
migrated and invaded breast cancer cells predicts early metastatic relapse and (2013) 2555–2564.
reveals Kruppel-like factor 9 as a potential suppressor of invasive growth in breast [95] R. Fajka-Boja, A. Marton, A. Toth, P. Blazso, V. Tubak, B. Balint, I. Nagy,
cancer, Breast Cancer Res. 1 (2014) 69–81. Z. Hegedus, C. Vizler, R.L. Katona, Increased insulin-like growth factor 1 pro-
[68] X.Y. Bai, S. Li, M. Wang, X. Li, Y. Yang, Z. Xu, B. Li, Y. Li, K. Xia, H. Chen, H. Wu, duction by polyploid adipose stem cells promotes growth of breast cancer cells,
Kruppel-like factor 9 down-regulates matrix metalloproteinase 9 transcription and BMC Cancer 18 (2018) 872.
suppresses human breast cancer invasion, Cancer Lett. 412 (2018) 224–235. [96] S. Mateo Sanchez, S.D. Freeman, L. Delacroix, B. Malgrange, The role of post-
[69] R.C. Simmen, J.M. Pabona, M.C. Velarde, C. Simmons, O. Rahal, F.A. Simmen, The translational modifications in hearing and deafness, Cell. Mol. Life Sci. 73 (2016)
emerging role of Kruppel-like factors in endocrine-responsive cancers of female 3521–3533.
reproductive tissues, J. Endocrinol. 204 (2010) 223–231. [97] D. Popovic, D. Vucic, I. Dikic, Ubiquitination in disease pathogenesis and treat-
[70] F. Yu, J. Li, H. Chen, J. Fu, S. Ray, S. Huang, H. Zheng, W. Ai, Kruppel-like factor 4 ment, Nat. Med. 20 (2014) 1242–1253.
(KLF4) is required for maintenance of breast cancer stem cells and for cell mi- [98] I.A. Voutsadakis, Ubiquitin- and ubiquitin-like proteins-conjugating enzymes (E2s)
gration and invasion, Oncogene 30 (2011) 2161–2172. in breast cancer, Mol. Biol. Rep. 40 (2013) 2019–2034.
[71] T. Li, H. Lu, C. Shen, S.K. Lahiri, M.S. Wason, D. Mukherjee, L. Yu, J. Zhao, [99] H. Zhou, Y. Liu, R. Zhu, F. Ding, Y. Wan, Y. Li, Z. Liu, FBXO32 suppresses breast
Identification of epithelial stromal interaction 1 as a novel effector downstream of cancer tumorigenesis through targeting KLF4 to proteasomal degradation,
Kruppel-like factor 8 in breast cancer invasion and metastasis, Oncogene 33 Oncogene 36 (2017) 3312–3321.
(2014) 4746–4755. [100] F. Ge, W. Chen, R. Yang, Z. Zhou, N. Chang, C. Chen, T. Zou, R. Liu, J. Tan, G. Ren,
[72] W. Jin, B.B. Chen, J.Y. Li, H. Zhu, M. Huang, S.M. Gu, Q.Q. Wang, J.Y. Chen, S. Yu, WWOX suppresses KLF5 expression and breast cancer cell growth, Chin. J. Cancer
J. Wu, Z.M. Shao, TIEG1 inhibits breast cancer invasion and metastasis by in- Res. 26 (2014) 511–516.
hibition of epidermal growth factor receptor (EGFR) transcription and the EGFR [101] D. Zhao, H.Q. Zheng, Z. Zhou, C. Chen, The Fbw7 tumor suppressor targets KLF5
signaling pathway, Mol. Cell. Biol. 32 (2012) 50–63. for ubiquitin-mediated degradation and suppresses breast cell proliferation,
[73] A.F. Chambers, A.C. Groom, I.C. MacDonald, Dissemination and growth of cancer Cancer Res. 70 (2010) 4728–4738.
cells in metastatic sites, Nat. Rev. Cancer 2 (2002) 563–572. [102] D. Zhao, X. Zhi, Z. Zhou, C. Chen, TAZ antagonizes the WWP1-mediated KLF5
[74] A. Ray, M. Alalem, B.K. Ray, Loss of epigenetic Kruppel-like factor 4 histone degradation and promotes breast cell proliferation and tumorigenesis,
deacetylase (KLF-4-HDAC)-mediated transcriptional suppression is crucial in in- Carcinogenesis 33 (2012) 59–67.
