Skip to main content

PRC2 mediated KLF2 down regulation: a therapeutic and diagnostic axis during tumor progression

Abstract

Surgery and chemo-radiotherapy are used as the common first-line treatment options in many cancers. However, tumor relapse is observed in many cancer patients following such first-line treatments. Therefore, targeted therapy according to the molecular cancer biology can be very important in reducing tumor recurrence. In this regard, a wide range of monoclonal antibodies against the growth factors and their receptors can offer more targeted treatment in cancer patients. However, due to the importance of growth factors in the normal biology of body cells, side effects can also be observed following the application of growth factor inhibitors. Therefore, more specific factors should be introduced as therapeutic targets with less side effects. Krüppel-like factors 2 (KLF2) belongs to the KLF family of transcription factors that are involved in the regulation of many cellular processes. KLF2 deregulations have been also reported during the progression of many tumors. In the present review we discussed the molecular mechanisms of KLF2 during tumor growth and invasion. It has been shown that the KLF2 as a tumor suppressor is mainly inhibited by the non-coding RNAs (ncRNAs) through the polycomb repressive complex 2 (PRC2) recruitment. This review is an effective step towards introducing the KLF2 as a suitable diagnostic and therapeutic target in cancer patients.

Background

Despite significant advances in cancer treatment, it is still considered as one of the main causes of human deaths globally [1, 2]. Surgery, hormone therapy, chemo-radio therapy, and targeted therapy are among the routine cancer treatment options. In some circumstances, the treatment plan may include a variety of therapeutic methods to maximize the therapeutic efficiency. Radiotherapy and surgery are the most effective treatments for non-metastatic and localized tumors but are ineffective in metastatic cancers. As a result, metastasis is the leading cause of cancer death, accounting for more than 90% of all cancer mortalities [3]. Since, anticancer drugs can reach any part of the body through the bloodstream, they are considered as the common treatment options for metastatic tumors [4]. However, chemotherapeutic side effects and multidrug resistance highlight the need for novel and effective targeted therapies based on molecular tumor biology [5, 6]. Such targeted therapies disrupt particular oncogenes and signaling pathways to trigger apoptosis and immune system stimulation with a lower side effects compared with chemotherapeutic modalities [7]. Transcription factors are the main transcriptional regulators [8]. Accordingly, understanding the role of transcription factors, downstream targets, and upstream regulators in tumor cells can help us to develop novel therapeutic approaches to overcome drug resistance [9]. Krüppel-like factors (KLFs) are a family of developmental transcription factors that participate in the modulation of cell growth and differentiation [10, 11]. KLF2 belongs to the KLF protein family that contains Cys2/His2 zinc-finger domains to interact with GC boxes in promoter sequences and exert as transcriptional activators or suppressors [12]. KLF2 can be regulated by ubiquitination, non-coding RNAs, and signaling pathways [13,14,15,16,17]. The polycomb repressive complex 2 (PRC2) belongs to the Polycomb proteins complex that inhibits gene expression through histone modification. PRC2 consists of EZH2, EED, and SUZ12 components. EZH2 catalyzes the H3K27me3 that results in transcriptional inhibition [18]. It has been reported that PRC2 complex has a key role in regulation of KLF2 expression in tumor cells via histone methylation in promoter region [19, 20]. KLF2 functions as a tumor suppressor or oncogene in different tumors [21, 22]. Therefore, in the present review, we discussed the molecular mechanisms of the KLF2 during tumor progression to introduce that as a reliable diagnostic and therapetutic target in cancer patients (Tables 1 and 2) (Fig. 1).

Table 1 Oncogenic ncRNAs inhibit the KLF2 by PRC2 recruitment
Table 2 KLF2 functions as a tumor suppressor or oncogene by the transcriptional regulation of target genes
Fig. 1
figure 1

KLF2 is mainly targeted by the oncogenic ncRNAs during tumor progression. (Created with BioRender.com)

Gastric, esophageal, and oral cancers

SUZ12 has a pivotal role in promotion of tumor cell proliferation and metastasis. Up regulation of SUZ12 has been observed in different types of human cancers [23,24,25]. There was significant SUZ12 up regulation in gastric cancer (GC) tissues that was associated with distant metastasis, tumor size, stage, and lower survival. SUZ12 induced GC cell proliferation and metastasis by KLF2 and CDH1 down regulations [26]. LSD1 is one of the components of CoREST transcriptional co-suppressor complex by demethylation of H3K4m1/m2 [27, 28]. Several studies demonstrated that LSD1 plays critical roles in cell growth, differentiation, EMT, and invasion [29,30,31]. Down regulation of LSD1 reduced GC cell proliferation and invasion while promoted apoptosis. LSD1 had an oncogenic role via inhibition of KLF2 through H3K4 demethylation [32]. Long non-coding RNAs (LncRNAs) are involved in X chromosome inactivation, self-renewal, differentiation, and apoptosis [33,34,35]. Deregulation of lncRNAs was also associated with several cancers by the modulation of gene expression through chromatin remodeling, histone modification, and microRNAs sponging [36, 37]. They are also correlated with tumor cell metastasis and poor prognosis [38, 39]. There was significant DLEU1 up regulation in GC tissues that was linked with the poor prognosis. Downregulation of DLEU1 suppressed the proliferation of GC cells by provoking cell cycle arrest. DLEU1 directly interacted with LSD1 in promoter regions of KLF2, consequently promoting H3K4me2 modification [20]. Suppression of ZFAS1 reduced GC cell growth while promoted apoptosis. ZFAS1 recruited the EZH2 and LSD1 to NDK2 and KLF2 promoters that inhibited their transcription through H3K27me3 and demethylation of H3K4me2. ZFAS1 had a critical role in inhibition of tumor suppressors by EZH2 and LSD1 recruitments in GC cells [19]. There were LINC00202 up regulations in GC tissues and cells. Downregulation of LINC00202 significantly decreased the GC cell proliferation. The KLF2 expression level was affected by the high level of LINC00202 that recruited the EZH2. LINC00202 attenuated the GC progression by KLF2 inhibition [40]. There was significant LINC01296 up regulation in esophageal squamous cell carcinoma (ESCC) tissues compared with normal tissues that was correlated with lymph node metastasis, TNM stage, and poor prognosis. Silencing of LINC01296 decreased ESCC cell proliferation and invasion. LINC01296 down regulated the KLF2 via binding to EZH2 in ESCC cells [41]. There was AFAP1-AS1 up regulation in GC tissues. AFAP1-AS1 induced GC cell proliferation and invasion through KLF2 targeting [42].

Epithelial-to-mesenchymal transition (EMT) is known as a critical biological process in which epithelial cells are altered into mesenchymal cells during particular physiological and pathological contexts to acquire invasive properties [43, 44]. CXCR4 is activated via binding to SDF1, which is a critical oncogene [45]. It has been shown that CXCR4 was significantly correlated with EMT in lung cancer [46]. MiR-32-5p up regulated the CXCR4 through KLF2 targeting, which induced cell proliferation and EMT process in oral squamous cell carcinoma (OSCC) cells [47]. β-catenin is the critical modulator of the Wnt/β-catenin signaling pathway that is involved in tumor progression [48]. Wnt pathway also participates in GC progression via EMT modulation [49]. HOXA11-AS induced GC cell progression by β-catenin up regulation via WDR5 interaction, KLF2 down regulation, and EZH2 mediated P21 inhibition [50]. PI3K/AKT signaling as the main down stream cascade of the growth factor receptors has a key role in tumor progression [51, 52]. PTEN functions as a tumor suppressor by the inhibition of PI3K/AKT [53]. It has been reported that KLF2 was significantly down regulated in GC tissues compared to normal tissues that was associated with overall survival. KLF2 reduced cell migration while promoted apoptosis by the suppression of PTEN/AKT signaling in GC cells. It promoted PTEN expression and inhibited AKT-mTOR signaling. KLF2 also induced apoptosis by regulating p16/CDKN2A and p27/CDKN1B [54].

Colorectal cancer

CUL4A belongs to the cullin family of proteins that functions as an oncogene by regulation of cell proliferation, differentiation, and apoptosis [55,56,57]. There was LINC00460 up regulation in colorectal cancer (CRC) tissues that was positively associated with lymph node involvement, stage, and tumor size. Downregulation of LINC00460 reduced CRC cell proliferation while promoted apoptosis. LINC00460 functioned as an oncogene by engaging EZH2 and H3K27me3 to the KLF2 promoter, consequently the inactivation of KLF2. LINC00460 negatively regulated miR-149-5p to up regulate CUL4A. LINC00460 inhibition repressed CRC progression through either EZH2/KLF2 and miR-149-5p/CUL4A pathways [58]. There was L22NC03-N64E9.1 up regulation in CRC tissues that was correlated with CRC progression. L22NC03-N64E9.1 induced CRC cell proliferation through down regulation of KLF2 via interacting with EZH2 [59].

The p53 is a tumor suppressor that plays an important role in mediating cell cycle arrest, apoptosis, and genomic stability [60]. It inhibits tumor progression via mediating transcription of different downstream target genes which are participated in apoptosis and cell-cycle arrest [61]. Several studies found that simvastatin had therapeutic influence on several types of cancers by NF-kB, AKT, JNK, and CASP3/Bcl-2/cIAP mediated apoptosis [62,63,64,65]. Simvastatin remarkably up regulated KLF2 in p53-muted colon cancer cells. KLF2 was demonstrated to intervene in the anti-proliferative impact and anti-metastasis consequence of simvastatin on mutp53 colon cancer cells. Anti-proliferative effects of KLF2 were revealed by p21 up regulation in mutp53 cancer cells [66].

CDKN2B belongs to the cyclin-associated kinase inhibitors that may form a complex with CDK4 or CDK6 and inhibits the activation of the cyclin-dependent kinase to suppress cell cycle. There was significant SNHG1 up regulation in CRC tissues that was associated with poor prognosis. SNHG1 up regulated the CCND2 through the miR-154-5p sponging. SNHG1 interacted with EZH2 for a PRC2-associated down regulation of KLF2 and CDKN2B [67]. HOXA-AS2 induced CRC cell proliferation by promotion of cell proliferation while inhibition of apoptosis. HOXA-AS2 epigenetically suppressed the p21 and KLF2 transcription via interacting with EZH2 and LSD1 [68].

HIF-1α is an important modulator of hypoxic response in cancer cells [69]. Accumulating evidence revealed that hypoxia plays a critical role in tumor progression, angiogenesis, distant metastasis, and cancer therapy [70, 71]. It has been demonstrated that HIF-1α can interact with the Notch target gene to regulate its signaling in cancer stem cells [72]. Notch-1 induces tumorigenesis in CRC and preserves cells from apoptosis [73]. KLF2 repressed CRC cell growth through suppressing the HIF-1α/ Notch-1 axis [74]. Exosomal miR-25-3p intervened in the construction of a pre-metastatic niche in nude mice through promotion of the vascular permeability and subsequent CRC metastasis. MiR-25-3p targeted the KLF2 and KLF4 in HUVECs that resulted in ZO-1, occludin, and Claudin5 down regulations while VEGFR2 up regulation [75].

Lung cancer

KLF2 inhibits the leukemia cell growth by p21 up regulation [76], while down regulates the Wee1 to promote apoptosis [77]. WW domain-containing protein 1 (WWP1) induces the ubiquitination and degradation of KLF2 [78]. Smurf1 plays important roles in regulating cell polarity and tumor progression via mediating BMP-Smad, RhoA signaling pathways [79, 80]. WWP1 and Smurf1/2 have been also demonstrated to mediate Smads degradation in TGF-b signaling pathway [81]. Smurf1 as the HECT-type ubiquitin ligase might promote the KLF2 degradation in lung tumor cells [13]. There were significant KLF2 down regulations in non small-cell lung cancer (NSCLC) tissues that was correlated with tumor size, tumor stage, lymphatic metastasis, and survival. KLF2 repressed NSCLC cell growth via p21 and p15 targeting [82]. There was LINC01133 up regulation in NSCLC cells that was correlated with poor prognosis. LINC01133 had an oncogenic role in NSCLC cells through associating with EZH2 and LSD1, and KLF2, P21, and CDH1 down regulations. LINC01133 promoted EMT via CDH1 down regulation in NSCLC cells [83]. It has been indicated that KLF2 was notably downregulated in NSCLC tissue samples that was correlated with NSCLC lymph node metastasis and advanced TNM stage. KLF2 remarkably inhibited tumor cell viability while induced apoptosis through the expression of p15 and p21 in NSCLC cells [84]. There was miR-572 up regulation in NSCLC samples that was significantly correlated with metastasis and prognosis. MiR-572 promoted NSCLC cell proliferation and migration via KLF2 targeting [85].

EZH2 is a catalytic subunit of PRC2 that has a histone methyltransferase function to mediate the H3K27me3 tails of different target genes [86]. LSD1 is a histone demethylase as the core subunit of the REST suppressor which particularly demethylases H3K4me1/2 [87]. LncRNAs can modulate the transcription of target genes by interacting with PRC2 [88]. LATS2 belongs to the LATS family of protein kinases that is involved in spindle construction and genome integrity [89, 90]. It has been documented that there was AGAP2-AS1 up regulation in NSCLC tissues that was correlated with poor prognosis. AGAP2-AS1 acted as an oncogene in NSCLC cells through the LATS2 and KLF2 down regulations. AGAP2-AS1 recruited the EZH2 and LSD1 to down regulate the LATS2 and KLF2 in NSCLC cells [91]. LINC00511 induced the NSCLC progression via LATS2 and KLF2 down regulations followning the recruitment of EZH2 and LSD1 to their promoter sequences, respectively [92]. XIST mediates cell proliferation and invasion via epigenetically inhibiting KLF2 in NSCLC cells. There was XIST up regulation in NSCLC tissues that was associated with poor prognosis and poor overall survival. KLF2 acts as tumor suppressor in NSCLC cells and its’ expression could be repressed through XIST via recruiting EZH2 to its promoter region [93]. MiR-126-5p down regulation was found in lung adenocarcinoma tissues that was correlated with poor prognosis. MiR-126-5p suppressed EZH2 to increase the expression level of KLF2 and decreased BIRC5 expression, that inhibited lung tumor cell proliferation, migration while increased radiosensitivity and apoptosis [94].

The ATP production by glycolysis is lower than oxidative phosphorylation that can be substituted by the higher glucose absorption in tumor cells. Glycolysis also supplies many nutrients to maintain the tumor cell proliferation [95, 96]. Tumor cells have a high level of the glutamine consumption to prepare their required energy for the cell proliferation and growth [97, 98]. It was shown that KLF2 significantly reduced the NSCLC cell proliferation through the reduced glutamine consumption following the glutamine transaminase down regulation [99].

Hepatocellular cancer

Hedgehog (Hh) signaling pathway participates in the promotion of tumor cell growth and metastasis [100]. Sonic (Shh), Desert (Dhh), and Indian (Ihh) encode secretory proteins that act as Hh ligands [101]. The secreted Hh ligand associates with Hip1, Patched 2 (Ptch2), and Ptch1 as transmembrane receptors through dissemination [102]. The Ptch receptor inhibits the effect of Smo in ligand loss. Activation of Smo may induce Gli1 transcription factor [103]. There was KLF2 down regulation in hepatocellular cancer (HCC) tissue that suppressed the cell growth and metastasis by repressing the Hedgehog/Gli1 signaling cascade. KLF2 competed with Gli1 to interact with HDAC1 to inhibit the Hedgehog signal [104].

TGF-β belongs to the TGF-β cytokine family that contains activin, nodal, and bone morphogenetic proteins [105]. TGF-β signaling is activated by TGF-β ligand that promotes Smad2/3 through phosphorylation. Then smad2/3/4 oligomeric complex enters into the nucleus to regulate the TGF-β target genes [106,107,108]. KLF2 has been found to reduce TGFβ/Smad signaling in endothelial cells through Smad7 up regulation [109, 110]. TGF-β promoted the expression of KLF2 in numerous HCC cells. KLF2 suppressed the TGF-β/Smad pathway by provoking the transcriptional activity of Smad3 and Smad4 [14].

FBXL19-AS1 promoted the HCC cell proliferation while inhibited apoptosis via KLF2 down regulation [111]. There was ANRIL up regulation in HCC tissues that was associated with tumor size and stage. It may modulate cell growth by epigenetic inhibition of KLF2 via interacting with PRC2. The expression of ANRIL could also be regulated by SP1. SP1-mediated ANRIL expression modulated the KLF2 expression. ANRIL suppressed KLF2 transcription through cooperating with EZH2 and SUZ12 in HCC cells and recruitment of PRC2 to the KLF2 promoter [112]. DUXAP8 was considerably up regulated in HCC that was correlated with poor prognosis. DUXAP8 induced HCC cell growth by KLF2 down regulation [113]. There was TUG1 up regulation in HCC tissues that was associated with tumor size and BCLC stage. The high expression level of TUG1 was promoted through SP1 and mediated HCC cell growth by epigenetically inhibiting KLF2 via interacting with PRC2 [15]. KLF2 was significantly up regulated in HCC tissues compared to surrounding normal liver tissues. KLF2 induced HCC cell proliferation through c-MYC targeting [114].