creasing vascular endothelial growth factor (VEGF) expression in breast cancer, J. [103] F. Ge, W. Chen, J. Qin, Z. Zhou, R. Liu, L. Liu, J. Tan, T. Zou, H. Li, G. Ren, C. Chen,
Biol. Chem. 288 (2013) 27232–27242. Ataxin-3 like (ATXN3L), a member of the Josephin family of deubiquitinating
[75] K. Akaogi, Y. Nakajima, I. Ito, S. Kawasaki, S.H. Oie, A. Murayama, K. Kimura, enzymes, promotes breast cancer proliferation by deubiquitinating Kruppel-like
J. Yanagisawa, KLF4 suppresses estrogen-dependent breast cancer growth by in- factor 5 (KLF5), Oncotarget 6 (2015) 21369–21378.
hibiting the transcriptional activity of ERalpha, Oncogene 28 (2009) 2894–2902. [104] J. Qin, Z. Zhou, W. Chen, C. Wang, H. Zhang, G. Ge, M. Shao, D. You, Z. Fan,
[76] P. Guo, X.Y. Dong, K.W. Zhao, X. Sun, Q. Li, J.T. Dong, Estrogen-induced inter- H. Xia, R. Liu, C. Chen, BAP1 promotes breast cancer cell proliferation and me-
action between KLF5 and estrogen receptor (ER) suppresses the function of ER in tastasis by deubiquitinating KLF5, Nat. Commun. 6 (2015) 8471.
ER-positive breast cancer cells, Int. J. Cancer 126 (2010) 81–89. [105] Y. Wu, J. Qin, F. Li, C. Yang, Z. Li, Z. Zhou, H. Zhang, Y. Li, X. Wang, R. Liu,
[77] R. Liu, J.T. Dong, C. Chen, Role of KLF5 in hormonal signaling and breast cancer Q. Tao, W. Chen, C. Chen, USP3 promotes breast cancer cell proliferation by
development, Vitam. Horm. 93 (2013) 213–225. deubiquitinating KLF5, J. Biol. Chem. (2019).
[78] J. Liu, T. Du, Y. Yuan, Y. He, Z. Tan, Z. Liu, KLF6 inhibits estrogen receptor- [106] C. Peng, C.C. Wong, The story of protein arginine methylation: characterization,
mediated cell growth in breast cancer via a c-Src-mediated pathway, Mol. Cell. regulation, and function, Expert Rev. Proteomics 14 (2017) 157–170.
Biochem. 335 (2010) 29–35. [107] P.O. Falnes, M.E. Jakobsson, E. Davydova, A. Ho, J. Malecki, Protein lysine me-
[79] R. Sarfstein, A. Belfiore, H. Werner, Identification of insulin-like growth Factor-I thylation by seven-beta-strand methyltransferases, Biochem. J. 473 (2016)
receptor (IGF-IR) gene promoter-binding proteins in estrogen receptor (ER)-posi- 1995–2009.
tive and ER-depleted breast cancer cells, Cancers (Basel) 2 (2010) 233–261. [108] D. Hu, M. Gur, Z. Zhou, A. Gamper, M.C. Hung, N. Fujita, L. Lan, I. Bahar, Y. Wan,
[80] A. Ali, I. Ielciu, H.M. Alkreathy, A.A. Khan, KLF17 attenuates estrogen receptor Interplay between arginine methylation and ubiquitylation regulates KLF4-medi-
alpha-mediated signaling by impeding ERalpha function on chromatin and de- ated genome stability and carcinogenesis, Nat. Commun. 6 (2015) 8419.
termines response to endocrine therapy, Biochim. Biophys. Acta 1859 (2016) [109] M.J. Dong, L.B. Wang, Z.N. Jiang, M. Jin, W.X. Hu, J.G. Shen, The transcription
883–895. factor KLF4 as an independent predictive marker for pathologic complete remis-
[81] G.H. El-Sokkary, S.H. Saber, S.P. Gao, H.F. Sun, L.D. Li, W.Y. Fu, W. Jin, UHRF1 sion in breast cancer neoadjuvant chemotherapy: a case-control study, Onco. Ther.
promotes breast cancer progression by suppressing KLF17 expression by hy- 7 (2014) 1963–1969.