Pancreatic cancer

CDK protein family has critical roles in cell cycle and gene expression regulation through interacting with transcription factors to mediate RNA polymerase II activity [115,116,117]. CDK8 as a part of the mediator complex, which contains cyclin C, MED12, and MED13, modulates transcription [118,119,120]. CDK8 induces the β-catenin expression in pancreatic cancer that enters into the nucleus to mediate the activation of angiogenesis-promoting transcription factors [121,122,123]. KLF2 is a critical downstream target of β-catenin that is involved in transcriptional regulation of several target genes [16, 17]. KLF2 was also known as a critical transcriptional modulator of endothelial inflammation, that could suppress VEGF-associated angiogenesis and tissue edema, and its upregulation may elevate the expression level of Semaphorin-3 F (SEMA3F) [124, 125]. The high expression level of CDK8 in pancreatic cancer was considerably associated with poor prognosis. KLF2 inhibited cancer cell angiogenesis. CDK8 was critical for tumor vessel progression in pancreatic carcinoma through the β-catenin-KLF2 axis [126]. KLF2 inhibited the growth and metastasis of pancreatic cancer cells by interaction with b-catenin that suppressed the activity of b-catenin/TCF complex [127].

P15 is a CDK inhibitor that functions through inhibition of CDK activation by CCND resulting in cell cycle G1 arrest. There was significant IRAIN up regulation in pancreatic cancer tissues that was associated with larger tumor sizes, higher TNM stages, and lymph node metastasis. IRAIN induced cell proliferation directly through interacting with EZH2 and LSD1 complexes and suppressing KLF2 and P15 in pancreatic cancer cells [128]. SNHG15 induced pancreatic cancer cell proliferation by repressing P15 and KLF2 expression via EZH2-related H3K27me3 [129]. There was significant DUXAP8 up regulation in pancreatic cancer tissues that was correlated with the larger size of the tumor, advanced clinical stage, and shorter survival rate. DUXAP8 promoted the cell proliferation and tumor progression in pancreatic cancer through p21 and KLF2 down regulations following interaction with EZH2 and LSD1 [130]. KLF2 also induced pancreatic tumor cell senescence through cooperating with FOXO4 and promoting the expression of p21 [131].

Breast, ovarian, and prostate cancers

Wee1 is a tyrosine kinase that regulates cell cycle progression. M-phasepromoting factor (MPF), which is a member of the CDC2 and cyclin B complex, could mediate the G2/M transition through cell cycle. MPF is required for mitosis and is also necessary for DNA damage-associated apoptosis. Wee1 negatively modulates the MPF complex via CDC2 phosphorylation that leads to mitosis deregulation and resistance to apoptosis [132,133,134]. KLF2 recruited the SP1/CPBP to down regulate the WEE1 that sensitized ovarian tumor cells toward the DNA damage mediated apoptosis [77]. It has been suggested that KLF2 can be introduced as a promising target to increase the sensitivity of breast cancer (BCa) to cisplatin via down regulation of WEE1 [135]. KLF2 promoted cell apoptosis while inhibited cell proliferation through p16, p21, and p27 up regulations and CCND1 and survivin down regulations in breast tumor cells [136]. Silencing of LINC00702 decreased the ovarian cancer (OC) cell proliferation. It facilitated the progression of OC through binding to EZH2 to suppress the transcription of KLF2 [137]. SNHG7 inhibition reduced the OC cell growth and invasive. SP1 up regulated the SNHG7 that interacted with EZH2 to inhibit KLF2 expression in ovarian tumor cells [138].

Matrix metalloproteinases (MMPs) are a multifunctional family of zinc-dependent endopeptidases that has a critical role in the degradation of the extracellular matrix (ECM). These extracellular molecules are secreted via cells that supply structural and biochemical associates with normal physiological cells. MMP2 has a critical role in tumor cell migration due to the degradation of collagens [139,140,141]. KLF2 was considerably down regulated in prostate cancer (PCa) tissues in comparison with normal margins. It suppressed the prostate tumor cell invasion through MMP2 inhibition [142]. KLF2 inhibits CRC cell proliferation by promoting HIF-1α/Notch-1 signal pathway [74]. There was GHET1 up regulation in PCa tissues that was negatively linked to KLF2 expression. GHET1 promoted the PCa progression through decreasing the level of KLF2 expression. Accumulating evidences demonstrated that HIF-1α and Notch-1 were also considerably down regulated through GHET1 inhibition in prostate tumor cells [143]. LINC00665 was significantly up regulated in PCa tissues and cell lines. LINC00665 down regulation reduced the PCa cell proliferation and migration. It facilitated the malignant progression of PCa by epigenetically suppressing the expression level of KLF2 via interaction with EZH2 and LSD1 [144].

Myeloma and osteosarcoma

Histone methylation is one of the main regulators of chromatin remodeling and gene expression that is required for several biological activities such as cell proliferation, DNA damage, and stress response [145, 146]. KDM3A belongs to the Jumonji histone demethylases family that acts as a coactivator for the androgen receptor and mediates the elimination of H3K9me1 and H3K9me2 [147]. It has a critical role as a modulator of spermatogenesis, self-renewal, metabolic gene expression, and sex resolvation [147,148,149,150]. IRF4 belongs to the interferon regulatory family of transcription factors that has a crucial role in mediating the plasma cell differentiation [151,152,153]. Silencing of KDM3A induced apoptosis in myeloma cells through KLF2 and IRF4 up regulations by H3K9 elimination. Down regulation of KLF2 promoted apoptosis and that KLF2 positively regulated the IRF4 promoter [154].

The epidermal growth factor-like protein-7 (EGFL7) stimulates endothelial cell survival, migration, and differentiation [155, 156]. Deregulation of EGFL7 has been frequently observed in multiple types of solid tumors and acute myeloid leukemia [157, 158]. Multiple myeloma (MM) cells can evade drug treatment via integrin-mediated cellular adhesion. ITGB3 promotes MM cell proliferation, protease secretion, and invasion [159,160,161]. EGFL7 is involved in angiogenesis by interaction with ITGB3 and Notch receptors [162]. EGFL7 induced MM growth through ITGB3 and KLF2 up regulations [163].

KLF2 functions as a tumor suppressor by PCNA and CCND1 down regulations while p21 up regulation [164]. It has been documented that SNHG6 was up regulated in osteosarcoma tissues that was correlated with tumor grade and shorter overall survival. Downregulation of SNHG6 repressed cell proliferation while increased apoptosis. There was a negative association between p21, KLF2, and SNHG6 and a positive association between CCND1 and SNHG6. SNHG6 facilitated the osteosarcoma cell proliferation via p21 and KLF2 modulations [165].

Conclusions

It has been reported that the KLF2 has mainly a tumor suppressor function that can be suppressed by the oncogenic ncRNAs following the PRC2 recruitment. On the other hand, ncRNAs can promote the tumor cell growth and proliferation by PRC2 mediated KLF2 targeting. Therefore, KLF2 and its ncRNA regulators can be introduced as appropriate therapeutic and diagnostic targets in cancer patients. Considering that the PRC2 complex as an KLF2 inhibitor has an oncogenic role, PRC2 inhibitors can indirectly inhibit the tumor growth and progression by KLF2 activation. On the other hand, it has been shown that lncRNAs promote PRC2-mediated KLF2 down regulation in tumor cells. Therefore, inhibition of lncRNAs/PRC2 axis can up regulate the KLF2 to reduce tumor progression. Besides the therapeutic importance, lncRNAs/PRC2/KLF2 axis can also be used as a diagnostic/prognostic marker in cancer patients. However, due to the pivotal role of KLF2 and PRC2 in normal cellular processes, targeted therapy against the PRC2/KLF2 axis results in side effects in normal cells and tissues. Therefore, it is required to use the novel methods to deliver the inhibitors of PRC2/KLF2 axis locally and specifically to the tumor tissue in order to reduce the side effects as much as possible. Indeed, further animal studies and clinical trials are needed to be able to use the lncRNAs/PRC2/KLF2 axis for diagnostic and therapeutic purposes in cancer patients.

Data Availability

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Abbreviations

BCa:

Breast cancer

CRC:

Colorectal cancer

Dhh:

Desert

EGFL7:

Epidermal growth factor-like protein-7

EMT:

Epithelial-to-mesenchymal transition

ESCC:

Esophageal squamous cell carcinoma

ECM:

Extracellular matrix

GC:

Gastric cancer

Hh:

Hedgehog

HCC:

Hepatocellular cancer

Ihh:

Indian

KLF2:

Krüppel-like factors 2

LncRNAs:

Long non-coding RNAs

MMPs:

Matrix metalloproteinases

MPF:

M-phasepromoting factor

MM:

Multiple myeloma

ncRNAs:

Non-coding RNAs

NSCLC:

Non small-cell lung cancer

OSCC:

Oral squamous cell carcinoma

OC:

Ovarian cancer

Ptch2:

Patched 2

PRC2:

Polycomb repressive complex 2

PCa:

Prostate cancer

SEMA3F:

Semaphorin-3 F

Shh:

Sonic

WWP1:

WW domain-containing protein 1

References

  1. Wei G, Wang Y, Yang G, Wang Y, Ju R. Recent progress in nanomedicine for enhanced cancer chemotherapy. Theranostics. 2021;11(13):6370–92.

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Siegel RL, Miller KD, Fuchs HE, Jemal A, Cancer Statistics. 2021. CA Cancer J Clin. 2021;71(1):7–33.

  3. Guan X. Cancer metastases: challenges and opportunities. Acta Pharm Sin B. 2015;5(5):402–18.

    PubMed  PubMed Central  Google Scholar 

  4. Chabner BA, Roberts TG Jr, Timeline. Chemotherapy and the war on cancer. Nat Rev Cancer. 2005;5(1):65–72.

    CAS  PubMed  Google Scholar 

  5. Moghbeli M. MicroRNAs as the critical regulators of cisplatin resistance in ovarian cancer cells. J Ovarian Res. 2021;14(1):127.

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Zangouei AS, Alimardani M, Moghbeli M. MicroRNAs as the critical regulators of doxorubicin resistance in breast tumor cells. Cancer Cell Int. 2021;21(1):213.

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Perez-Herrero E, Fernandez-Medarde A. Advanced targeted therapies in cancer: drug nanocarriers, the future of chemotherapy. Eur J Pharm Biopharm. 2015;93:52–79.

    CAS  PubMed  Google Scholar 

  8. Maston GA, Evans SK, Green MR. Transcriptional regulatory elements in the human genome. Annu Rev Genomics Hum Genet. 2006;7:29–59.

    CAS  PubMed  Google Scholar 

  9. Yao S, Fan LY, Lam EW. The FOXO3-FOXM1 axis: a key cancer drug target and a modulator of cancer drug resistance. Semin Cancer Biol. 2018;50:77–89.

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Preiss A, Rosenberg UB, Kienlin A, Seifert E, Jäckle H. Molecular genetics of Krüppel, a gene required for segmentation of the Drosophila embryo. Nature. 1985;313(5997):27–32.

    CAS  PubMed  Google Scholar 

  11. Miller I, Bieker JJ. A novel, erythroid cell-specific murine transcription factor that binds to the CACCC element and is related to the Krüppel family of nuclear proteins. Mol Cell Biol. 1993;13(5):2776–86.

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Bialkowska AB, Yang VW, Mallipattu SK. Krüppel-like factors in mammalian stem cells and development. Development. 2017;144(5):737–54.

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Xie P, Tang Y, Shen S, Wang Y, Xing G, Yin Y, et al. Smurf1 ubiquitin ligase targets Kruppel-like factor KLF2 for ubiquitination and degradation in human lung cancer H1299 cells. Biochem Biophys Res Commun. 2011;407(1):254–9.

    CAS  PubMed  Google Scholar 

  14. Li Y, Tu S, Zeng Y, Zhang C, Deng T, Luo W, et al. KLF2 inhibits TGF-β-mediated cancer cell motility in hepatocellular carcinoma. Acta Biochim Biophys Sin. 2020;52(5):485–94.

    CAS  PubMed  Google Scholar 

  15. Huang M-D, Chen W-M, Qi F-Z, Sun M, Xu T-P, Ma P, et al. Long non-coding RNA TUG1 is up-regulated in hepatocellular carcinoma and promotes cell growth and apoptosis by epigenetically silencing of KLF2. Mol Cancer. 2015;14(1):1–12.

    Google Scholar 

  16. Dejana E. The role of wnt signaling in physiological and pathological angiogenesis. Circul Res. 2010;107(8):943–52.

    CAS  Google Scholar 

  17. Phng L-K, Potente M, Leslie JD, Babbage J, Nyqvist D, Lobov I, et al. Nrarp coordinates endothelial notch and wnt signaling to control vessel density in angiogenesis. Dev Cell. 2009;16(1):70–82.

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Margueron R, Reinberg D. The polycomb complex PRC2 and its mark in life. Nature. 2011;469(7330):343–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Nie F, Yu X, Huang M, Wang Y, Xie M, Ma H, et al. Long noncoding RNA ZFAS1 promotes gastric cancer cells proliferation by epigenetically repressing KLF2 and NKD2 expression. Oncotarget. 2017;8(24):38227.

    PubMed  Google Scholar 

  20. Li X, Li Z, Liu Z, Xiao J, Yu S, Song Y, Correction. Long non-coding RNA DLEU1 predicts poor prognosis of gastric cancer and contributes to cell proliferation by epigenetically suppressing KLF2. Cancer Gene Ther. 2022;29(6):873.

    CAS  PubMed  Google Scholar 

  21. Zhang W, Levi L, Banerjee P, Jain M, Noy N. Kruppel-like factor 2 suppresses mammary carcinoma growth by regulating retinoic acid signaling. Oncotarget. 2015;6(34):35830.

    PubMed  PubMed Central  Google Scholar 

  22. Zou K, Lu X, Ye K, Wang C, You T, Chen J. Kruppel-like factor 2 promotes cell proliferation in hepatocellular carcinoma through up-regulation of c-myc. Cancer Biol Ther. 2016;17(1):20–6.

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Li H, Cai Q, Wu H, Vathipadiekal V, Dobbin ZC, Li T, et al. SUZ12 promotes human epithelial ovarian Cancer by suppressing apoptosis via silencing HRKSUZ12 promotes EOC via silencing HRK. Mol Cancer Res. 2012;10(11):1462–72.

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Martín-Pérez D, Sánchez E, Maestre L, Suela J, Vargiu P, Di Lisio L, et al. Deregulated expression of the polycomb-group protein SUZ12 target genes characterizes mantle cell lymphoma. Am J Pathol. 2010;177(2):930–42.

    PubMed  PubMed Central  Google Scholar 

  25. Iliopoulos D, Lindahl-Allen M, Polytarchou C, Hirsch HA, Tsichlis PN, Struhl K. Loss of miR-200 inhibition of Suz12 leads to polycomb-mediated repression required for the formation and maintenance of cancer stem cells. Mol Cell. 2010;39(5):761–72.

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Xia R, Jin F-y, Lu K, Wan L, Xie M, Xu T-p, et al. SUZ12 promotes gastric cancer cell proliferation and metastasis by regulating KLF2 and E-cadherin. Tumor Biology. 2015;36(7):5341–51.

    CAS  PubMed  Google Scholar 

  27. Lee MG, Wynder C, Cooch N, Shiekhattar R. An essential role for CoREST in nucleosomal histone 3 lysine 4 demethylation. Nature. 2005;437(7057):432–5.

    CAS  PubMed  Google Scholar 

  28. Shi Y-J, Matson C, Lan F, Iwase S, Baba T, Shi Y. Regulation of LSD1 histone demethylase activity by its associated factors. Mol Cell. 2005;19(6):857–64.

    CAS  PubMed  Google Scholar 

  29. Adamo A, Sesé B, Boue S, Castaño J, Paramonov I, Barrero MJ, et al. LSD1 regulates the balance between self-renewal and differentiation in human embryonic stem cells. Nat Cell Biol. 2011;13(6):652–9.

    CAS  PubMed  Google Scholar 

  30. McDonald OG, Wu H, Timp W, Feinberg AP. Genome-scale epigenetic reprogramming during epithelial-to-mesenchymal transition. Nat Struct Mol Biol. 2011;18(8):867–74.

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Lv T, Yuan D, Miao X, Lv Y, Zhan P, Shen X, et al. Over-expression of LSD1 promotes proliferation, migration and invasion in non-small cell lung cancer. PLoS ONE. 2012;7(4):e35065.

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Fang R, Xu J, Lin H, Xu X, Tian F. The histone demethylase lysine-specific demethylase-1–mediated epigenetic silence of KLF2 contributes to gastric cancer cell proliferation, migration, and invasion. Tumor Biology. 2017;39(4):1010428317698356.

    PubMed  Google Scholar 

  33. Fatica A, Bozzoni I. Long non-coding RNAs: new players in cell differentiation and development. Nat Rev Genet. 2014;15(1):7–21.

    CAS  PubMed  Google Scholar 

  34. Payer B, Lee JT. Coupling of X-chromosome reactivation with the pluripotent stem cell state. RNA Biol. 2014;11(7):798–807.

    PubMed  PubMed Central  Google Scholar 

  35. Flynn RA, Chang HY. Long noncoding RNAs in cell-fate programming and reprogramming. Cell Stem Cell. 2014;14(6):752–61.

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Fatima R, Akhade VS, Pal D, Rao SM. Long noncoding RNAs in development and cancer: potential biomarkers and therapeutic targets. Mol Cell Ther. 2015;3(1):1–19.

    CAS  Google Scholar 

  37. Khalili-Tanha G, Moghbeli M. Long non-coding RNAs as the critical regulators of doxorubicin resistance in tumor cells. Cell Mol Biol Lett. 2021;26(1):39.

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Zhang F, Zhang L, Zhang C. Long noncoding RNAs and tumorigenesis: genetic associations, molecular mechanisms, and therapeutic strategies. Tumor Biology. 2016;37(1):163–75.

    PubMed  Google Scholar 

  39. Rahmani Z, Mojarrad M, Moghbeli M. Long non-coding RNAs as the critical factors during tumor progressions among iranian population: an overview. Cell Biosci. 2020;10:6.