permethylating its promoter, Clin. Exp. Pharmacol. Physiol. 7 (2017) 1554–1565. [110] T. Nagata, Y. Shimada, S. Sekine, M. Moriyama, I. Hashimoto, K. Matsui,
[82] K. Takagi, Y. Miki, Y. Onodera, Y. Nakamura, T. Ishida, M. Watanabe, S. Inoue, T. Okumura, T. Hori, J. Imura, K. Tsukada, KLF4 and NANOG are prognostic
H. Sasano, T. Suzuki, Kruppel-like factor 5 in human breast carcinoma: a potent biomarkers for triple-negative breast cancer, Breast Cancer 24 (2017) 326–335.
prognostic factor induced by androgens, Endocr. Relat. Cancer 19 (2012) [111] D. Tong, K. Czerwenka, G. Heinze, M. Ryffel, E. Schuster, A. Witt, S. Leodolter,

8
J. Zhang, et al. Biomedicine & Pharmacotherapy 123 (2020) 109778

R. Zeillinger, Expression of KLF5 is a prognostic factor for disease-free survival and regulating Kruppel-like factor 5 transcription by Sp1, Sci. Rep. 8 (2018) 1138.
overall survival in patients with breast cancer, Clin. Cancer Res. 12 (2006) [117] Y. Lin, R. Liu, P. Zhao, J. Ye, Z. Zheng, J. Huang, Y. Zhang, Y. Gao, H. Chen, S. Liu,
2442–2448. J. Zhou, C. Chen, H. Chen, Discovery of novel mifepristone derivatives via sup-
[112] M.M. Reinholz, M.W. An, S.A. Johnsen, M. Subramaniam, V.J. Suman, J.N. Ingle, pressing KLF5 expression for the treatment of triple-negative breast cancer, Eur. J.
P.C. Roche, T.C. Spelsberg, Differential gene expression of TGF beta inducible Med. Chem. 146 (2018) 354–367.
early gene (TIEG), Smad7, Smad2 and Bard1 in normal and malignant breast [118] G.H. El-Sokkary, I.A. Ismail, Melatonin inhibits breast cancer cell invasion through
tissue, Breast Cancer Res. Treat. 86 (2004) 75–88. modulating DJ-1/KLF17/ID-1 signaling pathway, J. Cell. Biochem. 120 (2019)
[113] M. Subramaniam, J.R. Hawse, S.A. Johnsen, T.C. Spelsberg, Role of TIEG1 in 3945–3957.
biological processes and disease states, J. Cell. Biochem. 102 (2007) 539–548. [119] I.A. Ismail, H.S. Kang, H.J. Lee, H. Chang, J. Yun, C.W. Lee, N.H. Kim, H.S. Kim,
[114] R. 、Ebert, J. Meissner-Weigl, S. Zeck, J. Maatta, S. Auriola, S. Coimbra de Sousa, J.I. Yook, S.H. Hong, B.M. Kwon, 2-Hydroxycinnamaldehyde inhibits the epithe-
B. Mentrup, S. Graser, T.D. Rachner, L.C. Hofbauer, F. Jakob, Probenecid as a lial-mesenchymal transition in breast cancer cells, Breast Cancer Res. Treat. 137
sensitizer of bisphosphonate-mediated effects in breast cancer cells, Mol. Cancer (2013) 697–708.
13 (2014) 265. [120] S.H. Saber, I.A. Ismail, Low doses of Paclitaxel repress breast cancer invasion
[115] R. Liu, P. Shi, Z. Nie, H. Liang, Z. Zhou, W. Chen, H. Chen, C. Dong, R. Yang, S. Liu, through DJ-1/KLF17 signalling pathway, J. Cell. Biochem. (2018).
C. Chen, Mifepristone suppresses basal triple-negative breast Cancer stem cells by [121] Z. Li, J. Dong, T. Zou, C. Du, S. Li, C. Chen, R. Liu, K. Wang, Dexamethasone
down-regulating KLF5 expression, Theranostics 6 (2016) 533–544. induces docetaxel and cisplatin resistance partially through up-regulating Kruppel-
[116] R. Liu, X. Zhi, Z. Zhou, H. Zhang, R. Yang, T. Zou, C. Chen, Mithramycin A sup- like factor 5 in triple-negative breast cancer, Oncotarget 8 (2017) 11555–11565.
presses basal triple-negative breast cancer cell survival partially via down-

You might also like