    PubMed  PubMed Central  Google Scholar 

  40. Xu Q, Qiao S, Liu L, Xu J, Yang L, Chen X, et al. LINC00202 attenuates the progression of gastric cancer via suppressing expression level of KLF2. J BU ON: Official J Balkan Union Oncol. 2021;26(2):506–12.

    Google Scholar 

  41. Wang L, Meng D, Wang Y, Hu J. Long non-coding RNA LINC01296 promotes esophageal squamous cell carcinoma cell proliferation and invasion by epigenetic suppression of KLF2. Am J Cancer Res. 2018;8(10):2020.

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Yuan X, Li J, Cao Y, Jie Z, Zeng Y. Long non-coding RNA AFAP1-AS1 promotes proliferation and migration of gastric cancer by downregulating KLF2. Eur Rev Med Pharmacol Sci. 2020;24:673–80.

    PubMed  Google Scholar 

  43. Forghanifard MM, Rad A, Farshchian M, Khaleghizadeh M, Gholamin M, Moghbeli M, et al. TWIST1 upregulates the MAGEA4 oncogene. Mol Carcinog. 2017;56(3):877–85.

    CAS  PubMed  Google Scholar 

  44. Hamidi AA, Khalili-Tanha G, Nasrpour Navaei Z, Moghbeli M. Long non-coding RNAs as the critical regulators of epithelial mesenchymal transition in colorectal tumor cells: an overview. Cancer Cell Int. 2022;22(1):71.

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Zlotnik A. New insights on the role of CXCR4 in cancer metastasis. J Pathology: J Pathological Soc Great Br Irel. 2008;215(3):211–3.

    CAS  Google Scholar 

  46. Yin H, Wang Y, Chen W, Zhong S, Liu Z, Zhao J. Drug-resistant CXCR4-positive cells have the molecular characteristics of EMT in NSCLC. Gene. 2016;594(1):23–9.

    CAS  PubMed  Google Scholar 

  47. Qin SY, Li B, Chen M, Qin MQ, Liu JM, Lv QL. MiR-32‐5p promoted epithelial‐to‐mesenchymal transition of oral squamous cell carcinoma cells via regulating the KLF2/CXCR4 pathway. Kaohsiung J Med Sci. 2022;38(2):120–8.

    CAS  PubMed  Google Scholar 

  48. Yao H, Ashihara E, Maekawa T. Targeting the Wnt/β-catenin signaling pathway in human cancers. Expert Opin Ther Targets. 2011;15(7):873–87.

    CAS  PubMed  Google Scholar 

  49. Huang L, Wu R-L, Xu A-M. Epithelial-mesenchymal transition in gastric cancer. Am J Translational Res. 2015;7(11):2141.

    CAS  Google Scholar 

  50. Liu Z, Chen Z, Fan R, Jiang B, Chen X, Chen Q, et al. Over-expressed long noncoding RNA HOXA11-AS promotes cell cycle progression and metastasis in gastric cancer. Mol Cancer. 2017;16(1):1–9.

    Google Scholar 

  51. Zangouei AS, Barjasteh AH, Rahimi HR, Mojarrad M, Moghbeli M. Role of tyrosine kinases in bladder cancer progression: an overview. Cell Commun Signal. 2020;18(1):127.

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Moghbeli M, Makhdoumi Y, Soltani Delgosha M, Aarabi A, Dadkhah E, Memar B, et al. ErbB1 and ErbB3 co-over expression as a prognostic factor in gastric cancer. Biol Res. 2019;52(1):2.

    PubMed  PubMed Central  Google Scholar 

  53. Navaei ZN, Khalili-Tanha G, Zangouei AS, Abbaszadegan MR, Moghbeli M. PI3K/AKT signaling pathway as a critical regulator of cisplatin response in tumor cells. Oncol Res. 2021;29(4):235–50.

    PubMed  Google Scholar 

  54. Wang C, Li L, Duan Q, Wang Q, Chen J. Krüppel-like factor 2 suppresses human gastric tumorigenesis through inhibiting PTEN/AKT signaling. Oncotarget. 2017;8(59):100358.

    PubMed  PubMed Central  Google Scholar 

  55. Wang Y, Wen M, Kwon Y, Xu Y, Liu Y, Zhang P, et al. CUL4A induces epithelial–mesenchymal transition and promotes cancer metastasis by regulating ZEB1 expression. Cancer Res. 2014;74(2):520–31.

    CAS  PubMed  Google Scholar 

  56. Ren W, Sun Z, Zeng Q, Han S, Zhang Q, Jiang L. Aberrant expression of CUL4A is associated with IL-6/STAT3 activation in colorectal cancer progression. Arch Med Res. 2016;47(3):214–22.

    CAS  PubMed  Google Scholar 

  57. Sharma P, Nag A. CUL4A ubiquitin ligase: a promising drug target for cancer and other human diseases. Open Biology. 2014;4(2):130217.

    PubMed  PubMed Central  Google Scholar 

  58. Lian Y, Yan C, Xu H, Yang J, Yu Y, Zhou J, et al. A novel lncRNA, LINC00460, affects cell proliferation and apoptosis by regulating KLF2 and CUL4A expression in colorectal cancer. Mol Therapy-Nucleic Acids. 2018;12:684–97.

    Google Scholar 

  59. Lian Y, Yan C, Ding J, Xia R, Ma Z, Hui B, et al. A novel lncRNA, LL22NC03-N64E9. 1, represses KLF2 transcription through binding with EZH2 in colorectal cancer. Oncotarget. 2017;8(35):59435.

    PubMed  PubMed Central  Google Scholar 

  60. Joerger AC, Fersht AR. The p53 pathway: origins, inactivation in cancer, and emerging therapeutic approaches. Annu Rev Biochem. 2016;85:375–404.

    CAS  PubMed  Google Scholar 

  61. Labuschagne CF, Zani F, Vousden KH. Control of metabolism by p53–cancer and beyond. Biochim et Biophys Acta (BBA)-Reviews Cancer. 2018;1870(1):32–42.

    CAS  Google Scholar 

  62. Ghosh-Choudhury N, Mandal CC, Ghosh-Choudhury N, Choudhury GG. Simvastatin induces derepression of PTEN expression via NFκB to inhibit breast cancer cell growth. Cell Signal. 2010;22(5):749–58.

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Fang Z, Tang Y, Fang J, Zhou Z, Xing Z, Guo Z, et al. Simvastatin inhibits renal cancer cell growth and metastasis via AKT/mTOR, ERK and JAK2/STAT3 pathway. PLoS ONE. 2013;8(5):e62823.

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Cho SJ, Kim JS, Kim JM, Lee JY, Jung HC, Song IS. Simvastatin induces apoptosis in human colon cancer cells and in tumor xenografts, and attenuates colitis-associated colon cancer in mice. Int J Cancer. 2008;123(4):951–7.

    CAS  PubMed  Google Scholar 

  65. Gopalan A, Yu W, Sanders BG, Kline K. Simvastatin inhibition of mevalonate pathway induces apoptosis in human breast cancer cells via activation of JNK/CHOP/DR5 signaling pathway. Cancer Lett. 2013;329(1):9–16.

    CAS  PubMed  Google Scholar 

  66. Lu L, Huang W, Hu W, Jiang L, Li Y, Wu X, et al. Kruppel-like factor 2 mediated anti-proliferative and anti-metastasis effects of simvastatin in p53 mutant colon cancer. Biochem Biophys Res Commun. 2019;511(4):772–9.

    CAS  PubMed  Google Scholar 

  67. Xu M, Chen X, Lin K, Zeng K, Liu X, Pan B, et al. The long noncoding RNA SNHG1 regulates colorectal cancer cell growth through interactions with EZH2 and miR-154-5p. Mol Cancer. 2018;17(1):1–16.

    Google Scholar 

  68. Ding J, Xie M, Lian Y, Zhu Y, Peng P, Wang J, et al. Long noncoding RNA HOXA-AS2 represses P21 and KLF2 expression transcription by binding with EZH2, LSD1 in colorectal cancer. Oncogenesis. 2017;6(1):e288–e.

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Semenza GL. Targeting HIF-1 for cancer therapy. Nat Rev Cancer. 2003;3(10):721–32.

    CAS  PubMed  Google Scholar 

  70. Shen G, Li X, Jia Y-f, Piazza GA, Xi Y. Hypoxia-regulated microRNAs in human cancer. Acta Pharmacol Sin. 2013;34(3):336–41.

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Tian Q, Xue Y, Zheng W, Sun R, Ji W, Wang X, et al. Overexpression of hypoxia-inducible factor 1α induces migration and invasion through notch signaling. Int J Oncol. 2015;47(2):728–38.

    CAS  PubMed  Google Scholar 

  72. Wang Y, Liu Y, Malek SN, Zheng P, Liu Y. Targeting HIF1α eliminates cancer stem cells in hematological malignancies. Cell Stem Cell. 2011;8(4):399–411.

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Meng RD, Shelton CC, Li Y-M, Qin L-X, Notterman D, Paty PB, et al. γ-Secretase inhibitors abrogate oxaliplatin-induced activation of the Notch-1 signaling pathway in colon cancer cells resulting in enhanced chemosensitivity. Cancer Res. 2009;69(2):573–82.

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Wang H-G, Cao B, Zhang L-X, Song N, Li H, Zhao W-Z, et al. KLF2 inhibits cell growth via regulating HIF-1α/Notch-1 signal pathway in human colorectal cancer HCT116 cells. Oncol Rep. 2017;38(1):584–90.

    CAS  PubMed  Google Scholar 

  75. Zeng Z, Li Y, Pan Y, Lan X, Song F, Sun J, et al. Cancer-derived exosomal mir-25-3p promotes pre-metastatic niche formation by inducing vascular permeability and angiogenesis. Nat Commun. 2018;9(1):1–14.

    Google Scholar 

  76. Wu J, Lingrel JB. KLF2 inhibits Jurkat T leukemia cell growth via upregulation of cyclin-dependent kinase inhibitor p21WAF1/CIP1. Oncogene. 2004;23(49):8088–96.

    CAS  PubMed  Google Scholar 

  77. Wang F, Zhu Y, Huang Y, McAvoy S, Johnson WB, Cheung TH, et al. Transcriptional repression of WEE1 by Kruppel-like factor 2 is involved in DNA damage-induced apoptosis. Oncogene. 2005;24(24):3875–85.

    CAS  PubMed  Google Scholar 

  78. Zhang X, Srinivasan SV, Lingrel JB. WWP1-dependent ubiquitination and degradation of the lung Krüppel-like factor, KLF2. Biochem Biophys Res Commun. 2004;316(1):139–48.

    CAS  PubMed  Google Scholar 

  79. Zhu H, Kavsak P, Abdollah S, Wrana JL, Thomsen GH. A SMAD ubiquitin ligase targets the BMP pathway and affects embryonic pattern formation. Nature. 1999;400(6745):687–93.

    CAS  PubMed  Google Scholar 

  80. Wang H-R, Zhang Y, Ozdamar B, Ogunjimi AA, Alexandrova E, Thomsen GH, et al. Regulation of cell polarity and protrusion formation by targeting RhoA for degradation. Science. 2003;302(5651):1775–9.

    CAS  PubMed  Google Scholar 

  81. Morén A, Imamura T, Miyazono K, Heldin C-H, Moustakas A. Degradation of the tumor suppressor Smad4 by WW and HECT domain ubiquitin ligases. J Biol Chem. 2005;280(23):22115–23.

    PubMed  Google Scholar 

  82. Yin L, Wang J-p, Xu T-p, Chen W-m, Huang M-d, Xia R, et al. Downregulation of Kruppel-like factor 2 is associated with poor prognosis for nonsmall-cell lung cancer. Tumor Biology. 2015;36(4):3075–84.

    CAS  PubMed  Google Scholar 

  83. Zang C, Nie F-q, Wang Q, Sun M, Li W, He J, et al. Long non-coding RNA LINC01133 represses KLF2, P21 and E-cadherin transcription through binding with EZH2, LSD1 in non small cell lung cancer. Oncotarget. 2016;7(10):11696.

    PubMed  PubMed Central  Google Scholar 

  84. Jiang W, Xu X, Deng S, Luo J, Xu H, Wang C, et al. Methylation of kruppel-like factor 2 (KLF2) associates with its expression and non-small cell lung cancer progression. Am J Translational Res. 2017;9(4):2024.

    CAS  Google Scholar 

  85. Sun B, Zhao J, Shao Z. MiR-572 promotes the development of non-small cell lung cancer by targeting KLF2. Eur Rev Med Pharmacol Sci. 2022;26(9):3083–90.

    CAS  PubMed  Google Scholar 

  86. Völkel P, Dupret B, Le Bourhis X, Angrand P-O. Diverse involvement of EZH2 in cancer epigenetics. Am J Translational Res. 2015;7(2):175.

    Google Scholar 

  87. Shin J, Ming G-l, Song H. Molecular toggle switch of histone demethylase LSD1. Mol Cell. 2015;57(6):949–50.

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Sun C-C, Li S-J, Li G, Hua R-X, Zhou X-H, Li D-J. Long intergenic noncoding RNA 00511 acts as an oncogene in non–small-cell lung cancer by binding to EZH2 and suppressing p57. Mol Therapy-Nucleic Acids. 2016;5:e385.

    CAS  Google Scholar 

  89. Abe Y, Ohsugi M, Haraguchi K, Fujimoto J, Yamamoto T. LATS2–Ajuba complex regulates γ-tubulin recruitment to centrosomes and spindle organization during mitosis. FEBS Lett. 2006;580(3):782–8.

    CAS  PubMed  Google Scholar 

  90. McPherson JP, Tamblyn L, Elia A, Migon E, Shehabeldin A, Matysiak-Zablocki E, et al. Lats2/Kpm is required for embryonic development, proliferation control and genomic integrity. EMBO J. 2004;23(18):3677–88.

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Li W, Sun M, Zang C, Ma P, He J, Zhang M, et al. Upregulated long non-coding RNA AGAP2-AS1 represses LATS2 and KLF2 expression through interacting with EZH2 and LSD1 in non-small-cell lung cancer cells. Cell Death Dis. 2016;7(5):e2225–e.

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Zhu F, Zhang S, Wang L, Wu W, Zhao H. LINC00511 promotes the progression of non-small cell lung cancer through downregulating LATS2 and KLF2 by binding to EZH2 and LSD1. Eur Rev Med Pharmacol Sci. 2019;23(19):8377–90.

    PubMed  Google Scholar 

  93. Fang J, Sun C-C, Gong C. Long noncoding RNA XIST acts as an oncogene in non-small cell lung cancer by epigenetically repressing KLF2 expression. Biochem Biophys Res Commun. 2016;478(2):811–7.

    CAS  PubMed  Google Scholar 

  94. Han F, Huang D, Meng J, Chu J, Wang M, Chen S. Mir-126‐5p enhances radiosensitivity of lung adenocarcinoma cells by inhibiting EZH2 via the KLF2/BIRC axis. J Cell Mol Med. 2022;26(9):2529–42.

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Lu J. The Warburg metabolism fuels tumor metastasis. Cancer Metastasis Rev. 2019;38(1):157–64.

    CAS  PubMed  Google Scholar 

  96. Grasmann G, Smolle E, Olschewski H, Leithner K. Gluconeogenesis in cancer cells–repurposing of a starvation-induced metabolic pathway? Biochim et Biophys Acta (BBA)-Reviews Cancer. 2019;1872(1):24–36.

    CAS  Google Scholar 

  97. Matés JM, Campos-Sandoval JA, Márquez J. Glutaminase isoenzymes in the metabolic therapy of cancer. Biochimica et Biophysica Acta (BBA)-Reviews on Cancer. 2018;1870(2):158–64.

  98. Scalise M, Pochini L, Galluccio M, Console L, Indiveri C. Glutamine transport and mitochondrial metabolism in cancer cell growth. Front Oncol. 2017;7:306.

    PubMed  PubMed Central  Google Scholar 

  99. Xiao S, Jin-Xiang Y, Long T, Xiu-Rong L, Hong G, Jie-Cheng Y, et al. Kruppel-like factor 2 disturb non-small cell lung cancer energy metabolism by inhibited glutamine consumption. J Pharm Pharmacol. 2020;72(6):843–51.

    CAS  PubMed  Google Scholar 

  100. Shi Y, Sun X, He X. Overexpression of Aristaless-like Homeobox-4 inhibits proliferation, invasion, and EMT in hepatocellular carcinoma cells. Oncol Res. 2017;25(1):11.

    PubMed  PubMed Central  Google Scholar 

  101. Edeling M, Ragi G, Huang S, Pavenstädt H, Susztak K. Developmental signalling pathways in renal fibrosis: the roles of Notch, wnt and hedgehog. Nat Rev Nephrol. 2016;12(7):426–39.

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Epstein EH. Basal cell carcinomas: attack of the hedgehog. Nat Rev Cancer. 2008;8(10):743–54.

    CAS  PubMed  PubMed Central  Google Scholar 

  103. di Magliano MP, Hebrok M. Hedgehog signalling in cancer formation and maintenance. Nat Rev Cancer. 2003;3(12):903–11.

    Google Scholar 

  104. Lin J, Tan H, Nie Y, Wu D, Zheng W, Lin W, et al. Krüppel-like factor 2 inhibits hepatocarcinogenesis through negative regulation of the hedgehog pathway. Cancer Sci. 2019;110(4):1220–31.

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Morikawa M, Derynck R, Miyazono K. TGF-β and the TGF-β family: context-dependent roles in cell and tissue physiology. Cold Spring Harb Perspect Biol. 2016;8(5):a021873.

    PubMed  PubMed Central  Google Scholar 

  106. Derynck R, Budi EH. Specificity, versatility, and control of TGF-β family signaling. Sci Signal. 2019;12(570):eaav5183.

    CAS  PubMed  PubMed Central  Google Scholar 

  107. David CJ, Massagué J. Contextual determinants of TGFβ action in development, immunity and cancer. Nat Rev Mol Cell Biol. 2018;19(7):419–35.

    CAS  PubMed  PubMed Central  Google Scholar 

  108. Hill CS. Transcriptional control by the SMADs. Cold Spring Harb Perspect Biol. 2016;8(10):a022079.

    PubMed  PubMed Central  Google Scholar 

  109. Boon RA, Fledderus JO, Volger OL, Van Wanrooij EJ, Pardali E, Weesie F, et al. KLF2 suppresses TGF-β signaling in endothelium through induction of Smad7 and inhibition of AP-1. Arterioscler Thromb Vasc Biol. 2007;27(3):532–9.

    CAS  PubMed  Google Scholar 

  110. Yan X, Chen Y-G. Smad7: not only a regulator, but also a cross-talk mediator of TGF-β signalling. Biochem J. 2011;434(1):1–10.

    CAS  PubMed  Google Scholar 

  111. Chen Y, Yang L. FBXL19-AS1 aggravates the progression of hepatocellular cancer by downregulating KLF2. J BU ON: Official J Balkan Union Oncol. 2021;26(4):1333–9.

    Google Scholar 

  112. Huang M-d, Chen W-m, Qi F-z, Xia R, Sun M, Xu T-p, et al. Long non-coding RNA ANRIL is upregulated in hepatocellular carcinoma and regulates cell proliferation by epigenetic silencing of KLF2. J Hematol Oncol. 2015;8(1):1–14.

    PubMed  PubMed Central  Google Scholar 

  113. Jiang H, Shi X, Ye G, Xu Y, Xu J, Lu J, et al. Up-regulated long non-coding RNA DUXAP8 promotes cell growth through repressing Krüppel-like factor 2 expression in human hepatocellular carcinoma. OncoTargets and Therapy. 2019;12:7429.

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Zou K, Lu X, Ye K, Wang C, You T, Chen J. Krüppel-like factor 2 promotes cell proliferation in hepatocellular carcinoma through up-regulation of c-myc. Cancer Biol Ther. 2016;17(1):20–6.

    CAS  PubMed  PubMed Central  Google Scholar 

  115. Baker SJ, Reddy EP. CDK4: a key player in the cell cycle, development, and cancer. Genes & cancer. 2012;3(11–12):658–69.

    CAS  Google Scholar 

  116. Rea K, Sensi M, Anichini A, Canevari S, Tomassetti A. EGFR/MEK/ERK/CDK5-dependent integrin-independent FAK phosphorylated on serine 732 contributes to microtubule depolymerization and mitosis in tumor cells. Cell Death Dis. 2013;4(10):e815–e.

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Alarcón C, Zaromytidou A-I, Xi Q, Gao S, Yu J, Fujisawa S, et al. CDK8/9 drive smad transcriptional action, turnover and YAP interactions in BMP and TGFβ pathways. Cell. 2009;139(4):757.

    PubMed  PubMed Central  Google Scholar 

  118. Borggrefe T, Davis R, Erdjument-Bromage H, Tempst P, Kornberg RD. A complex of the Srb8,-9,-10, and-11 transcriptional regulatory proteins from yeast. J Biol Chem. 2002;277(46):44202–7.

    CAS  PubMed  Google Scholar 

  119. Larschan E, Winston F. The Saccharomyces cerevisiae Srb8-Srb11 complex functions with the SAGA complex during Gal4-activated transcription. Mol Cell Biol. 2005;25(1):114–23.

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Samuelsen CO, Baraznenok V, Khorosjutina O, Spåhr H, Kieselbach T, Holmberg S et al. TRAP230/ARC240 and TRAP240/ARC250 Mediator subunits are functionally conserved through evolution. Proceedings of the National Academy of Sciences. 2003;100(11):6422-7.

  121. Xu W, Wang Z, Zhang W, Qian K, Li H, Kong D, et al. Mutated K-ras activates CDK8 to stimulate the epithelial-to-mesenchymal transition in pancreatic cancer in part via the Wnt/β-catenin signaling pathway. Cancer Lett. 2015;356(2):613–27.

    CAS  PubMed  Google Scholar 

  122. Chan E, Gat U, McNiff JM, Fuchs E. A common human skin tumour is caused by activating mutations in β-catenin. Nat Genet. 1999;21(4):410–3.

    CAS  PubMed  Google Scholar 

  123. Dominguez I, Itoh K, Sokol SY. Role of glycogen synthase kinase 3 beta as a negative regulator of dorsoventral axis formation in Xenopus embryos. Proc Natl Acad Sci. 1995;92(18):8498–502.

    CAS  PubMed  PubMed Central  Google Scholar 

  124. Atkins GB, Jain MK. Role of Kruppel-like transcription factors in endothelial biology. Circul Res. 2007;100(12):1686–95.

    CAS  Google Scholar 

  125. Dekker RJ, Boon RA, Rondaij MG, Kragt A, Volger OL, Elderkamp YW, et al. KLF2 provokes a gene expression pattern that establishes functional quiescent differentiation of the endothelium. Blood. 2006;107(11):4354–63.

    CAS  PubMed  Google Scholar 

  126. Wei R, Kong L, Xiao Y, Yuan H, Song Y, Wang J, et al. CDK8 regulates the angiogenesis of pancreatic cancer cells in part via the CDK8-β-catenin-KLF2 signal axis. Exp Cell Res. 2018;369(2):304–15.

    CAS  PubMed  Google Scholar 

  127. Zhang D, Dai Y, Cai Y, Suo T, Liu H, Wang Y, et al. KLF2 is downregulated in pancreatic ductal adenocarcinoma and inhibits the growth and migration of cancer cells. Tumor Biology. 2016;37(3):3425–31.

    CAS  PubMed  Google Scholar 

  128. Lian Y, Wang J, Feng J, Ding J, Ma Z, Li J, et al. Long non-coding RNA IRAIN suppresses apoptosis and promotes proliferation by binding to LSD1 and EZH2 in pancreatic cancer. Tumor Biology. 2016;37(11):14929–37.

    CAS  PubMed  Google Scholar 

  129. Ma Z, Huang H, Wang J, Zhou Y, Pu F, Zhao Q, et al. Long non-coding RNA SNHG15 inhibits P15 and KLF2 expression to promote pancreatic cancer proliferation through EZH2-mediated H3K27me3. Oncotarget. 2017;8(48):84153.

    PubMed  PubMed Central  Google Scholar 

  130. Lian Y, Yang J, Lian Y, Xiao C, Hu X, Xu H. DUXAP8, a pseudogene derived lncRNA, promotes growth of pancreatic carcinoma cells by epigenetically silencing CDKN1A and KLF2. Cancer Commun. 2018;38(1):1–11.

    Google Scholar 

  131. Yuedi D, Houbao L, Pinxiang L, Hui W, Min T, Dexiang Z. KLF2 induces the senescence of pancreatic cancer cells by cooperating with FOXO4 to upregulate p21. Exp Cell Res. 2020;388(1):111784.

    PubMed  Google Scholar 

  132. Wang Y, Decker SJ, Sebolt-Leopold J. Knockdown of Chk1, Wee1 and Myt1 by RNA interference abrogates G2 checkpoint and induces apoptosis. Cancer Biol Ther. 2004;3(3):305–13.

    CAS  PubMed  Google Scholar 

  133. Elder RT, Yu M, Chen M, Zhu X, Yanagida M, Zhao Y. HIV-1 vpr induces cell cycle G2 arrest in fission yeast (Schizosaccharomyces pombe) through a pathway involving regulatory and catalytic subunits of PP2A and acting on both Wee1 and Cdc25. Virology. 2001;287(2):359–70.

    CAS  PubMed  Google Scholar 

  134. Rowley R, Hudson J, Young PG. The wee1 protein kinase is required for radiation-induced mitotic delay. Nature. 1992;356(6367):353–5.

    CAS  PubMed  Google Scholar 

  135. Li R, Chen J, Gao X, Jiang G. Transcription factor KLF2 enhances the sensitivity of breast cancer cells to cisplatin by suppressing kinase WEE1. Cancer Biol Ther. 2021;22(7–9):465–77.

    CAS  PubMed  PubMed Central  Google Scholar 

  136. Zhu K-Y, Tian Y, Li Y-X, Meng Q-X, Ge J, Cao X-C, et al. The functions and prognostic value of Krüppel-like factors in breast cancer. Cancer Cell Int. 2022;22(1):1–12.

    Google Scholar 

  137. Wang L, Ye T, Wu H, Chen S, Weng J, Xi X. LINC00702 accelerates the progression of ovarian cancer through interacting with EZH2 to inhibit the transcription of KLF2. Eur Rev Med Pharmacol Sci. 2019;23(3 Suppl):201–8.

    CAS  PubMed  Google Scholar 

  138. Bai Z, Wu Y, Bai S, Yan Y, Kang H, Ma W, et al. Long non-coding RNA SNGH7 is activated by SP1 and exerts oncogenic properties by interacting with EZH2 in ovarian cancer. J Cell Mol Med. 2020;24(13):7479–89.

    CAS  PubMed  PubMed Central  Google Scholar 

  139. Chen L, Yang H, Xiao Y, Tang X, Li Y, Han Q, et al. Lentiviral-mediated overexpression of long non-coding RNA GAS5 reduces invasion by mediating MMP2 expression and activity in human melanoma cells. Int J Oncol. 2016;48(4):1509–18.

    CAS  PubMed  Google Scholar 

  140. Huang D, Du X, Yuan R, Chen L, Liu T, Wen C, et al. Rock2 promotes the invasion and metastasis of hepatocellular carcinoma by modifying MMP2 ubiquitination and degradation. Biochem Biophys Res Commun. 2014;453(1):49–56.

    CAS  PubMed  Google Scholar 

  141. Lu L, Xue X, Lan J, Gao Y, Xiong Z, Zhang H, et al. MicroRNA-29a upregulates MMP2 in oral squamous cell carcinoma to promote cancer invasion and anti-apoptosis. Biomed Pharmacother. 2014;68(1):13–9.

    CAS  PubMed  Google Scholar 

  142. Wang B, Liu M, Song Y, Li C, Zhang S, Ma L. KLF2 inhibits the migration and invasion of prostate cancer cells by downregulating MMP2. Am J Men’s Health. 2019;13(1):1557988318816907.

    Google Scholar 

  143. Zhu Y, Tong Y, Wu J, Liu Y, Zhao M. Knockdown of LncRNA GHET1 suppresses prostate cancer cell proliferation by inhibiting HIF-1α/Notch‐1 signaling pathway via KLF2. BioFactors. 2019;45(3):364–73.

    CAS  PubMed  Google Scholar 

  144. Xue P, Yan M, Wang K, Gu J, Zhong B, Tu C. Up-regulation of LINC00665 facilitates the malignant progression of prostate cancer by epigenetically silencing KLF2 through EZH2 and LSD1. Front Oncol. 2021:1165.

  145. Greer EL, Shi Y. Histone methylation: a dynamic mark in health, disease and inheritance. Nat Rev Genet. 2012;13(5):343–57.

    CAS  PubMed  PubMed Central  Google Scholar 

  146. Black JC, Van Rechem C, Whetstine JR. Histone lysine methylation dynamics: establishment, regulation, and biological impact. Mol Cell. 2012;48(4):491–507.

    CAS  PubMed  Google Scholar 

  147. Yamane K, Toumazou C, Tsukada Y-i, Erdjument-Bromage H, Tempst P, Wong J, et al. JHDM2A, a JmjC-containing H3K9 demethylase, facilitates transcription activation by androgen receptor. Cell. 2006;125(3):483–95.

    CAS  PubMed  Google Scholar 

  148. Okada Y, Scott G, Ray MK, Mishina Y, Zhang Y. Histone demethylase JHDM2A is critical for Tnp1 and Prm1 transcription and spermatogenesis. Nature. 2007;450(7166):119–23.

    CAS  PubMed  Google Scholar 

  149. Loh Y-H, Zhang W, Chen X, George J, Ng H-H. Jmjd1a and Jmjd2c histone H3 lys 9 demethylases regulate self-renewal in embryonic stem cells. Genes Dev. 2007;21(20):2545–57.

    CAS  PubMed  PubMed Central  Google Scholar 

  150. Kuroki S, Matoba S, Akiyoshi M, Matsumura Y, Miyachi H, Mise N, et al. Epigenetic regulation of mouse sex determination by the histone demethylase Jmjd1a. Science. 2013;341(6150):1106–9.

    CAS  PubMed  Google Scholar 

  151. Mittrücker H-W, Matsuyama T, Grossman A, Kündig TM, Potter J, Shahinian A, et al. Requirement for the transcription factor LSIRF/IRF4 for mature B and T lymphocyte function. Science. 1997;275(5299):540–3.

    PubMed  Google Scholar 

  152. Sciammas R, Shaffer A, Schatz JH, Zhao H, Staudt LM, Singh H. Graded expression of interferon regulatory factor-4 coordinates isotype switching with plasma cell differentiation. Immunity. 2006;25(2):225–36.

    CAS  PubMed  Google Scholar 

  153. Klein U, Casola S, Cattoretti G, Shen Q, Lia M, Mo T, et al. Transcription factor IRF4 controls plasma cell differentiation and class-switch recombination. Nat Immunol. 2006;7(7):773–82.

    CAS  PubMed  Google Scholar 

  154. Ohguchi H, Hideshima T, Bhasin MK, Gorgun GT, Santo L, Cea M, et al. The KDM3A–KLF2–IRF4 axis maintains myeloma cell survival. Nat Commun. 2016;7(1):1–15.

    Google Scholar 

  155. Schmidt M, De Mazière A, Smyczek T, Gray A, Parker L, Filvaroff E, et al. editors. The role of Egfl7 in vascular morphogenesis. Novartis Foundation Symposium; 2007: Chichester; New York; John Wiley; 1999.

  156. Nikolić I, Stanković ND, Bicker F, Meister J, Braun H, Awwad K, et al. EGFL7 ligates αvβ3 integrin to enhance vessel formation. Blood the Journal of the American Society of Hematology. 2013;121(15):3041–50.

    Google Scholar 

  157. Hong G, Kuek V, Shi J, Zhou L, Han X, He W, et al. EGFL7: Master regulator of cancer pathogenesis, angiogenesis and an emerging mediator of bone homeostasis. J Cell Physiol. 2018;233(11):8526–37.

    CAS  PubMed  Google Scholar 

  158. Papaioannou D, Shen C, Nicolet D, McNeil B, Bill M, Karunasiri M, et al. Prognostic and biological significance of the proangiogenic factor EGFL7 in acute myeloid leukemia. Proc Natl Acad Sci. 2017;114(23):E4641–E7.

    CAS  PubMed  PubMed Central  Google Scholar 

  159. Ria R, Vacca A, Ribatti D, Di Raimondo F, Merchionne F, Dammacco F. Alpha (v) beta (3) integrin engagement enhances cell invasiveness in human multiple myeloma. Haematologica. 2002;87(8):836–45.

    CAS  PubMed  Google Scholar 

  160. Vacca A, Ria R, Presta M, Ribatti D, Iurlaro M, Merchionne F, et al. αvβ3 integrin engagement modulates cell adhesion, proliferation, and protease secretion in human lymphoid tumor cells. Exp Hematol. 2001;29(8):993–1003.

    CAS  PubMed  Google Scholar 

  161. Tucci M, De Palma R, Lombardi L, Rodolico G, Berrino L, Dammacco F, et al. β3 integrin subunit mediates the bone-resorbing function exerted by cultured myeloma plasma cells. Cancer Res. 2009;69(16):6738–46.

    CAS  PubMed  Google Scholar 

  162. Nichol D, Shawber C, Fitch MJ, Bambino K, Sharma A, Kitajewski J, et al. Impaired angiogenesis and altered notch signaling in mice overexpressing endothelial Egfl7. Blood. J Am Soc Hematol. 2010;116(26):6133–43.

    CAS  Google Scholar 

  163. Salama Y, Heida AH, Yokoyama K, Takahashi S, Hattori K, Heissig B. The EGFL7-ITGB3-KLF2 axis enhances survival of multiple myeloma in preclinical models. Blood Adv. 2020;4(6):1021–37.

    CAS  PubMed  PubMed Central  Google Scholar 

  164. Wu N, Chen S, Luo Q, Jiang Z, Wang X, Li Y, et al. Kruppel-like factor 2 acts as a tumor suppressor in human retinoblastoma. Exp Eye Res. 2022;216:108955.

    CAS  PubMed  Google Scholar 

  165. Ruan J, Zheng L, Hu N, Guan G, Chen J, Zhou X, et al. Long noncoding RNA SNHG6 promotes osteosarcoma cell proliferation through regulating p21 and KLF2. Arch Biochem Biophys. 2018;646:128–36.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

None.

Funding

None.

Author information

Authors and Affiliations

Authors

Contributions

NT, AM, ASZ, and IA were involved in search strategy and drafting. MM designed, revised, structured, and edited the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Meysam Moghbeli.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

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

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Taghehchian, N., Maharati, A., Akhlaghipour, I. et al. PRC2 mediated KLF2 down regulation: a therapeutic and diagnostic axis during tumor progression. Cancer Cell Int 23, 233 (2023). https://doi.org/10.1186/s12935-023-03086-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12935-023-03086-3

Keywords